Immunology Flashcards

1
Q

What is the ligand of CD28?

A

CD80/CD86

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

What are two key differences between CD28 and ICOS?

A

-CD28 is involved in initial activation, whereas ICOS helps maintain activity of differentiated T cells
-CD28 is expressed on all naive T cells and some memory cells while ICOS is expressed on memory and effector cells

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

What are the 3 signals and what do they do?

A

Signal 1: antigen-specific -> TCR activation
Signal 2: Co-stimulation -> proliferation and differentiation
Signal 3: Cytokines -> T cell differentiation

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

What happens when there is no signal 2?

A

T cell becomes anergic. This happens when there is an antigen but no APC presenting it

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

How may a self-reactive T cell develop?

A

A T cell isn’t screened against a peripheral self-antigen during development

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What causes anergy, and why is it benefitial?

A

Anergy happens when there is no co-stimulatory signal (signal 2). This helps provide tolerance, especially in the periphery. An APC must present the antigen for signal 2 to occur.

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

What is 4-1BB?

A

A co-stimulatory receptor that can act as CD28 when it is induced

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are two important inhibitory receptors?

A

CTLA4 and PD1

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

When is CTLA4 expressed?

A

Starting a few days after the start of an immune reaction due to it needing an accumulation of transcription factors

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

How does CTLA4 work?

A

It binds CD80/CD86 with a higher affinity than CD28 to prevent signal 2 co-stimulation (receptor competition) and gives a negative signal by dephosphorylating pathways via phosphatases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What does PD1 bind to?

A

PD-L1 and PD-L2 on APCs and tumor cells

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

Why is tumors expressing PD-L1 and PD-L2 significant?

A

It can prevent T cells from attacking it by preventing co-stimulation (since they also express antigens a.k.a signal 1)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

In what conditions does T cell exhaustion occur?

A

During persistent antigen presentation: Chronic viral infections, cancer/tumors, autoimmunity

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

What happens to T cells when they become exhausted?

A

They start gradually gaining inhibitory receptors, starting with PD-1.
They start losing their ability to make pro-inflammatory cytokines and lose their ability to prolifirate while also gradually becoming pro-apoptotic.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is the 3 signal model in T cell exhaustion?

A

1: Persistent antigen presentation
2: Negative co-stimulation
3: Chronic inflammation

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

What happens with transcription factors during T cell exhaustion?

A

TCF1 (helps proliferate) goes down at first as TOX goes up. TOX is supposed to go back down (as TCF1 goes back up) but doesn’t, making T cells TCF1 and TOX positive. Eventually they lose TCF1 and become terminally exhausted, losing the ability to proliferate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Why is T cell exhaustion benefitial?

A

It dampens immunopathology in i.e chronic infections, and prevents autoimmunity

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

What does CART stand for?

A

Chimeric antigen receptor T cell. It contains Both TCR (signal 1) as well as 4-1BB and CD28 for strong co-stimulation (signal 2)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is checkpoint inhibition?

A

Antibodies against inhibitory receptors CTLA4 and PD-1

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

What route do T cells leave the lymph nodes and spleen?

A

Through the efferent lymph

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

How do APCs leave the lympph nodes?

A

They don’t! They die there

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What regulates T cell exit?

A

Not chemokines, but lipids called S1P. Lymph nodes upregulate S1PR during dwell time in

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What cells synthesize S1P?

A

Endothelial cells and red blood cells. This makes S1P concentration high in bodily fluids and low in tissues, creating a gradient for T cells to follow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

How do T cells ensure they don’t leave the lymph node too soon?

A

They express CD69 as an early activation marker, which prevents re-expression of S1PR. CD69 is expressed less after a few divisions, allowing S1PR upregulation and exit after some time.

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

How does rolling work for T cells?

A

L-selectin weakly interacts with integrins, so glycoproteins on HEVs, causing slowing.

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

How does sticking work for T cells?

A

CCL21 expressed by HEVs interact with CCR7 on T cells now that it is slowed down. This interaction causes a conformational change in LFA-1, which can then bind to ICAM-1 with high affinity.

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

What happens after rolling?

A

Diapedesis (Transmigration)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

What is inside out signaling?

A

CCR7-CCL21 interaction causes a change in LFA-1 conformation on the T cell

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

What is outside in signaling?

A

LFA-1 (after a conformational change) on T cells binds to ICAM-1 on HEVs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What are peripheral Node Addressins?

A

Integrins. “postal code” sugars expressed by HEV

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

What homeostatic chemokines attract B cells and T cells, respectively, and what cells produce them and where?

A

CXCL13 is expressed by stromal cells in the B cell follicles.
CCL21 is produced by stromal cells in the paracortex and promotes T cell and dendritic cell migration.

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

What are the three mechanisms of attraction?

A

Chemotaxis, haptotaxis and chemo/haptokinesis

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

What is more common, chemotaxis or haptotaxis?

A

Haptotaxis

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

Why is chemotaxis not as effective as haptotaxis?

A

Diffusion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Where are integrins mainly expressed?

A

Endothelial cells

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

What determines the location at which a T cell can move into tissue after activation? Explain the process and name an example

A

When dendritic cells from a certain part of the body migrate to the lymph node and activate T cells, it ensures that the T cell expresses homing molecules specific for that location.. This ensures that the T cell only enters that area and in an efficient manner.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

What are the two blood supplies of the liver and what are their characteristics?

A

Portal vein - from the intestines, oxygen-poor and nutrient rich
Hepatic artery - oxygen rich

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

What are sinusoidal endothelial cells?

A

They line the vascular ducts in the liver and allow nutrients through to hepatocytes

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

What are the 3 main functions of the liver?

A

Metabolism
Detoxification
Synthesis

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

What biomolecules does the liver synthesize?

A

MBL and other complement system components
C-reactive protein
Soluble PRRs
Antimicrobial peptides
Clotting factors
Acute phase proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

How is acute phase protein production initiated?

A

When macrophages detect a threat, they secrete cytokines IL6, IL1, TNFa. These lead to an increase of acute phase proteins such as c-reactive protein.

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

What is a marker used to detect inflammation?

A

C-reactive protein

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

What forms does CD14 have?

A

Bound on macrophages in the liver and soluble

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

In what ways can liver tolerance become a problem?

A

Hepatitis B & C virus, malaria spores, tumor metastases and autoimmune reactions

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

Why are liver transplants more often successful than other transplants?

A

HLA-matching is not required

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

What does LSEC stand for?

A

Liver sinusoidal endothelium

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

Where do immune cells of the liver reside?

A

In the blood vessels between the portal and central veins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

How are waste products removed from the liver?

A

Liver sinusoidal endothelial cells remove waste using scavenger receptors and carbohydrate receptors -> endocytosis -> lysosomal degradation

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

What are the properties of LSECs as APCs?

A

-Weak with tolerogenic properties
-MHC I - non-productive activation of CD8 T cells
-MHC II - usually leads to Tregs
-Anti-inflammatory cytokine secretion: IL-10, TGF-ß
-PDL-1
-Large tolerizing surface

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

Why does activation through LSECs usually lead to induction of Tregs?

A

Liver sinusoidal endothelial cells mainly produce anti-inflammatory cytokines IL-10 and TGF-ß

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

Why is MHCI cross-presentation in LSECs non-productive?

A

There is a low level of co-stimulation (signal 2) and no inflammatory cytokines (signal 3)

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

What do the Fcy and Fcα receptors on Kupffer cells do?

A

Clear immune complexes and IgA-antigen complexes

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

What do complement receptors on macrophages do?

A

Allow for clearance of C3b-coated bacteria

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

What is Poly:I:C?

A

Synthetic dsRNA used to mimic TLR3 stimulation

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

What are the properties of Kupffer cells as APCs?

A

-Not very potent with tolerogenic properties
-Activation causes co-stimulatory molecule expression
-Induce expansion of Tregs
-Secrete IL-10, TGFß and prostaglandins

Prostaglandins can for example reduce IL-2 receptor expression

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

What type of phenotype do lymphocytes have in the liver?

A

Mostly HLA-DR+, so activated.
Also 35% are gamma-delta T cells and there are also NKT cells, so more innate T cell phenotype

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

What kind of infection is Hep A infection?

A

Acute, infectious hepatitis

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

What kind of infection is Hep B infection?

A

Persistent, serum hepatitis

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

What kind of infection is Hep C infection?

A

Persistent, non-A and non-B

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

Where do you normally see immune infiltration/inflammation during Hep B infection?

A

Around the portal tract area. Mostly T and B cells. There is very little liver damage until very late stage.

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

What type of virus is Hep A?

A

Picornavirus, RNA + sense

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

What type of virus is Hep B?

A

Hepadnavirus, DNA (partial double strand)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

What type of virus is Hep C?

A

Flavivirus, RNA - sense

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

Which of the 3 main Hep viruses have no envelope?

A

A

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

What age group usually gets chronically infected with Hep B and recovers less often?

A

Newborns

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

What age group usually develops chronic infection via Hep C?

A

Adults (20-40%)

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

When is the clinical outcome of HCV infection determined?

A

Within 24 weeks.

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

What is ALT?

A

An enzyme released into the blood during liver damage. Higher levels coincide with measured viral load of HCV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

What is peculiar about the adaptive response during HCV infection?

A

Delay. T cell proliferation starts after 10 weeks.

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

What inhibits killing of infected cells by NK?

A

HCV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What is peculiar about the adaptive response during HBV infection?

A

Most chronic HBV patients have a weak or absent HBV-specific T cell response. It comes up late and goes back down and stays low.
The relationship between viral load and T cells is inverse. Also liver infiltrating B cells are observed in active phases of chronic HBV.

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

What is a characteristic of T cells during chronic Hep B and C infection?

A

T cell exhaustion. High PD-1 expression.

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

What can restore T cell responses in HBV infection?

A

Nucleoside analogues - mimic nucleotides that get incorporated into growing DNA strands, impairing DNA polymerase

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

What are 4 ways to restore T cell responses in HBV infection?

A
  1. Reduce antigen load
  2. Block inhibitory receptors
  3. Block inhibitory pathways (IL-10, etc)
  4. Immunotherapeutic boosting of T cells (vaccines, cytokines, TLR agonists)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What is a parenchyma?

A

The functional structure of a tissue distinguished from the connective and supporting tissues

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What is spot necrosis?

A

A small area of necrosis

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

What are characteristics of an apoptotic hepatocyte?

A

Shrinkage and pyknosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

What are characteristics of balloon degeneration?

A

Swollen cytoplasm, eosinophilic enlarged nucleus -> karyoreksis

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

What are characteristics of bridging necrosis?

A

necrosis that extends between lobules, such as portal-portal or central-central bridging

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

What are two histopathological characteristics of a chronic HBV infection?

A

Ground glass (light eosinophilic cytoplasm, clear halo) and sanded nuclei (pale eosinophilic, finely granular)

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

What is the main difference between acute and chronic hepatitis?

A

The chronic component is in the portal area

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

What is the order of the segments of the small intestine?

A
  1. duodenum
  2. jejunum
  3. ileum
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

What is the order of the parts of the large intestine?

A
  1. cecum
  2. ascending colon
  3. transverse colon
  4. descending colon
  5. sigmoid colon
  6. rectum
  7. anus
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Where do secretions from the liver and pancreas empty into?

A

Duodenum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

How come stomach acid doesn’t affect the small intestine?

A

Duodenum’s brunner’s glands secrete bicarbonate which neutralizes it, and pancreatic juices and bile mixed in dilute it.

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

Where are the majority of nutrients absorbed?

A

Jejunum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

What parts of the intestine absorb water?

A

The ascending and transverse colon

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

What are M cells?

A

They are highly selective recognition cells that take up antigen to present to dendritic cells in the Peyer’s patches

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

What types of immune cells are in the mucosa outside of the lymhpoid structures?

A

Memory T and B cells, macrophages, dendritic cells and inter-epithelial lymphocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What lymphoid organs in the intestines are not structured?

A

Colonic patches or isolated lymhoid follicles

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

Where are intestinal goblet cells most numerous?

A

The colon

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Where can Paneth cells?

A

In the crypts of the small intestines

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

What is the function of Paneth cells?

A

They contain antimicrobial granules that, when secreted, keep the crypts clear of bacteria. It also functions as a signaling cell to surrounding cells/

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

WHat are GALTs?

A

Gut-associated lymphoid tissue

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What undergoes regional adaptation and why

A

Mesenteric lymph nodes, since they drain from different areas of the intestines, which are all different

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

Where is the highest microbiota diversity in the intestines?

A

The colon

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

What are two ways commensal bacteria can limit pathogen colonization?

A
  1. Pathogens are poorly adapted to compete for nutrients
  2. Commensals stimulate immune responses against pathogens
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

In what 5 ways is the intestinal epithelium a barrier?

A
  1. Microvillar extensions prevent attachment
  2. Epithelial tight junctions
  3. Secreted mucins
  4. Epithelial transcytosis of IgA
  5. Antimicrobial peptides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

In what 5 ways is the intestinal epithelium a barrier?

A
  1. Microvillar extension prevent attachment
  2. Epithelial tight junctions
  3. Secreted mucins
  4. Epithelial transcytosis of IgA
  5. Antimicrobial peptides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

In what 5 ways is the intestinal epithelium a barrier?

A
  1. Microvillar extension prevent attachment
  2. Epithelial tight junctions
  3. Secreted mucins
  4. Epithelial transcytosis of IgA
  5. Antimicrobial peptides
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What types of mucus layers are there in the intestines?

A

Small: single thin
Colon: Two (inner and outer)

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

What is a histopathological hallmark of spontaneous colitis?

A

Very long crypts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

What does idiopathic mean?

A

A disease that arises spontaneously or for which the cause is unknown

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

What is it called when something moves across a cell?

A

Transcytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

How do Paneth cells differ in their antimicrobial function from other epithelial cells?

A

They have a higher diversity of AMPs and have vesicles already ready to go

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

What is microbial sampling?

A

Controlled testing by M cells of bacteria present in the lumen of the intestine

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

In what three ways microbes are sampled?

