Final Flashcards

1
Q

What is immunology?

A

Study of hosts reactions to foreign substances that are introduced into the body

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is an antigen?

A

A substance that reacts with an antibodies or sensitized cells but may or may not be able to elicit an immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What is immunity?

A

Condition of being resistant to infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Who is the father of immunology?

A
  • Louise Pasteur
  • he created the first attenuated vaccine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is attenuation?

A
  • makes pathogens
  • takes place through heat, aging and chemicals
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is phagocytosis?

A

Cells that eat cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What is humoral immunity?

A
  • Protection from disease resulting from substances in the serum/plasma (antibodies and acute phase reactants)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the two types of the adaptive immune response?

A
  • cellular
  • humoral
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What is the function of humoral immunity?

A

Involves production of antibodies by B cells and plasma cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are antibodies?

A
  • serum proteins used by the immune system to identify and neutralize foreign objects such as pathogenic bacteria and viruses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What is innate immunity?

A

An individual ability to resist infection by means of normally present body functions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What are characteristics of the innate immunity?

A
  • nonspecific
  • no memory
  • immediate exposure
  • same response for all pathogens
  • influenced by nutrition, age, fatigue, stress, and genetic determinants
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is adaptive immunity?

A

Resistance that is characterized by specificity for each individuals pathogen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are characteristics of adaptive immunity?

A
  • delayed response
  • stronger response
  • specific
  • has memory
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is the function of WBCs?

A

Defend against invasion by bacteria, virus, fungi, and other foreign substances

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What are the 5 types of leukocytes?

A
  • monocytes
  • lymphocytes
  • basophil
  • eosinophil
  • neutrophils
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What WBC is part of the adaptive immunity?

A

Lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are the main cells found in tissues?

A
  • dendritic cells
  • macrophages
  • mast cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What are hematopoietic stem cells (HSC)?

A

All blood cell types arise from this cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Where are WBC produced? How much?

A
  • bone marrow
  • one and a half billion daily
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

What two distinct types of precursors does HSC give rise to?

A
  • common myeloid precursors (CMP)
  • common lymphoid precursors (CLP)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What do CMPs do?

A
  • give rise to WBC that participate in phagocytosis
  • also known as myeloid line
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What are characteristics of Neutrophils?

A
  • life span in several days
  • nucleus has 2-5 lobes (has lots of granules)
  • 50-70% of WBC
  • use wright stain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is the main function of neutrophils?

A

Phagocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What is diapedesis?

A

Movement through blood vessel wall

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What are chemotaxins?

A

Chemical messengers that cause cells to migrate in particular direction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What are characteristics of eosinophils?

A
  • 1-4% of WBC
  • eosin stain
  • bilobed/eccentrically located nucleus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What are the functions of eosinophils?

A

1) phagocytosis (not as efficient as neutrophils)
2) neutralize basophils and mast cells
3) kills parasites using cationic proteins
4) MOST IMPORTANT: regulation of adaptive immune response through cytokine release

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What are characteristics of basophils?

A
  • less than 1% of WBC
  • smallest
  • nucleus is bilobed, and has deep purple-blue granules (hard to see)
  • life span is a few hours
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What happens to basophils in the spleen?

A

Removed and destroyed by marcrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What are the 3 main functions of basophils?

A

1) induce and maintain allergic reactions
2) regulate some Th cell response
3) stimulate B-cells to produce IgE antibody

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

What is the function of histamine?

A

Contracts smooth muscle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What is the function of heparin?

A

Anticoagulant

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What are characteristics of monocytes?

A
  • Largest
  • irregular horseshoe or folded nucleus (occupies half or more of cell)
  • 2-10% of WBC
  • 30 hour lifespan
  • after 30 hours, travels to tissue and becomes macrophage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Why are neutrophils more efficient at phagocytosis than macrophages?

A

Macrophages are slower

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Why are neutrophils more efficient at phagocytosis than eosinophils?

A
  • less eosinophils
  • eosinophils lack digestive enzymes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What is the lifespan of macrophages?

A

A few months

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the functions of innate immunity?

A

1) phagocytosis
2) microbial killing
3) anti-tumor activity
4) intracellular parasite eradication
5) secretion of cell membrane

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

How do macrophages play an important role in the adaptive system?

A

By presenting phagocytosized antigens to T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Where are mast cells found?

A
  • located in variety of tissues (resembles a basophil)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

What are the functions of mast cells?

A

1)act to increase vascular permeability and increase blood flow to affected area
2) role in allergic reactions
3) APC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Describe the appearance of dendritic cells

A

Covered with long membraneous extensions that resemble nerve cell dendrites

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

What are dendritic cells?

A
  • APC that links innate and adaptive immunity and are critical for the induction of immune response
  • classified according to location
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

What is the most potent phagocytic cell?

A

Dendritic cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

What are characteristics of lymphocytes?

A
  • 20-40% of WBC
  • large, rounded and indented nucleus
  • similar size to RBC
  • has narrow ring
  • chromatin stains deep blue
  • sparse cytoplasm
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Why are lymphocyte granules unique?

A

They arise from HSC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

What are the 3 types of lymphocytes?

A
  • T cells
  • B cells
  • innate lymphoid cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

What is the most prominent innate lymphoid cells?

A

NK cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

What % of lymphocytes are B cells?

A

10-20%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What % of lymphocytes are T cells?

A

61-80%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What % of lymphocytes are NK cells?

A

10-15%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What is cluster of differentiation?

A

Protein found of cells surfaces that can be used to identify specific cell types and stages of differentiation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

What are the primary lymphoid organs

A

Bone marrow
Thymus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What is the largest tissue in the body?

A

Bone marrow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

What is bone marrow the main source of?

A

HSC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What can HSC develop into?

A
  • RBCs
  • lymphocytes
  • monocytes
  • granulocytes
  • platelets
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What occurs when lymphocytes remain in the bone marrow?

A

Mature and become NK cells or B cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

What occurs when the lymphocytes travel to the thymus?

A

Mature and become T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Describe the thymus

A

Filled with epithelial cells that play central role in differentiation process

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

How long does it take T cells to mature?

A

3 weeks

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

What are mature T cells released from?

A

Medulla thymus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

What are the secondary lymphoid organs?

A
  • spleen
  • lymph nodes
  • CALT
  • MALT
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Where does main contact of foreign antigens occur?

A
  • secondary lymphoid organs
  • T cells and B cells meet here as well
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Where do lymphocytes spend most of their time?

A

The tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

How do lymphocytes travel to blood stream?

A

Via. Thoracic duct

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

What is the largest lymphatic vessel?

A

Thoracic duct

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

What is lymphopoiesis?

A

Multiplication of lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Where is the spleen located?

A
  • upper left quadrant
  • below diaphragm
  • largest secondary organ
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is the function of the spleen?

A

Removes old/damaged cells and foreign antigens from the blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What is the function of red pulp?

A
  • to destroy old RBCs, platelets, and pathogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What is rich in red pulp?

A

Macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What is % of red pulp found in the spleen?

A

50%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

What % of white pulp is found in the spleen?

A

20%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

What is the main function of lymph nodes?

A
  • Filtration of interstitial fluid from around cells in tissues
  • important because it allows contact between lymphocytes and foreign antigens that have penetrated tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What are the layers of lymph node tissues?

A
  • outer cortex
  • para cortex
  • inner medulla
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What is lymph fluid?

A

A filtrate of blood and arises from the passage of water and low molecular weight solutes out of blood vessels walls and into the interstitial spaces between cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What cells could be found in a primary follicle of the lymph nodes?

A
  • stimulated B cells
  • macrophages
  • follicular dendritic
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What cells can be found in a secondary follicle of lymph nodes?

A

Antigen stimulated proliferating B cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

What is at the center of a secondary follicle of the lymph nodes called?

A

Germinal center

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What happens when B cells come in contact with antigens?

A

Plasma cells and memory cells form

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

Where can T cells be found in the lymph nodes?

A

Paracortex

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What is the main function of plasma cells?

A

Actively secrete antibodies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What is the main function of memory cells?

A

Can quickly change into plasma cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Where can MALT be found?

A
  • appendix
  • GI tract
  • ileum
  • peyers patch
  • respiratory tract
  • tonsils
  • urogenital tract
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

Why are macrophages and lymphocytes and localized in MALT?

A

Mucosal surface are main ports of entry for a foreign antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What cells cab be found in CALT?

A
  • dendritic cells
  • macrophages
  • monocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What are innate phagocytic cells?

A
  • dendritic cells
  • macrophages
  • monocytes
  • neutrophils
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

Describe NK cells

A
  • can kill virally infected or cancerous target cells without previous exposure to them
  • first line of defense against virally infected cells
  • no specificity
  • enhanced by exposure to cytokines: IL-12, INF-alpha, and INF-beta
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

How are T cells different from NK cells?

A
  • only NK cells are able to kill target cells without prior exposure to them
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What is the most potent phagocytic cell?

A

Dendritic cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What is the main function of T-cells?

A
  • produce cytokines that regulate both innate and adaptive immunity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

What blood cell kills parasite?

A

Eosinophils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

What are the 2 systems of the innate immunity?

A
  • external defense system
  • internal defense system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

Describe the external defense system of innate immunity

A
  • contains chemical, physical, and biological factors that work together to prevent most infections agents from entering the body
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

Describe the internal defense system of the innate immunity

A
  • triggered within minutes and clears invaders as quickly as possible
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

Describe the outer layer of the external defense barrier

A
  • outer layer of skin is the epidermis and contains several layers of tightly packed epithelial cells
  • proteins coated cell called keratin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What is the role of keratin in the epidermis?

A

Makes skin impermeable to most infectious agents

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What is the layer under the epidermis?

A

Dermis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

Describe dermis

A
  • thicker than epidermis
  • connective tissue with blood vessels, hair follicles, sebaceous glands, and WBC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

What cells are found in the dermis?

A
  • dendritic cells
  • macrophages
  • mast cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

Why does skin have several secretions?

A

Discourage growth of microorganisms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

What is secreted on the skin to maintain pH balance? What is the pH?

A
  • fatty acids and lactic acid
  • they prevent growth of microorganisms
  • ~5.6
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

what is psoriasis?

A
  • produced by skin
  • has antibacterial effects, especially for gram negative bacteria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What is the respiratory tracts external defense system?

A
  • mucous secretion block adherence
  • coughing and sneezing clear out pathogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What is the genitourinary tracts external defense systems?

A
  • flushing out urine
  • acidity helps to remove many potential pathogens
  • lactic acid of vagina maintains pH of 5
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What is the external defense for the digestive tract?

A

-hydrochloric acid keeps pH as low as 1 to kill pathogens brought in by food and drink

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

What are the lysosomes role in the external defense system?

A
  • attack cell wall of microorganisms, especially gram positive bacteria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What is microbiota?

A

A mix of bacteria that are normally found at species body sites and do not typically cause disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

Explain the internal defense system of innate immunity

A
  • composed of both cells
  • soluble factors that have specific and essential functions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

What is the function of phagocytes?

A
  • they engulf and destroy most foreign cells or particles that enter the body
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

What is the % of dendritic cells and macrophages found in tissues?

A

10-15%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

What are the most important cells in pathogen recognition?

A
  • dendritic cells
  • macrophages
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

What is the main function of patter recognition receptors (PRRs)?

A
  • able to distinguish pathogens from normally present molecules in the body by means of receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

What happens when PRRs binds to pathogen?

A

1) phagocytic cells activate and are better able to engulf and kill organisms
2) activated cells secrete proinflammatory cytokines and chemokines
3) cytokines and chemokines also trigger adaptive immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

Describe pathogen associated molecular patterns (PAMPs)?

A

Substances that allows PRRs to have ability to distinguish self from non-self

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

What TLRs are found in cytoplasm?

A
  • TLR1
  • TLR2
  • TLR4
  • TLR5
  • TLR6
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What TLRs are found in the endosomal compartment of cells?

A
  • TLR3
  • TLR7
  • TLR8
  • TLR9
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What are c-lectin receptors (CLRs)?

A
  • plasma membrane receptors that bind to Mannan and beta-glucans found in fungal cell walls
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

Describe serum amyloid A

A
  • major protein who concentration can increase X1000 in response to infection/injury
  • apolipoprotein synthesized in liver
  • acts like cytokines, chemical messenger (activates monocytes and macrophages
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

Why would serum amyloid A be increased?

A
  • chronic inflammation
  • atherosclerosis
  • cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

Describe complement

A
  • series of serum proteins that are normally present and contribute to inflammation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

What are the functions of complement?

A

1) opsonization
2) chemotaxis
3) lysis of cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

What is elastase?

A
  • an enzyme secreted by neutrophils during inflammation that can degrade elastin and collagen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What are the pro inflammatory cytokines?

A
  • TNF- alpha
  • interleukin-1beta
  • interleukin-6
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

Describe haptoglobin

A
  • alpha1-globulin
  • binds irreversibly to free hemoglobin
  • acts as antioxidant to protect against oxidative damage from free hemoglobin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

Describe fibrinogen

A
  • acute phase protein involved in coagulation pathway
  • small portion cleaned by thrombin to form fibrils that create a fibrils clot
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What do fibrin clots do?

A
  • increases strength with wound and stimulate endothelial cell adhesion and proliferation
  • creates barrier of microorganisms further into the body
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

Describe ceruloplasmin

A
  • consists of a single polypeptide chain
  • copper transporting protein in human plasma
  • acts as enzyme, converting the toxic ferrous ion (Fe^2+) to non toxic ferric form ( Fe^3+)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

What disease is characterized by depletion of ceruloplasmin?

A

Wilsons disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

What is inflammation?

A

Body’s overall reaction to injury or invasion by an infectious agent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What are the 4 cardinal signs of inflammation?

A
  • erythema (redness)
  • edema (swelling)
  • heat
  • pain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What is the acute inflammatory response?

A
  • acts it combat the early stages of infection and also begins a process that repairs tissues damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What cells are most active in phagocytosis?

A
  • dendritic cells
  • macrophages
  • monocytes
  • neutrophils
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What are selectins?

A
  • adhesion molecules on endothelial cells lining blood vessels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

what are integrins?

A
  • adhesion molecules on endothelial cell wall
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What is chemotaxis?

A
  • cells are attracted to site of inflammation by chemical substances such as soluble bacteria factors and acute phase reactants
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What enhances the binding process during phagocytosis?

A

Opsonins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

what are opsonins?

A

Serum proteins that attach to a foreign cell or pathogen and make it more susceptible to phagocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

What are phagosomes?

A

Pseudopodia fuse to completely enclose pathogen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What are phagolysosome?

A

Lysosomal granule fused to phagosome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

What are the two processes that eliminate pathogens?

A
  • oxygen-dependent pathway
  • oxygen-independent pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

Describe oxygen-dependent pathway

A
  • increase in oxygen consumption (oxidative burst) occurs in cell as the pseudopodia enclose particles within vacuole
  • generates considerable energy through oxidative metabolism
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

What is an important bacterial agent in oxygen dependent process?

A

Hydrogen peroxide

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

Describe oxygen-independent pathway

A
  • defensins kill gram-negative bacteria, many fungi, and some viruses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What are defensins?

A

Small cationic proteins that cleave segments without benefits of oxygen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

What is cytotoxicity?

A
  • the degree to which a substance can cause damage to a cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

What are two main binding receptors for NK cells?

A

1) inhibitory receptors
2) activating receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

What do inhibitory receptors do?

A

Deliver inhibitor signals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

What do activating receptors do?

A

Delivers signals to activate cytotoxic mechanism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What are perforins?

A

Proteins that form channels in target cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

What are granzymes?

A

Packets of enzymes tat may enter through channels and mediate cell lysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

Describe antibody-dependent cellular cytotxocity

A
  • NK cells recognize and lysis antibody-coated target cells
  • binding occurs through surface receptors; CD16 and CD32, which bind to Fc portion of immunoglobulins
  • destruction occurs outside of NK cells
  • important contributor to anti-tumor activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

Describe innate lymphoid cells

A
  • growing family of immune cells that develop from the common lymphoid progenitor but does not express markers of lymphocytes lineage
  • primarily found in mucosal sites
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

What can measurements of CRP levels be used for?

A
  • tracking progress of organ transplant
  • monitoring drug therapy with anti-inflammatory agents
  • determining active phases of RA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

What are the functions of C-reactive protein?

A

1) opsonization
2) complement activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

What is the function of serum amyloid A?

A

Activates monocytes and macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

What is the function of alpha1-antitrypsin?

A

Proteases inhibitor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

What is the function of fibrinogen?

A

Clot formation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

What is the function of haptoglobin?

A

Binds hemoglobin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

What is the function of ceruloplasmin?

A

Binds copper and oxidizes iron

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

What are the functions of complement C3?

A

1) opsonization
2) lysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

What causes increased vasodilation and vasopermability?

A

Inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

Are all immunogens antigens?

A

Yes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

Are all antigens immunogens?

A

No

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What is a distinct difference between immunogens and antigens?

A

Immunogens successfully stimulate an immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What is immunogenicity?

A

The ability of an antigen to stimulate an immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What factors affect the strength of immunogenicity?

A

1) macromolecule size
2) foreignness
3) ability to be processed and presented with MHC molecules
4) chemical composition
5) molecular complexity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

What are the most effective immunogens?

A
  • proteins (strongest)
  • polysaccharides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

Describe the structure of a proteins primary structure?

A
  • made up of subunits known as amino acids that are covalently linked together in polypeptide chains of various links
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

describe the structure of proteins secondary structure

A

Interactions between amino acids within primary structure causes chain to band/kink/loop creating 3D shapes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

What are the two common secondary structures found in proteins?

A
  • alpha-helices
  • beta-pleated sheets
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

Describe the alpha helices of the secondary structures of protein

A
  • peptide chains that twist to form a signal
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

Describe the beta-pleated sheet secondary structure of protein

A

Chains of undulating zig zags that have a planar shape

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

Describe the tertiary stucture of proteins

A

Secondary folds upon themselves once again, bringing distant regions of Amino acid chains together and embodying the 3D orientation of the entire molecule

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

Describe the quaternary structure of proteins

A

Two or ore polypeptide chains come together, forming a multimeric unit

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

What are polysaccharides less immunogenic than proteins?

A
  • their small size
  • T cells do not recognize carbohydrates
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What are not immunogenic by themselves?

A
  • pure nucleic acids
  • lipids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What is required for a substance to elicit an immune response?

A
  • must be subject to antigen processing
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What is antigen processing?

A
  • involves enzymatic digestion to create small peptides that can be completed by MHC molecules for presentation to responsiveness of an specific antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What is a hapten?

A
  • a substance that isn’t immunogenic by itself but is able to form a new antigenic determinant when combined with larger carrier molecule
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

Are haptens considered an antigen or an immunogen?

A

Antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

What is an epitope?

A
  • part of antigen that hosts immune system recognizes, eliciting the immune response to an invading pathogen
  • specifically binds to the corresponding antigen receptor on the immune cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What are the two types of epitopes that B cells recognize?

A

1) linear epitopes
2) conformation epitopes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

describe linear epitopes

A

Consist of sequential amino acids on a single polypeptide chain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

Describe conformational epitopes

A
  • results from the folding of one or more polypeptide chains, bringing together amino acids that may be distant from each other so that they are recognized together
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What is an autoantigen?

A
  • antigens that belong to host
  • does not evoke an immune response under normal circumstances
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What happens if an immune response does occur to autoantigens?

A

May result in autoimmune disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What is an alloantigen?

A

From other members of the hosts species and are capable of eliciting an immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

When are alloantigens important to consider?

A
  • tissue transplant
  • blood transfusion
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What is a heteroantigen?

A

From other speciesm such as other animals, plants or microorganisms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

What is a heterophile antigen?

A
  • heteroantigens that exist in unrelated plants or animals but are either identical or closely related in structures so that an antibody against either antigen will cross- react with each other
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

Describe current rapid screening tests for IM

A
  • detect heterophil antibodies present in the serum of infected patients that cross react with horse or bovine RBCs antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

where are MHC molecules found?

A

On all nucleated cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What do MHC molecules play a pivotal role in?

A
  • development both humoral and cell-mediated immunity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

What is the main immune function of MHC molecules?

A
  • bring antigens to cell surface for recognition by the T cells
  • only when the antigens is combined with the MHC molecules does the T cell activation occur
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

What is B cell recognition?

A

Surface receptors bind to antigen directly

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

What is T cell recognition?

A

Requires antigens to be cradled within MHC molecules

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

Where are class I MHC molecule genes found?

A

3 loci’s: A, B, C

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

Where are class II MHC molecule genes found?

A
  • D region: DR, DQ, and DP
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

Where are class III MHC molecules found?

A

Lies between the class I and class II regions on chromosome 6

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

describe class III MHC molecules

A
  • are secreted protein that have an immune function, but they are not expressed on cell surfaces
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

What is codominant?

A

All alleles that an individual inherits code for products that are expressed on cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

What is a haplotype?

A
  • group of alleles in an organism that are inherited together from a single parent
  • one haplotype is inherited from each parent
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

Where cells are class II MHC molecules found in?

A

Primarily on antigen-presenting cells (APCs)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

What are the most important class I MHC antigens to match for transplantation?

A
  • HLA-A antigens
  • HLA-B antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

What are the major class II molecules?

A
  • HLA-DP
  • HLA-DQ
  • HLA-DR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

Describe antigen presentation

A
  • peptide fragments derived from degraded proteins are transported to the plasma membrane, allowing recognition by T lymphocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

What does the class I MHC molecules mainly present?

A

CD8 T cells (cytotoxic)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
209
Q

What does the class II MHC molecules mainly present?

A

CD4 Th cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
210
Q

What is the function of class I MHC molecule?

A

Watchdogs of viral, tumor and intracellular bodies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
211
Q

What is the function of class II MHC molecule?

A
  • help to mount an immune response to bacterial infections or other pathogens usually found outside of cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

Describe modern transplant HLA testing

A
  • involves the use of molecular techniques to determine the MHC types of both donor and recipient
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

What could occur when an individual inherits certain HLA types?

A

May predispose individuals to the development of autoimmunity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
214
Q

What is rheumatoid arthritis?

A

Inflammation of multiple joints

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
215
Q

Describe type I diabetes

A

Increase in blood glucose because of destruction of insulin-producing cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
216
Q

What are two transporters associated with antigen processing?

A

TAP1
TAP2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
217
Q

What is immunogen?

A

A substance that reacts with an antibody or sensitized cell but always triggers an immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
218
Q

What is an adjuvant?

A
  • substances delivered simultaneously with an antigen to enhance the immune response; used in vaccines
  • also prevents antigen from diffusing more immune cells to the injection site of inoculation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q

What is an allele?

A

Alernate forms of a gene that code for a slightly different variety of the same product

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
220
Q

Where are class II MHC molecules synthesized?

A

Rough endoplasmic reticulum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
221
Q

Describe helper T cells

A
  • recognize antigen along with class II MHC protein
  • orchestrate the adaptive immune response, influencing activities of other immune cells
  • express CD4 receptors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
222
Q

What is the key portion of an antigen?

A

Epitope

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
223
Q

How do adjuvants work?

A

Acts by activating innate immune cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
224
Q

What is the most polymorphic gene system in humans?

A

HLA systems

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
225
Q

What is the function of cytotoxic T cells?

A

To kill cells infected with intracellular pathogens, such as viruses, bacteria and cancerous cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
226
Q

What is the function of Th cells?

A
  • control the immune response through the secretion of cytokines or signaling molecules that allow communication between immune cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

What are the roles of antibodies in immunity?

A

1) labeling targets for ingestion by phagocytes
2) rendering viruses and toxins inert through neutralization
3) blocking adhesion of microbes to body tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
228
Q

Describe clonal expansion when infection occurs

A

Only lymphocytes responsive to epitopes found on/in the invading pathogens are activated and proliferate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
229
Q

What are effector cells?

A

Either T cells that secrete cytokines or cytotoxic OR B cells that produce antibodies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
230
Q

Describe memory cells

A
  • enter quiescent state and become long-lived
  • lie in wait for reinfection with same microbe
  • when they encounter their specific epitope, they activate rapidly, resulting in faster response of greater magnitude that the primary response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

Where do thymocytes enter the thymus?

A

Cortico-medullary junction

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
232
Q

What are the stages of T cell differentiation?

A
  • double negative stage
  • double positive stage
  • mature T cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

What are the 3 pairs that occur in CD3 complex?

A
  • delta-epsilon
  • gamma-epsilon
  • tau-tau
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

What type of antigens is not recognized alone by T cell receptors?

A

Peptide antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
235
Q

Where does negative selection occur?

A

In the corticomedullary and medulla region of the thymus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
236
Q

When is a T cell considered mature?

A

When it exits the thymus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
237
Q

What is a naive T cells?

A
  • mature T cells that have not yet encounter d the specific peptide epitope recognized by their TCR
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
238
Q

What are all B cells derived from?

A

Hematopoietic stem cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
239
Q

What is antigen-independent phase?

A
  • first phase of B cell development in bone marrow that results in mature B-cells that have not been exposed to antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

What are the subpopulations of antigen independent phase?

A
  • pro-B cells
  • pre-B cells
  • immature B cells
  • mature B cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
241
Q

Describe the antigen dependent phase?

A

If B cells are stimulated by antigen, it undergoes transformation to a blast stage that forms memory cells and plasma cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
242
Q

Describe Pro B cells

A
  • stage in B-cell development in which rearrangement of the genes that code for the heavy-chain region of an antibody occurs
  • B-cell progenitors receive signals from bone marrow stromal cell to cell contact as well as soluble cytokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
243
Q

What is the most important event of Pro B cell phase?

A

Rearrangement of the B-cell receptors (BCR) genes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
244
Q

What do BCR and TCRs have in common?

A

1) composed of 2 different chains
2) have variable regions which determine their specificity
3) have constant regions, which allow for intracellular signaling activation of the lymphocytes expressing them
4) have a similar gene regions (V, D, J)
5) use similar mechanisms for gene rearrangement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
245
Q

What are the two chains in BCRs?

A

Light and heavy chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
246
Q

What genes in BCR contain multiple V, D, and J segements?

A

Heavy chain genes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
247
Q

What needs to occur for a Pro B cell to progress to the next phase of differentiation?

A

At least one heavy chain gene must undergo successful rearrangement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
248
Q

Describe the Pre-B cell phase

A
  • the stage of B cell development where the heavy chain part of the antibody molecule is present
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
249
Q

What are the two types of light chains possessed by humans?

A

Kappa
Lambda

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
250
Q

What is the first immunoglobulin produced?

A

IgM

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
251
Q

What signifies the Pre-B cells entry into the next phase, immature B cells?

A

The appearance of a functional BCR on the B cell surface

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
252
Q

What determines the antigen specificity of immature B cell and its IgM BCR?

A

The immunoglobulin variable region, found on both the light and heavy chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
253
Q

What happens to prevent BCRs from responding to self-antigens?

A

Negative selection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
254
Q

What occurs when B cells reach maturity?

A

B-cells respond to binding of antigen to the BCR by activation, proliferation, and antibody production

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
255
Q

What is the reaction of immature B cells when signaled?

A

Halting their development and undergoing apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
256
Q

Describe central tolerance

A
  • elimination of B cells that bear self-reactive receptors
  • it is estimated that more than 90% of B cells die in this manner
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
257
Q

What surface markers begin to make an appearance during the immature B cell phase?

A
  • IgM
  • CD21
  • CD40
  • class II MHC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
258
Q

When is a B cell considered mature?

A

Expresses a functional IgM BCR, survives selection by not reacting to self antigens and begins to display certain B cell markers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
259
Q

What occurs once a B cell is considered mature?

A
  • exit bone marrow and are carried in the blood to the spleen for the next stage of development
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
260
Q

What are the two types of mature B cells that develop in the spleen?

A
  • follicular B cells (majority)
  • marginal-zone cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
261
Q

Where can follicular cells be found?

A

Constantly recirculate between blood and secondary lymphoid organs in search of their specific antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
262
Q

Where can marginal-zone B cells be found?

A

Remain in spleen to respond quickly to blood-borne pathogens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
263
Q

Describe lymphoid follicles

A

Represent dense clusters of naiver B cells awaiting exposure to their specific antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
264
Q

Where are marginal-zone B cells located in the spleen?

A

Marginal sinus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
265
Q

What does a the presen se of both IgM and IgD on the cell membrane signify?

A

A mature B cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
266
Q

What are plasma cells role?

A

A differentiated B cell that actively secretes antibodies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
267
Q

What is a common resident of bone marrow and the germinal centers of peripheral lymphoid organs?

A

Plasma cells`

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
268
Q

What occurs when an infection occurs in the body tissues?

A
  • macrophages and dendritic cells are among the first immune cells to respond
  • APCs engulf pathogens at these distal sites of infection and carry associated antigen to local lymph nodes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
269
Q

What transforms a naive T cell to an activated T cells?

A

Combination of signals that arises when the TCR recognizes its specific peptide and CD28 is ligated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
270
Q

What are Th cells considered the most important cells of the adaptive response?

A
  • driving activities of other immune cells that act directly fight infection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
271
Q

What are the most prominent subsets of Th cells?

A

Th1
Th2
Th17

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
272
Q

What influences the type of Th cell subset that is produced after differentiation?

A

Influenced by the cytokines present during activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
273
Q

Describe role of Th1

A

-produce IFN-gamma, IL-2, and TNF-alpha which protect cells against intracellular pathogens by activating cytotoxic lymphocytes and macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
274
Q

Describe role of Th2

A

-essential role is to help B cells produce antibodies against extracellular pathogens and to generally regulate B cell activity
- produces: IL-4, IL-5, IL-6, IL-9, IL-10 and IL-13
- also may play role in allergies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
275
Q

Describe the role of Th17

A

Produce cytokines IL-17 and IL-22 which lead to the recruitment of granulocytes in response to an extracellular bacterial infection damage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
276
Q

Describe T regulatory cells (Treg cells)?

A
  • subset of T cell
  • posses the CD4 and CD25 antigens
  • play important role in suppressing the immune response to self-antigens and harmless antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
277
Q

How do Treg cells suppress the immune response to self and harmless antigens?

A

They proliferate of other T cell populations by secreting inhibitory cytokines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
278
Q

What are the two main distinct population created after cell division of Th cells?

