infection Flashcards

1
Q

Describe the nature of viruses.

A

Viruses are not considered living organisms and require a host cell to replicate.

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

Define bacteria.

A

Bacteria are single-celled organisms that are considered living.

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

Describe the cellular structure of fungi.

A

Fungi are mostly multicellular organisms with complex structures, including a cell wall made of chitin.

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

What is the replication process of viruses?

A

Viruses replicate inside host cells.

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

How do bacteria reproduce?

A

Bacteria reproduce independently through binary fission

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

Describe the reproductive methods of fungi.

A

Fungi reproduce through both sexual and asexual means, involving spores.

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

Compare the size of viruses, bacteria, and fungi.

A

Viruses are much smaller, bacteria are larger than viruses, and fungi are larger than bacteria.

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

Do viruses carry out metabolic processes without a host?

A

No, viruses cannot carry out metabolic processes without a host.

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

What are the effects of bacteria on humans?

A

Some bacteria are harmful (pathogenic), while others are beneficial.

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

How do fungi affect humans?

A

Fungi can cause infections, particularly in immunocompromised individuals.

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

Describe the structure of a virus.

A

A virus has genetic material (DNA or RNA) surrounded by a protein coat called a capsid.

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

What is the main difference between viruses and bacteria?

A

Viruses have genetic material surrounded by a protein coat, while bacteria are single-celled organisms with a cell wall.

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

Describe the different categories of microorganisms that cause infections.

A

Bacteria, viruses, fungi, protozoa, helminths (parasitic worms), and prions.

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

Name some viruses that can cause infections.

A

Influenza virus, Human Immunodeficiency Virus (HIV), Herpes simplex virus, SARS-CoV-2 (causing COVID-19).

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

Give examples of bacteria that can cause infections.

A

Streptococcus, Staphylococcus, Escherichia coli (E. coli), Mycobacterium tuberculosis.

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

What are some examples of fungi that can cause infections?

A

Candida albicans (causes yeast infections), Aspergillus, Cryptococcus.

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

List some protozoa that can cause infections.

A

Plasmodium (causes malaria), Giardia lamblia, Toxoplasma gondii.

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

What are helminths and give examples of parasitic worms that can cause infections?

A

Helminths are parasitic worms. Examples include roundworms, tapeworms, and flukes.

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

Describe the purpose of culturing microorganisms for diagnostic purposes.

A

To detect and identify the presence of infections.

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

Define prions and their association with diseases.

A

Prions are unusual infectious agents primarily associated with neurodegenerative diseases.

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

What are some common specimen types used for culturing microorganisms?

A

Blood, urine, sputum, stool, cerebrospinal fluid (CSF), wound swabs, throat swabs, nasal swabs, genital swabs, tissue biopsy.

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

How is blood used in culturing microorganisms for diagnostic purposes?

A

It is used to detect systemic infections.

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

What is the purpose of collecting urine as a specimen for culturing microorganisms?

A

It is commonly used to detect urinary tract infections.

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

Define sputum and explain its role in culturing microorganisms.

A

Sputum is mucus from the respiratory tract, and it is helpful in diagnosing respiratory infections.

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

What is the purpose of collecting cerebrospinal fluid (CSF) samples?

A

CSF samples are collected in cases of suspected central nervous system infections.

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

What are wound swabs used for?

A

Wound swabs are used to identify the causative organisms in wounds or skin lesions.

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

How are throat swabs used in the laboratory?

A

Throat swabs are used to identify bacteria causing throat infections like strep throat.

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

What is the purpose of collecting nasal swabs?

A

Nasal swabs are collected to identify nasal infections or carriers of certain bacteria.

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

Describe the use of genital swabs in laboratory testing.

A

Genital swabs are used to detect infections in the genital area

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

What type of samples are tissue biopsies?

A

Tissue biopsies are deeper tissue samples collected for analysis.

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

What factors determine the choice of specimen for testing?

A

The suspected site of infection and the type of microorganism being investigated determine the choice of specimen.

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

What is important in handling and collecting different specimen types?

A

Specific collection and handling procedures are important to ensure accurate results for each specimen type.

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

Describe the purpose of Gram staining in microscopy.

A

Gram staining helps categorize bacteria into Gram-positive and Gram-negative, providing initial information about their cell wall structure.

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

What is the benefit of using microscopy for rapid preliminary identification of bacteria?

A

Microscopic examination can offer a quick preliminary identification of bacteria, aiding in the selection of appropriate culture media and initial treatment decisions.

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

How can examining bacterial morphology under the microscope provide clues about the nature of the infection?

A

Examining bacterial morphology (shape, arrangement) under the microscope can provide clues about the nature of the infection and potential bacterial species involved.