A
  1. Passive diffusion
  2. Transcellular transport
  3. Macrophage uptake
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What type of antibodies are produced in the Peyer’s patches?

A

Mostly IgA but also IgG. Especially IgG is someone is IgA-deficient

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

What cells can disrupt tight junctions for antigen sensing and uptake?

A

Macrophages

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

How does microbial sensing by PRRs work?

A

Through TLRs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Why is sensing important?

A

So we can discern between harmless and harmful bacteria

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

How is sensing like a piano?

A

Microbes have many different structures that can be sensed by different PRRs, like fingers on a piano that hit keys in different strengths, creating unique cytokine cocktails. The microbiome interacting with us is the music.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

How does NF-kB maintain intestinal mucosal integrity?

A

Through its roles in regulating inflammation, promoting cell survival against inflammation (anti-apoptitic genes like Bcl-2) and proliferation, maintaining barrier function (ie. Upregulation of tight junction proteins and adherins), and regulating mucin production.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

Does NF-kB mean activation?

A

Not necessarily. It is essential for homeostasis in the intestines

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

What happens when you delete NF-kB in the intestinal epithelium?

A

Apoptosis and activation of macrophages underneath (due to infiltration), which leads to chronic intestinal inflammation

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

How are TLRs situated in the intestinal epithelium?

A

Mostly on the baso-lateral side, except for at the peyers patches. TLRs in the small intestine are also different than in the colon for example. TLR4 and 2 are also expressed a bit low.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

What is the TLR5 ligand?

A

flagella

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

What regulates the epithelial response pattern?

A

Inhibitory proteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

How do intestinal epithelial cells respond?

A

They downregulate different cells using anti-inflammatory cytokines unless necessary (hopefully)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

What is a consequence of mucosal tolerance regarding treatment?

A

Vaccination is very hard if you want to do a mucosal vaccination.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

What do adjuvants do?

A

Danger triggers that overcome homeostatic tolerance

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

Do dendritic cells or macrophages drain more to the LN?

A

DCs

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

What do dendritic cells do in the lymphoid tissue in the intestines?

A

Present to Naive T cells so they can eventually differentiate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

What is another term for Th1/Th17?

A

Tr1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

What types of T cells are in the lamina propria?

A

Effector memory T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

What adhesion molecule is required for cells to

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What do resident DCs in the mesenteric LN do?

A

They take up and present antigens that have diffused and drained in the lymph

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

What do subcapsular macrophages do?

A

They seave macromolecules out of the molecules that have drained with the lymph to the mesenteric LNs, digest them and transfer them to resident DCs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

How do differentiated T cells in the mesenteric LNs go back into the intestines?

A

Imprinting: They are imprinted to go back through chemokine receptors like CCR9, that respond to CCL25 from small intestines and adhere using α4ß7 to bind to MADCAM1.

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

What is α4ß7?

A

An adhesion molecule that allows cells that express CCR9 to enter the intestinal tissue

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

Where do Tregs differentiate in the intestines?

A

Peyers patches and mesenteric LNs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

What function do CD103+ DCs have?

A

They can convert vitamin A derived retinol into retinoic acid. In combination with TGFß from the environment, Treg induction occurs: Foxp3, CCR9, α4ß7, Treg.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

What is necessary for Treg induction in the intestines?

A

Vitamin A (retinol) -> retinoic acid by CD103+ DCs plus environmental TGFß

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

What type of branching system does the lungs have?

A

Dichotomous

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

What does the respiratory unit consist of?

A

The respiratory bronchioles and alveolar ducts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

What do the conducting airways consist of?

A

Trachea, segmental bronchi and nonrespiratory bronchioles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

What is found in the submucosa of the bronchus?

A

Cartilage and glands

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

What type of epithelium is that of the bronchus?

A

Pseudostratified

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

What is BALT?

A

Bronchus-associated lymphoid tissue

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

Does BALT have germinal centers?

A

Yes

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

What is the difference between BALT and lymph nodes?

A

LNs are more well defined and have a capsule with afferent and efferent lymphatic vessels for fluid circulation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

What are club cells?

A

Clara cells. They are nonciliated bronchiolar secretory cells that secrete surfactants, and are progenitors for other cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

Is the beating of cilia ATP dependent?

A

Yes

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

What causes ciliary dyskinesia?

A

Disorder of motor proteins within cilia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

What does ciliary dyskinesia lead to?

A

Mucus buildup with dirt and bateria

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

What influences mucus production?

A

DNA of damaged cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

What causes cystic fibrosis?

A

Mutations that affect chloride ion channels, preventing them from getting to the cell surface, leading to thick sticky buildup and flattening of cilia

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

What is metaplasia?

A

A change of one cellular phenotype to another

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

What is a risk factor for tumorlets?

A

Chronic or repeated inflammation, causing consistent increase in neuroendocrine bundles aka hyperplasia

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

What is the CTFR?

A

Ion transporter that regulates mucus viscosity

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

What does surfactant do?

A

Lower surface tension to be able to breathe more easily

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

How many type 2 pneumocytes are there per type 1 pneumocyte?

A

2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

What is the stem cell for type 1 pneumocytes?

A

Type 2 pneumocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

Why is the pleura being so thin important?

A

Negative pressure between the lung and the thorax

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

Why is pulmonary infection the most common type of infection?

A

Exposure/interface with the environment

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

What is a typical infection dynamic of viral pneumonia?

A

Usually self-limiting, but dominated by secondary bacterial infection in severe cases

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

What is one problem with histological diagnosis of viral pneumonia?

A

Limited morphological specificity

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

What group is most likely to get ill from CMV infection?

A

Immunocompromised people

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

What is histologically characteristic for DNA virus infections?

A

Large nuclei due to replication

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

What predicts the type of bacterial pneumonia?

A

The setting (community acquired, nosocomial, immunocompromised, etc)

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

What are the two pathological patterns in bacterial pneumonia?

A

Bronchopneumonia and lobar pneumonia

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

Species from which fungal phylum can cause disease in individuals with impaired immunity?

A

Mucormycetes

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

What is the definition of sarcoidosis?

A

A granulomatous disorder of unknown cause affecting multple organs

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

What organ is mostly involved in sarcoidosis?

A

Lungs, but generally more often organs more exposed to the air

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

How often do sarcoidosis patients develop progressive disease?

A

1/3 of patients

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

What surrounds granulomas?

A

T cells

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

What is the epidemiological pattern of sarcoidosis?

A

High rate in scandinavia but low morbidity, lower rates in other places but worse prognosis

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

What is the prevalence of chronic development of sarcoidosis in non-LS patients?

A

50%, about 20% with fibrosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

What are triggers of sarcoidosis?

A

Anorganic exposure, antigen exposure

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

What drives a granuloma?

A

Ongoing cytokine production by T helper cells around a trigger

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

What type of Th cells are present in pulmonary sarcoidosis?

A

Th17.1 cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

What is a biomarker of patients who develop chronic sarcoidosis?

A

Higher Th17.1 proportion to other Th cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

What is CTLA4?

A

A co-inhibitory receptor with higher affinity for CD80/CD86 than CD28

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

What is the CTLA4 expression level in memory Th cells in sarcoidosis, and what type of Th cells?

A

Reduced, Tregs and Th17

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

What does an increase in Th17 with reduced CTLA4 expression do?

A

Enhanced pro-inflammatory activity

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

What is double trouble in sarcoidosis?

A

Reduced expression of CTLA4 in Tregs (less inhibition) and Th17 (enhanced pro-inflammatory response)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What drug classes are associated with sarcoid-like reaction?

A

Checkpoint inhibitors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What is ICOS?

A

The memory and effector T cell version of CD28, which sustains proliferation and activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What is 4-1BB?

A

It can be activated in place of CD28 when absent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

Is CD80/CD86 constitutive or induced?

A

Induced: only expressed by activated APCs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

What does JAK inhibition do?

A

Targets multiple cytokines involved in sarcoidosis, since JAK is a kinase in many pathways involved with cytokine signaling

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

Is ICOS constitutive or induced?

A

Induced

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What happens when T cells become exhausted?

A

They slowly gain inhibitory receptors, starting with PD-1, and start losing their ability to make cytokines, proliferate and maintain. They end apoptotic.

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

In what conditions does T cell exhaustion occur?

A

Chronic viral infections, cancer/tumors, autoimmunity

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

What is the underlying cause of disease of interstitial lung disease?

A

Highly variable, over 200 known, including autoimmune disease, exposure-related, sarcoidosis, idiopathic, etc.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What is the initial site of damage in ILD associated with environmental factors or exposure?

A

Epithelial cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What happens initially in ILD when associated with autoimmunity?

A

Immune complex formation and vascular blockage

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

What is the late phase of ILD?

A

Fibrosis

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

Can ILD be reversible?

A

Only in the early phase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What is a major clinical need in fibrotic ILD?

A

To distinguish inflammation from fibrosis

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

What is a histological hallmark of ILD fibrosis?

A

Honeycombing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

What is hypersensitivity pneumonitis?

A

Immune-mediated response by susceptible and sensitized individuals to inhaled environmental antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

What is Acute HP?

A

Intermittent high level exposure to inducing antigen

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

What is chronic HP?

A

Chronic low level exposure to inducing antigen, which can be stable non-fibrotic but can sometimes progress to fibrotic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

How do connective tissue diseases play a role in ILD?

A

Autoimmune diseases like RA and SLE can lead to immune-mediated damage to lung tissue such as autoantibodies forming immune complexes and blocking and damaging capillaries

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

What is the pathogenic cytokine in pulmonary fibrosis?

A

IFNy, in the sense that it should inhibit Th2 but somehow doesnt

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

What cells are pro-fibrotic?

A

Th2

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

What is the dominant profile in pulmonary fibrosis?

A

Th2, but Th17 is also a bit pro-fibrotic

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

What are idiopathic pulmonary fibrosis risk factors?

A

50-70 years of age, predominantly men, often smokers, genetic predisposition

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What is a very pro-fibrotic cytokine?

A

TGFß

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

How is fibrosis induced?

A

Through TGFß:
-signals fibroblasts or epithelial cells to differentiate into myofibroblasts, which leads to production of extracellular matrix.
-Inhibits ECM degradation
-Inhibits lymphocyte proliferation and macrophage activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

What categories are genes associated with IPF subdivided into?

A

Telomere biology (telomerase) and surfactant metabolism and Muc5B

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

What removes senescent cells?

A

NK cells usually, however they are dysfunctional in IPF

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

What predicts IPF outcome?

A

Telomere length

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

Where are biologicals derived from?

A

Biological sources

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
206
Q

What is the efficacy of biologicals?

A

Overall much greater than other therapeutics

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
207
Q

What are the two main modes of action of biologicals

A

Soluble mediator binding or binding of cell surface molecules

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
208
Q

What are ways cell surface molecules binding biologicals can help in a disease?

A

Cell elimination and receptor blocking

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

How can soluble mediator binding by biologicals help in a disease?

A

Neutralization: cytokines, growth factors, etc

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

Are checkpoint inhibitors biologicals?

A

Yes

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

What is the mechanism of checkpoint inhibitors?

A

Anti-PD-1 prevent binding of co-inhibitory binding allowing T cells to bind

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
212
Q

Where can checkpoint inhibitors act?

A

During the priming phase and effector phase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
213
Q

Where is the priming phase and how can checkpoint inhibitors act there?

A

Prevent development of inhibitory receptors on T cells

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

What is -omab

A

Mouse derived mAb

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

What is -ximab

A

Chimeric mAb

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

What is -zumab

A

Humanized mAb

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

What is -umab?

A

Human mAb

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

What is the possible negative impact of ADA on disease activity?

A

Rapid clearance and loss of response, which can later have an impact on disease severity, which is why monitoring is really important

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
219
Q

What is the biologic trough level?

A

The amount needed of a biological to be effective

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

What is it called when you analyze ADA presence or biologic trough level?

A

Therapeutic drug monitoring

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

What does bioavailablity depend on?

A

Distribution, degradation and neutralization

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
222
Q

How are trough levels determined?

A

ELISA

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

Are all ELISA assays the same?

A

No

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

What can anti-thyroid antibodies predict?

A

Efficacy of anti PD-1 antibodes

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

What are biosimilars?

A

B brand unpatented biologicals

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

What is the risk of treating humans with gene therapy?

A

Introduction into the environment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
227
Q

What’s the difference between DM and ML?

A

DM involves an animal plus a genetically modified organism, whereas ML only involves a genetically modified organism

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

What is needed to be allowed to treat patients with gene therapy?

A

An IM-VM permit

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

Look up the 8 pillars of biosecurity

A

Awareness, personnel reliability, …

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

What are the three ladder steps of psoriasis?

A

Topical, photo, systemic

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
231
Q

What is important for pathogenesis in psoriasis?

A

Inflammatory cytokines, most notably IL23 (and IL17)

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

What is a potential side effect of biologics use?

A

Infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
233
Q

What type of adalunimab used in EMC?

A

Biosimilar

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
234
Q

Why can you rebound with biological use?

A

Immune reaction against the biological: ADAb

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

What is ADAb

A

Anti-drug antibodies, the change the bioavailability of a drug

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

What is necessary for long antibody halflife?

A

High affinity binding to the FcRn receptor

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

Why is it no use to give more of a drug than necessary?

A

No increased improvement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
238
Q

Why could an Ab gradually become less effective over long periods of time?

A

Changes in the disease

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

What if you have a low trough level and no to low anti antibodies?

A

The patient is likely not taking the drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
240
Q

What do you do if you don’t find a drug in nonresponders but do find ADAb?

A

Switch to a less immunogenic drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
241
Q

What are immunogenic drugs?

A

Drugs against which a patient has an immune response

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

What do you do when you have a nonresponder and find a high enough trough level?

A

Change to a different mode of action

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
243
Q

What is a calcineurin inhibitor

A

Tacrolimus

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

What is the main immunosuppressive drug

A

Tacrolimus

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

What is a pro of tacrolimus

A

Low level of rejection

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

What are 5 problems with tacrolimus

A

Narrow therapeutic window, nephrotoxicity, neurotoxicity, incidence of diabetes, a specific (infections)

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

What does belatacept do?