A

1) most Th cells begin to secrete to cytokines and may travel to infected tissues where their activities are most needed
2) a small percentage of Th cells generated after activation will differentiate into memory cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
279
Q

What are the two toxic substances found in cytotoxic T cell granules?

A
  • perforins
  • granzymes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
280
Q

How do T-dependent antigens get their name?

A

The follicular B cell response depends heavily on the activity of Tfh cells to promote an effective antibody response, antigens that provoke this type of response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
281
Q

How did T-independent antigens get their name?

A

Marginal-zone B cells don’t require the help of Tfh cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
282
Q

T-dependent antigens are most likely what?

A

Proteins, only type of antigen that can stimulate a T cell response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
283
Q

What are the two possible fates of daughter B cells that are produced during proliferative phase of T dependent response?

A

1) remain in contact with T cells and differentiate into IgM secreting plasma cells
2) others form germinal centers within follicles and participate in a series of processes that enhance the antibody response through time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
284
Q

What are the three overlapping processes of the germinal center formation require?

A

1) immunoglobulin isotope switching
2) activity maturation
3) memory cell generation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
285
Q

Describe IgG

A
  • predominant form of antibody found in blood
  • found in intestines and the bodies secretion
  • associated with allergies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
286
Q

What is isotope switching?

A
  • under the direction of T cells, germinal center B cells can change which class of antibody they express
  • also determines the class of antibody secreted once the B cell differentiates into a plasma cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
287
Q

What is affinity maturation?

A
  • immunoglobulin binds antigen with increasing strength (affinity) through the course of an immune response, resulting in the production of even more effective antibodies
  • this is accomplished through somatic hypermutation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
288
Q

What is somatic hypermutations?

A

Appearance of mutations in immunoglobulin gene variable regions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
289
Q

What causes daughter cells to be produced with slightly different antigen-binding abilities

A

Somatic hypermutation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
290
Q

For each generation of daughter cells produced within the germinal center, there are 3 basic populations of cells formed. What are these cells?

A

1) plasma cells
2) memory cells
3) B cells hat remain in the germinal center to continue the process of affinity maturation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
291
Q

What occurs once a memory B cell is re-exposed to an antigen?

A

It can rapidly respond with the production of high affinity, class switched antibody

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
292
Q

What are the Pro-B cells key CD markers?

A

CD10 and CD19

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
293
Q

What are the Pre-B cell key CD markers>?

A

CD10
CD19
CD20

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
294
Q

What are the immature B cell key CD markers?

A

CD10
CD19
CD20
CD21
CD40

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
295
Q

What are the mature B cell key CD markers?

A

CD19
CD20
CD21
CD40

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
296
Q

What are the Pro-B cell receptors?

A

None

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
297
Q

What is the Pre-B cell receptor

A

Pre-BCR: immunoglobulin heavy chain and surrogate light chain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
298
Q

What is the immature B cell receptors?

A

Functional BCR: IgM heavy chains and kappa or lambda light chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
299
Q

What is the mature B cell receptor?

A

Functional BCR: IgD or IgM gravy chains and kappa or lambda light chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
300
Q

What is the double negative key CD marker?

A

CD3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
301
Q

What is the double negative key CD marker?

A

CD3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
302
Q

What is the double negative T cell receptors?

A

None

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
303
Q

What are the double positive key CD markers?

A

CD3
CD4
CD8

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
304
Q

What are the double positive T cell receptors?

A

TCR alpha
TCR beta

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
305
Q

What are the single positive key CD markers?

A

CD3
CD4
CD8

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
306
Q

What are the single positive T cell receptors?

A

TCR alpha
TCR beta

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
307
Q

What is a characteristic of immune response to a T-independent antigen?

A
  • antigen are often polysaccharides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
308
Q

How does the humoral immunity produce antibodies?

A

By plasma cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
309
Q

What is negative selection?>

A
  • process that takes place among surviving DP T cells in the corticomedullary region and the medulla of the thymus
  • strong reactions with self-peptides other than MHC antigens triggers apoptosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
310
Q

What are thymocytes?

A

Lymphocyte precursors in the thymus that are committed to becoming T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
311
Q

Describe immature B cells

A

A phase in the growth of B cells characterized by the appearance of complete IgM antibody molecules on the cell surface

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
312
Q

What are variables?

A
  • contained by both alpha and beta chains on TCR
  • recognizes specific antigens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
313
Q

What is MHC restriction?

A

Selection of thymocytes that will only interact with the MHC antigens found on host cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
314
Q

What is a surrogate light chain?

A
  • consists of two short polypeptide chains that are noncovalently associated, along with two shorter chains, Ig-alpha and Ig-beta
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
315
Q

What is allelic exclusions?

A

Selection of an allele on one chromosome only

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
316
Q

What is clonal deletion?

A

Process of elimination of clones of cells that would be capable of an autoimmune responses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
317
Q

Describe Flow cytometry

A

Automated system for identifying cells based on the scattering of light as cells flow single file in a stream of fluid by a laser beam

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
318
Q

What is positive selection?

A

allows only DP cells with functional TCR receptors to survive

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
319
Q

What is a distinguishing feature of Pre-B cells?

A

Mu chains in the cytoplasm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
320
Q

What are the 3 characteristics of acute phase reactants?

A

1) rapid increase following infection
2) enhancement of phagocytosis
3) nonspecific indicators of inflammation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
321
Q

What are 3 characteristics of an immunogen?

A

1) large molecular weight
2) internal complexity
3) presence of numerous epitopes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
322
Q

Where are class II MHC ANTIGENS found

A

B cells
macrophage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
323
Q

What is an immunoglobulin?

A

Globular proteins (glycoproteins) that play a role in immunity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
324
Q

What is the composition of immunoglobulins?

A
  • 86-98% polypeptides
  • 2-14% carbohydrates
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
325
Q

What percentage of plasma proteins are immunoglobulins?

A

20%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
326
Q

Describe serum protein electrophoresis procedure

A

1) serum is placed on a agarose gel
2) electric current applied to separate the proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
327
Q

At what pH does electrophoresis separates most serum proteins by size and change?

A

8.6

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
328
Q

What do the results of electrophoresis appear as?

A

Five bands

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
329
Q

What are the slowest moving protein?

A

Immunoglobulins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
330
Q

What band of electrophoresis contains the most antibody activity’?

A

Gamma band

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
331
Q

What are the 5 classes of immunoglobulins?

A

IgG
IgM
IgD
IgE
IgA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
332
Q

Describe the two dimensional forms of IgG

A
  • two large peptide chains (heavy chains, unique)
  • two small peptide chains (light chains, kappa or lambda)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
333
Q

How do the kappa and lambda light chains differ?

A
  • differ by just a few amino acid substitutions along their length
  • no functional difference
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
334
Q

What are heavy and light chains held together by?

A
  • Disulfide bonds (s-s)
  • hydrogen bonds
  • hydrophobic forces
  • electrostatic attractions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
335
Q

What is the structural unit of all the immunoglobulin classes?

A

Tetrapeptide structure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
336
Q

Where is the variable region of an immunoglobulin located?

A

The first 110 amino acids of the molecule at the amino terminal

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
337
Q

Describe the variable region of an immunoglobulin

A
  • unique to each other antibody
  • allows the molecule to bind specifically to a particular antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
338
Q

Where is the constant region of the immunoglobulin located?

A

From amino acid 111 to the carboxyl terminal

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
339
Q

Describe the constant region of an immunoglobulin

A
  • are the same in each immunoglobulin class/subclass
  • responsible for the biological functions that play a role in immune defense against an antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
340
Q

What are the three constant regions of an immunoglobulin?

A

CH1
CH2
CH3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
341
Q

What are CH2 and CH3 responsible for?

A

For binding to complement and Fc receptors on phagocytic cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
342
Q

What are the prototypic enzymes?

A

Pepsin and papain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
343
Q

Describe Papain

A
  • cleaves the IgG molecule below the set of the disulfide bonds that holds the two heavy chains together, resulting in formation of 3 fragments: 2 Fab fragments and Fc fragment
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
344
Q

Describe Fab fragments

A
  • each have one light chain and one half of a heavy chain
  • located at amino-terminal end o the molecule
  • both are identical
  • have antigen-binding capability
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
345
Q

Describe the Fc fragment

A
  • consists of carboxyl-terminal halves of two heavy chain
  • spontaneously crystallizes at 4 C
  • does not bind antigen
  • contain constant regions CH2 and CH3
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
346
Q

Describe pepsin

A
  • cleaves IgG at the carboxyl-terminal side of the inter chain disulfide bonds
  • 2 fragments: F(ab’)2 fragment and Fc’
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
347
Q

Describe the F(ab’)2 fragment

A
  • contains all antigen binding ability in pepsin
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
348
Q

Describe Fc’ (fragment of pepsin)

A
  • in carboxyl-terminal portion of the molecule which is disintergrated into several smaller pieces
  • has no biological activity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
349
Q

Describe the hinge region of an immunoglobulin

A
  • segment of heavy chain located between the CH1 and CH2
  • rich in hydrophobic residues
  • high proline content that allows for flexibility
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
350
Q

What is flexibility important in the hinge region of immunoglobulin?

A
  • let’s two antigen binding sites operate independently and engage in angular motion relative to each other and to the Fc stem
  • initiation of complement cascade
  • binding to cells with specific receptors for the Fc portion of molecule
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
351
Q

What immunoglobulins have a hinge region?

A

IgG
IgD
IgA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
352
Q

Why do IgM and IgE gave flexibility even though they do not have a hinge region?

A

CH2 are paired in such a way as to confer flexibility to the Fab arms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
353
Q

What do immunoglobulins contain>?

A

4 polypeptide chains
Carbohydrate portion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
354
Q

Where is the carbohydrate portion of immunoglobulins located?

A

Between the CH2 domains of the two heavy chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
355
Q

What are the functions of the carbohydate portion of immunoglobulins?

A

1) increase solubility of immunoglobulin
2) provide protection against degradation of molecule
3) enhancing functional activity of Fc domain (most important)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
356
Q

What are the three antigenic determinants?

A

Isotype
Allotype
Idiotype

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
357
Q

describe isotype

A
  • unique amino acid sequences that are common to all immunoglobulins of given class/subclass
  • identical in all individuals or given species and differ from one species to another
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
358
Q

How can antibodies to human isotypes be prepared?

A

By immunizing animals with human serum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
359
Q

What are allotypes?

A
  • minor variations of amino acid sequences that are present in some individuals of the same species but not others
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
360
Q

What classes do allotypes occur in?

A
  • four Ig classes
  • one IgA subclass
  • lambda light chain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
361
Q

Where are allotypes located in an immunoglobulin?

A
  • found in the constant region
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
362
Q

What are idiotypes?

A
  • the variable portion of each immunoglobulin chain are unique to a specific-antibody molecule
  • essential to the formation of the antigen binding site
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
363
Q

Where are idiotypes located in the immunoglobulin?

A

At the amino-terminal ends of both heavy chains and light chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
364
Q

What is the predominant immunoglobulin in humans?

A

IgG

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
365
Q

What % of immunoglobulins are IgG?

A

70-75%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
366
Q

What immunoglobulin has the longest half life?

A

IgG

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
367
Q

Describe structure of IgG

A
  • monomer
  • consists of one tetrapeptide unit
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
368
Q

What are the 4 subclasses of IgG? What is their compositions?

A

IgG1: 66%
IgG2: 23%
IgG3: 7%
IgG4: 4%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
369
Q

What are IgG1 and IgG3 associated with?

A

Protein antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
370
Q

What are IgG2 and IgG4 associated with?

A

Polysaccharide antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
371
Q

What immunoglobulins can cross the placenta?

A

IgG (except IgG2)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
372
Q

What are the functions of IgG?

A

1) CH2 region is able to bind to complement
2) important mediator of opsonization
3) participates in antibody-dependent cellular cytotoxicity
4) ability to bind to bacterial toxins and viruses and neutralizes their activity
5) can cross placenta
6) agglutination reactions and precipitation takes place in vitro

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
373
Q

What does activation of complement result in?

A

An enhanced inflammatory response and destruction of foreign cells such as bacteria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
374
Q

What is the most efficient subclass of IgG to bind completeIgM? Why?

A

IgG3: it has the largest hinge region and the largest number of interchain disulfide bonds

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
375
Q

What is the 2nd most efficient subclass of IgG to bind complement
?

A

IgG1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
376
Q

What is opsonization?

A

The coating of a foreign antigen that leads to enhanced phagocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
377
Q

Why are IgG1 and IgG3 particularly good opsonins?

A

They bind most strongly to Fc receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
378
Q

Why is passive transfer of maternal IgG to the fetus important?

A

Providing immunity to the newborn during its first few months of life, when its immune system is immature

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
379
Q

Is IgG better at agglutination or precipitation? Why?

A

Precipitation because it involves small soluble particles, which are brought together more easily by the relatively small IgG molecule

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
380
Q

What else is IgM called? Where does it get this name?

A

Macroglobulin because it is the largest of all the immunoglobulin classes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
381
Q

Describe the structure of IgM

A
  • pentameter of five monomers held together by J chain (joining)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
382
Q

Describe J chain

A
  • glycoprotein that holds together monomers
  • cysteine residues form disulfide bonds that link the carboxyl-terminal ends of adjacent monomers in IgM and IgA
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
383
Q

What does each monomer of IgM contain?

A
  • Mu heavy chains and either kappa or lambda chains
  • Mu heavy chains possess one more constant domain (CH4) adding to it large size
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
384
Q

What % of immunoglobulins are IgM?

A

5-10%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
385
Q

What does IgM perform things more efficiently than IgG?

A

Because of IgMs multiple binding sites

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
386
Q

What immunoglobulin is most efficient at triggering the classical pathway?

A

IgM because a single molecule can initiate the reaction when complement binds to two adjacent CH region

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
387
Q

Where are IGN mainly found?

A

In the intravascular pool and not in other body fluids or tissues because of its large size

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
388
Q

What immunoglobulin is the first to appear after antigenic stimulation?

A

IgM

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
389
Q

What is the first immunoglobulin that appears in maturing infants?

A

IgM

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
390
Q

How many binding sites does IgM have?

A

10

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
391
Q

What % of immunoglobulins are IgA?

A

10-15%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
392
Q

Where does IgA migrate in electrophoresis?

A

Between the beta and gamma bands

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
393
Q

What immunoglobulins have complement fixation?

A

IgG
IgM

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
394
Q

Describe the basic structure of IgA

A
  • alpha heavy chain
  • contains one variable regions
  • contains three constant regions
  • dimer, two monomers held together by J chain
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
395
Q

What are the two subclasses of IgA?

A

IgA1
IgA2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
396
Q

How do IgA1 and IgA2 differ?

A

By 22 amino acids, 13 of which are located in hinge region (deleted in IgA2)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
397
Q

Where are IgA2 found?

A
  • secretions at mucosal surfaces
  • along respiratory, urogenital, and intestinal mucosa
  • appears in breast milk, saliva, tears, sweat, and colostrum
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
398
Q

Where are IgA1 found?

A

In serum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
399
Q

What is the major role of serum IgA?

A
  • anti-inflammatory agent
  • down regulate IgG mediated phagocytosis, chemotaxis, bactericidal activity, and cytokine release
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
400
Q

Why is the J chain essential for in IgA?

A

Essential for polymerization and secretion of IgA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
401
Q

Where are secretory IgA synthesized?

A

Plasma cells found mainly in mucosal-associated lymphoid tissue and is released dimeric form

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
402
Q

Describe secretary component

A
  • serves as specific receptors for IgA
  • precursor is present on surface of epithelial cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
403
Q

Where do plasma cells secret IgA>?

A

Subepithelial tissue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
404
Q

What happens once IgA bind to SC precursors?

A
  • Transcytosis
  • both of them are taken inside the cell and released at the opposite surface
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
405
Q

What is the main function if secretory IgA?

A
  • to patrol mucosal surfaces and act as a first line of defense by preventing farther into the body
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
406
Q

What is the important role of IgA?

A
  • neutralizing toxins produced by microorganisms and helps prevent bacterial and viral adherence to mucosal surfaces
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
407
Q

What immunoglobulin complex is trapped by mucus and coughed/sneezed out to protect the respiratory system?

A

IgA complex

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
408
Q

Where is IgA found in humans?

A

Breast milk

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
409
Q

How does breastfeeding help to maintain the health of newborns?

A

Passively transferring antibodies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
410
Q

What cells posses specific receptors for IgA?

A

Macrophages
Monocytes
Neutrophils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
411
Q

What % of immunoglobulins are IgD?

A

0.001%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
412
Q

What is the half day of IgD?

A

1-3 days

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
413
Q

What antibody migrates as fast as a gamma protein?

A

IgD

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
414
Q

Describe hinge region of IgD

A
  • unusually long
  • 58 amino acids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
415
Q

Why is IgD more susceptible to proteolysis that any other antibody?

A

Because of its long hinge region, this may be the reason for its short half life

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
416
Q

Where are most IgD found?

A

Surface of unstimulated B cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
417
Q

What is the second antibody that appears during an immune response?

A

IgD

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
418
Q

What are the roles of IgD?

A
  • B cell maturation and differentiation
  • not exactly understood
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
419
Q

Why is IgD an ideal early responder?

A

High level of surface expression and its intrinsic flexibility

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
420
Q

What is IgE best known for?

A

Very low concentration in serum and the fact that it has the ability to activate mast cells and basophils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
421
Q

What % of immunoglobulins are IgE?

A

0.0005%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
422
Q

Describe the IgE structure

A
  • epsilon heavy chain is composed of one variables and four constant domains
  • a single disulfide bond joins each epsilon chain to a light chain, and two disulfide bonds link the heavy chains to one another
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
423
Q

Does IgE participate in complement fixation, agglutination, or opsonization?

A

None of them

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
424
Q

Where can plasma cells that produce IgE be located?

A

Primarily in lungs and in the skin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
425
Q

Where can mast cells be located?

A
  • mainly in the skin
  • lining of the respiratory and alimentary tracts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
426
Q

What does the release of IgE mediators induce?

A
  • type I hypersensitivity
  • allergic reaction
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
427
Q

What are typical reactions with allergic reactions?

A
  • anaphylactic shock
  • asthma
  • diarrhea
  • hay fever
  • hives
  • vomiting
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
428
Q

Describe the role of IgE

A

Serve a protective role by triggering an acute inflammatory reaction that recruits neutrophils and eosinophils to the area to help destroy invading antigens that have penetrated IgA defenses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
429
Q

What occurs when the first time an individual is exposed to an antigen?

A

Primary antibody response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
430
Q

What are characteristics of primary antibody response?

A
  • IgM shows up first
  • lag phase between the encounter with the antigen and the production of detectable antibody
  • low affinity
  • amount of antibody produced are relatively low and decline during the span of a few weeks
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
431
Q

Describe the lag phase of the primary antibody response

A
  • lasts 4 and 7 days
    -T and B cells are being activated to respond to the antigen by the T-dependent mechanisms of antibody production
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
432
Q

What does the lag phase of the primary antibody response result in?

A

Results in the generation of antibody-secreting plasma cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
433
Q

What do activated B cells from the primary response develop into?

A
  • plasma cells
  • expand into clones of memory cells (long lived)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
434
Q

Describe secondary antibody response?

A

If memory cells are exposed to the same antigen weeks, months, or even years later, they can rapidly differentiate into plasma cells and larger amounts of antibody are produced

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
435
Q

What are characteristics of the secondary antibody production?

A
  • shorter lag time: 1-2 days
  • high affinity
  • production of low levels of IgM that rapidly declibe
  • higher levels of other immunoglobulin isotypes, mainly IgG levels decline slowy and persists for longer periods
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
436
Q

Describe clonal selection hypothesis

A
  • body has numerous close of lymphocytes, each possessing surface receptors with a unique specificity
  • when body is exposed to antigen, the antigen selectively binds to receptors on cells to proliferate and mount antibody responses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
437
Q

When can a gene be transcribed and translated into functional antibody molecule?

A

Not until it undergoes rearrangement, assisted by special recombinase enzymes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
438
Q

What does gene rearrangement involve?

A

Cutting and splicing process that removes much of the intervening DNA, resulting in a functional gene that codes for a specific antibody

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
439
Q

Where does rearrangement of the gene begin?

A

The heavy chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
440
Q

What are the genes that code for variable region of the heavy chain divided into?

A

Divided into 3 groups;
- V genes (variable) —> 45 genes
- D genes (diversity) —> 23 genes
- J genes (joining) —> 6 genes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
441
Q

Where are all the V,D and J genes present?

A

The germ line DNA of a bone marrow stem cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
442
Q

Joining of V, D, J and constant region of a heavy chain is a two step process. What are the steps?

A

1) In Pro-B cells, one D gene and one J gene are randomly chose and are joined by means of a recombines enzyme after the intervening DNA is deleted
2) in Pre-B cells, a V gene is joined to the DJ complex, resulting in a rearranged VDJ gene

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
443
Q

What is allelic exclusion?

A

The genes on the second chromosome are not rearranged because the rearrangement of DNA on one chromosome 14 was successful the first time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
444
Q

What occurs is the first rearrangement of heavy chains is unsuccessful?

A
  • rearrangement of the second set of genes on the other chromosome 14 occurs
  • this mechanism maintains clonal specificity by ensuring each B cell only expresses a single antigen receptor
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
445
Q

What happens after rearrangement of the genes of the heavy chains?

A
  • the variable and constant regions are joined
  • occurs at the RNA level, thus conserving the DNA of the constant regions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
446
Q

What are the markers of the Pre-B cells?

A

Mu heavy chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
447
Q

What does the presence of both C”Mu” and C”delta” regions of heavy chains allow?

A

Allows for mRNA for IgD and IgM t be transcribed at the same time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
448
Q

When do light chains rearrangement occur?

A

After Mu chains appear

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
449
Q

Describe VJ joining of light chains

A
  • accomplished by cutting out one intervening DNA
  • results in the V(k) and J(k) segments becoming permanently joined to one another on the rearranged chromosome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
450
Q

When does lambda chain synthesis occur in a light chain?

A

Only it a nonfunctioning gene product arises from Kappa rearrangement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
451
Q

What does the lambda locus consist of?

A

Approximately:
- 30 V(lambda) segments
- 4 J(lambda) segments
- 4 C(lambda) segments

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
452
Q

If functional heavy and light chains are not produced but a B cell, what occurs?

A

Cell dies by apoptosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
453
Q

Light chains are joined with Mu chains to form what?

A
  • a complete IgM antibody
  • first appears in immature B cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
454
Q

What happens once IgM and IgD are present on the surface membrane?

A

The B cell is fully mature and capable of responding to antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
455
Q

What are essential for initiating the VDJ recombination during B cell maturation?

A

Recombinase enzymes
RAG-1
RAG-2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
456
Q

What is the role of RAG-1 and RAG-2?

A

Recognize specific recombination signal sequences in the DNA that flank all immunoglobulin gene segments

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
457
Q

What is junctional diversity?

A
  • V, D and J segments doesn’t always occur at a fixed position, so each sequence can vary by small number of nucleotides
  • major contributor to diversity in the variable-region genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
458
Q

Where do immunoglobulins get their variation diversity from?

A

1) junctional diversity
2) different heavy chains can combine to different light chains
3) somatic hypermutation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
459
Q

What is class switching?

A

A the immune response progresses, B cells, may become capable of producing antibody of another class

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
460
Q

What is switch recombination?

A
  • a portion of the constant region DNA is deleted and the remaining C(h) genes are placed adjacent to the variable to the variable region genes
  • allows the same VDJ region to be coupled with a different C regions to produce-antibody of a different class but having identical specificity for antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
461
Q

What are monoclonal antibodies?

A
  • derived from singe antibody-producing cell that has reproduced many times to form a clone
  • all the cells in the clone are identical and the antibody produced is exactly the same
    -directed agains a specific epitope
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
462
Q

what is hybridoma?

A
  • fusion of activated B cell and myeloma cell
  • myeloma cell chosen lacks essential enzymes and cannot produce own antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
463
Q

How does production of hybridomas begin?

A

By immunizing a mouse with the desired antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
464
Q

What is the traditional process of mouse antibody production?

A

1) immunize a mouse with a specific-antigen
2) harvest spleen cells from the mouse spleen
3) combine spleen cells with myeloma cells in the presence of PEG
4) select fused cells and screen for presence of desired antibody
5) grow positive cells in larger quantities

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
465
Q

What was used before monoclonal antibodies?

A
  • antibody reagent could only be produced by immunizing animals such as horses or goats with the desired antigen and isolating polyclonal antibodies from the animal serum
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
466
Q

What are the primary advantages of the monoclonal antibody reagent?

A
  • provide decreased lot-to-lot variaton
  • increased specificity toward a single epitope, rather than multiple epitopes of an antigen
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
467
Q

What is a major limitation of using mouse monoclonal antibody as therapeutic agents?

A
  • highly immunogenic for humans, inducing the development of human anti-mouse antibodies (HAMAs) that can cause severe hypersensitivity reactions
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
468
Q

Where are isotypes located?

A

Constant region

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
469
Q

Where are allotypes located?

A

In the constant regions of the IgG subclasses, one IgA subclass, and light chain

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
470
Q

Where are idiotypes located?

A

Amino terminal regions of heavy and light chains

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
471
Q

Where do antigens bind on immunoglobulins?

A

Hypervariable regions that contain CDRs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
472
Q

What are complement fixation functions?

A

1) enhances inflammatory response
2) uses positive feedback cycle to promote phagocytosis
3) enlists more defensive elements

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
473
Q

Explain the process of complement fixation

A

1) antibodies bound to cells change shape and expose complement binding sites
2) this triggers complement fixation and cell lysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
474
Q

Describe neutralization of antibody action

A

Antibodies bind to and block specific sites on viruses or exotoxins, thus preventing these antigens from binding to receptors on tissue cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
475
Q

Describe agglutination of antibody action

A
  • makes antigen-antibody complexes that are cross-linked, causing clumping
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
476
Q

Describe precipitation of antibody action?

A
  • soluble molecules are cross-linked into large insoluble complexes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
477
Q

What enhances phagocytosis at the antigen-antibody complex?

A
  • neutralization
  • agglutionation
  • precipitation
  • complement
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
478
Q

What enhances inflammation of antigen-antibody complex? co

A

Complement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
479
Q

What are cytokines?

A
  • chemical messengers that regulate immunity
  • small proteins that bin to and activate receptors located on the target cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
480
Q

Describe regulations of cytokines?

A
  • Important in determining where cytokine action will be effective or inappropriate
  • massive overproduction and dysregulation of cytokines can lead to yperctokinemia
  • results in hypotension, fever, edema, multi-organ failure, or death
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
481
Q

What is pleitotropy?

A

Single cytokine with many different actions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
482
Q

What is redundancy?

A
  • occurs when different cytokines activate some of the same pathway and genes
  • can be explained by the fact many cytokines share receptors subunits
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
483
Q

How is cytokine activity classified?

A

According to the distance traveled between the producing cell and its targets cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
484
Q

What is autocrine?

A

Cytokines that bind to receptors on the same cell from which they were secreted

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
485
Q

What is a paracrine?

A

Cytokines that act one cells within tissue region surrounding their cellular source

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
486
Q

What is an endocrine?

A

Some cytokines diffuse into the bloodstream, allowing them to influence cells far from the cells that produce them

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
487
Q

What is synergistic?

A

Interactions that complement and enhance each other

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
488
Q

What is antagonism?

A

If one cytokine counteracts the action of another

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
489
Q

What is a cytokine cascade?

A

Many cytokines induce the production of additional cytokines by targeting cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
490
Q

What is the antagonist of IL-4?

A

TNF-alpha

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
491
Q

What synergies with IFN-gamma?

A

TNF-alpha

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
492
Q

What is hypercytokinemia?

A
  • also referred to as cytokine storm
  • massive overproduction and dysregulation of cytokines can result in hyper stimulation of the immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
493
Q

What are the major proinflammatory cytokines?

A
  • TNF-alpha
  • IL-1
  • IL-6
  • IFN-gamma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
494
Q

What are the major anti-inflammatory cytokines?

A
  • TGF-beta
  • IL-10
  • IL-13
  • IL-35
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
495
Q

What is the most important role of IL-1, IL-6, and TNF-alpha?

A

Recruiting effector cells, such as neutrophils and monocytes, into inflamed tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
496
Q

What are the best characterized cytokines of the IL-1 family?

A

IL-1alpha
IL-1beta
IL-1RA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
497
Q

Describe IL-1 alpha

A
  • retained within the cell cytoplasm
  • only released after cell death
  • presence of IL-1alpha helps attract inflammatory cells to areas where cell and tissues are being damaged or killed
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
498
Q

Describe IL-1beta

A

-mediates most paracrine (local) and hormonal (sytstemic) activity
- before secretion, IL-1beta must be cleaved intracellularly to an active form

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
499
Q

Why is IL-1beta referred to as endogenous pyrogens?

A

It’s ability to induce fever

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
500
Q

What purpose does a fever have?

A

1) inhibit of many bacteria and fungi.
2) increase the microbicidial activities of macrophages and neutrophils
3) contribute to feeling of discomfort and fatigue, compelling individuals to rest

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
501
Q

What are the role of IL-1beta?

A

1) activation of phagocytosis
2) induce fever
3) production of acute-phase reactant

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
502
Q

What are TNF products of?

A

Macrophages and lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
503
Q

Describe CD40 ligand

A

Essential for signaling between T and B cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
504
Q

What is the most important TNF of inflammation?

A

TNF-alpha

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
505
Q

What is a major inducer of TNF-alpha production?

A

Microbial substances such as LPS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
506
Q

What is LPS?

A

A component of the cell wall of gram-negative bacteria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
507
Q

The activity of IL-6 is highly ________.

A

Pleiotropic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
508
Q

What are innate roles of IL-6?

A
  • stimulates the production of acute-phase proteins by liver hepatocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
509
Q

What are the adaptive roles of IL-6?

A
  • increases activation of B and T cells
  • causes B cells to proliferate and differentiate into plasma cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
510
Q

What are chemokines?

A

Subgroup of cytokines that influence the motility and migration of their tartlet cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
511
Q

What are the 4 families of chemokines?