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

What other elements can microscopy reveal besides bacteria?

A

Microscopy can also reveal the presence of other elements, such as white blood cells, which may indicate an inflammatory response to bacterial infection.

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

Define microscopy.

A

Microscopy is a technique that allows direct visualization of bacterial cells in clinical specimens.

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

Describe the process of culturing in microbiology.

A

Culturing involves placing clinical specimens on specific nutrient media to encourage bacterial growth.

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

What is the purpose of isolation in culturing?

A

Isolation allows for the identification of individual bacterial colonies.

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

How does culture help in determining antibiotic susceptibility?

A

Culture is essential for determining the antibiotic susceptibility of bacterial isolates.

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

Why is quantification important in culture?

A

Quantification allows for assessing the severity of the infection by measuring bacterial load.

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

What is the main difference between microscopy and culture in microbiology?

A

Culture is often considered the gold standard for Microscopy provides rapid insights, while culture allows for isolating, identifying, and characterizing bacteria in detail.

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

Describe the role of culture in selecting antimicrobial therapy.

A

Culture helps guide the selection of the most effective antimicrobial therapy based on antibiotic susceptibility testing

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

Describe sterile sites.

A

Sterile in the body are areas that are generally free from microorganisms, including bacteria, viruses, fungi, and other pathogens.

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

What are the characteristics of sterile sites?

A

Sterile sites are typically protected by natural barriers, such as intact skin or mucous membranes, that prevent the entry of microorganisms.

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

Give examples of sterile sites in the body.

A

bloodstream, cerebrospinal fluid, and the interior of organs and tissues that are normally free from microorganisms.

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

How are sterile sites protected from microorganisms?

A

Sterile sites are protected by natural barriers, such as intact skin or mucous membranes, that prevent the entry of microorganisms.

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

Describe non-sterile sites.

A

Non-sterile sites are areas of the body that naturally harbor microorganisms or are exposed to the external environment, making them prone to microbial colonization.

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

What are some examples of non-sterile sites?

A

Some examples of non-sterile sites include the skin, mouth, gastrointestinal tract, respiratory tract, and genitourinary tract.

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

What is the role of the body’s microbiota in non-sterile sites?

A

The body’s microbiota in non-sterile sites play important roles in various physiological processes, such as digestion, immune system development, and protection against pathogenic invaders.

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

Describe the purpose of Polymerase Chain Reaction PCR).

A

Amplifies viral nucleic acids for detection.

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

What is the purpose of Reverse Transcription PCR (RT-PCR)?

A

Used for RNA viruses by converting RNA to complementary DNA before amplification.

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

Define Quantitative PCR (qPCR).

A

Quantifies the amount of viral nucleic acid present.

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

What is the function of Enzyme-Linked Immunosorbent Assay (ELISA)?

A

Detects viral antigens or antibodies produced in response to the virus.

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

Describe the process of Western Blot.

A

Identifies specific viral proteins through antigen-antibody interactions.

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

What is the purpose of Neutralization Assays?

A

Measures the ability of antibodies to neutralize viral activity.

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

Explain the process of Viral Culture.

A

Involves growing viruses in cultured cells to observe cytopathic effects.

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

What are the advantages of Viral Culture?

A

Allows for virus isolation and characterization.

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

Describe direct immunofluorescence (DFA).

A

Direct immunofluorescence (DFA) is a technique used to detect viral antigens directly in clinical samples.

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

What is the purpose of indirect immunofluorescence (IFA)?

A

The purpose of indirect immunofluorescence (IFA) is to detect antibodies against viral antigens in patient serum.

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

Define next-generation sequencing (NGS).

A

Next-generation sequencing (NGS) is a technique that identifies viral genomes directly from clinical samples without prior knowledge of the virus.

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

Describe lateral flow assays.

A

Lateral flow assays are rapid diagnostic tests that provide quick results for specific viral infections and are common in point-of-care settings.

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

What is the technique used in fluorescence in situ hybridization (FISH)?

A

Fluorescence in situ hybridization (FISH) is a technique used to locate viral nucleic acids in cells or tissues.

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

Do metagenomic sequencing techniques require prior knowledge of the virus?

A

No, metagenomic sequencing techniques identify viral genomes directly from clinical samples without prior knowledge of the virus.

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

What are the limitations of electron microscopy in viral detection?

A

Electron microscopy is limited by the need for high viral concentrations.

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

Describe the clinical presentation in parasitology.

A

Clinical presentation involves considering patient symptoms and physical signs to guide suspicion of a parasitic infection.

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

What is the importance of epidemiological history in parasitology?

A

Epidemiological history, including geographical location and exposure history, can provide valuable clues in narrowing down possibilities of a parasitic infection.