A

Blocks both CD80/86 (polyclonal) -> costimulation

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

Imlifidase mode of action

A

Cleaves IgG

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
249
Q

What are three new forms of immunosuppression?

A

Anti IL-6, imlifidase and CAR-T cells

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

In what way is tacrolimus better than cyclosporin?

A

Better kidney function

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

What is surfactant produced by?

A

Type II pneumocytes

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

What was the PANTHER trial?

A

They used prednisone and two other therapeutics to try to knock out the immune system of IPF patients, causing many people to die

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

Does anti-microbial therapy improve IPF symptoms?

A

No. You take away the trigger but not the immune response and damage already done

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

Is there antifibrotic therapy and (how) does it work?

A

Yes, Ninedanib (also other ILD types): and Pirfenodone (only IPF). Both kinase inhibitors for certain cytokines such as TGFß. Quality of life is still not great once already fibrotic

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

What is the most prominent type of asthma?

A

Type 2 asthma

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

What are two ways to treat asthma?

A

Corticosteroids and Bronchodilators (ß2-agonists)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
257
Q

What do corticosteroids target?

A

Almost every immune cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
258
Q

What do bronchodilators target?

A

Smooth muscle cells of the airway walls

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

What are two down sides to corticosteroid use against asthma?

A

Can cause severe side effects, and sensitivity and response is highly dependent on disease phenotype

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
260
Q

What causes disease exacerbations in asthma?

A

Respiratory viral infections

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
261
Q

What is a doenside of ß2 agonist use?

A

It only targets one symptom and does not treat the inflammation

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

What happens to tissues in chronic asthma?

A

Irreversible tissue remodelling, goblet cell hyperplasia

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

What causes the rapid constriction of the airway in asthma?

A

Mast cell degranulation (histamine)

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

What time of onset of asthma is Th2-associated and which is non-Th2 associated?

A

Childhood is Th2 (T2 high), usually allergy driven.
Adult is non-Th2 (T2 low), obesity and smoking associated, the later neutrophilic. Also smooth muscle mediated

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

What cytokines and adaptive and innate cells does T2 high asthma involve?

A

IL-4, IL-5, IL-13 and IL-9, Th2 and ILC2 and honestly almost every immune cell, even B cells

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

What does IL-5 lead to?

A

Tissue eosinophilia

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

What does IL-4 lead to?

A

IgE

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

What do Th17 cytokines lead to in asthma?

A

T2 low asthma -> neutrophil recruitment

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
269
Q

What cells are involved in T2 low asthma?

A

Th1, Th17, macrophages and ILC1

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

What type of asthma is characterized by neutrophilic inflammation and involved IFNy and IL-17?

A

T2 low

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
271
Q

What type of asthma is characterized by eosinophilic inflammation, type 2 cytokines and allergen-specific B cells and IgE?

A

T2 high

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

What type of receptor do mast cells have?

A

Igε

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

What are alarmins?

A

Innate cytokines produced by epithelial cells that affect DCs and activate ILCs

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

What do dendritic cells do in T2 high astghma?

A

Take up allergen, migrate to LN, active naive T cells, which proliferate and differentiate into Th2 cells.

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

What are the two ways type 2 cytokine production is induced in T2 high asthma?

A

DCs to Th2 and alarmins to ILC2

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

What does IL-13 do?

A

Disturb epithelial homeostasis by affecting the tight junctions and causing goblet cell hyperplasia, cause activation and migration of DCs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
277
Q

Where are neuroendocrine cells mostly found?

A

Bronchioles

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

Where are goblet cells mainly found?

A

Trachea and bronchi, a bit in bronchioles

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

Where are submocosa glands found?

A

Trachea and bronchi

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

Where are club cells found?

A

Trachea, bronchi and bronchioles

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

What do ILC2 cells respoind to?

A

alarmins produced by the epithelium

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

What cells do ILC2s interact with?

A

Almost everything you can think of

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

What cell makes the most IL-25?

A

The tuft cell

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

What is IL-13 essential for?

A

DC activation andmigration in protease-induced allergic airway inflammation

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

What is a marker for activated ILC2s?

A

CD-45RO

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
286
Q

What is high levels of CD-45RO+ ILC2 associated with?

A

Asthma severity and unresponsiveness to corticosteroids

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
287
Q

What happens with chromatin in asthma patients?

A

It is open at genes encoding for ILC2

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
288
Q

How can viral infections exacerbate asthma symtpoms?

A

They can turn into cytotoxic T cells that produce Th2 type cytokines. It’s weird because type 1 cytokines usually surpress Th2 responses.

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

What is the cytokine production pattern of CD8 T cells in severe asthma patients?

A

Type 2 cytokine production by CD8 T cells is highest in severe asthma patients

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

What is the paradox in CD8 T cells responses in asthma?

A

After viral infections, CD8 T cells, which usually produce IFNy which counteracts Th2 responses, can produce type 2 cytokines, also in patients that are unresponsive to corticosteroids

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

What is inflammatory bowel disease?

A

An intolerance to microbiota in the intestines

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

What is dysbiosis?

A

There are large shifts in the microbiota present in the GI tract

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

What are 4 factors that can contribute to dysbiosis?

A

Genetics, lifestyle (diet, stress), early colonization (during birth), medical practices (vaccination, antibiotics, hygiene)

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

What part of the intestines does UC affect?

A

The colon, always one adjacent superficial area

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

What part of the intestines does Crohn’s disease affect?

A

Can affect any area in the GI tract with healthier areas and areas with lesions

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

What are cobblestones and what are they a hallmark of?

A

Crohns. They are healthy pieces of tissue buldging out due to scar tissue that pushes softer tissue out

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

What is transmural?

A

Affecting all layers (of the bowel wall in Crohn’s)

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

Do you see granulomas in IBD?

A

In 60% of Crohn’s patients

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

What are two big histological distinguisher sbetween Crohn’s and UC?

A

Granulomas (though only in 60% of patients) and transmural vs superficial

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

What are typical histological signs in Crohn’s?

A

Patchy, transmural, lymphocyte infiltratin, Granuloma presence (60%), ulcer formation

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

What are typical histological signs of UC?

A

Superficial, lymphocyte infiltration, empty goblet cells, presence of ulcerations and crypt absesses (cells within crypt lumen)

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

SNPs in what types of genes are involved in IBD?

A

Immune-related (Innate, adaptive, HLA, epithelium, etc)

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

How can we classify IBD to battle heterogeneity in IBD?

A

Genetic susceptibility in groups of genes associated with the same pathways. This way we can see what pathways are essential, what type of inflammation emerges from particular defects and what microbial changes can we relate to this?

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

What is Chronic Granulomatous Disease?

A

Defective neutrophil function which leads to IDB caused by defective defense.

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

What is IL-10 RA deficiency?

A

Defective regulation of IL-10 signaling which leads to IBD cause by hyperactive host defense

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

Do IBD patients usually only have one SNP?

A

No, it often is SNPs in multiple genes which makes treatment very difficult

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

What is NOD2?

A

A NOD-like receptor (PRR) expressed by monocytes, macrophages, DCs and gut epithelial cells. Activation leads to NF-kB activation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
308
Q

What is NOD2 polymorphisms associated with?

A

Crohn’s disease in the ileum.

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

How does NOD2 affect Paneth cells?

A

Promotes the production of AMPs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
310
Q

How does NOD2 affect stem cells?

A

Induces survival through resistance to oxidative stress

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

How does chronic NOD2 stimulation affect APCs?

A

Downregulates pro-inflammatory cytokine production through reduced TLR2 signals

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

How does NOD2 affect macrophages?

A

Inhances killing through inflammasome mediated caspase-1 activation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
313
Q

What type of signalling is crucial for epithelial barrier function in the gut?

A

NF-kB

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

What happens if NF-kB is knocked out?

A

Chronic severe intestinal inflammation and increased TNF-mediated cell death (apoptosis), leading to infiltration

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
315
Q

What drives colitis in IL-10 deficient mice?

A

Helicobacter (microbiota in general)

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

What drives small intestine inflammation in IL-10 deficient mice?

A

Dietary antigens

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

Why does IL-10 deficiency in Tregs mainly affect the colon and not the small intestines?

A

There are other cells in the small intestine that can produce IL-10 such as Tr1, so there is more of a division of labor.

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

What is celiac disease?

A

Gluten induced inflammation of the GI tract

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

What is inflammed in celiac disease?

A

The proximal intestines, causing perturbation of nutrient uptake

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

Is there a cure for celiac?

A

No

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

Does celiac have genetic susceptibility?

A

Yes, HLA-DQ2 (95% of cases) or HLA-DQ8.

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

Does HLA-DQ2 lead to celiac?

A

No, only a minority get celiac, though 95% of celiac cases have HLA-DQ2

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

What are histological hallmarks of celiac disease?

A

Duodenal crypt hyperplasia and duodenal villous atrophy

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

What types of T cells can be found in by gluten-inflammed intestinal tissue?

A

CD8 intraepithelial lymphocytes and gliadin-specific IFNy-secreting CD4 T cells in the lamina propria

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
325
Q

What can be found in the serum of celiac patients?

A

autoantibodies against tissue transglutaminase 2

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

What drives celiac disease chronicity?

A

Gliadin-specific memory CD4 T cells

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

Why is HLA-DQ2 associated with celiac?

A

Deamidated gluten peptides fit in HLA-DQ2

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

How is celiac diagnosed?

A

anti-tissue transglutaminase 2 antibodies in the serum combined with a positive HLA-DQ2 or 8 test

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

Explain pathogenesis of celiac.

A
  1. Gluten is ingested
  2. Gluten passes the epithelial barrier, possibly through diffusion
  3. Deamidation by tissue transglutaminade 2
  4. Presentation by HLA-DQ2 of 8 to T cells
  5. Inflammatory T cell differentiation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
330
Q

Do anti-tissue transglutaminase 2 antibodies contribute to celiac?

A

No

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

Why are anti-tissue transglutaminase 2 antibodies produced?

A

The gliadin-tissue transglutaminase 2 complex is likely no longer seen as “self”

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

What is the most important cytokine in celiac disease?

A

IL-15 (but there is also IFNα and IL-21)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
333
Q

What are possible inducers of IL-15?

A

Virus/intracellular pathogens, IFNα, gliadin peptides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
334
Q

What are two adaptive responses during celiac disease?

A
  1. Inflammatory gliadin-specific HLA-DQ-restricted CD4 T cells secrete IFNy
  2. Plasma B cells secrete anti-tissue transglutaminase 2 (TG2) antibodies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
335
Q

What are two innate responses during celiac disease?

A
  1. Innate epithelial cells are stressed and secrete IL-15 and express NK-ligands
  2. CD8 MHCI intraepithelial lympocytes (IELs) lyse epithelium
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
336
Q

How does the pathogenesis of celiac work regarding IELs?

A
  1. CD4 T cell derived IFNy causes epithelial cell stress and activates cytotoxic IEL
  2. IL-15 production by stressed epithelial cells co-stimulates cytotoxic IEL
  3. Cytotoxic IELs kill epithelial cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
337
Q

What is the role of IL-10 in mice regarding celiac?

A

Mice require IL-10 to maintain tolerance against gluten-derived gliadin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
338
Q

What does the mucosa refer to?

A

Everything from the lumen to the muscularis mucosa

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

What is the normal villi-crypt ratio?

A

3:1 (to 5:1) in adults, 2:1 in children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
340
Q

What is the normal amount of intraepithelial lymphcytes in the villi of the duodenum and jejunum?

A

Up to 1 in 4 (25 per 100)

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

Where are biopsies to test for celiac taken from?

A

Duodenal bulb and the distal portion of the duodenum

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

What are 2 pre-requisites for accurate diagnosis with celiac via biopsy?

A

The tissue fragments need to be well-oriented and they must be performed when the patients consumes a gluten-containing diet

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

What are the 4 cardinal histological markers for celiac disease?

A
  1. lower villi:crypt ratio (atrophy of the villi)
  2. crypt hyperplasia
  3. increase of interepithelial lymphocytes (IE lymphocytosis)
  4. chronic inflammatory cell infiltration in the lamina propria
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
344
Q

What does atrophy mean?

A

To waste away due to degeneration of cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
345
Q

How is celiac severity scored?

A

Proportion of lymphocytes and levels of crypt hyperplasia and villous atrophy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
346
Q

What two things are part of normal ileum architecture?

A
  1. Increased proportion of goblet cells compared to other parts of the small intestine
  2. Presence of Peyer’s Patches
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
347
Q

What 3 things are part of normal colon architecture?

A
  1. Crypts lined with columnar cells
  2. Crypts oriented parallel to one another and perpendicular to muscularis mucosa
  3. Lamina propria is mostly lymphocytic and plasmacytic, with varying numbers of eosinophils and mast cells
348
Q

What are differential diagnoses?

A

Possible diagnoses

349
Q

How should a pathologist assess the colon for IBD?

A

Between different sites, between biopsies from the same site and within each biopsy

350
Q

What are three patterns of inflammation in an intestinal biopsy?

A

Diffuse, patchy, focal

351
Q

How can you determine whether there is evidence of basal lymphoplasmacytosis?

A

Infiltration of cells in an area between the crypts and the muscularis mucosa (there should not be space between those)

352
Q

What does a crypt abcess look like?

A

Immune cells in the lumen of crypts

353
Q

What is it called when there is space between the crypts and the muscularis mucosa filled with immune cells?

A

Basal lymphoplasmacytosis

354
Q

What do crypts look like in Crohn’s disease?

A

Distorted: splitting or loss, etc

355
Q

What is it called when there are immune cells between the epithelial cells of the crypts?

A

Cryptitis

356
Q

Are paneth cells usually found in the crypts of the colon?

A

No. If they are it is called Paneth cell metaplasia

357
Q

Should mucus (goblet) cells be present or visible in the colon?