A

1) CXC
2) CC
3) C
4) CX3C

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
512
Q

Describe CXC chemokines

A

Contain single amino acid between the first and second cysteines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
513
Q

Describe CC chemokines

A

The cysteines are found together, with no intervening amino acid

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
514
Q

Describe C chemokines

A

Has only a single cysteine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
515
Q

Describe CX3C chemokines

A

Have 3 amino acids between the cysteines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
516
Q

What is the most important effect of chemokines?

A
  • recruitment of leukocytes from the blood into infected or damaged tissues
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
517
Q

What are interferons?

A

Soluble substances produced by virally infected cells that interfere with the ability or viruses to replicate by making host cells less hospitable to viral takeover

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
518
Q

What are the 3 groups of IFNs?

A

Type I
Type II
Type III

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
519
Q

What are the most important cytokines in response to viral infections?

A

IFN- alpha
IFN-beta
(Both type I)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
520
Q

What are the functions of type I IFN?`

A

1) interfere with viral replication
2) activate NK cells
3) enhances expression of class I MHC proteins on target cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
521
Q

What is the most important innate defense mechanism against viral infection?

A

Combination of IFN-alpha and IFN-beta and NK cell killing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
522
Q

What secretes the majority of the cytokines in the adaptive immunity?

A

T cells, especially T helper Cells (CD4+)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
523
Q

What are the 4 sub populations of T helper cells?

A

Th1
Th2
Th17
Treg cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
524
Q

Describe differentiation of Th cells

A

The cells undertake a process during which they are transformed into one of the four subclasses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
525
Q

What causes differentiation to the Th1 lineage?

A

Dendritic cell production of cytokine IL-12

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
526
Q

What drives antibody mediated immunity?

A

IL-4

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
527
Q

What arises in the presence of IL-23?

A

Th17 cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
528
Q

What are produced in response to TGF-beta?

A

Treg cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
529
Q

what is the hallmark of Th1 cells?

A

High level expression of the proliferative cytokine IL-2 and IFN-gamma, a type II IFN

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
530
Q

What are the roles of Th1 cells?

A

1) expand to combat infection with viruses and intracellular bacteria
2) promote cell-mediated immunity with cytotoxic T cells, and activate macrophages
3) cause antigen-activated B cells to produce IgG1 and IgG3 antibodies capable of opsonization and fixing complement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
531
Q

What is another name for IL-2?

A

T-cell growth factor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
532
Q

What are the roles of IL-2?

A

1) drives proliferation and differentiation of both T and B cells
2) enhances the lytic activity of NK cells
3) causes naive Th to differentiate into Th1 cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
533
Q

What is the primary cytokine of the Th1 response?

A

IFN-gamma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
534
Q

What are the roles of IFN-gamma?

A

1) influences expression of more than 200 target-cell genes
2) stimulates antigen presentation by class I and class II MHC
3) potent activator of macrophages
4) involved in regulation and activation of CD4+ Th1 cells, CD8+ cytotoxic T cells and NK cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
535
Q

What are the most influential Th2 cytokines?

A

IL-4
IL-10

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
536
Q

What is the IL-4 activity on naive T cells?

A
  • turns on genes that generate Th2 cells and turns off genes that promote Th1 cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
537
Q

What are Th2 cells responsible for regulating?

A

Allergies
Autoimmune disease
Parasites

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
538
Q

What does IL-5 do along with IgE?

A

Drives differentiation and activation of eosinophils in both allergic immune response and responses to parasitic infection

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
539
Q

What does IL-4 and IL-13 have in common?

A
  • induce worm expulsion
  • favor IgE class switching
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
540
Q

How do IL-4 and IL-13 differ?

A

IL-13 plays anti-inflammatory role by inhibiting activation and cytokine secretion by monocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
541
Q

What are the roles of IL-10?

A

1) has anti-inflammatory and suppressive effects on Th1 cells
2) inhibits antigen presentation by macrophages and dendritic cells
3) down regulates immune response by counteracting IFN-gamma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
542
Q

What is the antagonist of IFN-gamma?

A

IL-10

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
543
Q

How can Treg cells be identified?

A

Expression of CD4, CD25, and FoxP3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
544
Q

What are Treg cells essential?

A

For establishing peripheral tolerance to a wide variety of self-antigens and harmless antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
545
Q

What are the roles of Treg cells?

A

1) establish peripheral tolerance to self-antigens and harmless antigens
2) produce suppressive cytokines: TGF-beta and IL-10
3) down regulate immune response to prevent chronic inflammation (main)
4) may prevent tumor cells from attack by inhibiting cancer-fight cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
546
Q

What does Th17 secrete?

A

IL-17 family cytokines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
547
Q

What cytokines play a role in finalizing the commitment to Th17?

A

IL-23

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
548
Q

What are the roles of Th17?

A

1) host defense against bacterial and fungal infection at mucosal surfaces
2) secretion of IL-17
3) promote recruitment of neutrophils
4) promote release of antimicrobial peptides
5) when dysregulated, may promote pathogenesis of several inflammatory dieases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
549
Q

What are the APC cytokines?

A

IL-12
IL-23

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
550
Q

What are the primary mediators of hematopoiesis called? Why?

A
  • IL-3
  • EPO
  • G-CSF
  • M-CSF
  • GM-CSF
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
551
Q

Why does IL-3 act one bone marrow stem cells?

A

To begin differentiation cycle

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
552
Q

What happens if M-CSF is activated?

A

The cells become macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
553
Q

What is the role of M-CSF?

A

Increases phagocytosis, chemotaxis, and additional cytokine production in monocytes and macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
554
Q

What happens if G-CSF is activated?

A

Cells become neutrophils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
555
Q

What are the roles of G-CSF?

A

1) enhances function of mature neutrophils
2) affects the survival, proliferation, and differentiation of all cell types in neutrophil lineage
3) decreases IFN-gamma production
4) increases production IL-4 in T cells
5) mobilizes multi potent stem cells from the bone marrow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
556
Q

What does GM-CSF do?

A

Acts to drive differentiation toward other WBC types

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
557
Q

What does EPO do?

A

Regulate RBC production in the bone marrow

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
558
Q

What is EPO mainly produced by?

A

The kidneys

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
559
Q

What is recombinant EPO?

A
  • administered to improve RBC counts for individual with anemia and those with cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
560
Q

What is anti-cytokine therapy?

A

Aims to break vicious cycle of chronic inflammation by targeting the interactions between specific cytokines cytokines and their cognate receptors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
561
Q

Describe newer anti-cytokine agents

A

Greatly improved using recombinant DNA techniques to generate humanized monoclonal DNA antibodies that are much less immunogenic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
562
Q

What are 3 cytokine assay formats?

A

1) ELISAs
2) microbead assays
3) ELISpot assays

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
563
Q

Describe ELISAs

A
  • can detect many pro- and anti- inflammatory cytokines in one reaction
  • allow for simultaneous detection of several cytokines from serum or plasma in single a test run
  • has “spots”
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
564
Q

Describe microbead assays

A
  • allow for the simultaneous detection of multiple cytokines in a single tube
  • each bead type has own fluorescent wavelength. When combined with fluorescent secondary antibody bound to a specific cytokine, allows for detection stop to 100 different analysis in one tube
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
565
Q

Describe ELISpot

A
  • similar to ELISA performed on in vitro activated peripheral WBC
  • allow the detection and enumeration of individual cytokine-specific capture antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
566
Q

What is the ELISpot procedure?

A

1) PBMCs are added to the wells of antibody-coated micro plate and treat with synthetic protein antigen
2) incubation period: Th1 cells secrete cytokines as they rest as the rest at the bottom of the microplate well
3) PBMCs and any soluble substances (cytokines) are washed from wells
4) detection antibody antibody specific to the target cytokine, is added to wells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
567
Q

What is the role of ELISpot detection antibody?

A

To produce a signal indicating where target cytokine is bound to capture antibody

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
568
Q

What do the “spots” represent in ELISpot?

A

Tiny, elliptical silhouettes of captured IFN-gamma speckling plate bottom

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
569
Q

What is PCR product?

A

Made using fluorescent-labeled primer and can be hybridized to either solid or liquid phase arrays

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
570
Q

Describe the solid-phase array of PCR

A
  • have up to 40,000 spots containing specific single-stranded DNA (ssDNA) oligonucleotides representing individual genes
  • fluorescence of a spot indicates that gene was expressed in the cell and that cell was producing the cytokine
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
571
Q

Describe liquid arrays of PCR

A
  • uses same beads as microbeads arrays but have oligionucleotides on surfaces instead of antibodies. Show up to 100 different complementary DNAs to be identified
  • combination of bead fluorescence and fluorescence of labeled cDNA produces an emission spectrum that identifies the cytokine gene that was expressed in cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
572
Q

What are 4 approaches to anti-cytokine therapy?

A

1) disrupting interaction between cytokines and their cognate receptors
2) using monoclonal antibodies that function as cytokines agonists
3) hybrid proteins containing cytokine receptor binding sites attacked to Ig constant regions to block cytokine activity
4) blocking IL-17 function

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
573
Q

What are Th2 cells involved in?

A

Antibody-mediated responses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
574
Q

What are Th1 induced by?

A

Dendritic cell production of IL-12

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
575
Q

What are Th2 induced by?

A

IL-4

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
576
Q

What are Th17 induced by?

A

IL-23

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
577
Q

What are treg cells induced by?

A

TGF-beta

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
578
Q

What are the roles of chemokines?

A

1) enhance motility and promote migration of many types of WBC toward source of the chemokines (chemotaxis)
2) modulate adhesion of WBCs to endothelial cells lining the blood vessels, facilitating extravasation into the tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
579
Q

Describe TNF-alpha

A
  • LPS is a major trigger of TNF-alpha production
  • secreted by activated monocytes and macrophages
  • cause vasodilation and increased vasopermeability
  • activates T cells by inducing expression of MHC class II molecules, vascular adhesion molecules and chemokines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
580
Q

What are the roles of complement?

A

1) causes lysis of foreign cells
2) acts as opsonins
3) increase vascular permeability
4) recruit monocytes and neutrophils to the area of antigen concentration
5) trigger secretion of immunoregulatory molecules that amplify the immune response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
581
Q

What is complement?

A

A series of more than 50 soluble and cell-bound proteins that interact to enhance host defense mechanisms against foreign cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
582
Q

What are the three pathways of complement system activation?

A

1) classical
2) alternative
3) lectin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
583
Q

Describe classical pathway

A
  • involves 9 proteins that are trigger primarily by antigen-antibody complexes
  • major role in natural defense system
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
584
Q

What classes of immunoglobulins activate the classical pathway?

A

IgM
IgG1
IgG2
IgG3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
585
Q

Describe recognition unit of the classical pathway?

A

Formation of C1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
586
Q

Describe activation unit of classical pathway

A

Once C1 is bound, C4, C2, and C3 are activated

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
587
Q

Describe membrane attacks complex (MAC)?

A
  • comprises of C5 through C9
  • completes lysis of foreign particles
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
588
Q

Describe C1 of classical pathway

A
  • consists of 3 subunits: C1q, C1r, and C1s
  • subunits require calcium to maintain structure
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
589
Q

What happens once the classical pathway is activated?

A

C1r cleaves a thioester bond on C1s, which in turn, activates it.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
590
Q

What does it mean when C1s activated?

A

Recognition stage ends

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
591
Q

What marks the beginning of the activation unit of the classical pathway?

A

Begins when C1s cleaves C4

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
592
Q

What marks the end of the activation unit in the classical pathway?

A

The production of the enzyme C3 convertase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
593
Q

What is the second most abundant complement protein?

A

C4

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
594
Q

What is the combination of C4b and C2a called?

A

C3 convertase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
595
Q

What occurs if C3 does not bind quickly?

A

Cleaved into two fragments, C3a and C3b

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
596
Q

What is the central constituent of the complement system?

A

C3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
597
Q

What is the most significant step of complement system?

A

Cleavage of C3 to C3b

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
598
Q

Describe structure of C3

A
  • consists of two polypeptide chains, alpha, beta
  • alpha chain highly reactive thioster beta
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
599
Q

What occurs after cleavage of C3 in the classical pathway?

A

About 200 molecules are split for every molecule of C4bC2a

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
600
Q

C3b also serves as a powerful _______

A

Opsonin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
601
Q

What has a specific receptors for C3b? Explain

A

Macrophages, primed to phagocytize antigen that has bound to C3b

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
602
Q

Where does C3b bind in classical pathway? What does this create?

A
  • within 40 nm of the C4bC2a
  • C4C2aC36 is created. Also called C5 convertase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
603
Q

Describe the structure of C5

A
  • contains two polypeptide chains, alpha, and beta, which are linked by disulfide bonds to form a molecule
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
604
Q

What are the two fragments is C5 split into?

A

C5a and C5b

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
605
Q

What occurs when sC5b-9 is formed in the classical pathway?

A
  • MAC produce a pole of 70 to 100A that allows ions pass in and out of the membrane
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
606
Q

What diseases are associated with abnormal C1q, C1r, and C1s?

A

SLE, recurrent infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
607
Q

What is the function of C1q?

A

Binds to Fc region of IgM and IgG, synaptic pruning

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
608
Q

What is the function of C1r?

A

Activates C1s

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
609
Q

What is the function of C1s?

A

Cleaves C4 and C2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
610
Q

What is the function of C4?

A

Part of C3 convertase (C4b)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
611
Q

What is the function of C2?

A
  • binds to C4b —> forming C3 convertase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
612
Q

What is the function of MBL?

A

Binds to mannose

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
613
Q

What is the function of MASP-1?

A

Unknown

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
614
Q

What is the function of MASP-2?

A

Cleaves C4 and C2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
615
Q

What is the function of factor B?

A

Binds to C3b to form C3 convertase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
616
Q

What is the function of factor D?

A

Cleaves factor B

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
617
Q

What is the function of properdin?

A

Stabilizes C3Bb-C3 convertase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
618
Q

What is the function of C3?

A

Key intermediate in all pathways

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
619
Q

What is the function of C5?

A

Initiates MAC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
620
Q

What is the function of C6?

A

Binds to C5b in MAC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
621
Q

What is the function of C7?

A

Binds to C5bC6 in MAC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
622
Q

What is the function of C8?

A

Starts pore formation on membrane

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
623
Q

What is the function of C9?

A

Polymerizes to cause cell lysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
624
Q

What are the steps of the classical pathway?

A

(Recognition unit)
1) the zygomens C1r and C1s are converted into active enzymes as binding of C1q occurs
2) autoactivation of C1r results from change that takes place as C1q is bound
3) when activated, C1r cleaves thioster bond on C1s, which activates it.
(Activation unit)
4) C1s cleaves C4 to create C4b and C2 to make C2a
5) C3 convertase (C4b2a) is formed
6) C3b binds to C4b2a to form C5 convertase (C4b2a3b)
(MAC)
7) C5 convertase splits C5 into C5a and C5b
8) C5b attaches to the cell membrane to initiate formation of the MAC (C5b6789)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
625
Q

How does the lectin pathway play an important role in infancy?

A

Defense mechanism during the interval between the loss of maternal antibody and the acquisition of as full-fledged antibody response to pathogens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
626
Q

What are the 3 recognition molecules of the lectin pathway?

A
  • Lectin
  • ficolins
  • collectins (CL-L1)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
627
Q

What is the key protein of the lectin pathway? Describe this protein

A
  • Mannan binding lectin (MBL)
  • binds to mannose or other related sugars in a calcium-dependent manner to initiate this pathway
  • considered acute phase protein because produced in liver and normally present in serum
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
628
Q

What are the steps of the lectin pathway ?

A

1) binds to mannose or ribose sugar to initiate pathway
2) complex associates with MASP-1, MASP-2, and MASP-3. Complex act like C1qrs
3) MASP-2 cleaves C4 and C2
4) C3 convertase (C4b2a) is formed
5) C3b binds to C4b2a to form C5 convertase (C4b2a3b)
6) C5 convertase splits C5 into C5a and C5b
7) C5b attaches to the cell membrane to initiate formation of the MAC (C5b6789)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
629
Q

What are the steps of alternative pathway?

A

1) C3 hydrolysis water to produce C3b which binds Factor B, and together they attach to target cell surface
2) B is cleaved by Factor D into the fragments Ba and Bb. Bb combines with C35 to form C3bBb,an enzyme with C3 convertase activity
3) more C3 is cleared, forming more C3bBb. This enzyme is stabilized by properdin and, it continues to cleave additional C3
4) it a molecule of C3 remains attached to the C3bBbP enzyme, the convertase now has the capability to cleave C5. The C5 convertase now has the capability to cleave C5. The C5 convertase this consists of C3bBbP3b
5) C5b attaches to the cell membrane to initiate formation of MAC (C5b6789)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
630
Q

What is the role of the alternative pathway?

A

Functions largely as an amplification loop for activation started for activation started from the classical or lectin pathways

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
631
Q

What are some triggering substances for the alternative pathway?

A
  • bacterial cell walls
  • some parasites
  • fungal cell call
  • viruses/ viral infected cells
  • tumor cell line
  • yeasts
  • these all serve as binding site complex C3bBb
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
632
Q

What proteins are involved in the alternative pathway?

A
  • C3
  • C5-C9
  • factor B
  • factor D
  • P (properdin)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
633
Q

Describe the alternative pathway

A

Acts as a natural defense system

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
634
Q

What protein is not stable in plasma in the alternative pathway?

A

C3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
635
Q

What has the ability to bind to factor B?

A
  • iC3 (C3b)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
636
Q

What happens when C3b binds to factor B in the alternative pathway?

A
  • factor D with magnesium can cleave B into Ba and Bb
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
637
Q

Describe Factor D

A
  • plasma protein that circulates in active enzyme form
  • serine protease
  • only substrate is bound to factor B
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
638
Q

What occurs once B is cleaved into Ba and Bb in the alternative pathway?

A

1) Ba goes off into plasma
2) Bb binds to the surface of the triggering cellular antigen
3) if it does not bind quickly, it disintegrates.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
639
Q

What does C1 inhibitor (C1-INH) inhibit?

A
  • activation of the first stages of the classical and lectin pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
640
Q

What is MCP also known as?

A

CD46

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
641
Q

What is DAF is also known as?

A

CD55

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
642
Q

What is Factor I?

A
  • serine protease that inactivates C3b and C4b when bound to one of these regulators
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
643
Q

What is immune adherence?

A

The ability of cells to bind complement-coated particles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
644
Q

Where are MCPs found?

A
  • found on the cell membrane of all epithelial and endothelial cells except erythrocytes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
645
Q

What is the most efficient cofactor for factor I?

A

MCP

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
646
Q

Describe DAF

A
  • a membrane glycoprotein that has wide tissue distribution
  • found on peripheral blood cells, endothelial cells, fibroblasts, and numerous types of epithelial cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
647
Q

What does the presence of DAF on host cells protect them from?

A

Bystander lysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
648
Q

What is bystander lysis?

A
  • main mechanism used in discrimination of self from nonself because foreign cells do not posses this substance
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
649
Q

What is the principle soluble regulator of alternative pathway?

A

Factor H

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
650
Q

What is S protein?

A
  • soluble control of protein that acts at a deeper level of complement activation
  • terminal pathway
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
651
Q

What does Factor H do in the alternative pathway?

A
  • acts by binding to C3b to prevent binding of Factor B
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
652
Q

What is the main function of C1-INH?

A

Dissociates C1s and C1r from C1q

653
Q

What is the main function of Factor I?

A

Cleaves C3b and C4b

654
Q

What pathways does C1-INH participate in?

A

Classical
Lectin

655
Q

What pathways does factor I participate in?

A

Classical
Lectin

656
Q

What are the main functions of Factor H?

A

1) cofactor with factor I to inactivate C3b
2) prevents binding of B to C3b

657
Q

What pathway does Factor H participate in?

A

Alternative

658
Q

What is the main function of C4BP?

A

Acts as a factor with factor I to inactivate C4b

659
Q

What pathways does C4BP participate in?

A

Classical
Lectin

660
Q

what is the main function of S protein?

A
  • prevents attachment of C5b67 complex to cell membranes
661
Q

What pathways does S protein participate in?

A

Classical
Alternative
Lectin

662
Q

What is the main function of MIRL?

A

Prevent insertion of C9 into cell membrane

663
Q

What is the main function of MCP?

A

Cofactor for factor I cleavage of C3b and C4b

664
Q

What is the main function of CR1?

A

Cofactor for factor I, mediates transport of immune complexes

665
Q

What is the main function of CR2?

A
  • B cell co-receptor for antigen with CD19
666
Q

What is the main function of CR3?

A

Adhesion and increased activity of phagocytic cells

667
Q

What is the main function of CR4?

A

Adhesion and increased activity of phagocytic cells

668
Q

What are 4 biological manifestations of complement?

A
  • amplifies inflammatory response
  • role in uptake and presentation of antigens in adaptive immune response
  • facilitates B cell activation
  • role in neural development
669
Q

What is present when the inflammatory response is amplified by complement?

A

1) anaphylatoxins (C5a, C3a) - increase vascular permeability
2) chemotaxins (C5a) - attracts phagocytic cells to enhance phagocytosis
3) opsonins (C3b, C4b, iC3b, C3dg) - coat damaged or foreign cells to enhance phagocytosis

670
Q

How can complement be harmful?

A

1) it is activated systemically on a large scale, as in gram-negative septicemia
2) it is activated by tissue necrosis and results in obstruction of the blood supply, as in myocardial infarction
3) lysis of RBCs occurs as in cold agglutinin disease or autoimmune hemolytic anemia

671
Q

Describe C3 glomerulopathy

A

Inflammation of the Renal glomeruli tat maybe due to autoantibodies that cause C’ dysregulation (C3 nephrotic factors; C3NeFs

672
Q

When detecting complement abnormalities, what are the immunologic assays of individual components?

A
  • automated nephelometry or immunoturbidity
  • radial immunodiffusion (RID)
673
Q

What are classical pathway assays?

A

1) hemolytic titration (CH50) assay
2) CH50 test for lysis of liposomes
3) ELISA to detect C9 epitopes exposed after binding in MAC

674
Q

What are the alternative pathway assays?

A

1) AH50
2) ELISA to detect C3bBbP or C3bP

675
Q

What is the CH50 test procedure?

A

1) serum to be tested is diluted serially and added to sensitized sheep RBC
2) tubes are incubated at 37 C
3) centrifuged to pellet the unlysed cells

676
Q

What is CH50 defined as?

A

As reciprocal of the dilution that causes lysis of 50% of the cells used in the assay

677
Q

Describe the complement fixation test

A
  • destroy any complement present in patient serum by heating serum to 56 C for 30 minutes
  • dilutions of serum are combined with known antigen and a measurement amount of Guinea pig complement
  • if patient antibody is present it will bind with the reagent antigen and complement be bound `
678
Q

What are the steps of complement fixation test?

A

1) add sheep RBCs that are coated with a hemolysin (will lyse if any complement is present)
2) incubate and centrifuge tubes
3) read for hemolysis
4) it hemolysis is present —> no patient antibody/negative results
If hemolysis is absent —> patient antibody to complement/positive results

679
Q

What are limitations of complement fixation test?

A

1) results are expressed as highest dilution showing no hemolysis
2) controls must be used
3) more sensitive than agglutination and precipitation, less sensitive than labeled immunoassay
4) errors

680
Q

What pathways are affected by abnormal C1q, C1r, and C1s?

A

Classical —> low

681
Q

What pathways are affected by abnormal C4 and C2?

A

Classical —> low
Lectin —> low

682
Q

What pathways are affected by abnormal MBL and MASP-2?

A

Lectin —> low

683
Q

What pathways are affected by abnormal B, D, and P?

A

Alternative —> low

684
Q

What pathways are affected b abnormal C3, and C5-C9?

A

All three —> low

685
Q

What pathways are affected by abnormal C1-INH?

A

Classical —> low
Lectin —> low

686
Q

What pathways are affected by abnormal Factor H and I?

A

Al three —> low

687
Q

What are levels of C3 and C4 routinely measured by?

A
  • automated nephlometry or immunoturbidimetry
688
Q

Describe nephlometry?

A

Measures the concentration of an individual complement protein according to the amount of light scattered by the antigen-antibody mixture

689
Q

Describe immunoturbimetry

A

Based on reduction in light transmission resulting from immune complex formation

690
Q

What is the difference between the CH50 and AH50 assays?

A
  • magnesium chloride is added to buffer
  • ethylene glycol tetraaactetic acid added to buffer
  • calcium left out
691
Q

Describe the AH50 assay

A
  • buffer chelates calcium, blocking classical pathway activation
  • rabbit RBC are used as indicator
692
Q

Describe ELISA testing in alternative pathway

A
  • can detect C3bBb or C3bP complexes in very small quantities
  • micrometer wells typically coated with bacterial lipopolyssachardes to trigger activation of the alternative pathways
693
Q

What are the assays that test for complement activation?

A
  • ELISA or multiplex versions of immunoassays
694
Q

Th cells affect the in innate immune response by inducing production of which cytokines?

A

IL-2
IL-4

695
Q

Describe macrophage-colony stimulating factor (M-CSF)

A
  • if activated, the cells become macrophages
  • increase phagocytosis, chemotaxis, and additional production in monocytes and macrophages
696
Q

What is autocrine stimulation?

A

Affecting the same cell that secreted it

697
Q

Describe colony stimulation factor?

A

Stimulates formation of colonies of calls in the bone marrow

698
Q

What is integrin?

A

Cell adhesion molecules on leukocytes

699
Q

Describe erythropoietin (EPO)

A

CSF that regulates RBC production in bone marrow

700
Q

Describe granulocytes colony stimulating factor (G-CSF)

A
  • if activated, the cells become neutrophils band affects survival, proliferation, and differentiation of all cells types in the neutrophil lineage
701
Q

Describe endocrine activity

A

System in affects

702
Q

Describe adaptive T regulatory 1 cells

A
  • CD4+ T cells tha are induced from antigen activated naive T cells in presence of IL-10
703
Q

Describe granulocyte-macrophages colony stimulating factor (GM-CSF)

A
  • cytokines produced by T cells and other cell lines that stimulates an increased supply of granulocytic cells and macrophages
704
Q

What is the main difference between IgE ad IgG?

A

IgE has one more constant region

705
Q

What is the best assay to measure a specific cytokine?

A

ELISA assay

706
Q

What is precipitation?

A

Combine soluble antigen and stable antibody to produce insoluble complexes

707
Q

What is agglutination?

A

Antigen-antibody complexes cross-link, causing clumping

708
Q

What is affinity?

A

Initial forces of attraction that exists between a single Fab site on antibody and single epitope or determinant site on corresponding antigen

709
Q

What is cross-reactivity?

A
  • the more the cross-reacting antigen resembles the original antigen, the stronger the bond will between the antigen and binding sites
  • cross-reacting antigens have lower affinity
710
Q

What is serology?

A

Study of fluid components in the blood, especially antibodies

711
Q

describe serum

A
  • liquid portion of blood minus coagulation factors
  • most frequently encounter specimen in immunologic testing
  • can be separated from other components of blood via centrifuge
712
Q

What color tube tops are serum collected in?

A

Red top or tiger top

713
Q

What specimens would be rejected?

A
  • hemolyzed
  • clotted specimen
  • wrong tube or container
  • tube not labeled
  • inadequate quantity
  • time mislabeled
714
Q

What are the 3 phases of laboratory testing?

A
  • preanalytical
  • analytical
  • postanalytical
715
Q

What are the examples of preanalytical testing?

A
  • Specimen collection
  • transport
  • processing
716
Q

What are examples of analytical testing?

A

Testing

717
Q

What are some examples of postanalytical phase?

A
  • testing results
  • transmission
  • interpretation
  • follow up
  • retesting
718
Q

What phases of the laboratory testing constitutes for 90% of the errors that occur?

A
  • pre-analytical
  • post-analytical
719
Q

What are the 3 phases of antigen/antibody complexes?

A

1) primary phenomenon
2) secondary phenomenon
3) tertiary phenomenon

720
Q

Describe the primary phenomenon phase of complexes

A
  • combination of binding site on antibody with a single epitope on antigen
  • reversible: occurs in milk-seconds
  • not easily detectable-hard to measure
721
Q

Describe the secondary phenomenon phase of antigen/antibody complexes

A
  • precipitation, agglutination, and complement fixation
  • easily detectable
  • most serological tests based
722
Q

Describe the tertiary phenomenon phase of antigen/antibody complexes

A
  • inflammation, phagocytosis, depression of immune complexes, immune adherence, chemotaxis (all in vivo)
723
Q

What is complement fixation?

A

Triggers the classical complement pathway to detect antibody or antigen in the patients serum

724
Q

what is avidity?

A
  • is the sum of the attractive forces between forces between an antigen and an antibody
  • strength with which a multivalent antibody binds a multivalent antigen
725
Q

What does avidity measure?

A

Overall stability of an antigen-antibody complex

726
Q

The more bonds that form between antigen and antibody, the higher the ______

A

Avidity

727
Q

What is the law of mass action?

A

Free reactants are in equilibrium with bound reactants

728
Q

What is the law of mass action equation?

A

K=(Ag/Ab)/(Ab x Ag)

729
Q

In the law of mass action, what does the K value depend on?

A

The strength of binding between antibody and antigen

730
Q

The higher the K value…..

A
  • the larger the amount of antigen-antibody complexes
  • the more visible or easily detectable the reaction
731
Q

In the law of mass action equation, what does the [Ag/Ab] represent?

A

Concentration of the the antigen-antibody complex (mol/L)

732
Q

In the law of mass action equation, what does [Ab] represent?

A

Concentration of free antibody (mol/L)

733
Q

In the law of mass action equation, what does [Ag] represent?

A

Concentration of free antigen (mol/L)

734
Q

What does a precipitation require antigens and antibodies to have?

A
  • multiple binding sites for one another
  • equivalent concentration
735
Q

What are the 3 groups of the precipitation curve?

A
  • zone of equivalence
  • prozone
  • postpone
736
Q

For optimal precipitation, reactions must be run in what phase of the precipitation curve?

A

Zone of equivalence

737
Q

Describe the zone of equivalence of the precipitation curve

A
  • number of multi-valent sites of antigen and antibody are approximately equal
738
Q

Describe the prozone of the precipitation curve?