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

How can symptoms and signs vary in parasitic infections?

A

Symptoms and signs can vary widely depending on the type of parasite and the affected organ system.

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

What is the role of laboratory testing in parasitology?

A

Laboratory testing, such as microscopy, is essential for detecting and identifying parasites in clinical specimens.

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

Describe serological tests for diagnosing parasitic infections.

A

Serological tests involve detecting antibodies or antigens in the patient’s blood to diagnose certain parasitic infections.

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

What are molecular methods used for in diagnosing parasitic infections?

A

Molecular methods, such as Polymerase Chain Reaction (PCR), are used for the direct detection and identification of parasite DNA or RNA.

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

What is the purpose of culture in diagnosing parasitic infections?

A

In some cases, parasites are cultivated in the laboratory to observe their life cycle stages.

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

How can imaging studies be used to diagnose parasitic infections?

A

Radiological imaging, such as X-rays, CT scans, or MRI, may be used to visualize parasitic infections affecting organs or tissues

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

Describe how travel and exposure history can help identify potential sources of parasitic exposure.

A

Travel and exposure history can provide information about the regions visited and activities engaged in, which can help identify potential sources of parasitic exposure.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

What is the significance of combining clinical, epidemiological, and laboratory methods in the diagnosis of parasitic infections?

A

Combining clinical, epidemiological, and laboratory methods in the diagnosis of parasitic infections increases accuracy and improves the chances of identifying the specific parasite causing the infection.

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

How can the response to therapy support the diagnosis of a parasitic infection?

A

Improvement in symptoms and clearance of parasites after appropriate treatment supports the diagnosis of a parasitic infection.

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

Describe the purpose of basic infection control measures.

A

Basic infection measures are essential practices aimed at preventing the spread of infectious agents in various settings, including healthcare facilities, community settings, and households.

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

How does handwashing help prevent the spread of pathogens?

A

Handwashing with soap and water for at least 20 seconds helps remove and kill pathogens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

What is an alternative to handwashing when soap and water are not available?

A

Using alcohol-based hand sanitizers with at least 60% alcohol is an effective alternative to handwashing.

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

When should disposable gloves be worn as part of infection control measures?

A

Disposable gloves should be worn when in contact with blood, bodily fluids, or contaminated surfaces.

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

What is the purpose of using masks in infection control?

A

Masks are used to prevent the inhalation or spread of respiratory droplets.

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

What should be done when coughing or sneezing as part of respiratory hygiene?

A

Cover the mouth and nose with a tissue or elbow, and dispose of used tissues properly while practicing immediate hand hygiene.

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

What areas should be regularly cleaned and disinfected in healthcare settings?

A

Surfaces and equipment in healthcare settings should be regularly cleaned and disinfected.

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

What surfaces should be frequently cleaned in households and public places as part of infection control?

A

Frequently-touched surfaces in households and public places should be regularly cleaned and disinfected.

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

What is the purpose of waste management in infection control?

A

Proper disposal of medical waste and other infectious materials to prevent the spread of infections.

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

Describe safe injection practices.

A

Using aseptic techniques administering injections to prevent the transmission of infections.

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

How are isolation precautions implemented in healthcare settings?

A

By placing patients with certain infections in dedicated rooms or using special precautions to prevent the spread of infectious agents.

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

What is the importance of education and training in infection control?

A

Providing education and training to healthcare workers, patients, and the general public on infection control practices.

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

Define vaccination and its role in preventing the spread of diseases.

A

Vaccination is the administration of a vaccine to stimulate the immune system and prevent the spread of vaccine-preventable diseases.

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

Describe quarantine and isolation measures in preventing the spread of infections.

A

Implementing quarantine measures for individuals exposed to infectious agents and isolating those who are infected to prevent further transmission.

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

What is the purpose of monitoring and surveillance in infection control?

A

Regular monitoring and surveillance of infection rates and adherence to infection control measures.

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

Why are these infection control measures crucial?

A

These measures are crucial in controlling the spread of infections, protecting individuals in healthcare and community settings, and minimizing the impact of infectious diseases on public health.

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

What are the 3 bacterial shapes

A

cocci (spherical), bacilli (rod-shaped), and spirilla (spiral-shaped).

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

How does bacterial size contribute to identification?

A

Certain size characteristics can be associated with specific bacterial groups.

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

Define bacterial morphology.

A

Bacterial morphology refers to the shape of bacteria, which is an important feature for identification.

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

What is the arrangement of bacterial cells called?

A

The arrangement of bacterial cells is called the diplo arrangement, which consists of pairs of cells.

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

Describe the difference between Strepto and Staphylo bacteria.