A

Yes. If not it’s called mucin depletion

358
Q

In summary, what are the 4 things to look for in IBD (histologically?)

A
  1. Architectural changes
  2. Chronic inflammation of the lamina propria
  3. Acute inflammation
  4. Epithelial changes
359
Q

What is found only in Crohn’s disease and not in ulcerative colitis?

A

Granulomas

360
Q

How long does it take for pedriatric IBD to resolve?

A

It’s incurable and lifelong

361
Q

Is Adult IBD or pediatric IBD more severe?

A

Pediatric

362
Q

What is the inflammation pattern in Crohn’s versus UC?

A

Crohn’s: whole GI, transmural
UC: Colon only, diffuse, mucosal inflammation

363
Q

What is the first diagnostic tool to be used to diagnose IBD?

A

Endoscopy

364
Q

What are four things to measure for in blood for Crohn’s disease diagnosis?

A

CRP (acute inflammation), Erythrocyte sedimentation rate (ESR, marker of systemic inflammation), low albumin (protein loss/malnourishment), high calprotectin

365
Q

What can you find in UC patients in the blood?

A

Low iron/hemoglobin (anemia) due to bleeding

366
Q

What are 4 procedures to diagnose IBD?

A
  1. Endoscopy
  2. Bloodwork
  3. Stool analysis
  4. Maybe ultrasound (constriction in Crohn’s)
367
Q

What does infliximab do?

A

Anti-TNF antibody

368
Q

How can Infliximab be used in Crohn’s and what does it do?

A

Infliximab is best used right after diagnosis to prevent further irreversible destruction of the intestines.
It induces mucosal healing by blocking TNF.

369
Q

How can we predict whether patients will be able to switch to dietary intervention after Infliximab or have to maintain Infliximab use?

A

Patients with low pre-treatment levels of Infliximab-modulated proteins are more likely to be able to stop Infiliximab treatment after remission and switch to dietary intervention

370
Q

What is autografting?

A

Transfer of living cells, tissues or organs from one part of the (same) body to another

371
Q

What is allografting?

A

Transfer of living cells, tissues or organs from one individual to another

372
Q

What is xenografting?

A

Transfer of living cells, tissues or organs from one species to another

373
Q

What is the allo-immune response?

A

An immune response to non-self antigens from members of the same species (allo-antigens)

374
Q

What two types of rejection are there?

A

Cellular rejection and antibody-mediated rejection

375
Q

In what two ways can antigens be presented to B cells?

A

Directly to the B cell, or through helper T cells

376
Q

What 3 types of allo-antigens are there?

A
  1. Major histocompatibility complex (MHC) (or HLA) antigens
  2. Minor histocompatibility complex antigens (miHA/non-MHC antigens)
  3. Blood group antigens
377
Q

What type of antigen causes the strongest and fastest rejection?

A

MHC. It is the main cause of grant rejection

378
Q

What type of antigen causes slow and weak graft rejection?

A

miHA or non-MHC antigens. It requires different antigen processing.

379
Q

What mismatched cells can be recognized by CD8 cells?

A

All nucleated cells

380
Q

What mismatched cells can be recognized by CD4 cells?

A

Antigen presenting cells and activated CD4 cells

381
Q

How are minor allo-antigens presented?

A

They are taken up and presented by recipient HLA molecules to recipient T cells, like all other peptides

382
Q

What are minor-allo antigens?

A

All non-MHC proteins that differ in amino acid composition between toe donor and recipient outside of MHC due to mutations or genetic polymorphisms

383
Q

Does presentation of red blood cell antigens require T cell help?

A

No, they immediately activate B cells

384
Q

What types of antibodies do you have if you are blood group A?

A

Anti-B

385
Q

What type of antibodies do you have if you are blood group B?

A

Anti-A

386
Q

What type of antibody do you have if you are blood group AB?

A

None against blood groups

387
Q

What type of antibody do you have if you are blood group O

A

Both Anti-A and Anti-B

388
Q

What is the most important allo-antigen recognition pathway?

A

Direct allo-recognition, of intact foreign HLA molecules on donor APCs by recipient T cells

389
Q

What is cross-reactivity in allo-recognition?

A

When T cell receptors can recognize peptides presented by intact foreign HLA molecules, causing an immune response

390
Q

What is peptide-independent binding?

A

When the recipient TCR recognizes the donor MHC but not the peptide

391
Q

What is peptide-dependent binding?

A

When the recipient TCR recognizes only the peptide but not the donor MHC presenting it

392
Q

What is foreign peptide:self MHC binding?

A

When the recipient TCR recognizes a foreign peptide on the recipient (own) MHC

393
Q

What is indirect allo-recognition?

A

When a donor peptide is taken up by a recipient APC and presented to the recipient TCR (through self-MHC)

394
Q

What is the semi-direct recognition pathway?

A

When an APC of the recipient presents a donor peptide on a donor HLA to a recipient TCR

395
Q

How can donor matching be tested in the lab?

A

Mixed lymphocyte reaction with T cell activation or cytokine production as read-outs

396
Q

What are two signs of kidney transplant rejection?

A

A rise in creatinine and descreased urine production

397
Q

What is done after creatininine and urine tests to prove kidney rejection?

A

Biopsy and histology

398
Q

What are measurable signs of liver rejection?

A

Rise in serum aminotransferases (ASAT, ALAT), alkaline phosphatase, bilirubin

399
Q

How is heart rejection tested?

A

Biopsy. There is no other biochemical way.

400
Q

What is a 4th target for organ transplant rejection?

A

Injury

401
Q

What happens when there is a blood group mismatch?

A

Blockages of capillaries, hyperacute rejection within minutes or hours. Causes severe damage by antibodies of the recipient

402
Q

What is hyperacute rejection?

A

Blood group mismatch causes severe damage to the transplant, causing organ loss within minutes or hours

403
Q

Can there already be pre-existing anti-donor HLA antibodies?

A

Yes, due to pregnancy, blood transfusions or earlier transplantations

404
Q

What are the 2 requirements for organ transplantation?

A
  1. ABO system compatibility
  2. Negative cross match (except for liver)
405
Q

How does tissue injury play a role in rejection?

A

reactive oxygen species, cellular damage, imflammatory cytokines, immune cell attraction and attachment of immune cells to inflamed endothelium

406
Q

Why can’t cardiac death donors donate?

A

Stopped circulation leads to lack of oxygen supply to the organ

407
Q

Why can’t brain dead donors donate?

A

Cytokine storm

408
Q

How can tissue injury be minimized?

A

Machine preservation

409
Q

What is the type of rejection based on?

A

The type of immune cells involved and how quickly rejection occurs

410
Q

What is hyperacute rejection?

A

Immediate lysis of donor cells due to ABO or donor HLA specific antibodies

411
Q

What type of rejection can take weeks or years?

A

Humoral rejection: B cell-mediated (antibody-mediated) rejection

412
Q

How can humoral rejection be detected?

A

C4d staining in capillaries

413
Q

What is CMV seropositivity a risk factor for?

A

Antibody-mediated rejection

414
Q

How can you treat acute cellular rejection?

A

High doses of methylprednisolone, otherwise T cell depletion

415
Q

How can you treat acute antibody-mediated rejection?

A

Plasmapheresis (antibody depletion), IVIg, otherwise Rituximab (anti-CD20) therapy

416
Q

What is rituximab?

A

Anti-CD20 therapy, so B cell depletion

417
Q

What are signs of chronic kidney rejection?

A

Increase of serum creatinine, proteinuria, hypertension

418
Q

What are two reasons chronic organ rejection happens?

A
  1. Low levels of immune attack against the organ
  2. Chronic calcineurin inhibitor (Tacrolimus) nephrotoxicity
419
Q

What is needed for improved immunosuppressive therapy?

A

Something as effective or more effective than Tacrolimus, but less nephrotoxic

420
Q

What Class I HLAs are mostly looked at in transplants?

A

A and B

421
Q

What Class II HLAs are mostly looked at in transplants?

A

DR

422
Q

When is Class I C and Class II DQ and DP also looked at for matching?

A

Stem cell transplants

423
Q

Why is strict stem cell matching important?

A

Because there is a huge risk of rejection but also that the new immune system also attacks the recipient body

424
Q

What are two clinical consequences of mismatching in solid organ transplant?

A
  1. Host versus graft (HvG) -> rejection
  2. Graft versus host (GvH) -> GvHD
425
Q

In what setting can you get host versus graft and thus rejection?

A

Usually HLA mismatching in solid organ transplants

426
Q

In what setting can you get graft-versus-host and this graft versus host disease?

A

In hematopoetic stem cell transplation when the stem cells differentiate into immune cells that attack the recipient immune system due to HLA mismatching

427
Q

What can cause sensitization before transplantation?

A

Pregnancy, previous transplants and blood transfusion

428
Q

What is sensitization in transplantation?

A

Formation of anti-HLA antibodies such as against paternal HLA molecules during pregnancy, making it more difficult to find a suitable donor

429
Q

What happens when anti-HLA antibodies encounter a mismatched organ?

A

Hyperacute rejection, which happens within minutes or hours

430
Q

What has reduced the incidence of hyperacute rejection?

A

Serological crossmatching

431
Q

What causes de novo antibody formation after transplantation?

A

Humoral alloimmune responses

432
Q

What can de novo antibody formation after transplantation lead to?

A

Acute humoral rejection and chronic transplant dysfunction

433
Q

What are 4 HLA screening methods used in the lab?

A

HLA typing, HLA antibody screening, crossmatching and donor search

434
Q

What is a luminex platform?

A

A multiplex ELISA with color-coded beads with specific HLA oligonucleotides

435
Q

What types of samples are used in crossmatching assays?

A

Patient serum and donor cells from the blood or spleen + complement

436
Q

What is flow crossmatch?

A

Same as normal crossmatching, but with an anti-IgG label instead of complement to measure using flow cytometry

437
Q

What are 5 reasons to do a renal biopsy?

A
  1. Provides a diagnosis (toxicity, rejection, viral, donor disease)
  2. Guides treatment (changes in more than half of the cases)
  3. Predicts prognoses
  4. Reveals pathogenesis
  5. Validates outcome (as endpoint result in clinical trials)
438
Q

What are 6 pitfalls in nephropathology?

A
  1. Morphology not specific
  2. Sample size
  3. Sample location might not show primary lesion
  4. Chronic changes can obscure origin or pathology
  5. Differences in pathologist experience
  6. Sampling error (might miss lesion)
439
Q

Name 4 examples of clinical information that is important to know when looking at nephropathology?

A

Anti-donor HLA antibodies
Original disease
Drug therapy
Time post transplant

440
Q

What is the Banff classification?

A

Standardized grading system to grade severity and type of rejection

441
Q

What is needed for an accurate classification according to Banff?

A

A minimum of 7 glomeruli and 2 arteries, but depends on the disease

442
Q

What are the two main types of rejection in the Banff classification?

A

Antibody-mediated rejection and T-cell mediated rejection

443
Q

What are the three levels of TCMR (histologically)

A

-bTCMR (borderline)
-I Tubulo-interstitial
-II Vascular rejection

444
Q

What can be found in AMR?

A

Antibodies, monocytes, neutrophils, and NK cells

445
Q

What morphological change is found in AMR?

A

Microvascular inflammation

446
Q

What is needed for an AMR diagnosis?

A

C4d+ and donor-specific antibodies (DSA+)

447
Q

What is BK nephrology?

A

Nephropathology caused by a polyomavirus, characterized histologically by enlarged nuclei (caused by viral replication in the nucleus)

448
Q

What is being done to improve diagnosis of transplant rejection?

A

microRNA profiling

449
Q

What 3 types of immunosuppression are there?

A

Induction therapy, maintenance therapy and anti-rejection therapy

450
Q

When is induction therapy given?

A

Shortly before and after the transplant surgery to fully knock out the immune system

451
Q

What type of induction therapy is given in a standard kidney transplantation?

A

Signal 3-blocking antibody (cytokines)

452
Q

What is blocked by the induction therapy used in The Netherlands?

A

Anti-CD25, which clocks the IL-2 receptor

453
Q

Why not use signal 1 antibodies?

A

A lot of side effects due to complete T cell depletion (infection susceptibility)

454
Q

What are 3 standard maintanence drugs given?

A

Tacrolimus (chronic calcineurin inhibitor), mycophenolate mofotil (MPA/MMF) and prednisolon

455
Q

What does Tacrolimus do?

A

Blocks downstream of signal 1

456
Q

How does prednisolon work?

A

It works anti-inflammatory and immuniosuppressive by inhibiting mRNA expression when interacting with corticosteroid receptor, mimicing cortisol. It inhibits T cell growth and differentiation.

457
Q

What is the standard immunosuppressive regimen in kidney transplantation in The Netherlands?

A
  1. High dose immunosuppression: Induction with Anti-CD25 (basiliximab), Prednisolon, Tacrolimus and MMF
  2. Taper down prednisolon for 3 months, also tacrolimus and MMF
  3. Tacrolimus and MMF only, no more prednisolon
458
Q

What are 3 additional immunosuppressive therapies in case of rejection?

A
  1. Methylprednisolon (always, high dose intravenously)
  2. Alemtuzumab (T and B depletion)
  3. IVIg (little evidence that it works)
459
Q

What are 3 major adverse effects of immunosuppressive drugs?

A

Infections, malignancies and cardiovascular disease

460
Q

Which immunosuppressive drug needs to be monitored closely and why?

A

Tacrolimus due to the small therapeutic window and interactions with other drugs

461
Q

How can drugs be monitored?

A

Measuring trough levels

462
Q

Which drug affects antiviral immunity the most?

A

Prednisolon

463
Q

What are 3 advantages of cellular therapies?

A
  1. Less side effects
  2. Long term effects (cells stick around)
  3. Cellular therapy may create a balance between activation and suppression, potentially in a donor antigen specific manner
464
Q

What cells have regulatory properties?

A

Myeloid cells, Mesenchymal stem cells (MSC) and lymphoid cells

465
Q

In what two ways can myeloid regulatory cells play a role in homeostasis?