A
  • antibody excess
  • antigen combine with one or two antibodies
  • no cross-linking
739
Q

What could cause a false negative result in the prozone phase of the precipitation curve?

A

High antibody concentration

740
Q

Describe the postzone phase of the precipitation curve

A
  • antigen excess
  • small aggregates are surrounded by excess antigen
  • no lattice network formed
741
Q

What could cause a false negative result in the postzone phase of precipitation curve?

A

Presence of small amount of antibody

742
Q

What are immunoturbidimetry and nephelometry used to test for?

A
  • complement
  • C-reactive protein
  • immunoglobulins
  • other serum proteins
743
Q

What is the principle of immunoturbidity and nephelometry?

A

Light that is scattered at an angle is measured, indications the amount of antigen or antibody present

744
Q

What is the radial immunodiffusion used to test for?

A
  • complement
  • immunoglobulins
745
Q

What is the principe of radial immunodiffusion?

A

Antigen diffuses out into gel that is infused with antibody. Measurement of the radius indicates concentration of antigen

746
Q

What is Ouchterlony double diffusion used to test for ?

A

Complex antigens, such as fungal antigens

747
Q

What is the principle of Ouchterlony double diffusion?

A

Both antigen and antibody diffuse out from wells in a gel. The lines of precipitate formed indicate he relationship of antigens

748
Q

What does immunoelectrophoresis test for?

A

Differentiation of serum proteins

749
Q

What is the principle of immunoelectrophoresis?

A

Electrophoresis of serum is followed by diffusion of antibody from the wells

750
Q

What is the immunofixation electrophoresis used to test for?

A

Over or under production of antibody

751
Q

What is the principle of immunofixation electrophoresis?q

A
  • electrophoresis of serum is followed by direct application of antibody to the gel
752
Q

What machines could be used for immunoturbidity?

A
  • spectrophotometer
  • automated clinical chemistry analyzer
753
Q

What machines is used for nephelometry?

A

Nephelotometer

754
Q

Describe nephelotometer

A
  • contains photodector located between 30-90 degrees from light source
  • amount of light scatter increases as the number of immune complexes increases and is in index of the concentration of antigen or antibody in solution
755
Q

What is passive immunodiffusion?

A

When no electric current is used to speed up the migration of the reactants

756
Q

What can affect the rate of diffusion in passive immunodiffusion?

A
  • size of particles
  • temperature
  • gel viscosity
757
Q

What is the end point (Mancini) method?

A

Reaction goes to completion

758
Q

What is the kinetic (Fahey) method?

A
  • faster
  • measurements taken before reaction is complete
759
Q

What is the RID end point method results?

A

The square of the diameter is proportional to the antigen concentration

760
Q

What are the three possible patterns of Ouchterlony diffusion?

A
  • identity (arc)
  • partial identity (one line extends)
  • nonidentity (line cross X)
761
Q

What is visualized when immunofixation electrophoresis (IFE) is performed?

A

Increased or decreased production of antibody classes and to differentiate monoclonal and polyclonal immunoglobulins

762
Q

What are agglutinins?

A

Produced by antibodies

763
Q

What are the 2 steps of agglutination?

A

1) sensitization
2) lattice formation

764
Q

Describe sensitization step of agglutination

A
  • initial bonding
  • antigen and antibody unite through bonding of antigenic determinants sites
  • fast and reversible
765
Q

Describe the lattice formation of agglutination

A
  • creation of large aggregates
  • visible aggregates form as multiple antigen and antibody molecules bind to create a stable lattice
766
Q

What is Coombs reagent?

A
  • anti-human immunoglobulin
  • frequently used in blood bank
  • binds to Fc portion of IgG
767
Q

Briefly describe immunoturbidity

A
  • automated methods
  • measures reduction in light intensity
768
Q

Briefly describe nephelometry

A
  • automated methods
  • measures light scatter as immune complexes form
769
Q

What are 4 types of agglutination reactions?

A

1) direct agglutination
2) passive agglutination
3) reverse passive agglutination
4) agglutination inhibition

770
Q

Describe direct agglutination

A
  • Uses particles with naturally occurring antigens to test for antibodies in patient serum
  • detects bacteria (widal test) and RBCs
771
Q

Describe Widal test

A
  • a rapid screening test used to determine the possibility of typhoid fever
  • uses salmonella O and H antigens
  • direct agglutination test
772
Q

What test are ran fro RBCs?

A
  • hemagglutination
  • ABO typing
    (Direct agglutination)
773
Q

Describe passive agglutination

A
  • uses particles that are coated with antigens not normally found on their surfacesL erythrocytes, gelatin, latex, and silicates
774
Q

What does passive agglutination test for ?

A
  • RA
  • antibodies to Group A streptococcus antigens
  • antibodies to viruses such as rubella
  • heterophile antibody in IM
775
Q

Describe reverse passive agglutination process

A

1) antibody is attached to the carrier particle
2) agglutination occurs is antigens present in the patient sample

776
Q

What can reverse passive agglutination test for?

A

-Staphylococcus aureus
- streptococcus Group A or B.
- rotavirus
- cryptococus neoformans

777
Q

Describe agglutination inhibition

A

-based on competition between particle and soluble antigens for limited antibody-combining sites
- used to detect hapten antigens (illicit drugs)
- lack of agglutination —> positive reaction

778
Q

What is an agglutination inhibition procedure?

A
  • hemagglutination inhibition —> uses RBCs
  • used to detect antibodies to certain viruses that can bind to RBCs and agglutinate them
779
Q

What virus can hemagglutination inhibition be used to test for?

A
  • rubella
  • influenza
  • respiratory syncytial virus
780
Q

What is the principle of direct agglutination?

A

Antigen is naturally found on a particle

781
Q

Describe results of direct agglutination

A

Agglutination indicates presence of patient antibody

782
Q

What is the principle of passive (indirect) agglutination?

A

Particle are coated with antigens not normally found on their surfaces

783
Q

Describe the results of passive (indirect) agglutination

A

Agglutination indicates the presence of patient antibody

784
Q

What is the principle of reverse passive agglutination?

A

Particles are coated with reagent antibody

785
Q

Describe results of reverse passive agglutination

A

Agglutination indicates the presence of patient antigen

786
Q

What is the principle of agglutination inhibition?

A
  • haptens are attached to carrier particles
  • particles compete with patient antigens for a limited number of antibody sites
787
Q

Describe results of agglutination inhibition

A

Lack of agglutination is a positive test, indication the presence of patient antigen

788
Q

What is the principle of hemagglutination inhibiton?

A

RBCs spontaneously agglutinate in presence of certain viruses

789
Q

Describe the results of hemagglutination inhibiton

A

Lack of agglutination is a positive test indication the presence of patient antibody

790
Q

Describe monospot tests

A

Used to detect the heterophile antibody characteristics of IM by its ability to agglutinate horse or bovine RBCs

791
Q

Describe cold agglutinins

A
  • produced by patients with certain autoimmune disorders, malignancies, or infections, are antibodies that agglutinate human type O RBCs when incubated at cold temperatures
792
Q

Describe PETINA

A
  • “particle-enhanced turbidmetric inhibiton immunoassay”
  • patient sample is incubated with latex beads coated with analytes will prevent antibody turbidity results
  • measures small analytes such as therapeutic drugs
  • homogenous competitive immunoassay
793
Q

Describe the results of PETINA

A
  • if low amount of analyte is present in the sample, the reagent antibody will bind to the latex beads, resulting in aggregate formation and high level of turbidity
  • when concentration of analytes high, analyte will bind to the reagent antibody and prevent it from binding to the latex beads, resulting is less turbidity
794
Q

How ca most cross-reactivity be avoided?

A

Through use of monoclonal antibody directed against an antigen determinant that is unique to antigen

795
Q

What are advantages of agglutination reactions?

A
  • RA did
  • easy to perform
  • no expensive equipment
  • relatively sensitive
796
Q

When does maximum binding at antigen and antibody occur?

A

When the aggregate number of multivalent sites of antigen and antibody are approximately equal

797
Q

How does precipitation differ from agglutination?

A

Precipitation involves a soluble antigen, whereas agglutination involves a particulate antigen

798
Q

Describe labeled immunoassays

A
  • designed for antigen and antibodies that may be small in size or present in very low concentrations
  • use a reactant labeled with a detection molecules to monitor the amount of specific binding that has take place
799
Q

What is an analyte?

A

A biomarker

800
Q

How are labeled immunoassays differ from unlabeled techniques?

A

A detection molecule (label) in the test system

801
Q

What is a common technique to many labeled immunoassay?

A

Enzyme-mediated catalysis of a reagent substrate to generate a light signal

802
Q

describe heterogenous immunoassays

A
  • involve physical separation of bound and free components
  • most commonly involve binding to a solid phase
  • magnetic separation or centrifugation maybe used
803
Q

Describe homogenous immunoassays

A

Do no require a physical separation step

804
Q

Describe competitive immunoassays

A
  • all reactants are mixed together simultaneously
  • labeled and unlabeled antigen compete for a limited number of binding sites on reagent antibody
  • have high specificity
805
Q

What does competitive immunoassays measure?

A

Small antigens that are relatively pure (drugs and hormones)

806
Q

What are the steps of competitive immunoassay procedure?

A

1) unknown concentration of analyte in patient sample competes with labeled analyte for binding sites on immobilized antibody
2) wash to remove unbound material
3) after substrate is added, a colored product (signal) is generated with an intensity proportional to the amount of enzyme-labeled analyte bound to the antibody

807
Q

What is the principle of noncompetitive immunoassays

A
  • patents analyte is allowed to bind with an excess amount of labeled reagents
  • Also known as capture, sandwich or immunogenic assays
808
Q

What are the steps of noncompetitive immunoassay procedure?

A

1) reagent antigen immobilized on solid phase binds antibody in patient sample
2) wash to remove unbound materials
3) add enzyme-labeled (detection) antibody that binds to human immunoglobulin
4) wash to remove unbound materials
5) add substrate and measure signal. Signal is directly proportional to concentration of antibody in patient sample

809
Q

Describe radioimmunoassay (RIA)

A
  • first immunoassay developed
  • uses radioactive labels - 125I is most popular
  • emits gamma radiation, which is detected by a gamma counter
  • is extremely sensitive and precise
  • measures trace amounts of analyte ( hormones, serum protein, and drugs) that are small in size
810
Q

What are disadvantages of radioimmunoassay?

A

1) working with radioactive substance (hazardous)
2) disposal of low level radioactive waste
3) short shelf for some reagents
4) testing in clinical labs is limited

811
Q

Describe enzyme immunoassays (EIAs)

A
  • Highly sensitive assay that uses enzymes as labels, which react with suitable substates to produce breakdown products that maybe chromogenic, fluorogenic, or luminescent
  • available in competitive and noncompetitive formats
812
Q

Briefly describe heterogenous EIAs

A

Requires a step to physically separate free analyte from bound analyte

813
Q

Describe competitive heterogenous EIAs

A
  • involve enzyme-labeled antigens competing with unlabeled patient antigen for binding sites or antibody molecules
  • have high specificity
814
Q

What is competitive heterogenous EIAs used for?

A

For measuring small antigens that are relatively pure (drugs and hormones)

815
Q

What are advantages of noncompetitive heterogenous EIAs

A
  • high sensitivity
  • high specificity
  • simple to perform
  • low cost
816
Q

Describe ELISA

A
  • “enzyme linked immunosorbent assay”
  • used to measure antibody production to infectious agents that are difficult to isolate and form autoantibody testing
  • easily applied to point of care and home testing
  • detects HIV, Hep B, and Hep C, rubella virus
817
Q

What is the principle of indirect ELISA?

A

Primary antibody specific for the antigen binds to the target, and a labeled secondary antibody against the host species of the primary antibody binds to the primary antibody

818
Q

What are the immuoassays that detect antibodies?

A

Indirect ELISA

819
Q

What are immunoassays that detect antigens?

A

Capture immunoassays

820
Q

Why is it referred to as “indirect” ELISA?

A

Because the enzyme-labeled reagent does not participate in the initial antigen-antibody reaction

821
Q

Describe capture immunoassays

A
  • EIAs that detect antigen in patient sample
  • best suited for antigens that have multiple determinants (cytokines, proteins, tumor marker, microbial antigens)
  • can be used to detect immunoglobulins of a particular class
822
Q

What is the principle of capture immunoassay?

A

Capture the antibody to an ELISA plate and allow the analyte of interest to bind to the capture antibody, then secondary antibody is added and binded

823
Q

Describe the high dose hook effect

A
  • antigen interference
  • excess patient antigen causes falsely decreased detection
  • analyte concentration appears to below or normal when it is actually high
  • also called Postpone effect
824
Q

When does the high dose hook effect occur?

A

When there are not enough capture antibody sites for antigen binding because the majority of binding sites are filled, so the remainder of patient antigen has no place to bind and gets removed during the wash step1

825
Q

What are some interferences of antibodies?

A
  • autoantibodies —> rheumatoid factor can cause a false positive, produced in vivo
  • heterophile antibodies —> usually cause false-negative results. Example: human and mouse antibodies (HAMA)
826
Q

Describe homogenous EIAs

A
  • less sensitive than heterogenous assays
  • rapid and simple to perform
  • include EMIT and CEDIA
  • does not require washing or separation step
  • when antibody binds to specific determinant sites on the antigen, the active site on the enzyme is blocked, resulting in a measurable loss of activity
827
Q

What are homogenous EIAs used for?q

A

To determine low-molecular weight analytes in serum and urine, hormones, therapeutic drugs, and drugs of abuse q

828
Q

Describe enzyme multiplied immunoassay technique (EMIT)

A
  • original homogenous design
  • enzyme active site is blocked when antibody binds to enzyme
  • does not allow substrate to react with enzyme
  • based on the principle of change in enzyme activity as specific antigen-antibody interaction occurs in solution
829
Q

What are the steps of EMIT?

A

1) reagent antigen bound to an an enzyme tag commonly G6PDH
2) when reagent antibody binds to specific sites on the enzyme-antigen pair, the active site on the enzyme is blocked. Results in loss of activity

830
Q

Describe cloned-enzyme donor immunoassay (CEDIA)

A
  • produce the beta-galatosidase enzyme in two parts: acceptor and donor
  • when the acceptor-donor pair combine, enzymatic activity is restored; and reagent substrate is catalyzed to generate a product signal that is measured by photometry
831
Q

Describe chemiluminescent immunoassays

A
  • highly sensitive
  • heterogenous or homogenous
  • automated immunoassays
  • detects antibodies or antigens
  • the emission of light cause by a chemical reaction, typically an oxidation reaction, producing an excited molecule that decays back to its original ground state
832
Q

Describe fluorescent immunoassays

A
  • uses Fluor chrome as the label
  • these compounds absorb energy from incident light and convert to to a longer wavelength electrons return to the ground state
833
Q

Describe fluorescein

A

Absorbs light at 490-495 nm and emits green light at 520 nm

834
Q

Describe rhodamine

A

Absorbs light at 550 nm and emits red lights at 585 nm

835
Q

Describe fluorochromes

A

Fluorescent compound that can absorb energy from an incident source and convert to that energy into light of a longer wavelength and lower energy as excited electrons return to ground state

836
Q

What is the direct immunofluorescence assay procedure?

A

1) antibody with a fluorescent tag is added to tissue or cells fixed onto a slide
2) after incubation and a wash step, the slide is read using a fluorescence microscopic

837
Q

Describe direct immunofluorescence assays

A
  • used to identify pathogens in patient samples
  • fluorescent-labeled antibodies bind to CD antigens
  • can be used to identify lymphocytes and other cells by flow cytometry
  • used for cells or tissues
838
Q

What is the principle of indirect immunofluorescence assay?

A

1) patient serum is incubated with microscopic slide to which a known antigen is attached
2) the slide is washed, then an anti-human immunoglobulin labeled with a fluorescent tag is added
3) A sandwich is formed with the first antibody, which localizes the fluorescence
* can be used to identify patient antibody (ANAs)

839
Q

What is indirect immunofluorescence assay used to detect?

A
  • antibody ID
  • syphilis testing (treponemal antibodies, viral antibodies and autoantibodies)
840
Q

describe multiplex immunoassay (MIA)

A
  • fluorescent immunoassay that allows multiple antibodies or antigens to be detected simutaneously
841
Q

What is the principle of multiplex immunoassay (MIA)?

A

1) to detect antibodies, patient serum is incubated with polystyrene beads conjugated to different antigens
2) antibody binding is detected by addition of a fluorescent- tagged anti-human immunoglobulin
3) beads containing bound antibody are identified by flow cytometry and can be distinguished by their unique shade of red

842
Q

What is a benefit of MIA?

A

Allows for multiple antibodies to be detected simutaneously

843
Q

How can MIA be used?

A
  • ANA testing
  • Detection of antibodies in transplant patients donor antigens
844
Q

Describe fluorescent polarization immunoassay (FPIA)?

A
  • determines concentrations of therapeutic drugs and hormones
  • based on charge in polarization of fluorescent light emitted from a labeled molecule when it is bound by antibody
  • If the labeled molecule is bound to antibody, the molecule emits increased amount of polarized light
  • homogenous
845
Q

Describe is the rapid immunoassays procedure?

A

1) patient sample is added to the test membrane and combines with labeled antigen or antibody conjugated to colored latex or colloidal gold particles
2)immune complexes are formed that migrate across the membrane to produce a colored reaction

846
Q

Describe immunochormatography

A

Combine any methods of immunoassays into one step

847
Q

How do heterogenous immunoassays differ from homogenous immunoassays?

A

Heterogenous immunoassays require a separation step

848
Q

What is the principle of FPIA?

A

Fluorescent-labeled antigen competes with patient antigen for a limited number of soluble antibody-binding sites

849
Q

What do nucleic acids do?

A

The carry genetic information codes for protein structure

850
Q

What are some primary characteristics of nucleic acids?

A
  • Nucleic acid complementary
  • melting temperature
851
Q

What is an advantage to molecular testing?

A

Advantageous because nucleic acids can be detected earlier than antibodies during the course of an illness

852
Q

Describe DNA

A
  • Nucleic acid that carries the primary genetic information within chromosomes
  • contains sugar deoxyribose
853
Q

Way are the purine bases of DNA?

A

Adenine
Guanine

854
Q

What are the pyrimidine bases of DNA?

A

Cytosine
Thymine

855
Q

Describe RNA

A
  • helps convert the genetic information encoded within DNA into proteins
  • contains sugar ribose
856
Q

What are the purine bases of RNA?

A

Adenine
Guanine

857
Q

What are the pyrimidine bases of RNA?

A

Cytosine
Uracil

858
Q

What are the types of RNA?

A
  • messenger RNA (mRNA)
  • transfer RNA (tRNA)
  • ribosomal RNA (rRNA)
  • noncoding RNAs
859
Q

What are nucleotide composed of?

A
  • deoxyribose OR ribose sugar
  • nitrogen base
  • a phosphate group
860
Q

Describe nucleotides

A

Units that make up DNA and RNA

861
Q

What are the two strands of the DNA double helix are held together by?

A

Hydrogen bonds between their nucleotide bases

862
Q

What is complementary to guanine in DNA?

A

Cytosine

863
Q

What is adenine complementary to adenine in DNA?

A

Thymine

864
Q

How many hydrogen binds are between guanine and cytosine?

A

3

865
Q

How many hydrogen bonds are between adenine and thymine?

A

2

866
Q

What is the length of double stranded DNA macromolecule measured in?

A

Base pairs

867
Q

What is the length of a single stranded RNA measured in?

A

Bases

868
Q

What is a megabase pair/megabase?

A

When there is one million base pairs/bases

869
Q

What is chromosome?

A

Double helix of DNA

870
Q

How many chromosomes are in a cells nucleus?

A
  • 46
  • two copies each of the 22 autosomes or non-sex chromosomes
  • plus two copies of X chromosomes in females OR one X chromosome and Y chromosome for males
871
Q

What are genes?

A

Sequences of nucleotides in chromosomes that carry information for either a protein or non coding genes making up the diploid genome

872
Q

What is genome?

A

Entirety of DNA in the cell

873
Q

Describe DNA replication

A
  • two DNA strands separate
  • each strand is a template for synthesis of a complementary strand
  • DNA polymerase is needed for synthesis
  • each new strand is synthesized in the 5’ to 3’ direction as the original double strand unwinds
  • two identical double-stranded molecules results
874
Q

What is the “origin of replication” for DNA?

A

Defined sequence of nucleotides that start DNA replication

875
Q

What is the cell cycle?

A

While cells divide, they undergo a series of events in which increase in size and duplicate their DNA

876
Q

What are the four stages of the cell cycle?

A

G1
S
G2
M

877
Q

Describe S phase of the cell cycle

A

DNA replication occur here

878
Q

Describe the G2 phase of the cell cycles

A

DNA complement of the cell are doubled

879
Q

Describe the M phase of the cell cycle

A

One complement of chromosomes is divided into each of two daughter cells

880
Q

Describe the G1 phase of the cell cycle

A
  • each of the new daughter cells are here
  • movement from G1 to the S phase is strictly regulated
881
Q

What is lagging strand?

A

Copied discontinuously toward the replication fork

882
Q

Describe RNA synthesis

A
  • can start de novo, without primer
  • is catalyzed by RNA polymerase
  • occurs throughout the cell cycle
  • mRNA codes for the amino acid sequence of a protein
883
Q

Describe the protein synthesis and translation steps

A

1) mRNA is transcribed from DNA using RNA polymerase
2) the mRNA delivers the information to ribosomes, where protein synthesis takes place
3) as each amino acid is added, the peptide chain continues to grow
4) this translation process is accomplished with help of tRNA, which brings in individual amino acids

884
Q

What is a phenotype?

A

Observable properties of an organism (blue eyes, height)

885
Q

What is a codon?

A

3 base recognition sequence

886
Q

Describe mutations and variants

A
  • changes in the nucleotide sequence of DNA
  • some are associated with disease
887
Q

What is the difference a mutation and variant?

A
  • variants are inherited
  • mutations are spontaneous changes in somatic cells
888
Q

Describe polymorphisms

A
  • alterations in DNA or protein sequences shared by at least 2% of population
  • MHC is highly polymorphic region in the human genome
889
Q

Describe electrophoresis

A
  • involves movement of particles under the force or an electrical current
  • electrophoresis of nucleic acids commonly takes place in a semisolid matrix called a gel
  • nucleic acids are negatively charged and migrate toward the positive pole (anode)
  • smaller nucleic acid chains migrate faster than large chains
890
Q

Describe capillary electrophoresis

A
  • a more sensitive, semi-automated method that separates particles in a gas, liquid or gel
  • DNA chains carry a fluorescent label that is excited by a laser to produce peaks of fluorescence
891
Q

Describe molecular analysis

A

Nucleic acid test designed to detect changes (mutations and polymorphism) in the DNA sequence

892
Q

What are the four main approaches to nucleic acid analysis?

A

1) strand cleavage methods
2) hybridization methods
3) amplification methods
4) sequencing methods

893
Q

What do the strand cleavage methods use to cleave DNA at specific locations?

A

Restriction endonuclease enzymes

894
Q

Describe the strand cleavage methods of molecular analysis

A
  • separate cleaved fragments by size and charge using electrophoresis, revealing potential variations
  • used to investigate small genomes, such as those of microorganisms or plasmids
  • detect mutations and polymorphisms
895
Q

Describe CRISPR-Cas9

A
  • newer method that uses an enzyme to alter DNA at used-defined locations identified by a small guide RNA molecule
  • used for DNA analysis, gene therapy, and genome editing
  • a strand cleavage method
896
Q

Describe the hybridization methods of molecular analysis

A
  • involves the binding two complements nucleic acids, most notably a template and probe
  • used on large, complex genomes
897
Q

What are some examples of hybridization methods of molecular analysis?

A
  • Souther blot
  • microarray
  • bead array
  • in situ hybridization
898
Q

What are probes?

A

Short nucleic acids that bind to complementary sequences

899
Q

What is hybridization?

A

Involves binding of two complementary strands of nucleic acids

900
Q

Describe western blot

A
  • detection of proteins and protein modifications
  • separated by gel electrophoresis and blotted to membrane
901
Q

Describe microarrays

A
  • allow for multiple targets or sample to be analyzed simultaneously
  • the test sample is labeled with a fluorescent dye and added to a glass slide containing thousands of specific unlabeled probes
  • fluorescent spots appear where complementary binding occurs
902
Q

What are types of microarrays?

A
  • comparative genomic array
  • RNA expression arrays
  • SNP arrays
903
Q

Describe comparative genomic arrays

A

Detect amplifications or deletion in DNA

904
Q

Describe RNA expression arrays

A
  • detect that have been actively transcribed into mRNA
905
Q

Describe RNA expression arrays

A
  • detect genes that have been actively transcribed into mRNA
906
Q

Describe SNP arrays

A

Detect single nucleotide differences in test samples, some of which may be associated with a particular disease.

907
Q

What is genomics?

A

Analysis of hundreds to thousands of targets or whole genomes, rather than single genes

908
Q

Describe bead arrays

A
  • used for multiplex detection of proteins and nucleic acids
  • tissue typing
  • respiratory virus panels
  • beads may have antibodies or single- stranded oligonucleotides attached
909
Q

Describe in situ hybridization

A

Labeled probes hybridize to tissues or cells on glass sides

910
Q

What are the types of in situ hybridization?

A
  • immunochemistry
  • fluorescence in situ hybridization
911
Q

Describe immunohistochemistry

A

Uses enzymes-labeled antibodies to identify protein targets

912
Q

Describe fluorescence in situ hybridization (FISH)

A
  • uses fluorescent labeled probes to detect specific DNA sequences
  • detects chromosomes abnormalities
913
Q

What are some examples of the amplification method?

A
  • PCR
  • target amplification
  • transcription based amplification
  • strand displacement amplification
  • signal amplification
914
Q

describe polymerase chain reaction (PCR) procedure

A

1) denaturation —> DNA is separated into single strands by heat (95 C)
2) annealing —> primer attach (60 C)
3) extension —> complementary by nucleotides are added to 3’ end (~70 C)

915
Q

Describe target amplification

A
  • amplification of PCR products, under optimal conditions, each cycle results in doubling product
  • in this manner, a target sequence present that are fewer than 100 copies can be detected
  • resulting DNA fragments (amplicons) are detected using various methods
916
Q

briefly describe PCR

A

In vitro DNA replication procedure

917
Q

What is key for specificity of a PCR reaction?

A
  • oligonucleotide primer —> synthetic single stranded nucleic acids, usually 18 to 30 in length
918
Q

What is the instrument used to carry out the amplification program?

A

Thermal cycler
- chamber-based
- block-based

919
Q

What is the a common approach used to detect mutations and polymorphisms?

A

SSP-PCR

920
Q

Describe quantitative PCR (qPCR)

A
  • determines the amount of a specific sequence in the original sample
  • both DNA and RNA target can be measured by qPCR
921
Q

What are the four probe types of qPCR?

A
  • fluoroscence resonance energy transfer (FRET)
  • Taqman
  • molecular beacon
  • scorpion probes
922
Q

What are some commonly used applications of qPCR?

A
  • detection of microorganisms, especially viruses and other pathogens, tumor associated gene expression and tissue typing
923
Q

Describe reverse-transcriptase PCR (RT-PCR)

A
  • amplifies cDNA made from an RNA template
  • HIV, HCV
924
Q

Describe transcription-based amplification (TMA)

A
  • RNA is the target as well as the primary product
  • product detected by chemiluminescence with acridinium ester
  • isothermal process
  • useful in testing got RNA viruses
  • temperature cycling not required
925
Q

Describe probe amplification

A
  • number of target nucleic acid sequences does not change
  • instead, primers are extended or ligated into many copies of detectable probes
926
Q

What are some examples of probe amplification?

A
  • strand displacement amplification (SDA)
  • loop-mediated isothermal amplification (LAMP)
  • molecular inversion probe (MIP)
927
Q

Describe strand displacement amplification (SDA)

A
  • after initial denaturation step, the reaction proceeds at one temperature
  • nick formed in strand by restriction endonucleus enzyme
928
Q

Describe loop-mediated isothermal amplification (LAMP)

A
  • high specific and sensitivity for target DNA
  • PCR primer carry sequences at 5’ end that will self hybridize, forming loops that self prime in cyclic manner
  • advantage —> shortened Run time
929
Q

What is the LAMP used to detect?

A
  • HIV
  • cytomegalovirus
  • staphylococcus aureus
  • E. Coli
930
Q

Describe molecular inversion probe (MIP)

A
  • high sensitive
  • probe ends bind to target sequences so that, in the presence of target, probes ends are brought together and ligated to form circles
931
Q

What is MIP used to detect?

A
  • staphylococcus aureus
  • streptococcus mutans
  • influenza virus
  • RNA typing
932
Q

Describe signal amplification

A
  • large amounts of signals are bound to the target sequences
  • example: branched DNA
933
Q

Describe branched DNA (bDNA)

A
  • has been used to detect certain viruses
  • series of short, single stranded DNA probes are used to capture the target nucleic acid and to bind to multiple reporter molecules, loading target nucleic acid with signal
934
Q

Describe DNA sequencing

A
  • direct determination of the order of nucleotides in a DNA chain
  • the most specific method of identifying genetic mutations or polymorphisms
  • methods include Sanger sequencing, pro sequencing, and next-generation sequencing (NGS)
935
Q

Describe Sanger sequencing

A
  • uses all components of PCR plus fluorescent-labeled dideoxynucleatides (ddNTPS) to synthesize DNA complementary to the target
  • when correct ddNTPs is added, DNA synthesis stops and fragments of varying sizes are produced
  • results are read by gel or capillary electrophoresis
  • less sensitive than other methods
936
Q

Describe pyrosequencing

A
  • based on the generation of light when nucleotides are added to a growing strand of DNA
  • not as versatile as Sanger sequencing, but less labor intensive
937
Q

Describe next-generation sequencing (NGS)

A
  • can sequence large numbers of templates simultaneously (massively parallel sequencing)
  • using bioinformatics, the short sequences are assembled to determine the complete sequence, which is compared to known database
  • more error prone than Sanger sequencing
938
Q

what are applications of next-generation sequencing (NGs)?