A

Strepto bacteria form chains of cells, while Staphylo bacteria form clusters of cells.

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

What are some of the growth requirements for bacteria?

A

Bacteria require specific environmental conditions such as temperature, pH, oxygen levels, and nutrient availability for growth.

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

Define Gram staining and its importance in bacterial identification.

A

Gram staining is a technique that classifies bacteria into Gram-positive and Gram-negative based on their cell wall structure. This differentiation is critical for bacterial identification and guides treatment decisions.

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

How is acid-fast staining used in the diagnosis of diseases?

A

Acid-fast staining is used to identify bacteria with waxy cell walls, such as Mycobacterium species. It is particularly useful in the diagnosis of diseases like tuberculosis.

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

What does endospore staining highlight?

A

Endospore staining highlights the presence of endospores, which are resistant structures formed by certain bacteria like Bacillus and Clostridium.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

Describe the structure of the bacterial cell membrane.

A

The cell membrane is composed of a phospholipid bilayer with embedded proteins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

What is the function of the cell membrane?

A

The cell membrane regulates the passage of substances into and out of the cell.

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

Define the cell wall.

A

The cell wall surrounds the cell membrane and provides structural support.

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

Describe the structure of the Lipopolysaccharide (LPS) Layer in Gram-Negative Bacteria.

A

The LPS Layer is a complex molecule with lipid and carbohydrate components.

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

What is the function of the Lipopolysaccharide (LPS) Layer in Gram-Negative Bacteria?

A

The LPS Layer contributes to the structural integrity of the outer membrane, acts as an endotoxin, and plays a role in host-pathogen interactions.

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

Describe the structure of the nucleoid.

A

The nucleoid is the region in the bacterial cell where the chromosomal DNA is located.

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

What is the function of the nucleoid?

A

The nucleoid houses the genetic material and is involved in DNA replication and transcription.

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

Describe the structure of ribosomes.

A

Ribosomes are molecular complexes of protein and ribosomal RNA (rRNA).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

What is the function of ribosomes?

A

Ribosomes are the site of protein synthesis (translation).

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

Define bacteriophage.

A

A bacteriophage is a virus that infects and replicates within bacteria.

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

What is the structure of a bacteriophage?

A

A bacteriophage is composed of genetic material (either DNA or RNA) surrounded by a protein coat (capsid).

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

Describe the function of a phage.

A

Injects its genetic material into a bacterial cell, hijacking the cellular machinery for replication and producing new phage particles.

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

What is the function of flagella in bacteria?

A

Provide motility by rotating like propellers.

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

Define pili (fimbriae) in bacteria.

A

Short, hair-like projections that aid in attachment to surfaces or other cells.

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

What is the role of penicillin-binding proteins (PBPs) in bacteria?

A

Involved in the synthesis of the bacterial cell wall and the target of beta-lactam antibiotics, including penicillin.

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

Describe the structure of bacterial spores.

A

Specialized, highly resistant structures formed by some bacteria, such as Bacillus and Clostridium.

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

What is the function of bacterial spores?

A

Enable bacteria to survive harsh conditions (heat, radiation, chemicals) by entering a dormant state and germinate into active bacterial cells when conditions become favorable.

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

Describe the role of penicillin-binding proteins (PBPs) in antibiotic action.

A

They are the target of beta-lactam antibiotics, including penicillin, which inhibit cell wall synthesis and lead to bacterial cell death.

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

Describe the three domains of life.

A

The three domains of life are Bacteria, Archaea, and Eukarya

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

What is the hierarchical classification of bacteria?

A

The hierarchical classification of bacteria progresses through the levels of class, order, family, genus, and species.

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

Define binomial nomenclature.

A

Binomial nomenclature is a system of naming species using a two-part name: the genus name followed by the species name.

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

What is the function of porins in Gram-negative bacteria?

A

Porins in Gram-negative bacteria are protein channels in the outer membrane that allow the passage of small molecules.

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

Describe the outer membrane of Gram-negative bacteria.

A

The outer membrane of Gram-negative bacteria contains lipopolysaccharides (LPS) and proteins

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

Do Gram-negative bacteria have a thick or thin peptidoglycan layer in their cell walls?

A

Gram-negative bacteria have a thin peptidoglycan layer in their cell walls.

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

What is the role of lipopolysaccharides (LPS) in Gram-negative bacteria?

A

Lipopolysaccharides (LPS) in Gram-negative bacteria act as endotoxins and can trigger an immune response. found in the outer membrane

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

What is the periplasmic space in Gram-negative bacteria?

A

The periplasmic space is a compartment between the inner and outer membranes of Gram-negative bacteria that contains enzymes and other molecules.