A

Either directly or by affecting Tregs

466
Q

What is pharmacokinetics?

A

What a body does to a drug

467
Q

What is pharmacodynamics?

A

What a drug does to the body

468
Q

What does ADME stand for?

A

Absorption, distribution, metabolism and elimination

469
Q

What is the absorption in an infusion?

A

almost 100%

470
Q

What is oral absorption dependent on?

A

How much the medication is absorbed and the form of the medicine (delayed-response, enteric-coating, etc.)

471
Q

What is bioavailability?

A

The fraction of an administered dose of an unchanged drug that reaches the systemic circulation. It is stated as a percentage in respect to intravenous form.

472
Q

What is F or BB?

A

Bioavailability

473
Q

What is Vd?

A

The volume of distribution

474
Q

How do you calculate the Vd?

A

Ab (drug quantity in the body)/C (blood concentration) or F*Dose/plasma concentration

475
Q

Where can a drug be distributed into?

A

Hydrophilic tissues (blood, muscles), lipophilic tissues (dermis, nerves, fat), brain (through BBB), placenta/breastmilk

476
Q

What does the volume of distribution mean?

A

The apparent or hypothetical volume in which the drug is dispersed

477
Q

Is the Vd of plasma high or low?

A

Low, around 5 liters

478
Q

What kind of drugs do you need if it has to be in the plasma?

A

High molecular weight or those that bind to plasma proteins exclusively

479
Q

What kind of drugs will distribute in the extracellular fluid?

A

Low molecular weight but hydrophilic

480
Q

What types of drugs will distribute in the intracellular fluid?

A

Those that bind strongly to tissues.

481
Q

What is the extracellular fluid Vd?

A

Equal to total water volume: ~42 L

482
Q

What is the water proportion of newborns compared to adults?

A

Much higher, around 80%. For Male adults 60 and Female adults 50

483
Q

What is metabolism?

A

Chemical conversion in the body (biotransformation)

484
Q

Why must a drug be metabolised?

A

To be converted into a water soluble form to be excreted by the kidneys

485
Q

What can genetic polymorphisms of CYP enzymes do?

A

They can speed up, slow down or otherwise change the normal breakdown of a drug

486
Q

What can drug-drug interactions do?

A

They can speed up, slow down or otherwise change the normal breakdown of a drug

487
Q

What happens when a fast metabolizer takes codeine and why?

A

Codeine is a pro-drug, which means it needs to metabolized into morphine before it can take effect. High metabolizers thus have a faster and stronger morphine effect, since more morphine goes into the system faster

488
Q

What happens when a fast metabolizer takes codeine and why?

A

Codeine is a pro-drug, which means it needs to metabolized into morphine before it can take effect. Slow metabolizers thus have a slower and weaker morphine effect, since more morphine goes into the system much slower

489
Q

How can a drug be excreted?

A

Via the urine, bile or feces

490
Q

What do you do with drug administration if someone has a decreased kidney function?

A

Dose must be decreased, since the drug will stay in the system longer.

491
Q

What are intrapatient reactions?

A

Differences in reactions of a patient to the same medication in multiple or repeated dosings

492
Q

What are interpatient reactions?

A

Differences between a group in how individuals react to a medication

493
Q

What is Tmax?

A

The time at which the blood concentration is at its highest

494
Q

What is Cmax?

A

The maximum concentration

495
Q

What is clearance?

A

The volume of plasma cleared of the drug per unit time

496
Q

What is renal clearance?

A

Via the kidneys

497
Q

What is hepatic clearance?

A

Out of the liver

498
Q

What is T1/2?

A

The time necessary to decrease the blood concentration of a medication by 50%

499
Q

What can prolong T1/2?

A

renal impairment of intoxification

500
Q

What is the general rule of when a medication is out of the blood?

A

4-5x T1/2

501
Q

When is a drug level steady?

A

3-5x T1/2

502
Q

What is AUC and when can it increase?

A

The area under the curve when concentration is plotted against time. It can increase when clearaqnce is decreased, which leads to accumulation and increases chances of side effects and toxicity

503
Q

What is the therapeutic range?

A

The window of time during which a drug is effective but not (as) toxic

504
Q

What is pharmacogenetics?

A

DNA analysis to explain or predict the response of a patient to drug therapy

505
Q

What are three main factors that determine how fast a drug is metabolized?

A

The dose, kidney function and liver metabolizing capacity (through CYP450 enzymes)

506
Q

Cytochrome P450 enzymes are responsible for the metabolism of what drugs?

A

80% of all drugs

507
Q

What causes someone to be an ultrarapid metabolizer?

A

3 copies of the CYP450 enzyme in question

508
Q

When is someone an intermediate metabolizer?

A

Usually one allele has a polymorphism associated with poor metabolism

509
Q

What causes poor metabolism?

A

Both alleles have polymorphisms associated with poor metabolism

510
Q

What CYP450 enzyme metabolizes antidepressants among many other drugs?

A

CYP2D6

511
Q

What do you have to be careful of when as patient does not react to prescribed medication?

A

They might not be adhering, it does not necessarily mean they are ultra-rapid metabolizers

512
Q

What effect can CYP2D6 polymorphisms have on codeine or tramadol?

A

They can make it so the patient has no pain relief

513
Q

What is voriconazol?

A

Antifungal

514
Q

What enzyme metabolizes Voriconazol?

A

CYP2C19

515
Q

Why are CYP polymorphisms important in transplant patients?

A

Some polymorphisms can cause poor or fast metabolism or Tacrolimus, causing either acute rejection when metabolized too fast (low concentration) or nephrotoxicity when metabolized too slow (high concentration). This is especially important since Tacrolimus has such a small therapeutic window.

516
Q

What does CYP2C19 do to voriconazol?

A

It inactivates it

517
Q

What CYP inactivates Tacrolimus?

A

CYP3A5 when expressed, most people are non-expressers

518
Q

What are the three meningeal layers that envelop the brain and spinal chord, from apical to basal?

A

Dura, arachnoid mater and pia mater

519
Q

What is the subarachoid space?

A

It is the space between the arachoid mater and pia mater that contains the cerebral spinal fluid (CSF) and blood vessels, the latter of which penetrates into the brain and spinal chord tissues as the blood-brain barrier.

520
Q

What does it mean when you can find inflammatory cells in the CSF?

A

The patient has meningitis

521
Q

Where is CSF produced?

A

In the choroid plexus

522
Q

What is cerebritis?

A

Inflammation of the brain tissue (encephalitis)

523
Q

What can cause brain abcesses?

A

Endocarditis, lung infections, abdominal infections (i.e. peritonitis)

524
Q

What is the first most fatal aspect of meningitis?

A

Brain swelling -> compressed vessels, leaking and infarctions

525
Q

What is a brain abcess?

A

A non-preformed space in the body that contains (dead) leukocytes, encapsulated by fibroblasts

526
Q

What is alzheimers disease characterized by?

A

Accumulation of amyloid-beta plaques and tau tangles

527
Q

What role do microglia play in alzheimers?

A

They can clear amyloid-beta plaques through phagocytosis, releasing IL-10 and TGFß for anti-inflammatory purposes, but can also lead to chronic inflammation and degeneration by releasing pro-inflammatory IL-12 and IL23

528
Q

What happens in gliomas?

A

An immunosupressive microenvironment is created that gliomas exploit to be able to evade and continue growing. TILs are recruited but impaired, microglia and macrophages polarize to M2 and accidentally support growth and supress immune responses, Tregs are involved, and gliomas express PD-L1.

529
Q

Is the optic nerve part of the CNS or PNS?

A

CNS

530
Q

What are 5 ways tumors avoid the immune system?

A
  1. Low immunogenicity (low receptor expression)
  2. Tumor treated as self antigen (no co-stimulation)
  3. Antigenic modulation
  4. Tumor-induced immune supression
  5. Tumor-induced privilged site
531
Q

What can pass the blood brain barrier?

A

Small molecules, and large molecules only in a very controlled way

532
Q

What is the glia limitans?

A

A second tight border on the other side of the perivascular space from the BBB that tightly controls entry and exit of cells and molecules into the parenchyma

533
Q

What is the phenotype of CNS T cells?

A

Generally more inhibitory, such as Tregs

534
Q

What is the natural habitat of CNS T cells?

A

The perivascular space

535
Q

Why doesn’t the parenchyma iliccit immune responses?

A

Microglia are much less able to express traits necessary for T cell activation (tolerogenic)

536
Q

What 3 things causes this tolerogenic phenotype in microglia?

A

They require multiple cytokines for activation, are moderate APCs and express few co-stimulatory molecules

537
Q

What can neurons do to prevent inflammation?

A

They express high levels of CD200R (the perivascular space is loaded with anti-inflammatory CD200)

538
Q

What is one of the main points of entry of T cells?

A

The choroid plexus

539
Q

What controls diapedesis of activated immune cells?

A

Inflamed endothelium

540
Q

What are the 5 ways the CNS is immune priviliged?

A
  1. Parenchymal APCs cannot provide effective activation
  2. Parenchyma produces anti-inflammatory signals
  3. Lymphocyte presence is strictly compartimentalized
  4. BBB
  5. T cells need to be re-activated after crossing BBB
541
Q

What viral infection is a prerequisite for developing MS?

A

Epstein-Barr virus

542
Q

What cells are affected by Epstein-Barr Virus?

A

Memory B cells

543
Q

What are 4 risk factors of MS?

A

Female (2:1)
Genetics
MHCII HLA-DRB1 variant
Environmental risks (vitamin D, smoking, EBV)

544
Q

Is MS similar across the lifespan?

A

No. Even babies can get MS. Most common age of onset is in early 30s though

545
Q

What does MS look like in younger people?

A

Attacks that people can recover from.

546
Q

What is the dominant phenotype of MS in older people?

A

Slow progression of disability, steady accumulation of neurological dysfunction, resistant to almost all drugs that are given to people that get attacks.

547
Q

What can be tested to monitor MS or learn about it?

A

Genetics, Blood, CSF, brain biopsies, autopsies

548
Q

Do you normally have B cells in your brain?

A

No, hardly any

549
Q

Are there naive T cells in the CNS?

A

No, no reason to search for antigens there

550
Q

WHat types of T cells do you see in the CSF?

A

Memory and effector CD4 cells, and some memory CD8 cells

551
Q

What is the dominant phenotype of T cells in the perivascular space compared to in the CSF?

A

CD69+ (early activation marker)

552
Q

Where are CD69- CD103- CD8 T cells mainly found with regards to the CNS?

A

The border organs (blood, choroid plexus and meninges

553
Q

Where are CD69+ CD103- and CD69+ CD103+ CD8 T cells mostly found with regards to the CNS?

A

CSF and white matter

554
Q

What could be the initiator of an MS attack?

A

Can be several mechanisms including:
1. peripheral antigens without presentative activation of CNS patrolling cells, or
2. presentation of CNS antigens in the periphery
3. Tregs are less functional and cannot control immune activation

555
Q

What do disease modifying therapies in MS act on?

A

Processes in the lymph nodes, such as T cell activation, clonal expansion, migration, cell depletion or blocking from crossing barriers

556
Q

What is the general lymphocyte phenotype in the blood of people with MS compared to healthy patients?

A

CD4s have a more cytotoxic phenotype, more memory B cells with a CSF infiltrating phenotype (CXCR3 expression -> CCR10, highly expressed in CSF)

557
Q

What is the most immunogenic region in the CNS?

A

The perivascular space

558
Q

When can cells enter the parenchyma?

A

Once they have interacted with an APC in the perivascular space

559
Q

What is found in white matter lesions of early in MS?

A

A lot of T cells, B cells and myeloid cells

560
Q

Where is inflammation mostly found in early MS

A

Far more in white matter than grey matter

561
Q

What is characteristic of lesions in the brain of most people who have died due to MS?

A

The lesions are still populated by MHCII cells that are demyelinating and expanding, tissue resident memory T cells and B cells

562
Q

What is interesting about B cells at end stage MS?

A

There is still continuous production of IgG, which may contribute to the progression of MS

563
Q

What does the presence of follicle-like structures in the meninges tell you about the disease progression?

A

These B and T cell interaction hotspots (where IgG synshtesis is stimulates) is associated with early onset of MS, faster accumulation of disability and earlier death

564
Q

When are grey matter lesions more or a problem in MS?

A

Later stages

565
Q

What are the three different MS patterns?

A
  1. Relapsing-remitting MS
  2. Secondary progressive MS
  3. Primary progressive MS
566
Q

What precedes secondary progressive MS?

A

Relapsing-remitting MS

567
Q

What are the three pathological hallmarks of MS?

A
  1. Influx of peripheral immune cells
  2. Demyelination
  3. Neurodegeneration
568
Q

What is a permanent aspect of MS?

A

Neurodegeneration. Myelin can be restored.

569
Q

What happens to microglia in MS lesions?

A

They become activated and turn into macrophages, that then damage the myelin

570
Q

What is the EAE model?

A

A mouse model in which an autoimmune response is induced against myelin, by injecting myelin protein with immune activators

571
Q

What are the 2 stages of axonal degeneration?

A

1: Focal swelling and 2: fragmentation

572
Q

What predicts axonal fate?

A

Ca2+

573
Q

What is the source of increased Ca2+?

A

Extracellular influx

574
Q

How does extracellular Ca2+ enter the axon?

A

Physical membrane damage (loss of integrity), so leaky

575
Q

How is membrane integrity (or disruption) tested in EAE models?

A

Injecting dyes of different sizes that may cross the membrane if there is loss of integrity

576
Q

What does the influx of Ca2+ cause?

A

Swelling and defragmentation

577
Q

What drives cortical pathology?

A

The immune cells in the meninges (meningial inflammation) that stimulate and activate the microglia

578
Q

What is the phenotype of MS2 (MS specific) microglia that is not found in other neurodegenerative diseases?

A

High branching, low homeostatic markers and low inflammatory markers (HLAII low)

579
Q

What is the CMI?

A

A chronic meningeal inflammation model to study cortical pathology in progressive MS

580
Q

What is the current hypothesis of pathogenesis of cortical pathology (in 4 steps)?