A
  • ID of genetic variations in inherited diseases and tumor cells
  • HLA allele typing
  • analysis of mixed microbial populations
939
Q

Describe ion conductance sequencing

A

Nucleotide order is determined through release of hydrogen ions by DNA synthesis

940
Q

Describe whole exonerated sequencing

A

Used to identify variants responsible for inherited disease conditions

941
Q

Describe whole genome sequencing

A
  • primarily research tools
  • massive parallel sequencing has the capacity to investigate all known genetic loci alterations
942
Q

Describe targeted libraries (sequencing)

A

See==elected genes or gene regions q

943
Q

What is the basis of all molecular diagnostic testing?

A

The high specificity of detection of nucleic acid sequences through complementary base pairing

944
Q

Between promotor, cytosine, S phase, and primer which one is associated with RNA synthesis only?

A

Promotor

945
Q

What is the function of restriction endonucleuses?

A

The cleave DNA at specific sites

946
Q

What technique uses RNA-guided enzyme?

A

CRISPR

947
Q

What is the purpose of an amplification control in qPCR?

A

Avoid false negatives

948
Q

What are some hybridization techniques??

A

Microarray technology
Fluorescent in situ hybridization

949
Q

What is the principle of rapid immunochromatographic?

A
  • patient sample is added to test strip and migrates through the strip. Patient antigen complexes to antibody-labeled particles or competes with antigen-labeled particles across individual test lanes at unique detection zones
950
Q

What is immunophenotyping?

A

Identifying their surface and cytoplasmic antigen expression

951
Q

What is flow cytometry used to detect?

A
  • leukemia
  • lymphoma
  • AIDS
  • chronic granulomatous disease
  • leukocyte adhesion deficiency
  • fetal RBC
952
Q

What is a significant advantage of flow cytometry?

A

Because the flow rate of cells is so rapid, thousands of events can be analyzed in seconds, allowing for accurate detection of cells that are present in very small numbers

953
Q

What are the 4 parts of instrumentation of flow cytometry?

A

1) fluidics
2) laser light source
3) optics and photodetectors
4) computer

954
Q

Describe fluidics of flow cytometry

A
  • allows for cell transport, one at a time
  • each time a cell passes in front of a laser beam, light is scattered, and the interruption of laser signal is recorded
955
Q

Describe laser light source of flow cytometry

A
  • for cell illumination and identification
  • solid-state diode lasers are typically used
956
Q

Describe optics and photodetectors of flow cytometry

A

Signal detection

957
Q

Describe computers of flow cytometry

A

Data management

958
Q

What colors are lasers?

A
  • red
  • blue
  • violet
  • ultraviolet
  • yellow-green
959
Q

What are the two types of light scatter?

A

1) forward scatter (FSC)
2) side scatter (SSC)

960
Q

Describe forward scatter

A
  • an indicator of size
961
Q

Describe side scatter

A
  • indicative of granularity or the intracellular complexity of the cell
962
Q

What are the intrinsic parameters of the light sources of flow cytometry?

A
  • forward scatter
  • side scatter
  • light scattering properties s
963
Q

Describe extrinsic parameters in light source of flow cytometry?

A

Requires the addition of a fluorescent probe for their detection

964
Q

Describe principle of hydronomically focusing in flow cytometry

A
  • cells pass single file through the intersection of the laser light source
965
Q

What is the additive in the tube that is used for whole blood collection?

A

EDTA heparin can also be used

966
Q

Describe single-parameters histogram

A
  • the y-axis consists of the number of events
  • usually extrinsic parameters (fluorescent-labeled) antibody
  • operator sets a marker to isolate the positive event
  • will calculate the percentage of positive events within the designated markers
967
Q

Describe dual-parameters dot plot

A
  • both parameters (axis) are chosen by operator
  • the operator then draws a “gate” or isolates the population of interest for further analysis
  • gated cells are analyzed for fluorescence
  • suppurated into four quadrants
968
Q

Describe quadrant 1 of dual parameter dot plot

A

X-axis —> negative for fluorescence
Y-axis —> positive for fluorescence

969
Q

Describe quadrant 2 of dual parameter dot plot

A

X-axis —> positive for fluorescence
Y-axis —> positive for fluorescence

970
Q

Describe quadrant 3 of dual-parameter dot plot

A

X-axis—> negative for fluorescence
Y-axis—> negative for fluorescence

971
Q

Describe quadrant 4 of dual-parameter dot plot

A

X-axis —> positive for fluorescence
Y-axis —> positive for fluorescence

972
Q

What are some applications of flow cytometry?

A

1) identifies particular markers for diagnosis and monitoring of leukemia and lymphomas
2) enumerates peripheral blood. CD4+ T cells to classify stages of HIV disease and guide treatment
3) enumerates CD34+ cells in stem cells transplantation
4) determines DNA content or ploidy status of tumor cells
5) helps diagnosis inherited diseases

973
Q

What is the significance of the presence of CD5+?

A

Indication of mantle cell lymphoma (CLL)

974
Q

How is HIV is classified?

A

Enumeration of peripheral blood CD4+ T cells by flow cytometry

975
Q

What are the advantages of immunoassay automation?

A
  • reduce error
  • is more accurate and precise
  • requires fewer staff
  • saves on controls, duplicates, dilutes, and repeats
  • potential for better sample ID with use of bar coding
976
Q

What are two types of immunoassay analyzer?

A
  • batch
  • random-access analyzer
977
Q

Describe batch analyzers

A
  • can examine multiple samples
  • provide access to samples for formation of reaction mixture
  • permits one type of analysis at a time
978
Q

Describe random access analyzers

A
  • measure numerous analytes from multiple samples
  • can perform different tests on any one sample
979
Q

What is accuracy?

A

Tests ability to measure what it claims to measure

980
Q

What is precision?

A

Ability to consistently reproduce a result on repeated testing of the same sample

981
Q

What is analytic specificity?

A

An assays ability to generate a negative result when the analyze is not present

982
Q

What is analytic sensitivity?

A

The lowest measurable amount of an analyte

983
Q

What is a reportable range?

A

The range of values that will generate a positive results for the specimens assayed by the test procedure

984
Q

What is the reference interval?

A

Range of values found in healthy individuals

985
Q

What is the single most important requirement for samples to be analyzed on a flow cytometry?

A

Cells must be in a single-suspension

986
Q

The various signals generate by cells intersecting with a flow cytometry laser are captured by what?

A

Photomultiplier tubes

987
Q

What is flow cytometry most commonly used method?

A

Immunophenotyping of lymphoid and myeloid

988
Q

Describe hypersensitivity

A
  • exggerated immune response to a typically harmless antigen
  • results in tissue injury and disease
  • 4 types
989
Q

Describe the immediate reaction of hypersensitivity

A
  • develop minutes to hours after antigen exposure
  • type I, II, and III
990
Q

Describe the delay reaction of hypersensitivity

A
  • develop 24 to 48 hours after antigen exposure
  • type IV
991
Q

Describe type I hypersensitivity

A
  • cell bound antibodies react with antigens to release physiologically active substances
  • complement is not involved
  • known as “allergies”
992
Q

Describe type II hypersensitivity

A
  • free antibody reacts with antigen associated with cell surfaces
  • complement plays major role in producing tissue damage
993
Q

describe type III hypersensitivity

A
  • antibody reacts with soluble antigen to form complexes that precipitate in tissues
  • complement plays major role in producing tissue damage
994
Q

Describe type IV hypersensitivity

A
  • not seen until 24-48 hours
  • differs from the other three because sensitized T cells rather than antibody are responsible for symptoms
  • complement not involved
995
Q

What is another name for type I hypersensitivity?

A

Anaphylactic hypersensitivity

996
Q

What is another name for type II hypersensitivity?

A

Antibody-mediated cytotoxic hypersensitivity

997
Q

What is another name for type III hypersensitivity?

A

Complex-mediated hypersensitivity

998
Q

What is another name for type IV hypersensitivity?

A

Cell-mediated hypersensitivity

999
Q

What are the key components of type I hypersensitivity?

A
  • IgE
  • Mast cells
  • basophils
  • eosinophils
1000
Q

Describe sensitization phase of hypersensitivity

A
  • APCs process allergens and present them to Th cells
  • Th2 cells induce production of allergen-specific IgE
  • IgEb binds to FceRI receptors on most cells and basophils
1001
Q

Describe the activation phase of hypersensitivity I

A
  • allergen cross-link adjacent cell-found IgEs
  • mast cells and basophils degranulate
  • chemical mediators are released and bind to target organs
  • allergy symptoms produced
1002
Q

What are the type I preformed/primary mediators?

A
  • histamine
  • heparin
  • eosinophil chemomatic factor of anaphylaxis (ECF-A)
  • neutrophil chemomatic factor
  • proteases
1003
Q

What are the type I newly formed mediators?

A
  • platelets activating factor (PAF)
  • prostaglandin (PG) D2
  • leukotrienes (LT) —> B2, C4, D4, and E4
  • cytokines
1004
Q

What are some common allergens?

A
  • pollen
  • mold spores
  • animal danders
  • dust mites
  • insect venom
  • certain foods
  • certain dogs
  • latex
1005
Q

What are the clinical manifestations of type I?

A
  • Rhinitis (hay fever)
  • allergic asthma
  • food allergies
  • urticaria (hives) —> wheal and flare
  • eczema
  • systemic anaphylaxis —> potentially fatal
1006
Q

What are treatment for type I?

A
  • avoid allergens
  • drug therapy —> antihistamines, bronchodilators, most cell stabilizers, corticosteroids epinephrine
  • monoclonal anti-IgE antibody
  • allergy immunotherapy (AIT) —> administer gradually, increasing doses of allergen
1007
Q

Describe process of percutaneous and intradermal of type I

A

1) apply a panel of allergens to seperate sites on the skin
2) wait 15 to 20 minutes
* positive test = wheal and flare at the site of application

1008
Q

describe allergen specific IgE testing of type I?

A
  • RAST (radioallergosorbent test) was the first type of test to be used for specific IgE
  • enzyme methods are now used to detect IgE specific allergen in patient serum
  • safer than skin testing
1009
Q

Describe the total IgE test of type I

A
  • RIST (radioimmunosorbent test) was first type of test method for total IgE
  • enzymes are now use to detect total concentration of IgE in patient serum
1010
Q

What are allergens?

A

Refers to an inherited tendency to develop classic allergic responses to naturally occurring inhaled or ingested allergens

1011
Q

How long does it take for type I to occur?

A

30-60 minutes

1012
Q

What is passive cutaneous anaphylaxis ?

A

Redness and swelling at site that was injected with serum (from patient who is allergic to allergen) and later exposed to allergen

1013
Q

What is atopy?

A
  • refers to an inherited tendency to develop classic allergic responses to naturally occurring inhaled or ingested allergens
1014
Q

What regulates IgE production?

A

Th2 cells

1015
Q

What is predominant in people with allergens? What does this cause to be produce?

A
  • Th2
  • IL-4 and IL-13
1016
Q

What are IL-4 and IL-13 responsible for?

A
  • final differentiation that occurs in B cells, initiating the transcription of the genes that codes for the epsilon heavy chain of immunoglobulin molecules belonging to the IgE
1017
Q

describe allergic rhinitis

A
  • most common form a of atopy
  • hay fever
  • symptoms —> paroxysmal sneering , itchy eyes and nose, nasal congestion, and runny nose
1018
Q

What is angioedema?

A

Skin reactions that occur deeper in the skin

1019
Q

Describe allergy immunotherapy (AIT)

A
  • goal is to induce immune tolerance to a specific allergen by administering gradually increasing doses of the allergen over time
1020
Q

How is allergic reaction scored?

A
  • based on presence or absence of erythema
  • diameter of the wheal
1021
Q

What are the key components of type II?

A
  • IgG and IgM directed against cell surface antigen
  • complement
1022
Q

What effects can type II have on antibodies?

A
  • cell destruction
  • inhibition of cell function
  • increase in cell function
1023
Q

What can cause cell damage in type II hypersensitivity?

A
  • activation of classical pathway of complement and cell lysis
  • opsonization and phagocytosis of the cell
  • antibody
  • antibody-dependent cell-mediated cytotoxicity (ADCC)
1024
Q

How long does it take for type II hypersensitivity to kick in?

A

A few hours after exposure to antigen

1025
Q

What happens when sensitized mast cells and basophils are activated?

A

Release chemicals that induce symptoms

1026
Q

Describe mast cells of the sensitization phase of type I?

A
  • have abundant cytoplasmic granules that store numerous preformed inflammatory mediators
1027
Q

Describe bind of IgE to mast cells or basophils

A
  • increases half life from 2-3 days to 10 days at least
  • once IgE binds, functions as an antigen receptor on mast cells and basophils
1028
Q

What are effector cells of type I hypersensitivity?

A

Mast cells

1029
Q

Describe hemolytic disease of the newborn (HDN)

A
  • pregnant woman produces antibodies to Rh antigens (usually RhD) on fetal RBCs
  • IgG cross placenta and destroy fetal RBCs
1030
Q

Describe autoimmune hemolytic anemia

A
  • type II hypersensitivity
  • direct against self-antigens because this disease forms antibodies to their own RBCs
  • 2 kinds of antibodies —> warm reactive antibodies and cold agglutinins
  • symptoms —> jaundice, unexplained fever, lightheadedness, weakness, and malaise
1031
Q

What temperature does warm reactive antibodies react at?

A

37 C

1032
Q

What temperature does cold agglutinins react to?

A

Below 30 C

1033
Q

Describe the warm autoimmune hemolytic anemia

A
  • account for more than 70% of autoimmune anemias
  • characterized by formation of IgG antibody
1034
Q

Describe cold agglutinins

A
  • autoantibodies that cause RBCs to clump together at cold temperatures
  • below 37 C
  • belong to IgM class
1035
Q

What is found in those affected with cold agglutinins syndrome?

A
  • high titer of cold agglutinins of C3d resulting in a positive direct antiglobulin test
1036
Q

Describe paraoxysmal cold hemoglobinuria

A
  • can cause autoimmune hemolytic anemia
  • occurs after certain infections (measles, mumps, chickenpox, IM)
  • agglutinate at cold temperatures
  • activates complement at 37C to produce intermittent hemolysis
1037
Q

Describe mycoplasma pneumoniae

A
  • cold agglutinins found in approximately 55% of patients
  • cold agglutinins appear around week 2 or 3 after on set
  • reoccurring respiratory infections
  • symptoms —> enlarged spleen, jaundice, pallor, hemoglobinuria
1038
Q

describe infectious mononucleosis

A
  • 50% of patients with disease have anti-i present
  • antibody normally detectable in vitro up to a temperature of about 25C
1039
Q

Describe direct antiglobulin test (DAT)

A
  • detects RBCs coated with complement components or IgG antibody in vivo
  • patient RBCs are incubated with a poly-specific anti-human Ig directed against IgG and C’
  • if positive , the test is repeated with mono-specific anti-IgG, anti-C3b and anti-C3d
1040
Q

Describe indirect antiglobulin test (IAT)

A
  • Coombs test
  • test patient serum for antibodies to RBC antigens
  • positive test —> agglutination
1041
Q

Describe process of indirect antiglobulin test

A

1) incubate reagent RBCs with patient serum at 37C, wash to remove excess
2) add anti-human globulin

1042
Q

What is DAT used to detect?

A
  • Transfusion reactions
  • HDFN
  • autoimmune hemolytic anemia
  • drug induced hemolytic anemia
  • detects in vivo binding
1043
Q

What is IAT used for?

A
  • cross matching of blood to prevent a transfusion reaction
  • determine the presence of particular antibody in patient plasma
  • type patient RBCs for specific blood group antigens
  • detects in vitro binding
1044
Q

Describe type III hypersensitivity

A
  • keys components are IgGand IgM directed against a soluble antigen
  • small antigen- antibody complexes precipitate out and deposit in tissues
  • C’ binds —> vasodilation and vasopermability increase
  • macrophages and neutrophils migrate to affected areas and release lysosomal enzymes, resulting in tissue damage
1045
Q

How long does type III hypersensitivity take to occur?

A

A few hours after exposure

1046
Q

Describe Arthus reaction

A
  • skin reaction caused by type III hypersensitivity
  • localized inflammation characterized by redness and edema
  • peaks at 3 to 8 hours
1047
Q

Describe serum sickness

A
  • generalized type III hypersensitivity reaction
  • caused by passive immunization of humans with animal serum
  • produces antibodies against the foreign animal proteins against the foreign animal proteins in patients
  • cause immune complexes to form and deposit in tissues
  • symptoms —> headache, fever, nause, joint pain, rashes, lymphadenopathy
1048
Q

What are the laboratory tests of type III hypersensitivity?

A
  • testing ANAs
  • fluorescent staining of tissue sections to detect deposited immune complexes
  • testing for rheumatoid factor (anti-IgG)
  • testing complement levels
1049
Q

When are complementary levels decreased in serum?

A
  • during periods of high disease activity
1050
Q

What methods detect antinuclear antibodies in SLE and RA?

A
  • indirect immunofluorescence
  • ELISA
  • fluorescent microsphere multiplex immunoassay
1051
Q

What methods can detect RF?

A
  • latex agglutination
  • nephelometry
  • other immunoassays
1052
Q

Describe type IV hypersensitivity?

A
  • Th1 and macrophages are involved
  • APCs present antigen to naive Th cells, which differentiate into Th1 cells
  • Th1 cells release cytokines that attract and activate macrophages
  • macrophages induce inflammation
  • cytotoxic T lymphocytes are recruited and destroyed target cells
1053
Q

Describe hypersensitivity pneumonitis

A
  • allergic disease of lungs
  • caused by inhalation of bacterial and fungal spores
  • primarily mediated by sensitized T cells that respond to inhaled allergens
1054
Q

What are some examples of hypersensitivity pneumonitis?

A
  • farmers lung disease
  • birds breeders lung disease
  • humidifier lung disease
1055
Q

Describe contact dermatitis

A
  • low-molecular weight compounds contact the skin and act as haptens to sensitive Th1 cells
1056
Q

What are trigger for contact dermatitis?

A
  • poison ivy
  • poison oaks
  • Nickles salts
  • cosmetics
  • hair dye
  • latex
1057
Q

What are granoulomas?

A
  • cluster of cells produced by chronic persistence of antigen
  • can function to wall off the organism in contained area, preventing spread
1058
Q

What are the skin tests for type IV hypersensitivity?

A
  • patch test
  • skin testing for immunodeficiency
  • mantoux method
1059
Q

Describe patch test

A
  • antigen applied to skin surface
  • test for contact dermatitis
  • positive test —> redness with papules or blisters
1060
Q

Describe mantoux method

A
  • antigen injected intradermally
  • tests for TB exposure or T cell function
  • positive test —> induration
1061
Q

What are types of interferon gamma release assays?

A
  • quantification TB gold plus assay
  • T-spot TB test
1062
Q

Describe interferon gamma release assays (IGRA)

A
  • measures production of IFN- gamma by patient T cells stimulated with MTB antigens
1063
Q

Describe T-spot TB test

A
  • patient mononuclear cells are incubated with MTB antigens and tested for IFN- gamma by ELISPOT
1064
Q

What is the gold standard testing for contact dermatitis?

A

Patch test

1065
Q

What could a positive result in DAT show?

A
  • presence of IgG on RBCs
  • presence of C3b or C3d on RBCs
  • a transfusion reaction caused by preformed antibody
1066
Q

What is the difference between type I and type II?

A

Type II involves cellular antigens

1067
Q

A young woman developed red, itchy papules on her wrist 2 das after wearing a new bracelet. What is this reaction caused by?a

A

An inflammatory response induced by cytokines released from Th1

1068
Q

What are reactions to latex caused by?

A
  • type I
  • type IV
  • skin irritation
1069
Q

What is the principle of the tuberculin test?

A
  • PPD is injected into the forearm and stimulates a delayed type hypersensitivity response mediated by T cells in patients with prior exposure causes induration at the injection site within 48-72 hours
1070
Q

Describe autoimmune diseases

A
  • humoral and cell-mediarted immune responses directed toward self-antigens
  • cause tissue or organ damage
  • systemic or organ-specific due to loss of self tolerance
1071
Q

What is self tolerance?

A

Ability of the immune system to accept self-antigens and not initiate an immune response against them

1072
Q

What does autoimmunity result from?>

A
  • complex interactions between genetic and environmental factors
  • inheritance of certain genes can make some individuals some susceptible
  • hormones can influence autoimmunity
  • tissue injury
  • microbial infections
  • environmental factor that can alter gene expression
1073
Q

What are the microbial that can trigger autoimmunity?

A
  • molecular mimicry
  • epitope spreading
  • superantigens
1074
Q

Why does molecular mimicry trigger autoimmunity?

A
  • bacteria or viruses possess antigens that are like a self antigen
1075
Q

Why does epitope spreading trigger autoimmunity?

A
  • immune response to a microbe expands to activate, including self antigens
1076
Q

Why do superantigens trigger autoimmunity?

A

Can activate numerous clones of T cells, or through certain viruses, which can cause polyclonal B cells activation

1077
Q

What are systemic diseases of autoimmunity?q

A

SLE
RA
GPA
Others SARDS

1078
Q

What are organ specific diseases of autoimmunity?

A
  • autoimmune thyroid disease
  • type I diabetes
  • celiac disease
  • autoimmune liver disease
  • multiple sclerosis
  • myasthenia gravis
  • anti-glomerular basement membrane disease
1079
Q

Describe immunologic tolerance

A

A state of immune unresponsiveness that is direct against a specific antigen

1080
Q

Describe central tolerance

A

Occurs in central or primary lymphoid organs, thymus, and bone marrow

1081
Q

What is anergy?

A

Absence of the normal immune response to a particular antigen or allergen

1082
Q

Describe peripheral tolerance

A

Lymphocytes that recognize self-antigens in the secondary lymphoid organs are rendered incapable of reacting with those antigens

1083
Q

What are the classification of autoimmune disease?

A

1) systemic
2) organ specific

1084
Q

What are the 2 groups of system diseases?

A
  • systemic autoimmune rheumatic diseases (SARDs)
  • anti-neutrophil cytoplasmic antibodies (ANCAs)
1085
Q

Describe SARDs

A

Involve inflammation of the joints and their associated structures

1086
Q

Describe ANCAs

A

Characterized by inflammation of the blood vessel

1087
Q

Describe system lupus erythematous (SLE)

A
  • chronic sytstemic inflammatory disease that affects multiple organ systems
  • pathology caused by type III, tissue damage occurs at the sites in thee body were immune complexes have deposited
  • develop numerous autoantibodies
  • symptoms —> joint involvement, skin rash, renal involvement, weight loss, malaise, fatigue
1088
Q

What happens when immune complexes form within a patient with SLE?

A

Trigger
- complement activation
- chemotaxis of neutrophils
- inflammation

1089
Q

What autoantibodies develop in patients with SLE?

A
  • dsDNA and other nuclear components
  • lymphocytes
  • RBCs
  • platelets
1090
Q

Describe the skin rash that occurs on patients with SLE

A
  • a classic butterfly rash across the nose and cheeks
  • this is where lupus comes from, meaning wolf-like
1091
Q

Describe the LE cell

A
  • it is a neutrophil that have engulfed the antibody-coated nucleus of another neutrophil, mainly in vitro
1092
Q

What are the laboratory tests to diagnosis SLE?

A
  • CBC
  • urinalysis
  • CRP/ESR
  • complement quantitation
  • ANAs
1093
Q

What test is typically done first if SLE is suspected?

A
  • screening test for antinuclear antibodies (ANAs)
1094
Q

What is the most common cause of death is lupus?

A
  • renal failure and infection
1095
Q

Describe ANAs

A
  • directed against antigens in cell nuclei
  • present in more than 95% of lupus patients
  • includes
    —> anti-dsDNA
    —> anti-ssDNA
    —> anti-histones and nucleosomes
    —> antibodies to centromere or nuclear component
    —> anti-ENA
1096
Q

What antibody (ANA) is most specific for SLE?

A
  • dsDNA
1097
Q

What is a nucleolus?

A

Prominent structure within the nucleus where transcription and processing of ribosomal RNA and assembly of ribosomes take place

1098
Q

What testing is most widely used to detect ANAs?

A

Fluorescent antinuclear antibody

1099
Q

What is the FANA test principle?

A

1) incubate patient serum with hep-2 cells fixed no microscope slide
2) wash and incubate with fluorescein-labeled anti-human IgG
3) wash and view under-fluorescent microscope

1100
Q

Describe ENA Ouchterlony test

A
  • immunodiffusion test detects antibodies to specific ENA
  • patient serum in outer wells react with ENA antigens in center well
  • anti-sm is diagnostic for SLE
  • positive reactions indicated by immunoprecipation lines of serological identity
  • not as sensitive
  • long turn around times
1101
Q

What are the 3 main applications of IIF?

A

1) as initial test to determine the presence or absence of ANAs in patients with SARDs, aiding in diagnosis
2) to provide guidance in selection of follow-up tests based on the immunofluorescence patterns obtained
3) to determine ANA titer because moderate to high titers have been better correlated with disease

1102
Q

Why are HEp-2 cells used in IIF?

A
  • they have large nucleic with high antigen expression, allowing for high sensitivity and facilitating visualization of results.
1103
Q

What are the 4 major nuclear patter groups?

A

1) homogenous
2) speckled
3) centromere
4) nucleolar

1104
Q

Describe homogenous nuclear pattern

A
  • characterized by uniform staining of entire nucleus in interphase cells and of condensed chromosomal region in metaphase cells
  • associated with dsDNA, his tones, and nucleosomes
1105
Q

When is homogenous nuclear patterns presents?

A

Found in patients with
- SLE
- drug induced lupus
- AIH
- juvenile idiopathic arthritis

1106
Q

Describe the speckled nuclear pattern

A
  • characterized by discrete fluorescent specks throughout the nuclei of interphase cells
  • 3 groups: dense fine, tiny/fine, and large/course
1107
Q

What are fine or coarse speckled patterns associated with?

A
  • antibodies to ENAs
  • SLE
  • Sjorgen’s syndrome
  • SSc
  • other SARDs
1108
Q

What is the dense speckled pattern associated with?

A
  • antibodies to the DFS70/LEDGF antigen
  • correlated with absence of a SARD upon confirmatory test
1109
Q

Describe the centromere nuclear pattern

A
  • numerous discrete speckles are seen in nuclei of interphase cells and the chromatin of divding cells
  • 46 speckles, represent each chromosome
  • found mainly in patients with CREST syndrome
1110
Q

Describe the nucleolar nuclear pattern

A
  • prominent staining of the nucleoli within nuclei of interphase cells is seen in this pattern
  • seen in pateints with SSc but also can be present in other SARDs
1111
Q

Describe rheumatoid arthritis (RA)

A
  • chronic arthritis of the peripheral joints that can progress to joint deformity and disability
  • 25% of patients have osteoporosis
  • some patients also develop
  • subcutaneous nodules
  • pericarditis
  • interstitial lung disease
  • vasculitis
1112
Q

Describe pathology of RA

A
  • inflammation destroys the bone and cartilage
  • TNF-alpha plays key role in the process
  • overly active osteoclasts absorb the bone
  • autoantibodies combine with antigen to form immune complexes
1113
Q

What are some laboratory tests for RA?

A
  • Rheumatoid factor
  • anti-CCP
  • ANAs
  • ESRs, CRP and C’
1114
Q

Dscescribe rheumatoid factor

A
  • autoantibody (usually IgM) that reacts with Fc portion of IgG
  • found in approximately 80% of patients with RA, not specific for RA
1115
Q

Describe anti-CCP

A
  • autoantibody directed against cyclic citrullinated peptide
  • high specifc for RA
  • best for early detection of RA
1116
Q

What is pannus?

A

A sheet of inflammatory granulation tissue that grows into the joint space and invades the cartilage

1117
Q

What is the gold standard for ANA testing?

A

IIF

1118
Q

What are symptoms of RA?

A
  • transient joint pain in hands and feet
  • muscle spasms
  • limited movement
  • weight loss
  • fatigue
  • fever
  • malaise
1119
Q

What autoimmune disease is characterized by dry mouth and eyes?

A

Sjorgren syndrome

1120
Q

What is the most common cause for death in RA?

A

Cardiovascular disease

1121
Q

What are the criteria for Identifying RA?

A
  • number and type of joints involved
  • duration of symptoms
  • serology results for RF and anti-CCP
  • serum level of the acute phase reactant
  • CRP
  • ESR
1122
Q

Describe sjogren syndrome

A
  • dry eyes and mouth
  • ss-A and ss-B antibodies
  • characterized by chronic inflammation of the exocrine glands, most notably —> ocular and salivary gland
  • have primary and secondary categories
1123
Q

Describe systemic sclerosis (SSc)

A
  • fibrosis and vasculitis affecting skin, joints, other organs
  • includes CREST syndrome and many ANAs
  • symptoms
    —> tightening and hardening of skin
    —> musculoskeletal pain
    —> Raynaud’s phenomenon
    —> heartburn
1124
Q

Describe mixed connective tissue disease

A
  • overlap of limited cutaneous SSc and other SARDs; anti-u1-RNP
1125
Q

Describe inflammatory myopathies

A
  • polymytosis and dermatomyostis
  • progressive muscle weakness; many ANAs
  • characterized by chronic inflammation of the skeletal muscles and progressive muscle weakness
1126
Q

Describe granulomatosis with polyangitis

A
  • Wegners granulomatosis
  • rare autoimmune disease involving inflammation of small-to-medium sized blood vessels and respiratory tract
  • progresses to more systemic disease involving other organs
  • most patients have antibodies to neutrophil cytoplasmic antigens such as proteinase 3
  • neutrophil activation results in damage to vascular endothelium and Th1 response
1127
Q

What are symptoms of granulomatosis with polyangitis? (GPA)

A
  • collapsed nose bridge
  • hearing loss
  • fever
  • joint pain
  • anorexia/weight loss
  • persistent runny nose
  • pain and arthritis of large joints
  • rhinitis
  • oral or nasal ulcers
1128
Q

Describe anti-neutrophil cytoplasmic antibodies

A
  • produced against proteins in neutrophil granules
  • strongly associated with syndromes involving vascular inflammation
  • detected by IIF of ethanol-fixed leukocyte
  • ELISA and chemo luminescent assay are also available
1129
Q

Describe ANCA detected by IIF on ethanol fixed leukocytes procedure

A

1) patient serum incubated with microscope slide containing ethanol-fixed leukocytes
2) wash and add FITC-labeled anti-IgG conjugated
3) wash and view fluorescence in neutrophils under fluorescence in neutrophils under fluorescent microscope

1130
Q

What are the 3 syndromes involving vascular inflammation associated with ANCAs?