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

How do Gram-negative bacteria appear after Gram staining?

A

After Gram staining, Gram-negative bacteria appear pink or red under the microscope.

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

Describe the cell wall composition of Gram-positive bacteria.

A

Gram-positive bacteria have a thick layer of peptidoglycan in their cell walls and lack an outer membrane.

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

What color do Gram-positive bacteria appear after Gram staining?

A

Gram-positive bacteria appear purple or blue under the microscope after Gram staining.

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

What is the periplasmic space like in Gram-positive bacteria?

A

The periplasmic space is reduced or absent in Gram-positive bacteria.

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

What are teichoic acids and where are they found?

A

Teichoic acids are polymers that are often present in the cell wall of Gram-positive bacteria and play a role in cell wall structure.

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

How does the thick peptidoglycan layer in Gram-positive bacteria affect their sensitivity to antibiotics?

A

The thick peptidoglycan layer in Gram-positive bacteria makes them more susceptible to certain antibiotics as it provides a target for antibiotic action.

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

Describe the process of binary fission in bacteria.

A

Binary fission is a form of asexual reproduction in bacteria where the bacterial chromosome is replicated, followed by cell elongation and septum formation, resulting in the division of the cell into two genetically identical daughter cells.

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

What is the purpose of DNA replication in binary fission?

A

The purpose of DNA replication in binary fission is to produce two identical copies of the bacterial chromosome.

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

Define septum in the context of binary fission.

A

In binary fission, a septum refers to a partition that forms between the two DNA copies, eventually dividing the cell into two daughter cells.

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

How does binary fission contribute to the rapid increase in bacterial population?

A

Binary fission allows bacteria to quickly increase their population by dividing into two genetically identical daughter cells.

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

Describe the role of cell elongation in binary fission.

A

Cell elongation in binary fission is essential for accommodating the duplicated DNA before the cell divides into two daughter cells.

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

What is the origin of replication in binary fission?

A

The origin of replication in binary fission is the specific site on the bacterial chromosome where DNA starts to unwind and replication begins.

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

How do bacteria generate genetic variation?

A

Bacteria can generate genetic variation through several mechanisms, such as mutation, horizontal gene transfer, and genetic recombination.

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

What is the purpose of septum formation in binary fission?

A

The purpose of septum formation in binary fission is to physically divide the cell into two daughter cells, each containing an identical copy of the original DNA.

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

Describe the process of cell separation in binary fission.

A

Cell separation in binary fission refers to the two daughter cells separating from each other, becoming independent, genetically identical bacterial cells.

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

What is the significance of genetic variation in bacteria?

A

Genetic variation in bacteria promotes adaptability and evolution, allowing them to better survive and thrive in changing environments.

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

Describe genetic variation in bacteria.

A

Genetic variation in bacteria refers to the introduction of different genetic traits or functions through mechanisms such as mutation, horizontal gene transfer, plasmid transfer, recombination, and selective pressure.

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

What is mutation in bacteria?

A

Mutation in bacteria refers to random changes in the DNA sequence that can occur during replication or due to external factors like radiation or chemicals, resulting in altered traits or functions.

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

How does horizontal gene transfer occur in bacteria?

A

Horizontal gene transfer in bacteria can occur through mechanisms such as transformation (uptake of free DNA from the environment), transduction (transfer of DNA via bacteriophages), and conjugation (direct transfer of DNA through cell-to-cell contact using a pilus).

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

Define plasmid transfer in bacteria.

A

Plasmid transfer in bacteria refers to the transfer of small, circular DNA fragments called plasmids between bacteria, facilitating the spread of specific traits, such as antibiotic resistance.

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

Describe recombination in bacteria.

A

Recombination in bacteria refers to the process of homologous recombination between similar DNA sequences, leading to the exchange of genetic material and the creation of hybrid DNA molecules.

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

What is selective pressure in bacteria?

A

Selective pressure in bacteria refers to environmental factors, such as the presence of antibiotics or changes in nutrient availability, that can favor the survival and reproduction of bacteria with specific genetic traits.

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

How does transformation occur in bacteria?

A

Transformation in bacteria occurs when bacteria take up free DNA from the environment, allowing them to acquire new genes.

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

What is transduction in bacteria?

A

Transduction in bacteria is the transfer of DNA via bacteriophages, which are viruses that infect bacteria.

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

Describe conjugation in bacteria.

A

Conjugation in bacteria is the direct transfer of DNA through cell-to-cell contact using a structure called a pilus.

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

What is the purpose of molecular methods in detecting bacteria?

A

Molecular methods, such as PCR, are used to detect bacterial DNA or RNA, allowing for highly specific identification of pathogens even in low concentrations.