A
  1. Meningeal B cells secrete inflammatory factors
  2. Loss of microglia-mediated neuroprotection
  3. Impaired neuronal mitochondrial transport
  4. Neuronal necroptosis
581
Q

In what 4 ways does the skin act as a barrier?

A

Physical, microbiota, chemical, immune

582
Q

Why do people that suffer from food allergy also often suffer from itchiness?

A

There is a skin-gut axis due to similar homing molecules

583
Q

What are the main skin functions?

A

Protection/defense
Signal reception
Thermoregulation
Communication
Secretion
Absorption

584
Q

How does the skin thermally regulate the body?

A

Sweating, vasoconstriction/dilation

585
Q

What is sebum?

A

A sticky, oily substance produced by the sebaceous glands

586
Q

What can skin absorb?

A

Light, pharmaceutics

587
Q

What are the three main layers of the skin?

A

Epidermis, dermis, hypodermis

588
Q

What does the epidermis consist of?

A

Keratinocytes in 5 stratum layers:
Corneum
Lucidum
Granulosum
Spinosum
Basale

589
Q

What does the stratum basale consist of?

A

Basal cells (stem-cell like keratinocyte precursors), Merkel cells and melanocytes

590
Q

Where are langerhans cells located?

A

The stratum spinosum

591
Q

What do lamellar granules do in the skin?

A

Play a role in keratinization

592
Q

What is the function of the stratum lucidum?

A

Water resistance and barrier

593
Q

What does the stratum corneum consist of?

A

Dead cells

594
Q

What is the site of entry of microbiota?

A

The glands and hair follicles

595
Q

What prevents invasion of microbiota into the hair follicles and glands?

A

Monocytes and Tregs

596
Q

How do keratinocytes attach to the basal lamina?

A

Hemidesmosomes

597
Q

What does the basal lamina consist of?

A

Type IV collagen, laminin and proteoglycans

598
Q

What do desmosomes do?

A

They interconnect keratinocytes to each other, provide structural integrity but still allow passage of substances

599
Q

How is transport of substances limited in the epidermis?

A

Tight junctions, particularly in the stratum granulosum, enforcing cytoplasmic uptake

600
Q

Is all keratin expressed in skin cells the same?

A

No, different cells have different types of keratin made up of heterodimers of keratin with different functions (barrier, metabolism, differentiation, etc)

601
Q

How is photoprotection provided?

A

Melanocytes produce melanin and form melanosomes
Melanosomes are exocytosed
Melanosomes are endocytosed by keratinocytes
Exposure to sunlight causes migration of melanosomes to apical side of keratinocytes to protect

602
Q

What do Merkel cells do?

A

They are specialized epithelial cells especially present in thick skin. They are in close contact with sensory nerves and aid in perception of light touch

603
Q

What are the two dermis layers?

A

Papillary dermis and reticular dermis

604
Q

What is the papillary dermis?

A

It is highly vascularized and made up of a lot of loose connective tissue

605
Q

What is the reticular dermis?

A

Makes up the bulk of the dermis, made up of tight connective tissue

606
Q

What is the dermis populated by?

A

Next to collagen and elastic fiber, also fibroblasts, endothelial cells and innate and adaptive immune cells

607
Q

What is the vascularization pattern in the dermis?

A

Capillaries in papillary, arterioles and venules to arteries and veins in reticular dermis (thickest towards basal lamina)

608
Q

Is the skin a passive barrier?

A

No

609
Q

How is homeostasis maintained in the skin by the microbiota?

A

They themselves produce AMPs and they interact with keratinocytes and immune cells

610
Q

How do keratinocytes aid in innate immunity of the skin?

A

They express TLRs, signal for AMPs, produce inflammatory cytokines and chemokines to facilitate direct killing and recruit and modulate immune cells

611
Q

What does CCR7 do?

A

Aids in migration of langerhans cells to the lymph nodes

612
Q

What does CCR6 do?

A

It has CCL20 as a ligand, whch is produced by keratinocytes and other langerhans cells during inflammation to attract langerhans cells to the site

613
Q

What do langerhans cells do?

A

They have a sentinel function

614
Q

What are inflammatory reactions of langerhans cells?

A

Cytokine secretion (IL-1ß, TNF-α, IL-6, IL-12)
Chemokine secretion (recruitment)
IL-23: Th17 development

615
Q

What are tolerance reactions of langerhans cells?

A

IL-10 and TGFß
Co-stimulatory receptor downregulation

616
Q

How does TGFß play a role in skin homeostasis?

A

Wound healing: promotes keratinocyte proliferation, activates fibroblasts to produce ECM
Immune regulation: T cell differentiation into Tregs

617
Q

What do langerhans cells do in steady state?

A

They respond to commensals and self antigens by producing TGFß and IL-10 to induce antigen-specific Tregs
They also upregulate AXL to interact with apoptotic keratinocytes to facilitate clearance and trigger anti-inflammatory signaling and supress inflammatory signaling

618
Q

What cells represent the vast majority of dermal cells?

A

Phagocytes, mostly macrophages, but also dermal DCs and migrating and resident langerhans cells

619
Q

What is CGRP?

A

It is a neurotransmitter produced by nerves in the skin that is produced when sensing fungi or bacteria.

620
Q

What does CGRP do?

A

Downregulates langerhans APC function, increases IL-10 and IL-23 production in langerhans cells and downregulates production of pro-inflammatory cytokines by langers hans cells and dermal macrophages

621
Q

When is TAFA4 produced?

A

UV exposure by sensory neurons around hair follicles

622
Q

What does TAFA4 do?

A

Induces macrophage survival, induces IL-10 production which limits inflammatory response by newly arriving macrophages and induces healing

623
Q

Do mast cells have homeostatic functions or inflammatory functions?

A

Both!

624
Q

What plays a role in vaso-dilation and constriction?

A

TNFa produced by phagocytes and mast cells

625
Q

What do mast cells do in the skin?

A

-Vaso-dilation and activation
-Recruit inflammatory cells
-induce emigration and activation of resident leukocytes
-Peptide degradation
-Scratch reflex

626
Q

What do mast cells do in severe allergies?

A

Massive degranulation and prolonged inflammation, systemic reaction and tissue remodeling

627
Q

Can mast cells phagocytose?

A

Yes

628
Q

How can mast cells facilitate viral dissemination?

A

They can get infected and they can cause vascular leakage and thus viremia

629
Q

What do mast cells recruit during viral infections?

A

T cells and NK cells

630
Q

What do mast cells recruit during bacterial infection?

A

Neutrophils

631
Q

What do mast cells recruit during parasitic infection?

A

DCs

632
Q

What are extracellular traps?

A

The NETs of mast cells (against parasites)

633
Q

What are the transcription factors of ILCs?

A

Same as their helper counterparts

634
Q

What do CCR10+ ILCs do?

A

Facilitate effector and Treg homeostasis

635
Q

What do CCR6 ILCs do?

A

Facilitate barrier and microbiome homeostasis

636
Q

What ILC is involved in tissue repair?

A

ILC2

637
Q

What ILC plays a role in psoriasis?

A

ILC3 (Th17 does too)

638
Q

What cells play a role in contact hypersensitivity?

A

NKs and ILC1

639
Q

What is the host response (skin) against S. aureus?

A

TLR2 -> infl. cytokines -> NFkB -> AMPs and neutrophil recruitment
TLR2 -> inf. cytokines -> Th17, ydTcell, NK(T) -> AMPs and neutrophil recruitment

640
Q

Who produces AMPs in the skin?

A

Keratinocytes, leukocytes, adipocytes and commensals

641
Q

AMP production is increased or decreased in certain skin diseases, which ones?

A

Psoriasis: increased -> seldom bacterial infections
Atopic dermatitis: decreased -> higher susceptibility to infection and commensal imbalance

642
Q

How do AMPs work?

A

Lysis or sequestering iron

643
Q

Which T cell subsets are involved in the development of psoriasis and atopic dermatitis?

A
644
Q

What is the link between T cell transcription and cell plasticity?

A
645
Q

What are novel therapies for psoriasis and atopic dermatitis?

A
646
Q

What facilitates adhesion during T cell homing to the skin?

A

CLA and selectins on endothelial cells

647
Q

What is a chemokine receptor of T cells, B cells and DCs for homing to the skin?

A

CCR10

648
Q

What does TGFß help differentiate T cells into?

A

If also IL-6 -> Th17
If also IFNy -> Treg

649
Q

What makes T cells plastic?

A

They can be manipulated into changing their phenotype (fully or partially) to other T cell types through still available transcription factors associated with other types. It depends on the environment.

650
Q

What is the macroscopic hallmark of psoriasis?

A

Plaque formation

651
Q

What are the 4 histological features of psoriasis?

A

Acanthosis: thickening of the skin
Papillomatosis: Elongation of the epidermis into the papillary dermis
Hypogranulosis: Loss of stratum granulosum
Parakeratosis: Nuclei still present in stratum corneum cells
Also influx of T cells and neutrophils

652
Q

What are munro’s microabscesses?

A

Collections of neutrophils in the stratum corneum during psoriasis

653
Q

What happens during pre-psoriasis?

A

Innate and adaptive cell activation and recruitment, Th1, Th22 and Th17 -> AMP production

654
Q

What do epithelial cells do during psoriasis?

A

Hyperproliferation

655
Q

What triggers psoriasis?

A

Microorganisms, trauma, drugs combined with genetic susceptibility

656
Q

What cytokines are important in psoriasis plaque formation?

A

IL-23 and IL-17

657
Q

What type of treatment is effective against psoriasis?

A

Biologics targetting p19 subunit of IL-23, since p19 is specific for that cytokine and/or anti-p40 (IL-12 inhibitor) and/or anti-TNF. Some treatment is anti IL-17 or anti-IL-17

658
Q

What is atopic dermatitis initiated by?

A

Defective skin barrier in combination with genetic susceptibility

659
Q

What happens when the skin of an AD susceptible person is disrupted?

A

Activation -> Th2 type reaction including ILC2 (Th22 is also there) -> mast and eosinophil activation (histamines), IgE producing B cells.

660
Q

What T cells dominate the acute stage of atopic dermatitis?

A

Th2/Th22

661
Q

What T cells are activated as atopic dermatitis prgresses?

A

More Th2 and Th22 cells, but also Th1 and Th17 (though the question is if this is not just plasticity instead of recruitment)

662
Q

What types of itching pathways are there?

A

Histamine dependent and histamine-independent

663
Q

How is the histamine-dependent pathway initiated?

A

Histamines interact directly with histamine receptors on sensory neurons

664
Q

How is the histamine-independent pathway initiated?

A

Cytokines produced by Th2 cells (IL-4, IL-13 and IL-31) interact with their respective receptors on sensory neurons

665
Q

What is the synovial membrane made up of?

A

Type A (mAcrophage) and type B (fibroBlast) synoviocytes

666
Q

What are the layers of the joint lining?

A

The sublining, lining (fibroblast and barrier-forming macrophages) and the synovial fluid)

667
Q

How are type A synoviocytes connected?

A

Tight junctions

668
Q

What happens to the Type A lining during arthritis?

A

The lining is disrupted, enabling infiltration of myeloid cells

669
Q

What is the sublining composed of?

A

Different types of macrophages and fibroblasts (than those of the lining). There are many different fibroblast types in the sublining with different roles like proliferation, proinflammatory, and adhesion).

670
Q

What happens (diseasewise) in early RA?

A

Immune infiltration and proliferation of the synovial lining, blood vessel formation.

671
Q

What happens (diseasewise) in established RA?

A

Synovial lining is overlaying the cartilage, produced enzymes lead to cartilage and bone destruction

672
Q

What are chondrocytes?

A

Cartilage cells

673
Q

What do macrophages do in arthritis?

A

They release a lot of inflammatory cytokines as well as IL-10 and TGFß (to try to dampen the whole thing) and interact with T and B cells through soluble factors and direct contact

674
Q

What causes RA?

A

We don’t know, but environmental factors (smoking, stress, microbes) and genetic predisposition is involved

675
Q

What happens before clinical symptoms?

A

Pre-arthritis, which is a bit auto-immunelike but not yet arthritis. It might need an extra trigger (neurological, biochemical or microvascular events?) to progress.

676
Q

Can pre-arthritis be detected?

A

Yes, there are some antibodies that can be detected in some patients, such as IgG-RF and/or anti-CCP

677
Q

What are ACPAs directed against and what does binding do?

A

Citrullinated protein on osteoclast precursors. This causes further activation and maturation of osteoclasts and IL-8 production

678
Q

What do osteoclasts usually do?

A

Bone resorption which is necessary for maintenance, repair and remodeling

679
Q

How do ACPAs induce pain?

A

IL-8 produced by mature osteoclasts can interact with (sensory) nociceptor.
Recruitment of neutrophils can lead to inflammation and progression, which is also painful

680
Q

What does knocking out IL-12 in RA patients do, and why does knocking out IFNy not have the same effect?

A

Dampens symptoms, while knocking out IFNy makes symptoms worse. This is because knocking out IL-12 also knocks out IL-23 (due to the p40 subunit they share, so actually IL-23 is important here

681
Q

So what is driving RA autoimmune processes?

A

IL-17/IL-23 (so Th17), not Th1

682
Q

What does Th17 have an effect on?

A

Monocytes, B cells and fibroblasts

683
Q

What is the proportion of IL-17 in early RA patients, and why?

A

Higher compared to healthy controls, due to a CCR6 polymorphism

684
Q

What is the pro-inflammatory loop in RA?

A

Th17 ->TNFα and L-17 ->fibroblast ->IL6 and IL8 which act on Th17 and so on.

685
Q

Is Th17 plastic?

A

Ya

686
Q

What are possibel treatments?

A

Rituximab, CD80/86 inhibitors, JAK inhibitors, TNF inhibitors, IL6R inhibitors

687
Q

What are the two kinds of RA?

A

Antibody-mediated (70%) and non-antibody mediated (30%)

688
Q

How can antibodies contribute to inflammation?