A
  • GPA
  • microscopic polyangitis (MPA)
  • eosinophil granulomatosis with polyangitis (EGPA)
1131
Q

What are the two patterns that can be observed with ANCAs?

A
  • cytoplasmic (c-ANCA)
  • pernicular (p-ANCA)
1132
Q

Describe c-ANCA pattern

A
  • primarily caused by PR3-ANCA and appears as a diffuse, granular staining in cytoplasm of neutrophils
1133
Q

Describe p-ANCA pattern

A
  • fluorescence surrounds the lobes of the nucleus, blending them together so that individual lobes cannot be distinguished
1134
Q

What is the autoantigen of c-ANCA?

A

PR3 antigen

1135
Q

What disease is associated with c-ANCA?

A

GPA

1136
Q

What is autoantigen of p-ANCA?

A

Positively charged antigens, including MPO

1137
Q

What are disease associated with p-ANCA?

A
  • MPA
  • EGPA
1138
Q

What diseases can ANCA be detected in?

A
  • SARDs, such as SLE and RA
  • autoimmune gastrointestinal disease
  • liver disease
  • HIV
  • hepatitis C
  • malignancy
1139
Q

Describe autoimmune thyroid diseases (AITDs)

A
  • thyroid hormone synthesis is carefully regulated by endocrine feedback loop
  • autoantibodies produced in AITDs can lead to to decreased or increased production of thyroid hormones
1140
Q

What diseases are AITDs?

A
  • Hashimoto’s thyroiditis
  • Graves’ disease
1141
Q

What is the target tissue of Graves’ disease and hasimotos disease?

A

Thyroid gland

1142
Q

Describe Hashimoto’s thyroiditis

A
  • immune destruction of the thyroid gland produces hypothyroidism
  • lab results:
    —> normal or high TSH
    —> low free T4
    —> anti-TPO
    —> anti-Tg
  • symptoms
    —> formation of goiter
    —> brittle hair
    —> weight gain
    —> dry skin
    —> fatigue
1143
Q

What is hypothyroidism?

A
  • Decreased thyroid function
  • symptoms
    —> puffy face with edematous eyelids
    —> descreased sweating
    —> brittle hair
    —> pallor with yellow tinge
    —> dry skin
    —> weight gain
    —> fatigue
1144
Q

Describe Graves’ disease

A
  • AITD characterized by hyperthyroidism
  • TRAbs produced
  • low TSH and high FT4; anitbodies to TPO and Tg may be produced
1145
Q

What are autoantibodies of Hashimoto’s thyroiditis?

A
  • anti-thyroglobulin
  • anti- thyroid peroxides
1146
Q

What are the autoantibodies of Graves’ disease?

A
  • TSH receptor antibodies (TRAbs)
  • anti-thyroglobulin
  • anti-thyroid peroxides (TPO)
1147
Q

What is hyperthyroidism?

A
  • state of excess thyroid function
  • associated with Graves’ disease
1148
Q

What are the symptoms of Graves’ disease?

A
  • bulging eyes
  • nervousness
  • insomnia
  • depression
  • heat intolerance
  • sweating
  • heart palpitations
  • breathlessness
  • lower leg edema
  • cardiac dysrhythmias
1149
Q

What are most commonly used to detect thyroid antibodies?

A
  • sensitive ELISA
  • chemiluminescent immunoassays
1150
Q

What is the best indicator for Hashimoto’s thyroiditis?

A

Antibodies to TPO

1151
Q

What is highly indicative of Graves’ disease?

A

TRAbs

1152
Q

What are the 2 tests for TRAbs?

A
  • binding assay
  • bioassay
1153
Q

Describe binding assays for TRAbs

A
  • automated solid-phase ELISA
  • chemiluminescent immunoassays
  • labeled antibody for TSH receptor bound to solid phase
  • unable to distinguish between TSI and TRAbs
1154
Q

Describe bioassay for TRAbs

A
  • requires tissue culture
  • difficult to perform
  • specificity measure the function of TSI
1155
Q

Describe type I diabetes mellitus (T1D)

A
  • endocrine disorder characterized by hyperglycemia
  • type I destruction of beta cells in pancreas results in insulin deficiency
  • genetic susceptibility to the disease
1156
Q

What are the long term effects of type I diabetes mellitus?

A
  • cardiovascular disease
  • kidney dysfunction
  • nerve damage
  • blindness
  • infections
1157
Q

What would the results look like for a patient with type I diabetes mellitus?

A
  • increased glucose blood level
  • elevated HbA1
  • autoantibodies to GAD and IA-2, ICA
1158
Q

What is hyperglycemia?

A

A high level of glucose in the blood

1159
Q

What are the 4 types of diabetes?

A
  • type I
  • type II
  • gestational
  • neonatal/ pancreatitis
1160
Q

What leads to fibrosis criteria that is used to diagnose type I diabetes mellitus?

A

Progressive inflammation of the islets of langerhans in pancreas

1161
Q

What are the 4 criteria that is used to diagnosis type I diabetes mellitus?

A

1) fasting glucose greater than 126 mg/dL on more than one occasion
2) a random plasma glucose level of 200 mg/dL or more with classic symptoms of diabetes
3) oral glucose tolerance test of 200 mg/dL or more in a 2 hour sample with a 75 g glucose load
4) a hemoglobin A12 value (HbA1c) greater than 6.5%

1162
Q

Describe celiac disease?

A
  • affects small intestine and other organs
  • triggered by gluten
  • symptoms
    —>diarrhea
    —> abdominal pain
    —> bloating
1163
Q

What is the diagnosis of the celiac disease based on?

A
  • clinical symptoms
  • serological test findings
  • duodenal biopsy
  • presence of HLA-DQ8 or HLA-DQ2 haplotype
1164
Q

Describe autoimmune hepatitis

A
  • hepatocytes targeted
  • AIH-1 —> positive for SMA, ANAs
  • AIH-2 —> positive for LKM-1 or LC-1 antibodies
  • affects small interlobular bile ducts
1165
Q

What are the alleles are associated with AIH-1?

A

HLA-DR3 and HLA-DR4

1166
Q

What alleles are associated with AIH-2?

A

HLA-DRB1 and HLA-DQB1

1167
Q

What is necessary to confirm AIH?

A

Liver biopsy

1168
Q

Describe PBC

A
  • destruction of intrahepatic bile ducts; cholestasis
  • inflammation of portal vein in liver
  • accumulation of scar tissue that can lead to cirrhosis and liver failure
  • majority of patients, produce mitochondrial Abs (AMAs)
  • symptoms
    —> itchy skin
    —> greasy stool
    —> dry mouth and eye
    —> abdominal pain
    —> jaundice
    —> fatigue
1169
Q

What is the most common autoimmune liver disease?

A

PBC

1170
Q

What is cholestasis?

A

A condition in which flow of bile is slowed or blocked

1171
Q

What methods are these to detect AMAs?

A
  • IIF
  • immunoblotting with mitochondrial preparations from mammal tissues
  • ELISA
  • fluorescent microbead immunoassay
1172
Q

Describe multiple sclerosis (MS)

A
  • involves inflammation and destruction of the central nervous system
  • most patients produce antibodies against myelin basic protein
  • plaques form in while matter of the brain and spinal cord, causing destruction of the myelin sheath of axons
  • symptoms
    —> weakness in limbs
    —> facial palsy
    —> visual disturbance
    —> dizziness
    —> sensory abnormalties
1173
Q

What would be the laboratory findings of a patient with MS?

A
  • lesions on magnetic resonance imaging (MRI)
  • increased immunoglobulins in spinal fluid and increased IgG index
  • oligoclonal bands on protein electrophoresis of CSF
1174
Q

What are environmental factors that have been associated with MS?

A
  • reduced exposure to sunlight
  • vitamin D deficiency
  • cigarette smoking
  • infection with EBV after early childhood
1175
Q

Describe myasthenia gravis

A
  • affects neuromuscular junction, resulting in weak skeletal muscles
  • most patients have antibodies to acetylcholine receptors (ACHR) which block binding of ACH to receptor and transmission of nerve impulses that activate muscles
  • symptoms
    —> drooping eyelids
    —> inability to retract corners of the mouth
    —> difficulty speaking, chewing, and swallowing
    —> respiratory muscle weakness
    —> associated with several HLA antigen abnormalities
1176
Q

What other diseases are associated with presence of MG?

A
  • SLE
  • RA
  • pernicious anemia
  • thyroiditis
1177
Q

Describe laboratory testing for MG

A
  • RIA is used to detect antibody, based on assays that block the binding of receptors by anti-ACH receptor (ACHR) antibody
  • precipitation of receptors caused by combination with antibody is then measured
  • a quick immuno stick ELISA has also been developed for testing
1178
Q

What is the gold standard for detecting binding AChRs?

A

Quantitative radioimmunoassay that have radio-labeled snake venous

1179
Q

Describe anti-glomerular basement membrane disease

A
  • formlerly known as good pasture syndrome
  • patients produce autoantibodies to basement membranes living the renal glomeruli and lung alveoli
  • immune complexes bind to basement membranes, attract complement and cause damage by type II hypersensitivity
  • antibodies to GBM found in most patients
  • symptoms
    —> edema
    —> hypertension
    —> malaise
    —> fatigue
1180
Q

Describe testing for anti-GBM disease

A
  • detected by IIF on frozen kidney sections or ELISA for antibody to a 3-subunits
  • tissue-bound anti-GBM detected by direct immunofluorescence on kidney biopsy sections
  • produces a smooth, linear, ribbon-like fluorescence along the GBM
1181
Q

What allele has a strong association with anti-GBM disease?

A

HLA-DRB1-15

1182
Q

What detects GBM antibodies currently?

A
  • ELISA
  • fluorescent microbead immunoassay that uses recombinant alpha-3 (V) antigen substrates
  • confirmation —> western blot
1183
Q

What is symbiotic relationship?

A
  • hosts and microbes live together long term
  • indigenous microbiota
1184
Q

What is a commensalstic relationship?

A
  • no benefits or harm to either organism
1185
Q

What is mutualistic relationship?

A

Both host and microbes benefit

1186
Q

What is parasitic relationships?

A

Microbes cause harm to host

1187
Q

What are three types of symbiotic relationship?

A

Commensalisitic
Mutualistic
Parasitic

1188
Q

What does the relationship within our indigenous microbiota exist through?

A
  • co-evolution
  • co-adaption
  • co-dependency
1189
Q

What needs to happen for a microorganism to survive?

A
  • needs to stimulate a immune response
  • needs to colonize the host and acquire nutrients
1190
Q

What is infectivity?

A
  • Organisms ability to establish an infection
  • “contagious”
1191
Q

What is pathogenicity?

A
  • ability of an organism to cause disease
  • qualitative trait
1192
Q

Describe virulence?

A
  • extent of damage and pathology caused by an organism when it infects a host
  • quantitative trait
1193
Q

What does virulence factors increase an organisms pathogenicity?

A

1) organisms ability to establish itself on or in the host
2) invade or damage host tissue
3) evade host-immune response

1194
Q

Describe endotoxins

A
  • the lipid A portion of LPS in gram-negative cell walls
  • powerful stimulator of cytokine release
  • complement activator, which forms anaphylatoxins C3a and C5a, which causes vasodilation and increased vasopermability
1195
Q

Describe exotoxins

A
  • potent toxic proteins released from living bacteria (mostly gram-positive)
  • neurotoxins, cytotoxins, enterotoxins
  • known as most potent molecules to harm living organisms
  • extremely immunogenic
  • induce production of protective antibodies
  • may act like superantigen
1196
Q

What is virulence factor?

A

Bacterial properties that determine whether an organism is pathogenic and able to cause disease

1197
Q

What are plasmids?

A
  • self replicating extra-chromosomal DNA molecules that are located in the bacteria’s cytoplasm
  • contain limited number of genes
1198
Q

What are bacterial cells classified as?

A

Prokaryotic cells

1199
Q

What are human cells classified as?

A

Eukaryotic cells

1200
Q

What are the difference between prokaryotic and eukaryotic cells?

A
  • P—> bacterial chromosomes are found in bacterial cytoplasm. E—> enclosed in membrane-bound nucleus
  • P—> bacteria cell wall is outermost feature. E—> plasma membrane is outermost feature
  • P—> cell wall contains alototenymes
1201
Q

What is the primary component that provides shape and rigidity of bacterial cells?

A

Peptidoglycan

1202
Q

What are the two variations of bacterial cell wall?

A
  • positive gram
  • negative gram
1203
Q

What variations of bacteria contains lipopolysaccharide layer (LPS)?

A

Gram-negative

1204
Q

What are the 3 components of LPS?

A
  • outer core polysaccharide
  • inner core polysaccharide
  • lipid A
1205
Q

How does the capsule of bacteria contribute to the organisms ability to resist innate and adaptive immune response?

A

1) block the attachment of antibodies
2) inhibit activation of complement
3) act as a decoy when capsular material is released into the surrounding host environment

1206
Q

What is the most important feature of of the capsule?

A

Blocking phagocytosis by WBCs

1207
Q

What cytokines are produced by endotoxins?

A

IL-1
IL-6
IL-8
TNF
PAF

1208
Q

What are two defenses against bacteria?

A

Innate defenses
Adaptive defenses

1209
Q

Describe innate defense against bacteria

A
  • intact skin and mucosal surfaces serve as structural barriers
  • antibacterial defense peptides are lysozyme, defensins, ribonuclease
  • complement proteins, cytokines, acute-phase reactants
  • recognition of PAMPs by PRRs such as TLRs
1210
Q

Whta is the function of a ribonuclease?

A

Destroy RNA and have antimicrobial and antiviral activities

1211
Q

What are the 3 classes of defensins?

A

Alpha
Beta
Theta

1212
Q

What occurs when PRR engages with appropriate PAMP?

A

1) triggers release of immune mediators such as cytokines and chemokines
2) boosts production for various defensins and proteins
3) initiates phagocytosis

1213
Q

Describe adaptive defenses for bacteria

A
  • antibody production —> binding C’, osponization, neutralization of bacterial toxins
  • cell-mediated immunity —> CD4 T cells produce cytokines that induce inflammation; cytotoxic T lymphocytes attack host cells that contain intracellular bacteria
  • Uses granulosomes to prevent spread
1214
Q

What is the main defense against bacteria in adaptive immunity?

A

Antibody production

1215
Q

What is the binding of antibodies to invading bacteria referred to as?

A

Opsonization

1216
Q

What mechanism do bacteria do use to inhibit the innate immune response?

A

1) avoiding antibody
2) blocking phagocytosis
3) inactivating the complement cascade
4) blocking digestion
5) inhibiting chemotaxis
6) cleaving IgA

1217
Q

What mechanisms do microorganisms use to resist digestion?

A
  • block fusion of lysosome granules with phagosomes after being engulfed by phagocyte
  • production of extra cellular products after bacteria is phagocytosized
  • block action of complement
1218
Q

What are some laboratory detection tests of bacterial infection?

A

1) culture of causative agent
2) microscopic examination
3) detection of bacterial antigens
4) molecular detcection of bacterial DNA or RNA
5) serology

1219
Q

Describe cultures of causative agent

A
  • grow on broth or solid media
  • major means of diagnosis, but may take time or may not be possible
1220
Q

Describe microscopic examination for bacteria

A

Uses gram stain or special stains

1221
Q

What tests are for detection of bacterial antigens?

A

Rapid testing or special stains
- ELISA
- LFA
- LA

1222
Q

Describe serology testing for bacteria

A
  • detects antibodies to bacterial antigens
  • to detect and confirm when option are not available
  • to diagnosis infections with nonspecific symptoms
  • current infection indicated by IgM high titer of IgG, or fourfold rise in antibody
  • to determine a past exposure to an organism
  • to assess reactivation or re-exposure
1223
Q

What do the results of a serology test look like when a past exposure to an organism is present?

A

IgM
IgG+

1224
Q

What are the disadvantages of serology testing for bacterial infection?

A
  • delay between start of infection and production of antibodies
  • low antibody production by immunosuppressed patients
1225
Q

What are disadvantages of culturing for causative agents?

A
  • several bacterial pathogens that don’t have cultures available
  • may take too long
  • some organisms are difficult to grow
  • some organisms present a danger to start it handled improperly
1226
Q

Describe gram stain

A
  • gram-positive bacteria stains purple with crystal violet
  • gram-negative bacteria stains pink with safranin
1227
Q

What is the most widely known molecular technology?

A

PCR

1228
Q

Describe streptococci structure

A
  • are gram positive bacteria
  • spherical, Ovoid, or lancet shaped organisms that are catalase negative and are often seen in pairs or chains
  • divided into groups on basis of certain cell wall components, including two major proteins known as M and T proteins
1229
Q

Describe streptococci

A
  • transmitted person to person
  • additional virulence factors include exoantigens or exotoxins, proteins are secreted by the bacterial cells as they metabolize during the course of streptococcal infections
  • detection of host antibodies exotoxins is important in the diagnosis of sequelae such as glomerulonephritis and acute rheumatic fever
1230
Q

What is another s. Pyogenes?

A

Group A streptococcus (GAS)

1231
Q

Describe serotyping for GAS

A
  • used to identify a particular strain of GAS
  • involves ID of the M protein antigens by precipitation with type-specific sera
1232
Q

What are disadvantages of serotyping?

A
  • limited availability of type sera
  • new M proteins do not react with anti-sera
  • difficulty interpreting results
1233
Q

What is the most common and ubiquitous pathogenic bacteria?

A

GAS

1234
Q

what are virulence factors of GAS?

A
  • the M protein
  • has net-negative charge at the amino terminal end that helps to inhibit phagocytosis
  • M protein limits deposition of C3 on the bacterial surface (diminishing complement activation)
1235
Q

What are clinical manifestations of pus forming group A streptococcal (GAS) infection?

A
  • pharyngitis (“step throat”)
  • pyoderma (impetigo or skin infection)
1236
Q

What are the clinical manifestations non-pus forming group A streptococcal (GAS) infection?

A
  • scarlet fever
  • toxic shock syndrome
  • necrotizing fasciitis
1237
Q

What are the two major sites of strep A infection?

A
  • upper respiratory tract
  • the skin
1238
Q

What are symptoms of pharyngitis?

A
  • tonsillar exudates
  • petechial rash on the soft palate
  • severe sore throat
  • chills
  • fever
1239
Q

Describe impetigo

A
  • most common skin infection
  • vesicular lesions on extremities that become pustular and crusted
  • symptoms
    —> lesions
    —> otitis media
    —> cellultiis
1240
Q

Describe scarlet fever

A
  • usually associated with pharyngeal infections
  • initially appears on the neck and chest then spreads over the body
  • results from infection with strep A that elaborates SpeA and SpeC (can act as superantigens)
  • symptoms
    —> fever of 101 F or higher
    —> rash
    —> abdominal rash
    —> vomiting
    —> nausea
    —> headache
1241
Q

Describe necrotizing fasciitis

A
  • may occur when Strep A
  • such infection invades the muscles in the extremities or the trunk
  • exotoxins produced by s. Pyogenes cause rapid infection deep in fascia
1242
Q

Describe sequelae

A
  • conditions that are consequence of a previous disease or injury
  • results from the host response to infection
1243
Q

What are the two serious sequela that could be developed from Strep A?

A
  • acute rheumatic fever
  • poststreptococcal glomerulonephritis
1244
Q

Describe acute rheumatic fever

A
  • develops 1-3 weeks after pharyngitis or tonsillitis in 2-3% of infected individuals
  • mostly likely caused by immune responses to streptococcal antigens that cross-react with human heart tissue
  • only follow upper respiratory tract infections
1245
Q

Describe post streptococcal glomerulonephritis

A
  • may follow strep infection of the skin or pharynx
  • damages glomeruli
  • deposits of immune complexes containing streptococcal antigens in glomeruli
  • characterized by damage to the glomeruli in the kidneys
  • impaired because glomerular filtration rate is reduced
1246
Q

What are symptoms of acute rheumatic fever?

A
  • fever
  • joint pain
  • inflammation of heart
1247
Q

What are the symptoms of post streptococcal glomerulonephritis?

A
  • hematuria
  • proteinuria
  • edema
  • hypertension
  • malaise
  • backache
  • abdominal discomfort
  • impairment renal function
1248
Q

What laboratory tests are run for suppurative strep A?

A
  • culture on sheep blood agar
  • rapid assays to detect strep A
1249
Q

What are rapid assays used to detect suppurative strep A?

A
  • strep screen
  • LFA
  • assays are easy to perform
  • allow for a single sampling testing
  • more sensitive
  • cultured should be ran if rapid assays are negative
1250
Q

Describe serological detection of strep A sequelae

A
  • antistreptolysin O (ASO) test
  • classic hemolytic method for determining the ASO tier was the first test developed to measure streptococcal antibodies
  • based on the ability of patient antibodies to neutralize the hemolytic activity of streptolysin O
  • an ASO titer greater than 166 Todd units is considered a positive test
1251
Q

What are the serological detection test of nonsupprative strep A?

A
  • antistreptolysin O (ASO)
  • anti-DNase B
  • streptozyme test
1252
Q

Describe antistreptolysin O (ASO) test of nonsupprative strep A

A
  • nephelomatic methods currently used that measure light scatter produced by immune complexes containing streptolysin antigen
  • titer is elevated in 85% of patients with acute rheumatic fever
  • automatic and rapid
1253
Q

Describe anti-DNase B test of nonsuppurative strep A

A
  • produced by both rheumatic fever and impetigo patients
  • tested by EIA and nephelomatic methods
  • useful in patients suspected of having glomerulonephritis preceded by skin infection
  • highly specific for strep A
  • if anti-DNase antibodies are present, they neutralize reagent DNase B, which prevents it from depolymerizing DNA
  • tubes are graded by color, 4+ indicating intensity of color unchanged and 0 being complete color loss
1254
Q

Describe the streptozyme test

A
  • a slide agglutination screening test for detection of antibodies streptococcal antigens
  • sheep RBCs are coated with the 5 antibodies
  • hemagglutination is a positive test result
  • positive in 95% of patients with acute phase post streptococcal glomerulonephritis
  • detects antibodies to five streptococcal products:
    —> ASO
    —> anti-hyaluronidase (AHase)
    —> anti-streptokinase (ASKase)
    —> anti-nicotinamide-adenine dinucleotide (anti-NAD)
    —> anti-DNase B
1255
Q

What are some disadvantages of the streptozyme test?

A
  • the lest reproducible test for antibodies
  • more false negative and false positives have been reported
1256
Q

Describe heliobacter pylori

A
  • gram- negative microaeorphilic spiral bacterium with flagella
  • transmission likely by fecal-oral route
  • major cause of gastric and duodenal ulcers
  • can survive in acidic environment of the stomach because of production of urease, which provides a buffering zone around the bacteria
  • if untreated, can lead to gastric carcinoma or mucosa-associated lymphoid tumors
  • resides in mucus layer, b the gastric epithelium cells
1257
Q

What are detection tests for heliobacter pylori?

A
  • detect urease in stomach biopsy (CLO test)
  • urea breathe test
  • heliobacter pylori antigens
  • heliobacter pylori antibodies
  • ELISA is method of choice
  • IgG serum indicates active infection
1258
Q

What are virulence factor of H. Pylori ?

A

Culture

1259
Q

Describe urea breath test

A
  • patient ingests urea labeled with radioactive carbon or nonradioactice carbon broken down by the Reese’s enzyme of H. Pylori producing ammonia and bicarbonate
  • excellent sensitivity and specificity
1260
Q

An H. Pylori infection results in production of what?

A

IgG (test detect this class)
IgA
IgM

1261
Q

What is considered a successful treatment of H. Pylori infection?

A

Decrease in antibody titer more than 25%

1262
Q

Describe mycoplasma pneumonia

A
  • tiny bacteria that lack a cell wall
  • leading cause of respiratory droplets
  • belong to class mollicutes
  • incubation period is 1 to 3 weeks
  • infection has insidious onset
  • symptoms
    —>fever, headache, malaise, and cough
1263
Q

What are clinical manifestations of mycoplasma pneumonia?

A
  • walking pneomonia
  • Raynaud syndrome
  • Stevens-Johnson syndrome
1264
Q

What is a mycoplasma?

A
  • represents the smallest known free-living life forms
  • have small genome
1265
Q

What are laboratory diagnosis procedures for M. Pneumonia?

A
  • culture
  • antibody detection
  • cold agglutinins
  • molecular methods
1266
Q

Describe a culture of M. Pneumonia

A
  • gold standard but isn’t used anymore
  • produces mulberry colonies with a “fried egg” appearance on specialized media
1267
Q

Describe antibody detection for M. Pneumonia

A
  • most useful diagnostic assay
  • IgM —> recent-infection
  • IgG —> possible reinfection
  • most widely used —> EIAs
1268
Q

Describe cold agglutinins testing of M. Pneumonia

A
  • present in 50% of patients M. Pneumonia but not specific for the infection
  • cause RBC agglutination at 4 C
  • reversible at 37 C
  • not very specific or sensitive
1269
Q

Describe molecular methods of M. Pneumonia

A
  • will most-likely become gold standard
  • LAMP technology
1270
Q

Describe rickettsial infections

A
  • obligate intracellular gram-negative bacteria
  • spotted fever group (spf) —> Rocky Mountain spotted fever
  • typhus group (tg) —> epidemic typhus
  • organisms transmitted by arthropods (tick, mites, lice, or fleas) through biting after feeding on an infected animal
1271
Q

Describe Rocky Mountain spotted fever (RMSF)

A
  • caused by R. Rickettsii
  • transmitted by 3 species of ticks
  • symptoms
    —> rash that starts on hands and soles of feet and proceed to trunk
    —> headache
    —> nausea
    —> vomiting
    —> diarrhea
    —> death
1272
Q

What is RMSF diagnosed?

A
  • clinical presentation of symptoms
  • serology by IFA (gold standard)
1273
Q

How do RMSF organisms spread in the body?

A
  • via lymphatic and circulatory system
  • attach to and invade target cells (vascular endothelium)
  • organism multiply by binary fission
1274
Q

What is the main pathophysiological event caused by RMSF?

A

Endothelial cell damage which leads to increased vascular permeability, resulting in edema, hypovolemia, hypotension, and hypoalbuminemia

1275
Q

What is principle of the IFA test for detection of antibodies produced in RMSF?

A

1) specific antibodies in serum sample attach to the antigens fixed to microscope side
2) attached antibodies are stained with fluorescein-labeled anti-human immunoglobulin and visualized with a fluorescence microscope

1276
Q

What is the principle of LFA?

A
  • involves movement of a liquid sample containing labeled antibodies specific to analyte
  • if antigen-antibody complex is present, it is captured by another antibody at the end of the strip, resulting in a visible line
1277
Q

What are spirochetes?

A
  • long, slender, helically coiled bacteria containing axial filaments or periplasmic flagella
  • wind around the bacterial cell wall and are enclosed by an outer sheath
  • gram-negative
  • microaerophilic
  • corkscrew flexi on and motility
1278
Q

Describe spirochete diseases

A
  • caused by spirochete bacteria
  • localized skin infection, disseminates to numerous organs as disease progresses
  • cardiac and neurological involvement if disease remains untreated
1279
Q

What are the two major spirochete diseases?

A

Lyme disease
Syphilis

1280
Q

Describe syphilis

A
  • sexually transmitted disease caused by treponemal pallidum
  • rapidly destroyed by heat, cold, and drying
  • direct contact with open lesion needed
  • transmission to fetus during pregnancy
  • bloodborne transmission rare
  • most common spirochete disease
  • have TROMP membrane proteins
1281
Q

What are the pathogens in the palladium subspecies?

A
  • T. Pallidum subspecies pertenue
  • T. Pallidum subspecies endemicum
  • T. Carateum
1282
Q

Describe T. Pallidum subspecies pertenue

A
  • agents of yaws
  • found in tropics
1283
Q

Describe T. Pallidum subspecies endemicum

A
  • cause of nonvernereal endemic syphilis
  • found in desert regions
1284
Q

Describe T. Carateum

A
  • the agent of pints
  • found in central and South America
1285
Q

What are the stages of syphilis?

A

1) primary stage
2) secondary stage
3) latent stage
4) teritary stage

1286
Q

What is a characteristic of the primary stage of syphilis?

A

Development of chancre

1287
Q

What are symptoms of the secondary stage of syphilis?

A
  • rash
  • generalized lymphadenopathy
  • fever
  • malaise
  • pharyngitis
1288
Q

Describe the latent stage of syphilis?

A
  • asymptomatic
  • follows disappearance of secondary stage
  • patients are not infectious at this time with the exception of pregnant women
1289
Q

What are the symptoms of the tertiary stage of syphilis?

A
  • gummatous
  • cardiovascular
  • neurosyphilis
  • Tabes dorsalis (shuffling gate)
1290
Q

Whta stages of syphilis could spontaneous healing occur?

A

Primary
Secondary

1291
Q

Describe tertiary stage of syphilis

A
  • stage occurs most often 10-30 years following the secondary stage
1292
Q

What are gummas?

A
  • localized areas of granulomatous inflammation that are most often found on bones, skin or subcutaneous tissue
  • contains lymphocytes, epithelial cells and fibroblasts
  • may heal spontaneously with scarring
1293
Q

Describe cardiovascular complications in the tertiary stage of syphilis

A
  • involve ascending aorta
  • symptoms caused by destruction of elastic tissue in aortic arch
1294
Q

Describe congenital syphilis

A
  • occurs when a woman who has early syphilis or early latent syphilis transmit treponemae’s to the fetus
  • most affected in second and third trimester
  • causes death in 10% of cases
1295
Q

What are laboratory diagnostic procedures of syphilis?

A
  • direct detection
    —> demonstration of treponemal in active lesion
    —> dark-field microscopy
    —> fluorescent antibody staining
  • serological test
    —> nontreponemal
    —> treponemal
1296
Q

How can primary and secondary syphilis by diagnosed?