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

What are serological tests used for in the detection of bacteria?

A

Serological tests, like ELISA, detect bacterial antigens or antibodies produced in response to bacterial infections.

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

Define culturing in the context of bacterial detection.

A

Culturing is a method that involves growing bacteria on culture media, allowing for the isolation and identification of bacterial colonies.

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

Give an example of an RNA virus.

A

Influenza virus

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

Define enveloped viruses.

A

Viruses with an outer envelope derived from the host cell membrane

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

How do retroviruses replicate?

A

They use reverse transcription to convert RNA into DNA

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

What is the function of the capsid in viruses?

A

The capsid is a protein coat that surrounds and protects the viral genetic material, providing structural integrity and shape to the virus.

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

How do viruses compare in size to bacteria and eukaryotic cells?

A

Viruses are much smaller than bacteria and eukaryotic cells, typically ranging in size from about 20 to 300 nanometers.

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

Define host specificity in viruses.

A

Host specificity refers to the ability of viruses to infect specific types of cells or organisms, determined by the interaction between viral surface proteins and host cell receptors.

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

Describe the replication process of viruses.

A

Viruses rely on the host cell’s machinery for replication and can follow either the lytic cycle (resulting in cell lysis and release of new virions) or the lysogenic cycle (integration of viral DNA into the host genome).

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

Give examples of viruses that are pathogens.

A

Examples of pathogenic viruses include the flu virus, human immunodeficiency virus (HIV), and the common cold viruses.

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

What challenges do viruses pose in terms of evolution?

A

Viruses exhibit a high mutation rate and can undergo rapid evolution, contributing to the development of new strains and challenges in vaccine development.

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

Describe the purpose of viral culture.

A

Viral culture is a laboratory technique used to isolate and grow viruses in a controlled environment for the study of viral characteristics, production of viral stocks, and development of vaccines.

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

What is the importance of selecting suitable host cells in viral culture?

A

Choosing the appropriate cell line or tissue for culture is crucial as different viruses have specific host cell preferences.

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

Explain the importance of maintaining sterility in viral culture.

A

Maintaining sterility is essential in viral culture to prevent contamination. Aseptic techniques are employed to ensure the purity of the viral culture.

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

How is the inoculum prepared in viral culture?

A

The inoculum in viral culture is prepared using a sample containing the virus, such as a clinical specimen or a previously isolated viral stock.

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

What is the process of cell inoculation in viral culture?

A

In viral culture, the host cells are inoculated with the viral sample to allow for virus replication and growth.

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

Describe how viral attachment to cells can be enhanced.

A

Viral attachment to cells can be enhanced by adding the sample directly to the cell culture medium or by using techniques like centrifugation.

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

What is the purpose of incubation in viral culture?

A

The purpose of incubation in viral culture is to allow the virus to infect the host cells and initiate the replication cycle.

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

What are cytopathic effects (CPE) in viral culture?

A

Cytopathic effects are changes in the appearance of infected cells due to viral replication, such as cell rounding, detachment, and cell death.

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

What is the purpose of harvesting in viral culture?

A

The purpose of harvesting in viral culture is to collect the culture medium containing the released virus and, in some cases, disrupt the host cells to release intracellular virus particles.

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

How is the harvested virus identified and characterized in viral culture?

A

The harvested virus is identified and characterized through various methods, including microscopy, serological tests, and molecular techniques.

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

Describe the process of subculturing in viral culture.

A

Subculturing involves transferring a portion of the harvested virus to a new batch of host cells. This process can be repeated to maintain the viral culture or generate larger quantities for further studies.

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

Describe the impact of influenza viruses on public health.

A

Seasonal flu outbreaks can lead to widespread illness, hospitalizations, and deaths. Influenza A viruses are also responsible for occasional pandemics.

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

What is the impact of HIV on global health?

A

HIV/AIDS has had a profound impact on global health, with millions of people affected worldwide. It causes acquired immunodeficiency syndrome (AIDS) by attacking the immune system.

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

What are the potential consequences of chronic HBV infection?

A

Chronic HBV infection can lead to liver cirrhosis and hepatocellular carcinoma. Hepatitis B is a major global health concern.

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

How does chronic HCV infection impact the liver?

A

Chronic HCV infection can cause liver damage, cirrhosis, and hepatocellular carcinoma. Hepatitis C is a significant cause of liver-related morbidity and mortality.

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

Describe the impact of herpes simplex viruses

A

Herpes simplex viruses (HSV-1 and HSV-2) cause oral and genital herpes infections.

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

What are the types of influenza viruses?

A

Influenza A, B, and C viruses are the types of influenza viruses.

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

What diseases are associated with Human Papillomavirus (HPV)?