A

Activating complement and binding to Fc receptors on macrophages

689
Q

What is RF?

A

Rhematoid factors are autoantibodies directed against IgG Fc that can form enormous immune complexes

690
Q

What is ACPA?

A

Antibodies (general) against Citrullinated Protein antigens. It is a general term for different autoantibodies against citrullinated epitopes than can be inside or outside the joint

691
Q

What is anti-CCP?

A

Anti-cyclic citrullinated Peptide, which is an artificial peptide used to measure ACPA using ELISA (so anti-ACPA)

692
Q

What is citrullination?

A

Totally normal. Post-translational amino acid modification (arginine -> citrulline), can occur in every protein

693
Q

Why is smoking a risk factor?

A

Inflammation (lungs) results an increase citrullination and breakdown of tolerance in genetically predisposed individuals

694
Q

What 3 clinical outcomes are there in RA?

A

Self-limiting, presistent non-erosive and persistent-erosive. It is important to be able to determine who is who for treatment (or not)

695
Q

What are strong predictors of presistent RA?

A

IgM-RF and Anti-CCP

696
Q

What are the 3 roles of B cells in RA pathogenesis?

A

Antibody production, antigen presentation to T cells and cytokine production via cell-cell interaction

697
Q

What joints are involved in RA?

A

Small joints of hands and feet, symmetrical

698
Q

What are inflammation markers in the lab for RA?

A

C reactive protein (acute inflammation) and ESR (chronic inflammation)
Also RA and ACPA

699
Q

The earlier you treat a disease…

A

the better the outcome!
Window of opportunity

700
Q

What are glucocorticoids, what is the downside and how to go about that?

A

Part of initial RA treatment that can prevent radiological progression. They work very fast.
Downside: They cannot be taken for a long time, so they are used as bridging therapy until other therapies kick in

701
Q

What is Methotrexate?

A

The most important initial RA treatment. They inhibit IKK-NFkB pathway and thus TNF

702
Q

Where is the cell body of the sensory neurons?

A

Dorsal root ganglia

703
Q

Where it the cell body of the motor neurons

A

In the brain stem or spinal cord

704
Q

What part of the motor neuron is part of the PNS?

A

Only the axon

705
Q

What is the outside layer of connective tissue around a nerve?

A

The epineurium

706
Q

What surrounds the nerve fiber veriscles?

A

The perineurium

707
Q

What surrounds the individual nerve fibers?

A

Endoneurium

708
Q

Are all axons myelinated, and who produces myelin?

A

No, not all. Schwann cells are present in all nerve fibers but only some produce myelin. Unmyelinated cells are present in both the CNS and PNS

709
Q

What is myelin?

A

A lipid-rich sheath that is essential for fast and efficient transmission of signals. It is interrupted by Nodes of Ranvier which aid in transmission via action potential

710
Q

What cells are unmyelinated?

A

Cells important for regulating smooth muscles. These signals don’t need to be as fast

711
Q

What cells are myelinated?

A

Sensory and motor neurons involved in (acute) sensation and muscle contraction

712
Q

What is the difference between the CNS and PNS regarding Schwann cells?

A

in the PNS, one Schwann cell myelinates only one axon

713
Q

What happens when there is damage in the myelin or Nodes of Ranvier?

A

Conductions are slowed which leads to sensation or movement problems

714
Q

What are characteristics of the blood-nerve barrier regarding the immune system?

A

Endothelial cells create a tight barrier using tight junctions, but are leaky at nerve roots, ganglion cells and nerve terminals to allow for the passage of activated T cells, macrophages and antibodies

715
Q

What cells in the PNS can act as APCs?

A

Schwann cells and endothelial cells can express MHCII, also endoneurial macrophages

716
Q

What are endoneurial macrophages?

A

Specialized macrophages important for regulation of inflammation and promotion of healing such as during Wallerian degeneration

717
Q

What is Wallerian degeneration?

A

The degeneration of the part of an axon that is severed from the neuron cell body after injury, allowing for new space for the proximal part of the nerve to sprout and make new connections

718
Q

What makes peripheral nerves so vulnerable?

A

They are extremely large and require proper alignment of the membrane and myelin to work correctly

719
Q

What type of disease is GBS?

A

An acute peripheral nerve specific immune-mediated neuropathy?

720
Q

What are paraproteins?

A

A very prominent clone of antibody present in the serum, usually IgM but can also be IgG or IgA

721
Q

What age-group has highest incidence of GBS?

A

Higher usually, except in M. pneumoniae

722
Q

What are the neurological deficits in GBS? (4)

A

-Muscle paralysis
-Sensory deficits
-Autonomic dysfunction
-Areflexia

723
Q

What is areflexia?

A

Loss or absence of reflexes

724
Q

What is ataxia?

A

Lack of voluntary coordination of muscle movements, such as balance, coordination and speech

725
Q

What are the symptoms of autonomic deficits in GBS?

A

Tension fluctuations and cardiac arrhythmia

726
Q

Two categories of diagnostic criteria for GBS

A

-Clinical features
-Additional examinations

727
Q

What are the clinical features to look out for when considering GBS?

A

-Rapidly progressive (<4 weeks)
-Symmetrical paresis of arms and legs
-Reduced or absent reflexes

728
Q

What are the additional examinations you can perform when considering GBS?

A

-Blood (no abnormalities that may explain symptoms)
-Cerebrospinal fluid (no cells, increased protein level)
-Electrophysiology (demyelination, axonal degeneration)

729
Q

Sequence of events in GBS

A
  1. Progression
  2. Plateau phase
  3. Recovery phase
  4. Disability
730
Q

What are the two histological classifications of GBS?

A

-Demyelination
-Axonal degeneration

731
Q

How is demyelinating GBS called disease wise?

A

Acute inflammatory demyelinating polyneuropathy (AIDP)

732
Q

How is axonal degenerating GBS called disease wise?

A

Acute motor axonal neuropathy (AMAN)

733
Q

How can macrophages attack the myelin? (3)

A

-Phagocytosis of myelin debris and myelin sheaths
-Pro-inflammatory cytokines
-Antigen-presentation of myelin derived antigens to T cells

734
Q

How does complement activation work in GBS?

A

Antibodies are directed against glycolipids/gangliosides, which are present in membranes of all of your cells BUT highly enriched in the nerve system. Antibody::antigen complex activate complement.

735
Q

Where does compliment activation occur during GBS?

A

Along Schwann cell surfaces

736
Q

Describe the concept of molecular mimicry in GBS

A

Generation of an immune response against the infectious agent generates anti-ganglioside antibodies that cross-react with glycolipids present in peripheral nerves and nerve roots

737
Q

Which antibodies are associated with GBS?

A

IgM, IgG1

738
Q

Which ganglioside is most often attacked?

A

GM1

739
Q

Where is GM1 present?

A

-Myelin
-Axolemma
-Paranodes
-Nodes of Ranvier (peripheral motor nerves)

740
Q

What are the three categories of treatments in GBS?

A

Specific treatment
Supportive treatment
Rehabilitation

741
Q

What are the options for specific GBS treatment?

A

-Plasma exchange or plasma pheresis
-Intravenous immunoglobulins

742
Q

What are the options for supportive GBS treatment?

A

-Artificial respiration
-Prevention and treatment of complications (infections, pain)
-Physiotherapy

743
Q

Why is GBS not a typical classic auto-immune disease? (5)

A

-No predominance in females
-No association with other auto-immune diseases
-No relapsing-remitting or chronic disease course
-No association with specific HLA haplotypes
-No improvement after corticosteroids

744
Q

Why is GBS a typical post-infectious disease?

A

-2/3 of patients have symptoms of a recent respiratory or GI-infection
-Antibodies to glycolipids in serum are detected in 50-60% of the cases
-Association type of infection and clinical features
-Association type of infection and prognosis
- >95% has a monophasic disease course, which is typical for post-infeciton diseases

745
Q

What is the most common clinical and electrophysiological phenotype of GBS related to a preceding C.jejuni infection?

A

Pure motor clinical variant and the acute motor axonal neuropathy

746
Q

Which infections are associated with GBS?

A

C.jejuni
CMV
EBV
Mycoplasma pneumoniae
HEV

747
Q

Which infections commonly precede sensory motor GBS? What is their electrophysiological phenotype?

A

M. pneumoniae, CMV and EBV. Demyelinating (AIDP)

748
Q

True or False: “Clinical outcome is better if you had a preceding C.jejuni infection than if you had a preceding M.pneumoniae infection.”

A

False. You actually have a worse outcome

749
Q

What kind of data can you use to investigate an association between infections and GBS?

A

Epidemiological data
Case-control study

750
Q

What do you look for in a case-control study to study an association between Zika infection and GBS?

A

-Zika virus positive cases vs Zika virus negative cases –> compare %GBS cases
-GBS cases vs control cases –> compare % Zika virus positives

751
Q

How long do lymphocytes spend in the lymph nodes?

A

approximately 12 hours

752
Q

What to T cells do in the lymph nodes?

A

They move around along stromal cells and scan DCs to see if there are peptides that are being presented

753
Q

What if a T cell doesn’t find an antigen-presenting DC in the lymph node?

A

It goes into the efferent lymph and drains back into circulation

754
Q

What is the life cycle of a macrophage or DC?

A

They are formed in the bone marrow and move to the tissues directly. They scan peripheral tissues and take up whatever is there, move to the lymph nodes and present to T cells. They do not exit, they die in the lymph nodes.

755
Q

What regulates T cell exit?

A

soluble lipids: S1P

756
Q

What produces S1P?

A

Endothelial cells and red blood cells

757
Q

What prevents T cells from leaving the lymph nodes too soon?

A

Once in the lymph node, they will express CD69, which is often used as an early activation marker. CD69 prevents re-expression of S1PR, which prevents activated cells from leaving the lymph node too soon. After a few divisions, CD69 will be expressed less and allow for the upregulation of S1PR, allowing exit.

758
Q

How do B cells organize themselves into B cell follicles?

A

Marginal reticular cells express CXCL13, which is a homeostatic chemokine to promote B cell migration

759
Q

What do follicular dendritic cells do?

A

They express Fc receptors, complement receptors and CXCL13 for B cell migration and assist in germinal center development

760
Q

What do fibroblastic reticular cells do and where are they located?

A

FRCs are located in the cortical sinus where they express CCL21 and CCL19. These are homeostatic chemokines that promote T cell and dendritic cell migration (which express CCR7). They also express IL-7 for resting T cell survival.

761
Q

What are HEVs?

A

High endothelial venules express adhesion molecules for naive T cell migration from blood into the lymph nodes. Naive T and B cells can ONLY leave the blood through those. They are also found in Peyer’s patches

762
Q

How do HEVs facilitate lymphocyte entry into the LNs?

A
  1. They express LN homing molecules GlyCAM-1, ICAM-1 and CD34, and secrete CCL21.
  2. GlyCAM1 and CD34 weakly interact with L-selectin (LFA-1) on the surface of naive T cells.
  3. LFA-1 (already present) changes conformation through inside-out signalling (this necessity prevents inappropriate interactions elsewhere)
  4. New conformation of LFA-1 can now bind with high affinity to ICAM-1 (outside-in signalling), making the T cell stop rolling.
  5. Diapedesis (extravasation).
763
Q

What attracts T cells to the lymph nodes?

A

CCL21 and CXCL12

764
Q

What are the two chemokine classes?

A

Homeostatic and Inflammatory

765
Q

What do homeostatic chemokines do?

A

They attract immune cells and are always expressed by the lymphoid organs, which is why lymphocyte entry into the LNs is continuous. They ensure lymphoid organ maintenance and attract cells.

766
Q

What do inflammatory chemokines do?

A

Attract activated lymphocytes. Expression is induced by activated cells and can happen anywhere.

767
Q

When are inflammatory cytokine receptors expressed?

A

Only after activation

768
Q

What is chemo/haptokinesis?

A

It is not a gradient-mediated movement of cells, it is simply movement of cells independent of chemokines

769
Q

What is cell migration regulated by?

A

Chemokines and adhesion by integrins

770
Q

What are integrins?

A

Multimeric proteins made of α- and ß- chains. You always have combinations of αs and ßs.

771
Q

What are the 4 integrin classes?

A

Collagen-binding
RGD receptors
Laminin receptors
Leukocyte receptors

772
Q

Where are leukocyte receptor integrins mainly expressed?

A

Vascular cells

773
Q

Why are integrins important?

A

Homing of cells from the blood into tissues

774
Q

How do leukocytes know where to go?

A

When a DC comes from a certain location, a T cell activated by that DC expresses the homing molecule (i.e CCR9 for intestine) that correlated with the tissue of DC origin. This ensures that the T cell only enters that tissue and not other organs, as this would be highly inefficient when an immune response is necessary in a specific location.

775
Q

What are the molecules involved in T cell homing to the intestines?

A

CCL25 (CCR9 on T cells), P-selectin, MADCAM1

776
Q

What are the molecules involved in homing to the skin?

A

CCL17 (CCR4), E-selectin, P-selectin, ICAM1, VCAM1

777
Q

What is CCR10?

A

Binds to CCL27, which is produced by keratinocytes in the skin. This receptor-ligand interaction is important for the migration of T cells to the skin, particularly in the context of inflammatory skin diseases.

778
Q

What is CCR6?

A

Interacts with CCL20 (MIP-3α), expressed by epidermal keratinocytes and some dermal cells. CCR6 is found on various immune cells, including Th17 cells, contributing to their migration to the skin, especially in conditions like psoriasis.

779
Q

What is CCR4?

A

Expressed on Th2 cells, Tregs, and skin-homing memory T cells. Its ligands, CCL17 (TARC) and CCL22 (MDC), are expressed in the skin and play roles in attracting cells involved in allergic inflammation and maintaining tolerance.

780
Q

What cells express Fc receptors?

A

Every immune cell

781
Q

Does every Fc receptor bind to every Fc with the same affinity?

A

No, they differ per antibody subclass

782
Q

What does Fc stand for

A

Fragment crystallizable

783
Q

What does FCyRI bind to and what affinity?