A
  • presence of T. Pallidum in exudates from skin lesions
1297
Q

When do false negatives occur in dark-field microscopy when testing for syphilis?

A

1) a delay in evaluating the slides
2) insufficient specimen obtained
3) patient is pretreated with antibiotics

1298
Q

What testing should be performed for syphilis if no lesions are present?

A

Serological

1299
Q

Describe nontreponemal tests

A
  • detect antibody against cardiolipin (Reagin)
  • sensitive, inexpensive and simple to perform
  • includes:
    —> venereal disease research laboratory test (VDRL)
    —> rapid plasma Reagin test (RPR)
    —> both - look for flocculation
    —> screen - undiluted patient serum
    —> titer - test two-fold dilutions of patient serum
1300
Q

Describe VDRL test procedure

A

1) patient serum mixed on slide with cardiolipin-lecithin-cholesterol antigen suspension
2) rotated for 4 minutes at 180 RPM
3) viewed under a light microscope for flocculation
Results:
- reactive —> medium to large clumps
- weakly reactive —> small clumps
- nonreactive —> no clumps or slight roughness

1301
Q

What is Reagin?

A
  • a lipid released from membranes of cells damaged as a result of the infection
1302
Q

Describe the RPR test procedure

A

1) patient serum mixed on a card with charcoal particles coated with cardiolipin antigen
2) rotate 8 minutes, 100 RPM
3) observe for macroscopic flocculation

1303
Q

Briefly describe VDRL

A
  • quantitative and qualitative slide flocculation
  • all sera that is weakly reactive and reactive must be retested using quantitative slide test
1304
Q

Why are patients in the secondary stage of syphilis subject to a false positive?

A

Prozone phenomenon (antibody excess)

1305
Q

Briefly describe RPR

A
  • cardiolipin - containing antigen suspension is bound to charcoal particle, making results easier to read
  • all active tests should be confirmed by retesting using doubling dilutions in a quantitative procedure
  • more sensitive than VDRL in primary stage of syphilis
1306
Q

Describe treponemal tests for detecting syphilis

A
  • detect antibody to T. Pallidum
  • usually positive before nontreponemal tests
  • lack of sensitivity in congenital syphilis and neurosyphilis
  • fluorescent treponemal absorption (FTA-ABS)
  • T. Pallidum particle agglutination (TP-PA)
  • automated immunoassays
    —> ELISA
    —> CLIA
    —> MFI
1307
Q

Briefly describe FTA-ABS

A
  • an indirect immunofluorence test antibody to T. Pallidum
  • use Reiter strain
  • false positive results may occur in patients with SLE
1308
Q

Describe FTA-ABS test procedure

A

1) patient serum is incubated with sorbent to remove cross-reacting antibodies
2) absorbed patient serum is incubated with a microscopic slide fixed with T. Pallidum (Nichol’s stain)
3) folllowing a wash step, anti-human Ig conjugated with fluorescein is added
4) after second incubation, and wash, the slide is examined under a fluorescent microscope

1309
Q

Describe TP-PA test procedure

A

1) patient serum and controls are diluted and incubated with unsensitized gel particles or gel particles sensitized with T. Pallidum
- positive —> agglutination (smooth may covering surface of well)
- negative —> no agglutination (button)

1310
Q

Briefly describe TP-PA test

A
  • used colored gelatin particles coated with treponemal antigens
  • more sensitive in detecting primary syphilis
1311
Q

What types of EIAs are used to detect the antibodies to T. Pallidum?

A
  • one or two-step sandwich assays
  • one step competitive assays
  • immune capture assays
1312
Q

Describe sandwich assays of syphilis

A
  • antibodies in the patient sample to bind to recombinant T. Pallidum antigens coated onto microtiter plate wells
1313
Q

Describe the immune capture of syphilis

A
  • micrometer wells are coated with antibody to IgM or IgG and are reacted with patient serum
1314
Q

What are capture EIAs useful at diagnosing?

A

Congenital syphilis

1315
Q

Describe competitive EIAs of syphilis

A
  • treponemal antibody in the patient sample competes with an enzyme-labeled treponemal antibody conjugate for T. Pallidum antigens bound to microtiter plate wells
1316
Q

Describe CLIAs of syphilis

A
  • one-step sandwich assay
    —> higher sensitivity, faster performance, and more stable reagents
    —> patient sample is incubated with paramagnetic microparticles that have been coated with T. Pallidum anitgens linked to chemiluminescent derivative
1317
Q

describe PCR testing of syphilis

A
  • involves isolating and amplifying a specific sequence DNA
  • extremely sensitive, more than dark field microscopy
  • availability is limited
1318
Q

What is the main disadvantages of nontreponemal testing?

A

Subject to false positive

1319
Q

What should a reactive nontreponemal test be confirmed by?

A

A more specific treponemal test

1320
Q

When should treponemal test for syphilis by performed?

A
  • confirmatory test to distinguish false positive from true positive nontreponemal results
  • establish diagnosis in late latent or late syphilis
1321
Q

Why are nontreponemal test for syphilis performed?

A
  • screening for syphilis
  • monitoring the progress of the disease
  • determining the outcome of treatment
1322
Q

Describe traditional testing algorithm for syphilis

A

1) initial screening with nontreponemal (VDRL or RPR)
2) if reactive, perform treponemal test for confirmation (TP-PA or FTS-ABS). In nonreactive no further testing needed
3) if reactive, patient is positive for syphilis. If non reactive, previous result was false

1323
Q

Describe reverse sequence algorithm of syphilis

A
  • testing order reversed
    1) samples are screened by automated treponemal immunoassay
    2) if reactive, results are confirmed by nontreponemal testing
1324
Q

What are the advantages of reverse algorithm over traditional?

A

1) cost
2) automation
3) saves time
4) less errors
5) can detect more early, late and treated syphilis cases
6) high sensitivity

1325
Q

What should be performed for congenital syphilis?

A
  • perform nontreponemal tests on mother, and infant at birth
  • titers decline with successful treatment
1326
Q

What should be performed for congenital syphilis?

A
  • perform nontreponemal tests on mother, and infant at birth
  • IgM-specific treponemal assays to confirm
1327
Q

What should be performed if neurosyphilis is suspected?

A
  • perform CSF VDRL or ELISA on cerebrospinal fluid
1328
Q

What should be performed if neurpsyphilis is suspected?

A
  • perform CSF VDRL and ELISA on cerebrospinal fluid
1329
Q

What are the tests in order of reverse algorithm sequence?

A

1) perform EIA or CIA
2) if positive, perform quantitative RPR (or other nontreponemal tests)
3) if RPR positive, indicates past or present syphilis infection. If negative, perform TP-PA
4) If TP-PA is positive, indicates past or present inflection of syphilis

1330
Q

What test is recommended to confirm congenital syphilis?

A

Western blot

1331
Q

Describe Lyme disease

A
  • caused by the spirochete bacterium Borrelia burdorferi
  • multisystem illness involving the skin, nervous system, heart and joints
  • transmitted by ixodes ticks
  • main reservoirs white-footed mouse
  • most common vector-borne disease in the US
1332
Q

Describe the clinical manifestations of Lyme disease

A

Stage 1: localized rash and hallmark erythema migraines
Stage 2: early dissemination
Stage 3: later dissemination with arthritis, heart and nervous system involvement

1333
Q

What is the most prevalent neurological signs of Lyme disease?

A

Facial palsy

1334
Q

What are the types of Ixodes ticks?

A

1) ixodes scapularis —> northeast and Midwest US
2) Ixodes pacificus —> in the west
3) ixodes ricinus —> Europe
4) ixodes persulcatus —> Asia

1335
Q

Describe erythema migrans

A
  • localized rash stage
  • hallmark rash of early infection of Lyme disease
  • appears 2-14 days after bite
  • begins as a small red papule where bite occurred, then radio expands to form a large ring-like erythema and central area exhibits partial clearing
  • must be at east 5 cm in diameter to diagnose with Lyme disease
1336
Q

Describe the early dissemination stage

A
  • occurs via bloodstream in days to weeks following rush
  • skin, nervous system, heart, and joints involved
  • usually migratory pain occur in joints, tendons, muscles, and bones
1337
Q

Describe late dissemination stage of Lyme disease

A
  • can develop in untreated patients months to years after acquiring infection
  • major manifestation —> arthritis, peripheral neuropathy and encephalomyelitis
  • some develop fatigue, concentration and short term memory problems and muscoskeletal pains
1338
Q

Describe antibody response to Lyme disease

A
  • may not be detectable for 3-6 weeks
  • IgM occurs first, followed by IgG
  • IgG peaks at 3rd and 4th weeks of infection
1339
Q

Using the standard testing algorithm. What test (in order) are ran to diagnose Lyme disease?

A

1) screened with sensitive ELISA or IFA
2) it positive or borderline, Webstern blot test for IgM is performed to confirm results of patient with symptoms present less than 30 days. Western blot test for IgG is patient had symptoms that exceeded 30 days

1340
Q

What are characteristics of standard testing algorithm of Lyme disease?

A
  • costly
  • western blot can be difficult to interpret
  • highly specific
1341
Q

What are tests (in order) performed in the modified two-tiered algorithm?

A

1) symptomatic patients are tested with sensitive that uses purified borrelia peptide antigens
2) if positive or borderline, test with different ELISA method for confirmation

1342
Q

What are characteristics of the modified two-tiered algorithm?

A
  • easy to perform and interpret
  • comparable specificity to standard method
  • increased sensitivity to detecting early Lyme disease
1343
Q

Describe immunofluorence assay (IFA) of Lyme disease

A
  • first test used to evaluate the antibody response
    1) doubling dilutions of patients seem are incubated with commercially prepared slides coated with antigen from whole or processed borrelia spirochetes
    2) following wash step, anti-human globulin with fluorescent tag is added and reacts with any specific antibody bound to spirochetes on slide
    3) after second wash, slide is viewed under a fluorescent microscope
  • test considered positive only if titer of 1:256 or higher is obtained
1344
Q

What can produce false positives in IFA?

A
  • closely related organisms can cross react
  • autoimmune connective tissue diseases such as RA and SLE
  • fluorescent light pattern is very subjective
1345
Q

Describe EIA testing for Lyme disease

A
  • ELISA is inexpensive and yields timely results
  • automation
  • high volume testing
  • less sensitive in early Lyme disease
1346
Q

Describe western blot for Lyme disease

A
  • also called immunoblotting
  • very complex
  • patient serum is incubated with nitrocellulose membrane containing electrophoresed B. Burgdorferi antigens
  • positive IgM results —> 2-3 characteristic bands
  • positive IgG results —> 5-10 characteristic bands
1347
Q

Describe PCR testing of Lyme disease

A
  • involved probes for target DNA that is present only B. Borrellia
  • involves extraction of DNA, followed by amplification using specific primers, DNA polymerase and nucleotides
  • highly specifc
  • skin biopsy is not justifiable
  • used to diagnose difficult neurological and arthritic cases
1348
Q

Describe leptospirosis

A
  • caused by leptospirosis species
  • zoonotic infection
  • humans are exposed by mucous membrane contact with urine-contaminated water
  • causes febrile episode that can progress to severe disease involving renal, liver, pulmonary, and CNS
  • symptoms
    —> febrile episodes of headache
    —> myalgia
    —> nausea
    —> vomiting
    —> diarrhea
    —> renal involvement
    —> hepatic involvement
    —> pulmonary involvement
    —> CNS involvement
1349
Q

Whar are characteristics of the leptospira species?

A
  • Thin, flexible, and lightly coiled spirochetes
  • pointed ends with hook-like shape
1350
Q

Describe the stages of leptospirosis

A
  • incubation period —> 2-30 days
  • usually manifest 5-14 days after exposure
  • severe cases can lead to Weil’s disease
1351
Q

What are the laboratory tests performed for leptospirpsis?

A
  • specimens —> blood or serum for first week after, urine or serum
  • IgM screening by ELISA, immunoDOT, and LFA
  • MAT is gold standard for confirmation
1352
Q

What is the purpose of the Reiter treponemes in FTA-ABS?

A
  • prevents cross-reactivity with antibody with antibody to non-pathogenic treponemes
1353
Q

What test is recommended for testing cerebrospinal fluid for detection of neurosyphilis?

A

VDRL

1354
Q

What are the advantages of direct fluorescent antibody testing to T. Pallidum?

A
  • Reading is less subjective
  • monoclonal antibody makes the reaction very specific
  • slides can be prepared for later reading
1355
Q

What syphilis test detects specific treponemal antibodies?

A

FTA-ABS

1356
Q

What can be used to distinguish between IgG and IgM antibodies in syphilis?

A

Treponemal EIAs

1357
Q

What is toxic shock syndrome associated with?

A

Overproduction of cytokines

1358
Q

What are ANTIGEN noninvasive tests used to detect H. Pylori?

A
  • urea breath test
  • ELISA
  • LFA
  • DNA
1359
Q

What antibody is associated with M. Pneumonia?

A

Anti-I (cold agglutinins)

1360
Q

What disease is associated with anti-i?

A

Hemolytic anemia

1361
Q

What is as rapid urease method for H. Pylori?

A

CLOtest

1362
Q

What are virsuses?

A

Submicroscopic pathogens whose size is measured in nanometers

1363
Q

Describe the basic structure of a virus

A
  • a core of DNA or RNA packaged into a protien coat (capsid)
  • in some viruses, the capsid is surrounded by an outer envelope of glycolipids and proteins derived from the host-cell membrane
  • obligate, intracellular
1364
Q

What are the steps of the basic virus lifecycle?

A

1) attachment of their virus to a receptor on the host cell surface
2) penetration of virus into host cell through endocytosis
3) degradation of the viral capsid and subsequent release of viral nucleic acid
4) transcription to produce additional viral nucleic acid
5) translation of viral nucleic acid to produce viral proteins
6) assembly of the viral components to produce intact virions
7) budding off the host-cell membrane or host-cell lysis results
8) release of viral progeny

1365
Q

What are the defenses type against a virus?

A
  • innate defenses
  • humoral antibody responses
  • cell- mediated immunity
1366
Q

Describe innate defense against viruses

A
  • first like of protection
  • skin and mucous membrane barriers
  • recognition of PAMPs on virus- infected host cells
  • interferons alpha and beta
1367
Q

What occurs if the initial barrier of innate defense does not work?

A

Other innate defense are activated when cells of innate immunity recognizes PAMPS of surface or within virus infected host cells

1368
Q

What are other innate defenses against viruses? (Besides skin barrier)

A
  • viral cells are stimulated to produce IFN-alpha and IFN-beta after recognizing viral replication by inducing transcription of several gene that code for proteins with anti-viral activity
  • they also enhance activity of NK cells
1369
Q

How do NK cells fight against viruses?

A
  • bind to virus-infected cells and release proteins such as a perforin and granzymes, causing cells to die and release viruses and are now accesible to antibody molecules
1370
Q

What plays a key role in preventing the spread of viral infection through neutralization?

A

Antibodies

1371
Q

Describe antibodies role in preventing the spread of viral infection through neutralization

A
  • involves production of antibodies that are specific for a component of the virus that bind to the virus, the prevent it from attaching to and penetrating the host cell. IgA plays a large role in this.
  • IgG and IgM can bind to viruses in blood stream
  • IgG and IgM activate complement
  • IgG also promote phagocytosis of viruses and promote destruction of viruses through ADCC
  • IgM also viral particles by agglutinating them
1372
Q

What does intracellular viruses require?

A

Cell-mediated immunity

1373
Q

What cells have key roles in cell mediated immunity?

A
  • Th1 cells
  • cytotoxic T cells
1374
Q

Describe Th1 cells action that occur in cell mediated immunity

A
  • produce IFN-gamma, which induces an anti-viral state within the virus-infected cells
  • produce IL-2, which assists in development of effector cytotoxic T cells
1375
Q

Describe cytotoxic T cells actions that occur in cell mediated immunity

A
  • CD8+ cytotoxic T cells become programmed to expand in number and attack the virus infected cells
    CD8 is a co-receptor of T-cell receptor on cytotoxic T cells that must bind to viral antigen complexed with MHC class I on infected cells surfaces
1376
Q

What occurs after cytotoxic T cells bind to viral antigen complexed with MHC class I?

A
  • stimulate granules in the cytotoxic T cells to release perforin and granzymes entering the pores
1377
Q

What occurs once granzymes enter the viral infected cell?

A
  • activate apoptosis in the host cell, interrupting the viral- replication cycle and resulting release of assembled infectious virions
  • the free virions can then be bound by antibodies
1378
Q

Describe humoral antibody responses

A
  • antibodies attack free virus particles
  • viral neutralization, opsonization, C’ fixation, and ADCC
1379
Q

What are viral escape mechanisms? And examples?

A

1) mutations results in production in new viral antigen (influenza viruses undergo frequent genetic changes)
2) viruses block action of immune system components (HSV can bind C3b)
3) suppression of the immune response (CMV reduces MHC I)
4) immune function altered (EBV stimulates polyclonal B-cell activation)
5) latent state is established (VZV remains latent in nerve cells)

1380
Q

What laboratory test are ran to detect a viral infection?

A
  • serological tests
  • distinguish between current and past infection
  • antibody titers used to monitor course of infection
  • assess immune status
  • molecular
  • detect active infection
  • quantitative tests —> guide antiviral therapy
1381
Q

How are current and past infections detected?

A
  • IgM (+) and IgG (+/-) —> Current or recent infection. (Congenital)
  • IgM (-) and IgG (+) —> past infection
1382
Q

What does the presence of virus-specific IgG indicate?

A

Immunity to virus

1383
Q

What is the hepatitis virus?

A
  • hepatitis is the inflammation of the liver
  • caused by:
    —> radiation
    —> chemical toxins
    —> secondly to other disease
    —> cirrhosis
    —> drugs
    —> hypothermia
    —> bacteria
    —> fungi
    —> parasites
1384
Q

What are the hepatitis are transmitted by fecal-oral route?

A

Hep A (HAV)
Hep E (HEV)

1385
Q

What types of hepatitis are transmitted through parenteral route?

A

Hep B (HBV)
Hep D (HDV)
Hep C (HCV)

1386
Q

What are indicators of hepatitis?

A
  • flu-like symptoms early on
  • pain in upper right quadrant of abdomen
  • hepatomegaly and liver tenderness with profession
  • jaundice
  • dark urine
  • light feces
  • Elevated bilirubin and liver enzymes (ALT)
1387
Q

Describe hepatitis A

A
  • nonenveloped, single strand RNA virus
  • belongs to hepatovinis genus or picornarviridae family
  • acute hepatitis in majority of adults
  • infections in children are usually asymptomatic
  • formalin- activated vaccine
  • HAV immune globulin may be recommended for unimmunized persons exposed to virus
1388
Q

How is Hepatitis A transmitted?

A
  • fecal-oral route
  • close person to person contact
  • ingestion of contaminated food food or water
1389
Q

What is critical for establishing a diagnosis of hepatitis A?

A
  • serological tests antibody
1390
Q

What sheds from the feces from someone who has hepatitis A?

A
  • HAV antigen
1391
Q

What are hepatitis A most commonly detected by?

A

EIAs
Chemiluminescent microparticle immunoassays

1392
Q

When is IgM anti-HAV detectable in a patient with hepatitis A?

A

Onset of of clinical symptoms and declines to undetectable levels by 6 months

1393
Q

Describe tests for total HAV antibodies

A
  • detect IgM but predominantly IgG, which persists for life
1394
Q

Although IgM anti-HAV is primary marker to detect acute hepatitis. What do you have to look out for?

A

False-negative results due to early phase of infection

1395
Q

What is the most common format of these molecular methods of HAV?

A
  • RT-PCR
  • used to test samples of food or water suspected of transmitting virus
1396
Q

Describe Hepatitis E

A
  • nonenveloped, single stranded RNA virus with four genotypes
  • HEV-1 and HEV-2 are transmitted primarily through ingestion of feces-contaminated drinking water
  • HEV-3 and HEV-4 are transmitted mainly by consumption of infected pork
  • most asymptomatic or self-limiting infections
  • can detect HEV RNA in blood or stool during acute infection
  • belong to hepevirus; in the family hepediridae
1397
Q

What indicates an acute infection of Hep E?

A

IgM anti-HEV

1398
Q

What indicates past exposures of Hep E?

A

IgG anti-HEV

1399
Q

What does diagnosis of HEV?

A
  • serology to detect antibodies to the virus (EIAs)
  • molecular methods to HEV nucleic acids
1400
Q

What is used to detect later stages, past exposures, and identify seroprevalence of the infection in HEV?

A

Immunoassays for IgG anti-HEV

1401
Q

What is the gold standard for diagnosis of acute HEV infections?

A
  • qPCR, quantization of HEV nucleic acid
1402
Q

What testing can be done for immunocompromised patients that are suspected of having HEV?

A
  • molecular testing for HEV RNA (qPCR)
  • LAMP
1403
Q

Describe Hepatitis B

A
  • DNA virus with 8 genotypes (A-H)
  • belongs to hepadnaviridae family
  • acute infection —> symptoms increase with age
  • chronic infection —> 6 months or more, occurs in 90% of infected infants and 10% of infected adults
  • preventable with immunization
  • hepatitis B immune globulin (HBIG) may be given to unimmunized persons exposed to HBV
1404
Q

What does chronic hepatitis B increase the risk of?

A

Liver cirrhosis
Hepatocellular carcinoma

1405
Q

Describe hepatitis B surface antigen (HBsAg)

A
  • first HBV marker to appear (2-10 weeks after exposure)
  • protein on outer envelope of virus-like particles in blood
  • marker for active HBV infection
  • component of hepatitis B vaccine
1406
Q

Describe hepatitis B antigen (HBeg)

A
  • protein in core of HBV
  • marker of active viral replication
  • indicates high degree of infectivity
1407
Q

What are serological markers of HBV antigens?

A

HBsAg
HBeg

1408
Q

What are the antibodies markers for HBV?

A

Anti-HBc
Anti-HBe
Anti-HBs

1409
Q

Describe anti-HBc of HBV

A
  • directed against HEB core antigen
  • IgM anti-HBc consists mainly of IgG and can indicate a current or past-infection
1410
Q

What is a core window?

A
  • period when neither HBsAb can be detected in the serum of the patient only the anti-HBc
1411
Q

Describe anti- HBe of HBV

A
  • directed against HBeAg
  • indicates recovery from hepatitis B
1412
Q

Describe anti-HBs of HBV

A
  • directed against HBsAg
  • indicates immunity to hepatitis B
1413
Q

How is HBV transmitted?

A

Transmitted through parenteral or perinatal routes:
- sexual contact
- IV drug use
- during birth process

1414
Q

Describe chronic hepatitis B

A
  • Majority of infants, 10% of adults, one-third children
  • most likely in immunocompromised patients
  • results in liver inflammation and damage
1415
Q

Describe structure of the HBV

A
  • nucleocapsid core surrounded by outer envelope of lipoprotein
  • core of virus contains circular partially double-stranded DNA
1416
Q

How are serological markers of HBV used?

A
  • differential diagnosis of HBV
  • monitoring course of infection
  • assessing immunity to virus
  • screening blood products infectivity
1417
Q

Why is HBsAg important marker of HBV?

A
  • indicator of active infection
  • monitoring course of infection and progression
  • screening of donor blood
1418
Q

How are serological markers for hepatitis most commonly detected? Describe

A

Commercial immunoassays (EIAs and CLIA)
- typically automated
- excellent specificity and sensitivity
- false-negatives and false-positives occur
- positive results should be verified by repeat testing following by confirmation with additional assay (HBsAg neutralization test or molecular methods that detects that detect HBV DNA

1419
Q

What is the method of choice of to quantify HBV DNA?

A
  • qPCR
1420
Q

What are molecular methods used to detect HBV DNA?

A
  • PCR
  • qPCR
  • branched DNA signal amplification
1421
Q

What is considered a successful treatment for HBV?

A

1-log10 reduction in HBV DNA level`

1422
Q

Describe hepatitis D

A
  • RNA virus that requires presence of HBV with 3 genotypes
  • superinfection of chronic HBV carriers
    —> chronic liver disease with accelerated progression to cirrhosis and liver failure
  • co-infection with HBV —> usually results in acute, self limited hepatitis
1423
Q

In HDV, what is the marker for active viral replication?

A

HDV RNA

1424
Q

What do co-infections of HDV test results look like?

A

Positive for ani-HDV and IgM anti- HBc

1425
Q

what do chronic cases of HDV results look like?

A

Positive for anti-HDV and IgG anti-HBC

1426
Q

How is HDV transmitted?

A

Through parenteral or perinatal routes

1427
Q

What markers appearance would make cause to for HDV?

A
  • HBsAg
  • involves detection
  • antibodies and HDV RNA
1428
Q

What is testing used to distiniguish HBV and HDV from acute infection or superinfection?

A

Serology testing

1429
Q

What does the presence of IgG anti-HDV indicate?

A

Acute, chronic, or past infection

1430
Q

What molecular methods are used to detect HDV RNA?

A
  • RT-PCR
  • also provides quantitative results that can be used to monitor the response of patients to antiviral therapy
1431
Q

Describe hepatitis C

A
  • enveloped, single-stranded RNA virus with 7 genotypes
  • belongs to flavivirdae family and genus hepacivirus
  • most infections are asymptomatic at first but develop into chronic liver
  • genotyping is used to determine best therapy
1432
Q

How is HCV transmitted?

A
  • exposure to contanimated blood, sexual contact, and perinatal exposure
1433
Q

What is detection of anti-HCV IgG used for?

A

Screening and diagnosis

1434
Q

What is qualitative HCV RNA used for?

A

Confirmation

1435
Q

What are quantitative molecular tests used for?

A

Monitor viral load during antiviral therapy

1436
Q

What is the most common blood born infection in the US?

A

HCV

1437
Q

What is the HCV genotype that is most prominent in the united states?

A

Genotype 1

1438
Q

Why is difficult to create a vaccine for HCV?

A
  • the variability of HCV
  • its ability to undergo rapid mutations within its hosts
1439
Q

What does chronic HCV lead to?

A
  • cirrhosis of liver
  • increased risk of Hepatocellular carcinoma
  • rheumatoid conditions
  • glomerulonephritis
  • vasculitis
  • neuropathy
  • dermatological systems
1440
Q

What is anti-HCV most commonly detected by?

A
  • EIA and CLIAs that use recombinant and synthetic antigens
  • rapid immunoblot is alternative
1441
Q

When do antibodies become detectable in HCV?

A

8-10 weeks after exposure and remain positive for a lifetime

1442
Q

What could case a false positive result in serological testing of HCV?

A
  • because of cross-reactivity in persons with other viral infections or autoimmune disorders
1443
Q

What test is recommended for HCV RNA confirmation?

A

Nucleic acid testing

1444
Q

What do qualitative tests distinguish between in HCV?

A
  • presence or absence of HCV RNA
1445
Q

What are qualitative molecular assays used for HCV?

A
  • to confirm infection in HCV antibody positive patients
  • detect infection in antibody negative patients who are suspected of having HCV
1446
Q

What are the types of quantitative molecular assays from HCV? What are they used for?

A
  • RT-PCR
  • qPCR
  • bDNA application
    —> monitor amount of HCV RNA, or viral load, carried by patents before, during and after antiviral therapy in chronic patients
1447
Q

What is the goal of antiviral therapy of HCV?

A

When patient continuously tests negative for HCV RNA 12 or 24 weeks after therapy is completed

1448
Q

Describe genotyping of HCV

A
  • to determine exact genotype and subtype of viruses responsible for infection
  • ran on all HCV positive patient before antiviral therapy
  • important to identify genotype because they vary in their response to different antiviral drugs
1449
Q

What can genotyping for HCV be performed by?

A
  • PCR amplification and sequencing of target gene
  • PCR followed by identification of the target gene with genotype-specific probes
  • qPCR
1450
Q

What is the reference method of genotyping for HCV? Why?

A
  • PCR/direct sequencing (Sanger sequencing)
  • because it provides precise information about genomic variability of the virus during course of infection
1451
Q

Describe herpes virus

A
  • large complex DNA viruses that are surrounded by a protein capsid, an amorphous tegument, and an outer envelope
  • herpesviridae family —> includes 8 viruses
1452
Q

Describe Epstein-virus (EBV)

A
  • DNA herpes virs most commonly transmitted by intimate contact with salivary secretions
  • begins in oropharynx in B cells and epithelial cells and spreads through lymphoreticular system
1453
Q

What are diseases caused by Epstein-Barr virus?

A
  • IM
  • lymphoproliferative disorders
  • certain malignancies (burkitt lymphoma)
1454
Q

How can Epstein-Barr virus be transmitted?

A
  • salivary sectretions (most frequent)
  • blood transfusions
  • bone marrow
  • solid-organ transplant
  • sexual contact
  • perinatal exposure
1455
Q

What cells are infected in the oropharynx in a patient with EBV?

A

Epithelial cells
B cells

1456
Q

How does EBV enter the B cells?

A

By binding to surface CD21

1457
Q

What are early antigens (EAs) of EBV?

A
  • antigen produced during the initial stages of viral replication in the lyric cycle
  • can be classified into two groups: EA-D and EA-R
1458
Q

Describe EA-D

A

In EBV, early antigens that have a diffuse distribution in nucleus and cytoplasm

1459
Q

Describe EA-R

A

In EBV, early antigens restricted to cytoplasm only

1460
Q

Describe latent antigens of EBV

A
  • appear during the period of the lyric cycle following viral DNA synthesis
  • includes the viral capsid antigens in the protein capsid and antigens in the protein capsid and the membrane capsid in viral envelope
1461
Q

What antigens appear during the latent stage of EBV?

A
  • EBNA-1 through EBNA-6
  • LMP-1
  • LMP-2A
  • LMP-2B
1462
Q

What are the classical symptoms of IM?

A
  • fever
  • lymphodenopathypy
  • sore throat
  • fatigue
  • lasts 2-4 weeks
1463
Q

What are the laboratory findings of IM?

A
  • absolute lymphocytes
  • 20% or more atypical lymphocytes
  • heterophile antbodies —> IgM
1464
Q

What produces can be performed for detection of IM heterophil antibodies?

A
  • Monospot
  • paul-Bunnell test
  • rapid agglutination tests
  • immunochromatographic assays using purified bovine RBC extract as antigen
1465
Q

What can AID in diagnosis of IM?