A

Various types of HPV can cause genital warts and are associated with the development of cervical and other cancers.

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

Define Varicella-Zoster Virus (VZV) and its impact.

A

VZV causes chickenpox in childhood and can later re-emerge as shingles in adults. Vaccination has reduced the incidence of chickenpox.

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

What disease does the Measles Virus cause and what are its potential complications?

A

Measles is a highly contagious viral illness that can lead to severe complications, including pneumonia and encephalitis.

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

Describe the impact of Rotavirus.

A

Rotavirus is a common cause of severe diarrhea in young children worldwide. Vaccines have been introduced to reduce the burden of disease.

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

What is the concept of host range in viruses?

A

The concept of host range refers to the range of different host organisms that a particular virus can infect and successfully replicate within.

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

What is cell tropism in viruses?

A

Cell tropism refers to the preference of viruses for infecting specific types of cells within a given host organism. For example, a virus may have tropism for lung cells, liver cells, or immune cells within a human host.

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

How does the concept of host range influence the epidemiology and transmission of viral infections?

A

The host range of a virus is a crucial determinant of the types of diseases it can cause and influences the epidemiology and transmission of viral infections.

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

Define viral receptors and explain their role in determining host range.

A

Viral receptors are specific proteins on the surface of host cells that interact with viral surface proteins. The presence of compatible receptors is necessary for successful viral attachment and entry into host cells. The interaction between viral receptors and host cells determines the host range of a virus.

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

How can evolutionary factors influence the host range of a virus?

A

Evolutionary factors, such as mutations or genetic reassortment events, can affect the ability of a virus to infect new hosts. Changes in the genetic makeup of a virus can expand or limit its host range.

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

Describe the concept of virus-host coevolution.

A

Virus-host coevolution refers to the dynamic relationship between viruses and their hosts. As viruses evolve over time, they interact with their hosts and may undergo genetic changes that allow them to better infect or evade the host’s immune system. Similarly, hosts may develop immune responses to combat viral infections. This ongoing interaction between viruses and hosts drives their coevolution.

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

Define host range in the context of virology.

A

Host range refers to the range of different species that a virus can infect and replicate within.

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

Describe asymptomatic infection.

A

Asymptomatic infection refers to a viral infection where the individual does not show any noticeable symptoms but can still transmit the virus to others.

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

What are some examples of mild to moderate viral infections?

A

Examples of mild to moderate viral infections include the common cold, flu, and many childhood viral infections.

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

Define severe illness in the context of viral infections.

A

Severe illness refers to the development of serious and potentially life-threatening symptoms as a result of a viral infection, such as severe respiratory or systemic diseases.

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

How do the consequences of viral infections vary?

A

The consequences of viral infections can vary depending on factors such as the specific virus, the host organism, the individual’s immune response, pre-existing conditions, and the availability of medical interventions.

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

Describe chronic infection.

A

Chronic infection refers to a persistent infection caused by certain viruses that can last for an extended period or even a lifetime, leading to ongoing health issues and an increased risk of complications.

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

What is the impact of immunosuppression caused by certain viruses?

A

Immunosuppression caused by certain viruses, such as HIV, can suppress the host immune system, making the individual more susceptible to other infections.

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

Explain the concept of latency and reactivation in viral infections.

A

Latency refers to a state where the viral genome remains dormant within the host cells. Periodically, the virus may reactivate, leading to recurrent symptoms. Herpesviruses, like herpes simplex virus and varicella-zoster virus, exhibit latency and reactivation.

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

Describe the immune response to a viral infection.

A

The immune response to a viral infection is the host’s defense mechanism against the virus. It involves the activation of immune cells, such as T cells and B cells, which produce antibodies to neutralize the virus and eliminate infected cells.

199
Q

Describe viral latency.

A

Viral latency is a state in which a virus can persist within a host organism in an inactive or dormant form without active replication or production of new infectious virus particles.

200
Q

What are the key features of viral latency?

A

The key features of viral latency include a dormant state, no active replication, and host cell survival.

201
Q

Define the lytic cycle.

A

The lytic cycle is a viral replication cycle in which viruses actively replicate, cause cell damage, and produce new virions that can spread to other cells.

202
Q

How does viral latency differ from the lytic cycle?

A

Viral latency is a state of inactivity and dormancy, while the lytic cycle involves active viral replication, cell damage, and production of new virions.

203
Q

What happens to host cells during viral latency?

A

Host cells hosting latent viruses can survive for extended periods without apparent harm, unlike in the lytic cycle where host cell lysis and death often occur.

204
Q

What are some factors that can trigger the reactivation of latent viruses?