A

Monomeric IgG, high

784
Q

What do FcyRIIA, B, C and FCRIIIA and B bind to and what affinity?

A

medium to low affinity
Recognize IgG, but require immune complexes for activation

785
Q

What FcyRs have an activating motif?

A

All but FCyRIIB, which is the only one expressed by B cells

786
Q

What are the 9 roles of Fc receptors in immune responses?

A
  1. Clearance of Immune complexes
  2. Phagocytosis and antigen presentation
  3. Production of cytokines
  4. Antibody dependent cellular toxicity (ADCC)
  5. Modulation of cellular activation
  6. Mast cell degranulation
  7. Generation of high affinity antibodies
  8. Prevent autoimmunity by forming late auto-immune checkpoint
  9. Transportation/recycling of imunoglobulins
787
Q

In what ways can FcR binding lead to antigen presentation?

A
  1. Internalization, increased endosomal maturation, processing and loading on MHCII
  2. Immune complexes are protected from degradation in the early endosome, degraded by the proteosome and loaded onto MHCI in the ER
788
Q

How do FcRs deal with immune complexes with viruses that manage to escape vesicles?

A

FcRs such as TRIM21 detect them and target them for proteosomal breakdown to be loaded onto MHCI

789
Q

How long does it take for FcR activation to lead to cytokine production?

A

Can take hours

790
Q

What is antibody-dependent cellular cytotoxicity?

A

ADCC is a mechanism initiated by FcRs on NK cells.

791
Q

How does ADCC work?

A
  1. Antibodies bind antigens on the surface of target cells such as tumor cells or microbes
  2. FcRs on NK cells (FCyIII/CD16) recognize bound antibody
  3. Crosslinking of FcRss signals the NK cell to kill the target cell
  4. NKs release granzymes and perforin which killes the cell through apoptosis
792
Q

When is ADCC especially important?

A

Biological use such as anti-CD20 (Rituximab) which depletes B cells

793
Q

What is glycosylation?

A

A carbohydrate expressed on Fc is required for FcR binding.

794
Q

How does glycosylation affect Fc-FcR binding affinity?

A

Low fucose = higher binding

795
Q

What affects glycosylation?

A

Age, changes after immunization, during pregnancy and in some autoimmune disease

796
Q

How does glycosylation play a role in severe COVID-19 patients?

A

Afucosylated IgGs are a result of strong BCR cross-linking due to repetitive expression of spike. A lack of fucose leads to a >50x higher affinity for FcyRIII (NKs, macrophages, neutrophils, some T cells). This signal, combined with recognition of dsRNA by TLR3, create very strong activation and TLR3 response and thus alveolar macrophage response, leading to ARDS.

797
Q

What is meant by net effect in FcR interactions?

A

In case of immune complexes, activatory and inhibitory receptors can be activated at the same time. One pathway counteracts the other. When multiple receptors are involved, the net effect determines whether there is activation or inhibition. Cytokines or bacterial molecules like LPS influence the expression of these receptors, shifting the balance to activation or inhibition.

798
Q

How does mast cell degranulation by FcRs work?

A

Cross-linking via immune complexes leads to release of vasoactive substances such as histamines and chemoattractants. This is a really fast process.

799
Q

How do FcRs help in generation of high affinity antibodies?

A

Through expression on follicular DCs in germinal centers

800
Q

What 3 things affect Fcy binding affinity?

A

Glycosylation, polymorphisms and expression levels

801
Q

Most people have a stop codon in what FcR?

A

FcyRIIC, but some people have a mutation where there is an open reading frame

802
Q

What is FcyRIIB important for?

A

Peripheral tolerance. It has an inhibitory reaction. Activated B cells upregulate IIB to prevent it from going haywire (feedback loop). Only B cells with very strong affinity will have the necessary activation threshold to be selected for, and the added competition for growth factors and antigens prevents autoimmunity

803
Q

What do neonatal FcRs do?

A

Recycle IgG and transport them through mucosa and placenta

804
Q

What is FcE for and what does it do?

A

It is important for allergies. When cross-linked with IgE complexes, it results in degranulation which causes allergic symptoms

805
Q

How are fetuses sensitized for allergens?

A

IgE can be present in the amniotic fluid

806
Q

How does food allergy work?

A

IgE is internalized by binding to CD23 in the gut. Binding of IgE to FcERI on mast cells and DCs cause allergic inflammation and further IgE synthesis after B cell class switching. Inflammation mediated by IgE damages the intestinal epithelium, disrupts tight junctions, allowing allergens to pass and bind to sensitized mast cells and DCs, exacerbating the food allergy.

807
Q

What does FCαR do?

A

It is expressed by myeloid cells and binds only monomeric IgA. It results in a similar response to IgG: Phagocytosis, endocytosis, ADCC and respiratory burst (ROS)

808
Q

What diseases can FcRs contribute to?

A

ADE
Deficiency
SLE
Autoimmunity
IBD

809
Q

How can FcRs contribute to ADE?

A

ADE is thought to be mediated by antibodies that bind but do not neutralize a virus. The virus, when internalized through FcRs, can uncoat and replicate in the cell, potentially leading to more severe outcomes by leading to altered cytokine production, immune activation and potentially supression or dysregulation.

810
Q

What happens when mice have an FcyRIIB deficiency?

A

B cells cannot be adequately inhibited, causing increased humoral responses and increased collagen-induced arthritis severity. This is also thought to be behind SLE

811
Q

What are the two sides of the FcR balance scale?

A

Pathogen clearance vs. septic shock and autoimmunity (SLE)

812
Q

What does insufficient FcR-mediated clearance lead to?

A

Sustained myeloid cell activation and induction of autoimmunity

813
Q

What is a pitfall when studying FcRs?

A

Species specificity, of mice but not men

814
Q

What is the definition of epigenetics?

A

Epigenetic mechanisms transduce the inheritance of gene expression patterns without altering the underlying DNA sequence but by adapting chromatin, which is the physiological form of our genetic information. In summary: chromatin affects gene expression

815
Q

What role does epigenetics play in myeloid cells?

A

The transcriptome contains housekeeping genes that are expressed by all of the cell types, but there is also cell type specific genes that are transcribed by some and not others, which results in a different phenotype.

816
Q

How is transcription initiated?

A

The TATA box binds to its transcription factor and recruits RNA pol II, which binds to the promoter along with one or more general transcription factors.

817
Q

Where is the promoter with respect to the transcription starting site?

A

Upstream and/or partially downstream, can have some coding region overlap.

818
Q

What do promoters do?

A

They define where transcription of a gene begins

819
Q

What happens when RNA pol II binds?

A

It unwinds around 14bp of the DNA to form the RNA-polymerase-poromoter complex, exposing the “transcription bubble”.

820
Q

What initiates transcription?

A

Enhancers (a gene can have multiple), which are non-coding DNA regions distant from the promoter.

821
Q

How do enhancers interact with the promoter?

A

Chromosome loops are formed to allow for close proximity. The loops are stabilized with anchored architectural proteins, and specific transcription factors that bind along with RNA pol II, also bind to the enhancer.

822
Q

How can epigenetics impact mice with the Agouti gene?

A

The Agouti gene should only be expressed during hair follicle development, which is influenced by ingestion of folic acid during pregnancy. Folic acid provides the building blocks with methylation, which prevents expression of the gene in cells. When this does not happen properly, the gene is not silenced and leads to obesity and a light fur color in mice.

823
Q

How can epigenetics cause tortoiseshell color in cats?

A

Cells individually inactivate one X chromosome through hypermethylation.

824
Q

What are 4 epigenetic mechanisms?

A

DNA methylation
Nucleosome positioning
Histone modification
3D genome folding

825
Q

What is DNA methylation?

A

DNA methyltransferase adds a methyl group to usually cytosine, causing some transcription factors to no longer be able to bind. When a promoter is heavily methylated, this leads to silencing.

826
Q

What can demethylate promoters?

A

TET enzymes

827
Q

What is imprinting in the context of epigenetics?

A

Mostly involved in embryonic growth. One gene copy is methylated and the other is not based on whether is was maternal or paternal, allowing for a balance of gene expression (i.e. length).

828
Q

What happens when imprinting is lost?

A

Usually life incompatibility

829
Q

What is closed chromatin?

A

DNA with a high nucleosome content.

830
Q

What are histone modifications?

A

Post-translational modifications. Acetylation mediated by HAT allows for DNA to have space, giving room for binding. The opposite is mediated by HDAC enzymes (deacetylation).
Phosphorylation and others can also modify histones.

831
Q

How does 3D genome folding contribute to epigenetics? Name 2

A
  1. It determines the proximity of enhancers to promoters
  2. In recombination for lymphocyte receptors and antibodies, all V genes need to have equal opportunity, so folding should be as such that they are all in close proximity to the recombination center.
832
Q

What are 2 key players in epigenetics?

A

Transcription factors and gene regulatory elements

833
Q

How many transcription factors are encoded in the genome?

A

More than 1000

834
Q

What do transcription factors do?

A

They lead to the transcription of genes

835
Q

How are transcription factors produced?

A

Through signaling iniciated by i.e cytokines

836
Q

What are gene regulatory elements?

A

They are pieces of non-coding DNA (almost all transcription is regulated by non-coding DNA): enhancers, silencers and insulators.

837
Q

What is the role of epigenetic mechanisms lymphocyte diversity? Name 2 examples

A
  1. Transcription factors are required for the adaptation of different T cell phenotypes depending on what is necessary.
  2. T cell exhaustion is defined by up- and downregulation of genes
838
Q

How does epigenetics shape T cell maturity?

A

Memory T cells already have their chromatin accessible through demethylation and acetylation sothat the necessary genes can immediately be transcribed.
Naive T cells still need to undergo epigenetic changes.

839
Q

How can (disturbed) epigenetics cause disease? Name 4.

A
  1. Enhancer mutation of Pax5 in a B cell leads to loss. It stays alive but becomes to plastic, allowing it to change to a T or NK when exposed to different signals. The plasticity poses a high risk for leukemic transformation.
  2. Loss of imprinting
  3. Cancer: silencing of supressor genes and genomic instability
  4. Gene misfolding can cause developmental syndromes
840
Q

How can we exploit epigenetics for therapeutics (for example)

A

Ex: To inhibit things that promote transcription of hyperexpressed genes via methylation

841
Q

What is signal transduction?

A

Any process by which the cell converts one kind of signal stimulus into another, which involves ordered sequences of biochemical reactions inside the cell

842
Q

What are the different types of receptors and signalling molecules? (5)

A

Linkers
Protein Tyrosine Kinases
Protein Tyrosine Phosphatases
Lipid-metabolizing enzymes
Guanine nucleoride exchange factors

843
Q

What are linkers?

A

They don’t have any enzymatic activity, but have components that interact with other signaling proteins

844
Q

What are Protein Tyrosine Kinases?

A

They have enzymatic activity/domain and can phosphorylate their target

845
Q

What are Protein Tyrosine Phosphatases?

A

They have enzymatic activity/domain and can dephosphorylate their target

846
Q

What are lipid-metabolizing enzymes?

A

They have enzymatic activity and can cross and bind to the cell membrane

847
Q

What are the 4 steps of signal transduction?

A

Signal perception, Intracellular transduction, Cellular response and Termination

848
Q

What is signal perception?

A

When a ligand binds to a receptor, it activates other proteins bound to the membrane by recruiting signaling proteins

849
Q

What are 3 ways proteins are activated after signal perception?

A
  1. The intracellular domain is phosphorylated which creates docking sites
  2. Ras proteins bound to the membrane are activated by a linked receptor, which leads to a conformational change and downstream interaction
  3. Signaling proteins bind directly to the plasma membrane
850
Q

Are phosphorylation sites activating or inhibiting?

A

They can be either

851
Q

What 2 types of linkers are there?

A

Scaffolds and adaptors

852
Q

What are scaffolds?

A

Linkers that can be phosphorylated

853
Q

What are adaptors?

A

They cannot be phosphorylated but have binding sites that can connect 2 other signaling proteins so they interact

854
Q

What do kinases do?

A

They add a phosphate group from ATP (->ADP) to an amino acid residue, leading to phosphorylation

855
Q

What is a kinase cascade?

A

A cascade that can amplify signal logarithmically (Ca2+ can also do that)

856
Q

What do phosphatases do?

A

Dephosphorylate targets. They can also activate them through conformational changes, allowing binding of other proteins

857
Q

How long do cellular responses last after signalling?

A

They can take hours or days, sometimes throughout the entire lifetime of the cell.

858
Q

What (3) types of cellular responses are there?

A

Gene transcription, cytoskeleton changes (to aid in cell migration) and metabolism

859
Q

What are the 3 mechanisms by which signaling can be terminated?

A
  1. (De)phosphorylation
  2. Proteosomal degradation (~15aa chains can be reused)
  3. Lysosomic degradation
860
Q

How does Notch signalling work?

A
  1. Notch binds to stromal cell receptors
  2. Intracellular part of Notch is cleaved
  3. Enters nucleus with Coenzyme A
  4. T cell fate is induced
861
Q

How can signalling be defective in the context of Notch?

A

Notch is oncogenic in mature T cells

862
Q

What happens when there is no BTK?

A

BTK is a kinase with a function in B cell development. When there is none, no B cells can be developed, leading to X-linked agammaglobulinemia (XLA)

863
Q

What happens when BTK is always activated?

A

Chronic lymphocytic leukemia

864
Q

What happens when BTK is increased?

A

Autoreactive cells in mice survive

865
Q

Why are there so many off-target effects in treatment involving B and T cells?

A

BCR and TCR (scaffold) pathways are extremely similar (even the same phosphorylation sites), causing cross-reactivity in used therapeutics.

866
Q

Why are there in general so many off-target effects in therapeutics?

A

Signaling molecules are often involved in more than one pathway

867
Q

Why can B cells still survive when BTK is targetted in CLL (always activated BTK)?

A

They can survive by rewiring since there is so much redundancy, so off-target effects don’t really matter. In the mean time CXCL4 inhibition in tumor cells cause the tumors to leave the LN and die.