A
  • testing for EBV- specific antibodies
  • helpful is patient that test negative for heterophil antibodies or determine past exposure
  • detected by IFAs, blot techniques, ELISA, CLIA or flow cytometric microbead immunoassays
1466
Q

What with the gold standard of EBV serology methods?

A
  • IFAs
  • labs prefer to use EIAs or CLIA tests because the are automated and easier to interpret
1467
Q

What is the most useful marker for acute IM?

A

IgM antibody to the VC

1468
Q
A
  • presence of IgM anti-VCA and anti-EA-D
  • absence of anti-EBNA
1469
Q

What types of tests are best to detect EBV in immunocompromised patient?

A

Molecular tests

1470
Q

How is cytomegalovirus transmitted?

A
  • DNA herpes virus is transmitted through oral secretions, genital secretions, congenitally, or by transfusion/transplantation
1471
Q

Describe cytomegalovirus

A
  • healthy individuals may be asymptomatic or develop a mononucleosis-like syndrome
  • in immunocompromised persons, CMV can disseminate to lungs, liver, GI tracts, CNS, and eyes and cause life-threatening infections
  • healthy individuals cause congenital defects and decreased survival in infants
1472
Q

What are types of testing for CMV?

A
  • direct virus detection
  • serology
1473
Q

Describe direct virus detection of CMV

A
    • viral culture
  • sample placed with a cell type that the virus being tested for can infect
  • if the cells show changes, known as cytopathic effects, then the culture is positive
  • ID of CMV antigens
  • molecular tests for CMV DNA
1474
Q

Describe serology methods for CMV

A
  • used to screen blood and organ donors; pregnant women
  • presence of IgG anti-CMV indicates infection
  • low avidity antibodies indicate recent infection
  • most beneficial in determining past exposures
  • usually semi-or fully automated EIAs
1475
Q

What is the most common cause of congenital infections?

A

CMV

1476
Q

What indicates CMV?

A

Characteristics cytopathtic effects (CPEs) that produce enlarged, rounded, refractile cells

1477
Q

Describe the CMV antigemenia assay

A
  • uses immunocytochemical or immunofluorescent staining to detect the CMV lower-matrix protein pp65 in infected leukocytes from peripheral blood or cerebral spinal fluid
  • 2-4 hours to perform
1478
Q

Describe VZV

A
  • DNA herpes virus
  • cause of: varicella (chicken pox) and here’s zoster (shingles)
  • preventable by vaccine
1479
Q

How is VZV transmitted?

A
  • by inhalation of infected respiratory secretions or aerosols from skin lesions
1480
Q

Describe varicella

A
  • highly contagious illness
  • blister-like rash
  • intense itching
  • fever
1481
Q

What are the activities of VZV in the primary infection?

A
  • thought to travel from the skin lesions and blood to sensory neurons, where it deposits its DNA and establishes a lifelong latent state in the dorsal root, autonomic and cranial ganglia
1482
Q

Describe herpes zoster

A
  • painful vesicular rash caused by the virus moves down the sensory nerve to the dermatome supplied by that nerve
1483
Q

What is the definitive diagnosis of VZV?

A
  • identifying VZV in skin lesions, vesicular fluids or tissue
  • used for atypical cases
1484
Q

What is the method of choice to detect VZV DNA?

A

QPCR

1485
Q

How is serology testing useful with VZV?

A
  • determines immunity to VZV in health-care worker, pregnant women, and organ transplant candidates
1486
Q

What cells can be detected in VZV patients?

A

Tzanck cells

1487
Q

What is quantitative PCR useful with VZV?

A

Monitoring response of immunocompromised patients to antiviral drugs

1488
Q

Describe serology testing for VZV

A
  • most of them detect total VZV antibody, which consists of primarily of IgG
  • most reliable and sensitive method —> FAMA (reference method)
  • most commonly used—> ELISA
1489
Q

Describe rubella virus

A
  • enveloped, single-stranded RNA virus
  • cause of German measles
  • can be prevented with immuzation
  • genus rubivirus and belongs to family Togaviridae
  • after incubation, virus replicates in upper respiratory tract and cervical lymph nodes, then travels through blood stream
1490
Q

How is rubella transmitted?

A
  • respiratory droplet
  • across the placenta
1491
Q

What can rubella virus cause?

A
  • deafness
  • eye defects
  • cardiac abnormalities
  • mental retardation
  • motor disabilities
  • miscarriage
    Stillbirth in infants
1492
Q

What are the symptoms of rubella virus?

A
  • erythematous (macropapular rash). Appears first on face, then spreads to the trunk and extremities
  • low grade fever
  • malaise
  • swollen glands
  • upper respiratory infection lasting 1-5 days
  • 50% infected with Rubella are asymptomatic
  • arthritis
1493
Q

What populations have severe consequences with rubella virus? Why?

A
  • pregnant women, especially in first trimester
  • may cause miscarriage, stillbirth, or congenital rubella syndrome
1494
Q

What can occur if infants are born with congenital rubella syndrome (CRS)?

A
  • many abnormalities
    —> deafness
    —> eye defects
    —> cardiac abnormalities
    —> mental retardation
    —> liver and spleen damage
    —> motor disabilities
1495
Q

What type of methods are ran for rubella virus?

A
  • serology
  • viral culture
  • molecular methods
1496
Q

Describe serology testing of rubella virus

A
  • presence of IgG is used to screen for immunity
  • congenital infection is indicated by rubella-specific IgM or fourfold rise in IgG
  • low avidity antibodies indicate recent infection
1497
Q

Describe viral culture of the rubella virus?

A
  • diagnosis can be obtained this way
  • viral growth is slow
  • may not produce characteristic CPE
1498
Q

If CPE is absent in the viral culture of rubella. What should be done next?

A
  • viral nucleic acid can be identified by RT-PCR or viral proteins can be detected with IFA or EIA
1499
Q

What is the most widely used test for rubella virus?

A
  • molecular methods —> RT-PCR
  • used to detect rubella RNA
1500
Q

What are the most common methods used to confirm rubella?

A
  • serology tests (rapid and low cost)
  • detect rubella antibodies
  • ELISA- most widely used
  • hemagglutination inhibiton (HI)
  • latex agglutination
  • immunoassays
1501
Q

Describe ELISA testing of rubella

A
  • specific solid phase capture ELISAs can be used to detect IgM rubella antibodies
1502
Q

What assays can simultaneously detects measles, mumps, rubella, and varicella?

A
  • automated chemiluminescence assays
  • multiplex bead immunoassay
1503
Q

How is a primary rubella infection indicated?

A
  • presence of rubella-specific IgM antibodies
    OR
  • four-fold or greater rise in rubella specific IgG antibody titers collected at least 10-14 days apart
1504
Q

What can cause a false negative in rubella?

A

Obtained sample too earlier

1505
Q

Describe test ran to confirm positive IgM results of rubella

A
  • EIA —> measures avidity of rubella IgG antibodies, which helps distinguish between recent and past infections
  • low antibody avidity —> recent infection
  • high antibody avidity —> past infections
1506
Q

Describe laboratory testing of congenital rubella infection

A
  • begins with serological evaluation of the mothers antibodies and measurement of rubella-specific IgM antibodies in fetal blood, cord blood, or neonatal serum
    -any positive IgM results should be confirmed by viral culture, RT-PCR amplification, or demonstration of persistently high titers of rubella IgG antibodies after 3-6 months of age
1507
Q

Describe rubeola

A
  • RNA virus single-stranded RNA virus
  • genus morbillivirus in the paramyxoviridae family
  • after initial infection of the epithelial cells in upper respiratory tract, rubeola virus is disseminated through the blood to multiple sites in body, such as the skin, lymph nodes and liver
  • preventable by immunization
  • diagnosis is usually based on clinical presentation and confirmed by serology
1508
Q

How is rubeola transmitted?

A

Through respiratory droplets

1509
Q

What does rubeola cause?

A
  • measles
  • subacute sclerosing pancephalitis (SSPE)
1510
Q

What are the symptoms of rubeola?

A
  • fever
  • cough
  • runny nose
  • conjunctivitis
  • koplik spots appear on mucous membrane of inner cheeks or lips
1511
Q

What is the rubeola virus characterized by?

A
  • erythamatous (maculopapular eruption) that begins at hairline, then spreads to the face and neck and gradually moves down to trunk, arms, hands, legs, and feet
1512
Q

What complications can measles result in?

A
  • Diarrhea
  • otitis media
  • croup
  • bronchitis
  • pneumonia
  • encephalitis
1513
Q

What is SSPE associated with?

A

Extremely high titers of rubeola antibodies

1514
Q

What is preferred method to detecting IgM antibodies in rubeola?

A
  • IgM capture ELISA method
  • 3-4 days after symptoms
1515
Q

What is the preferred method of detect IgG antibodies of rubeola?

A
  • ELISA
  • detectable 7-10 days after symptoms and persists for life
  • IgG antibodies indicate immunity
1516
Q

When are molecular methods used to detect rubeola RNA?

A
  • in which serological tests are inconclusive or inconsistent
  • can be used to genotype the virus
1517
Q

What is the preferred molecular method for detecting rubeola?

A

RT-PCR

1518
Q

Describe mumps virus

A
  • Single stranded RNA virus
  • paramyxoviridae family, genus ribuavirus
  • most common clinical manifestations is parotitis
  • replicates initially in nasopharyngeal and regional lymph nodes
  • preventable by immunization
  • diagnosis is usually based on clinical presentation
  • confirmation is done by culture or RT-PCR
1519
Q

How is mumps transmitted?

A

Respiratory droplets, saliva formites

1520
Q

How does the mumps virus function?

A
  • virus spreads from blood to various tissues:
    —> meninges of brain
    —> salivary glands
    —> pancreas
    —> testes
    —> ovaries
  • produces inflammation at site
1521
Q

What is parotitis?

A
  • inflammation of the parotid glands
  • occurs in 30-40% of patients with mumps
1522
Q

What is the gold stranded for detecting the mumps virus?

A
  • culture
1523
Q

What is the primary diagnostic test for mumps? Why?

A
  • RT-PCR
  • because it is more sensitive than serology
1524
Q

Describe the serological testing of mumps

A
  • provides simple means of confirming mumps diagnosis but has some important limitations
  • EIAs and IFA mostly only measure IgG mumps virus antibodies
  • most commonly used —> ELISA
  • use of solid-phase IgM capture assays reduces incidence of false positive because of rheumatoid factor C
1525
Q

What is a current or recent infection of mumps indicated by?

A
  • presence of mumps-specific IgM antibody in a single serum sample or by at least a four-fold rise in specific IgG antibody between two specimens collected during the acute and convalescent phases of illness
1526
Q

Describe human T-cell lymphoytropic virus type I and type II (HTLV-1 & HTLV-2)

A
  • closely related retroviruses that preferentially infect T- lymphocytes (usually CD4+)
  • serological tests for antibodies to HTLV-I/II are used to diagnose infections and screen blood donors
  • have RNA as their nucleic acid and enzyme reverse transcriptase
  • has 3 structural genes: gag, pol, and env and region called pX
  • ELISA and CLIA are used to screen
    -western blot or LIA are used for confirmation of positive results
1527
Q

What does HTLV-1 cause?

A
  • adult T-cell leukemia/lymphoma(ATLL) and HAM/TSP
1528
Q

How are HTLV types I and II transmitted?

A
  • mainly blood borne
  • sexual conduct
  • perinatal (especially breastfeeding)
1529
Q

What is “gag”?

A

Codes for viral core proteins

1530
Q

What is “pol”?

A

Codes for viral enzymes

1531
Q

What is “env”?

A

Encodes proteins in viral envelope

1532
Q

What is pX?

A

Encodes several regulatory proteins, including tax and Rex

1533
Q

What is reverse transcriptase function of HTLV?

A
  • to transcribe the viral RNA into DNA
1534
Q

What are the 4 subtypes of ATLL? What are they characterized by?

A
  • acute
  • T-cell non-Hodgkins lymphoma
  • chronic
  • smoldering
  • a monoclonal proliferation of mature T cells that express the surface markers CD3, CD4 and CD25
1535
Q

How is HAM/TSP characterized?

A
  • slowly progressive weakness and stillness of legs
  • back pain
  • urinary incontinence
1536
Q

What has been associated with HTLV-1?

A
  • variety of autoimmune and inflammatory disorders
  • uveitis (intraocular inflammation of the eyes)
  • infective dermatitis
  • myositis (inflammation of the muscles)
  • arthropathy (inflammation of the joints)
1537
Q

When do HTLV antibodies develop?`

A
  • 30-90 days after exposure to the virus and persists for life
1538
Q

When is a sample considered positive for HTLV-1 performed with western blot?

A
  • if visible bands are produced for one of the env proteins (either gp46 or gp62/68) AND one of the gag proteins (either p19, p24, or p53)
1539
Q

Describe PCR methods for HTLV testing

A
  • detects HTLV type I and II DNA
  • can be used to monitor the proviral load in patients during therapy
1540
Q

Describe toxoplasmosis

A
  • found in feces of house cats and rodents (other mammals)
  • transmitted:
    —> accidental ingestion of oocyst (fecal contamination of meat, raw milk,etc)
    —> transplacental transmission
1541
Q

How does one individual prevent congenital toxoplamosis?

A
  • avoid touching mucous membranes of mouth and eyes while handling raw meat
  • wash hands and kitchen surfaces after coming in out with raw meat
  • not allow flies and cockroaches access to food
  • avoid contact (or wear gloves) when handling cat litter boxes or gardening
1542
Q

What are signs and symptoms of toxoplasmosis?

A
  • usually asymptomatic organism that can multiply in any organ of body
  • similar to mono ( if symptoms are mono)
  • spontaneous recovery
1543
Q

Describe congenital infections of toxoplasmosis

A
  • of most concern
  • result in CNS malformation and mortality of the neonate
  • 75% have no symptoms at birth, disease is dormant, discovered only when neurological problems such as blindness occurs
1544
Q

What is the method of choice for a laboratory diagnosis for toxoplasmosis?

A

EIA

1545
Q

Describe TORCH testing

A
  • consists of tests for antibodies to four organisms that cause congenital infections transmitted from mother to fetus
    T- toxoplasmosis
    O- others
    R- rubella
    C- CMV
    H- herpes simplex virus
1546
Q

What are the “others” of TORCH?

A
  • syphilis
  • HBV
  • coxsackie virus
  • EBV
  • VZV
  • human parvovirus
1547
Q

What viruses affect the repiratory system?

A
  • adenovirus
  • influenza
  • mumps
  • measles
  • RSV
1548
Q

What virus affects the skin?

A

Arbovirus

1549
Q

What virus effects the HI tract?

A

Rotovirus

1550
Q

What is the role of CTLs in immune responses against viruses?

A

Destroy virus-infected host cells

1551
Q

A newborn suspected of having a congenital viral infection should be tested for virus-species antibodies of which class?

A

IgM

1552
Q

The serum of an individual who received all doses of the hepatitis B vaccines should contain what?

A

Anti-HBs

1553
Q

Heterophile antibodies are routinely detected by their reaction with what?

A

Bovine erythrocytes antigens

1554
Q

What is the method of choice for detecting VZV infection in immunocompromised hosts?

A

RT-PCR

1555
Q

Describe human immunodeficiency virus (HIV)

A
  • causes AIDS (acquired immunodeficiency syndrome)
  • two types: HIV-1 and HIV-2
1556
Q

Describe HIV-1 (will be referred as that for the rest of the set)

A
  • cause most HIV infections worldwide
  • 4 groups —> M, O, N, P
  • 9 subtypes in group M —> A, B, C, D, F, G, H, J, K
  • groups M is responsible for most cases
  • predominant subtype —> C
  • most prevalent is US —> B
  • belongs to genus lentivirinae of virus family retroviridae
  • decrease in cell production is a hallmark feature
1557
Q

Describe HIV-2

A
  • originated in west Africa
  • causes fever cases
  • less pathogenic
  • lower transmission rate
1558
Q

What are ways HIV can be transmitted?

A
  • sexual contact involving exchange of body fluids —> responsible for majority of cases
  • contact with blood or other body fluids
  • perinatal —> before , during and after (breast milk)
1559
Q

What body fluids are infective with HIV?

A
  • blood
  • semen and vaginal secretions
  • synovial, pleural, peritoneal and pericardial
  • CSF
  • breast milk
1560
Q

What are characteristics of HIV

A
  • retrovirus
  • contains two copies of ssRNA
  • reverse transcipatase transcribes the viral RNA into DNA
  • surrounded by a protein coat (capsid)
  • outer envelope of glycoproteins embedded in a lipid bilayer
1561
Q

Why is HIV considered a retrovirus?

A

Because it contains RNA as its nucleic acid and a unique enzyme called reverse transcriptase

1562
Q

Describe the HIV structure

A
  • spherical particle
  • includes 3 structural genes: gag, pol, and env
  • 100-200 nm in diameter
  • contain inner core with two copies of single stranded RNA surrounded by protein coat (capsid) and an outer envelope of glycoproteins embedded in lipid bilayer
  • glycoproteins are knob-like structures that are involved in binding the virus to host cells during infection
1563
Q

Briefly describe replication of HIV

A
  • attachment of hIV to host cell
  • main target: CD4 Th
  • coreceptor required
1564
Q

What are the steps of HIV replication?

A

1) binding- HIV binds to receptors on surface of CD4 cell
2) fusion- HIV envelope and CD4 cell membrane fuse and allow virus to enter cell
3) reverse transcription- inside cell, HIV uses and releases reverse transcriptase, which HIV RNA into HIV DNA. This allows HIV to enter nucleus of CD4 cell
4) integration- inside nucleus, HIV releases integrase. Uses intergrase to insert its viral DNA into the DNA of the CD4 cell
5) Replication- HIV uses machinery of CD4 cell to make long chains of HIV proteins. These proteins are building blocks for more HIV
6) Assembly- new HIV proteins and HIV RNA move to surface of cell and assemble into immature HIV
7) Budding- immature HIV pushes out of host CD4 cell surface. New HIV release proteases, which break down immature HIV, forming mature HIV

1565
Q

What is T-tropic or X4 strains of HIV?

A

They preferentially infect T-cells

1566
Q

What is M-tropic or R5 strains of HIV?

A

Can infect T-cells and macrophages

1567
Q

What chemokine receptor is required for HIV to enter T-lymphocytes?

A

CXCR4

1568
Q

What chemokine receptor is required for HIV to enter macrophages?

A

CCR5

1569
Q

When does HIV not virally replicate?

A

During the latent stage

1570
Q

What are the type of immune responses for HIV?

A
  • innate
  • humoral antibody production
  • cell-mediated immunity
1571
Q

describe the innate defenses against HIV

A
  • NK cells mediate cytolysis of HIV infected cells
  • dendritic cells stimulate release of cytokines that have antiviral effects
1572
Q

Describe humoral antibody production against HIV

A
  • antibodies are detected by 6 weeks after infection
  • antibodies produced later may prevent HIV from infecting host cells and participate in ADCC
1573
Q

Describe cell-mediated immunity against HIV

A
  • T cells produce cytokines with antiviral activity
  • CTLs destroy HIV-infected host cells
1574
Q

What are the HIV escape mechanisms?

A

1) genetic mutations rapidly occur, generating new viral mutants with altered antigens
2) down regulates expression of MHC-1 molecules on infected host cells
3) can survive as a patent provirus for prolonged periods
4) As result, HIV persists and destroys the immune system
5) CD4 T cells are the prime targets of destruction, resulting in reduced effectiveness of antibody and cell-mediated immune responses

1575
Q

How is HIV suspected to kill or render function of CD4 Th cells?

A

1) loss of plasma-membrane integrity because of viral budding
2) destruction by HIV-specific CTL
3) viral induction of apoptosis

1576
Q

What is the central role in the immune system for CD4 T cells?

A

Regulating the activities of B and T cells

1577
Q

What is the acute stage of HIV characterized by?

A
  • rapid burst of viral replication before the development of HIV-specific immune responses
  • high levels of viremia is found in this stage
  • decrease in CD4 T-cell number
1578
Q

What are the symptoms of acute stage of HIV-1?

A
  • flu like symptoms
  • IM-like symptoms
  • many are asymptomatic during this stage
1579
Q

Describe HIV latent stage

A
  • decrease in viremia
  • increase in CD4 T-cell number
  • symptoms are subtle or absent
1580
Q

Describe the last stage of HIV

A
  • full blown AIDS
  • characterized by profound immunosuppression with very low numbers of CD4 T cells
  • resurgence of viremia
  • patients demonstrate neurological symptoms
  • appearance of life-threatening opportunistic infections and malignancies
1581
Q

What are the symptoms for infants with AIDS?

A
  • failure to thrive
  • persistent oral candidiasis
  • hepatosplenomegaly
  • lymphadenopathy
  • recurrent diarrhea
  • recurrent bacterial infections
1582
Q

Describe antiretroviral therapy (ART)

A
  • drugs that block various steps of the HIV replication cycle
  • nucleoside analog reverse transcriptase inhibitors
  • proteases inhibitor
  • integrase inhibitors
  • fusion inhibitors
  • CCR5 antagonists
  • post attachment inhibitors
  • are most effective when used in combination
  • has significantly improved morbidity and mortality of HIV-infected persons and has reduced the rate of perinatal transmission
1583
Q

What are ways to prevent HIV?

A
  • screening blood/organ donors for HIV
  • education of the general public on HIV transmission, safety measures
  • precautions for health-care workers
  • vaccine-being researched
1584
Q

Describe screening and diagnosis of HIV

A
  • previous algorithms and tests methods
  • current algorithm and test methods
1585
Q

Describe previous testing algorithm of HIV

A
  • screen for HIV-1/HIV-2 antibodies by ELISA or rapid EIA
  • confirm positive test results by repeating ELISA, followed by western blot
1586
Q

Describe western blot interpretations of HIV

A
  • no band: negative
  • positives are harder to interpret
  • CDC says must have antibody against two or three of the following bands:
    —> p24
    —> gp41
    —> gp120/160 (these bands are very close together and is hard to distinguish between the two.
1587
Q

Describe the current CDC testing algorithm for HIV

A
  • initial screening: HIV-1/2 antigen/anitbody combination immunoassay
  • if positive, run HIV-1/2 differentiation immunoassay
  • HIV-1(+) & HIV-2 (-): HIV-1 antibodies present
  • HIV-1(-) & HIV-2(+): HIV-2 antibodies present
  • HIV(+) & HIV-2(+): HIV-1 and HIV-2 antibodies present
  • if both HIV negative, run HIV-1 NAT
    —> HIV-1 NAT(+): acute HIV-1 infection
1588
Q

What is the principle of the fourth generation HIV-1 antibody/HIV-2 antibody/p24 antigen combination immunoassay?

A

1) incubate patient serum with solid phases onto which HIV-1 antigens, HIV-2 antigens and antibody to HIV-1 p24 have been bound
2) following incubation, HIV-1 or HIV-2 antibodies in the patient sample will bind to their respective antigens
3) HIV-1 p24 antigen in the patient sample will bind to anti-p24 solid phase
4) wash, then add conjugate consisting of labeled anti-24 labeled HIV-1/HIV-2 antigens
5) following, incubation, wash and addition of trigger solutions or substrate/stop solution, relative light units or optical absorbance are measured
6) confirm positive result with rapid EIA

1589
Q

What are some ways the HIV is monitored?

A
  • peripheral blood CD4 T-cell counts
  • quantitative viral load assays
  • drug resistance and tropism
1590
Q

Why do false-positives occur int he 4th generation HIV-1/2 antibodies and p24 antigen test?

A
  • heat inactivation
  • repeated freezing and thawing
  • presence of heterophil antibodies
  • passive immunoglobulin administration
  • administration of an experimental HIV vaccine to patient
1591
Q

What is seroconversion?

A
  • the change of a serological test result from antibody negative to antibody positive
1592
Q

What are the two markers used to monitor HIV for disease progression and guide the treatments?

A
  • the peripheral blood CD4 T-cell count (best indicator of immune functions)
  • HIV-1 RNA level (viral load)
1593
Q

Describe CD4 T-cell enumeration

A
  • CD4 T-cell numbers are the best indicator of immune function i HIV-infected individuals
  • incubate peripheral blood with fluorescent-labeled anti-CD4; analyze results by flow cytometry
  • in untreated patients, CD4 T cell number declines progressively, and CD4 T: CD8 T cell ratio is less than 1:1
  • CD4 T cell count of less than 200/Ul indicates stage 3 HIV
  • a significant decline in CD4 T cell count over time may indicate a need to change ART or administer prophylactic therapy for certain infections
1594
Q

What is the gold standard for enumerating CD4 T cells?

A

Immunophenotyping with data analysis by flow cytometry

1595
Q

Describe the CDC classification list of HIV

A

0-5
- 0: early infection of patient who tested positive for HIV in initial screening but has a negative or indetermine confirmatory test
- 1-3: based on peripheral blood CD4 T-cell count or percentage. If this information or percentage. If this information is missing, classified as unknown

1596
Q

Describe quantitative viral load assays

A
  • measure amount of HIV RNA circulating in patient plasma
  • methods: qPCR and bDNA
  • HIV RNA detectable about 11 days after infection
  • successful therapy with ART results in the decline in the viral load to an undetectable antigen
  • patients with persistently increased viral load should undergo drug resistance testing and may need a possible change in ART
1597
Q

Why are viral load tests used?

A
  • help predict disease progression
  • monitor patient response to ART
  • guide treatment plan
1598
Q

Describe the viral load assays of HIV RNA

A
  • based on amplification methods that increase the number of HIV RNA copies
  • most common: PCR and bDNA assay
1599
Q

What are the 2 PCR methods that have been developed to detect HIV nucleic acid?

A
  • RT-PCR (not usually used)
  • qPCR
1600
Q

What is the basic principle of RT-PCR for HIV RNA?

A
  • to amplify a DNA sequence that is complementary to a portion of the HIV RNA genome
1601
Q

What are the disadvantages of RT-PCR for HIV RNA?

A
  • limited dynamic range
  • highly susceptible to cross-contamination with extraneous nucleic acid
1602
Q

What is the basic principle of bDNA assay?

A
  • based on amplifying the detection signal generated in the reaction
  • accomplished by using a solid-phase sandwich hybridization assay that incorporates multiple sets of oligonucleotide probes and hybridization steps that produce a series of branched molecules
1603
Q

What are disadvantages of bDNA?

A
  • requires larger sample volume
  • lacks internal controls
  • lower specificity
1604
Q

Describe genotype resistance assays of HIV

A
  • performed in clinical laboratory settings
  • HIV reverse transcriptase and protease genes from RNA in patient plasma are amplified by RT-PCR
  • products are sequenced and analyzed with software for mutations
  • results are reported as: Resistance, possible resistance, no evidence or resistance
1605
Q

Describe phenotype resistance assays of HIV

A
  • determine ability of HIV from clinical samples to grow in the presence of antiretroviral drugs
  • involve sophisticated technologies only performed by specialized reference laboratories
1606
Q

Describe tropism testing of HIV

A
  • genotypic or phenotypic assays are performed to determine if the patient has virus that will bind to the CCR5 co-receptor and be responsive to CCR5 antagonists
1607
Q

Decribe testing of infants younger than 18 months in HIV

A
  • all pregnant women should be tested for HIV
  • maternal antibodies in infant serum can complicate serological tests results
  • molecular methods are used for diagnosis
  • Qualitative HIV-1 DNA PCR using infants peripheral blood mononuclear cells is the preferred method
  • serological testing at 12-18 months of age may be used to confirm the diagnosis
1608
Q

HIV virions bind to host T cells through which receptors?

A

CD4
CCR5

1609
Q

What is a typical of the latent stage of HIV infection?

A
  • proviral DNA is attached to cellular DNAq
1610
Q

Why would false negative results in a test from HIV?

A

Collection of the test sample before seroconversion

1611
Q

What does the conjugate used in the 4th-generation immunoassays for HIV consists of?

A
  • HIV-1 and HIV-2 specific antigens plus antibody to p24
1612
Q

What is the first detectable antibody in serum after infection with hepatitis B?

A

HBc

1613
Q

What is the difference between HAV and HBV incubation periods?

A
  • HAV —> short
    HBV —> long
1614
Q

How do you interpret these results?
Anti-HAV (IgG) —> neg
Anti-HAV (IgM) —> neg
HBsAg —> neg
Anti-HBc —> pos
Anti-HBs —> neg
HBe —> neg

A

HBV in core window phase

1615
Q

What is the most common cause of congenital infections?

A

CMV

1616
Q

What populations does VZV cause severe complications in?

A
  • immunocompromised patient
  • pregnant women
1617
Q

What is the clinical importance of CMV?

A

Life threatening for immunocompromised patients and infants

1618
Q

A patient has IgG and IgM antibodies against viral capsid antigens of EBV. What does this patient have?

A

Current EBV infection

1619
Q

What are heterophil antibodies routinely detected by their reaction with this agent?

A

Bovine RBC antigens

1620
Q

What does the presence of IgM anti-rubella antbodies in serum from an infant born with a rash?

A

Congenital infection with rubella virus

1621
Q

What are the most commonly used tests to detect mumps virus?

A

RT-PCR
ELISA

1622
Q

What is the method of choice for detecting VZV in immunocompromised hosts?

A

RT-PCR

1623
Q

Describe Tzank smear

A

Scrap lesion with scalpel but put on slide and stain, the look for giant multi-nucleated cells

1624
Q

What does p24 encode for?

A

Gag

1625
Q

How should these results be interpreted for HIV?
ELISA: pos
Repeat ELISA: neg
Western blot: no bands

A

Negative for HIV

1626
Q

What type of cells can HIV infect?

A
  • primarily lymphocytes
  • monocytes
1627
Q

What does the HIV core consists of?

A

Two identical strands of RNA

1628
Q

Why is HIV called a retrovirus?

A

Can transcribe RNA into DNA

1629
Q

A 22 year old male sees his physician for an annual checkup. He reported having flu-like symptoms including fever, sore throat, and lymphadenopathy several months ago. He has lost 10 pounds but otherwise fee fine. HIV testing is recommended and ordered
- ELISA —> pos
- western blot —> POs
- CD4+ T-cell count is 500/ml
What disease and stage is this?

A

HIV positive in latency stage 1