A

Reactivation of latent viruses can be triggered by factors such as stress, immune suppression, hormonal changes, or other infections.

205
Q

Define proviral latency.

A

Proviral latency is a form of latency where the viral DNA integrates into the host cell genome and remains transcriptionally silent.

206
Q

Do all viruses have the potential to reactivate from latency?

A

No, not all viruses have the potential to reactivate from latency. However, many viruses, especially herpesviruses, are known for establishing latency and can reactivate.

207
Q

Describe the reactivation of latent HIV.

A

Reactivation of latent HIV is a significant challenge in achieving a cure for HIV infection. In this state, the virus remains transcriptionally silent and can be triggered to resume active replication.

208
Q

Describe the advantages of viral latency for the virus.

A

Viral latency allows the virus to evade the host immune response by minimizing viral replication and reducing the production of viral antigens.

209
Q

What are the clinical implications of latent infections?

A

Latent infections can lead to recurrent symptoms or the spread of the virus to new host cells when the virus reactivates under certain conditions.

210
Q

Why is understanding viral latency important?

A

Understanding viral latency is crucial for developing strategies to control and treat persistent viral infections, as reactivation from latency is associated with symptom recurrence and transmission to new hosts.

211
Q

Describe the link between viruses and cancer.

A

Viruses can contribute to the development of cancer through various mechanisms, and infections with oncogenic viruses are associated with an increased risk of certain types of cancers.

212
Q

What are some examples of oncogenic viruses?

A

Some examples of oncogenic viruses are Human Papillomavirus (HPV), Hepatitis B Virus (HBV), Hepatitis C Virus (HCV), Epstein-Barr Virus (EBV), and Human T-cell Lymphotropic Virus-1 (HTLV-1).

213
Q

Which types of cancer are associated with Human Papillomavirus (HPV) infection?

A

HPV infection is associated with cervical cancer, anal cancer, and oropharyngeal cancer.

214
Q

How does Human Papillomavirus (HPV) contribute to the development of cancer?

A

HPV can integrate its DNA into the host genome, disrupting cell cycle regulation and promoting uncontrolled cell growth.

215
Q

What is the purpose of anti-viral therapy?

A

To target the virus without harming host cells and treat viral infections.

216
Q

Describe the principles of anti-viral therapy.

A

Inhibiting viral replication, blocking entry into host cells, or enhancing the host’s immune response.

217
Q

What factors are considered when selecting the right antiviral?

A

Virus type, its life cycle, and the patient’s health.

218
Q

Why is early initiation, adherence to prescribed regimens, and monitoring for side effects important in antiviral therapy?

A

They are crucial for optimal outcomes in antiviral therapy

219
Q

What are the indications for anti-viral therapy?

A

Viral infections where specific antiviral medications can target the virus without harming host cells.

220
Q

Describe how viral infections can be prevented.

A

Viral infections can be prevented through hygiene practices, vaccination, and maintaining healthy habits.

221
Q

. What are the key methods for eradicating viruses globally?

A

The key methods for eradicating viruses globally include vaccination, treatment, and international efforts.

222
Q

Define eradication of a virus.

A

Eradication of a virus refers to the complete elimination of the virus from a specific population or globally.

223
Q

Describe active immunisation

A

Active immunisation involves stimulating the immune system to produce an immune response against a specific pathogen through the use of vaccines.

224
Q

What is the purpose of vaccines in active immunisation?

A

Vaccines contain weakened or inactivated forms of the pathogen or its components to stimulate the immune system and provide protection upon future exposure.

225
Q

Define passive immunisation.

A

Passive immunisation involves the direct transfer of pre-formed antibodies or immune cells to an individual to provide immediate but temporary protection.

226
Q

Give an example of passive immunisation.

A

An example of passive immunisation is the administration of immune globulins or antitoxins.

227
Q

How do active and passive immunisation differ?

A

Active immunisation stimulates the immune system to produce an immune response, while passive immunisation involves the direct transfer of pre-formed antibodies or immune cells.

228
Q

Describe the process of vaccination

A

Vaccination involves administering a vaccine to stimulate the immune system, providing protection against specific diseases.

229
Q

Describe what contraindications to vaccination are.

A

Contraindications to vaccination are conditions or circumstances that suggest a person should not receive a particular vaccine due to potential risks.

230
Q

Define anaphylaxis.

A

Anaphylaxis is a severe allergic reaction that can occur in response to a component of a vaccine

231
Q

How does an immunocompromised status affect vaccination?

A

People with weakened immune systems, either due to illnesses like HIV/AIDS or immunosuppressive treatments, may be advised against certain vaccines.

232
Q

What is the potential risk of vaccinating during pregnancy?

A

Some vaccines are contraindicated during pregnancy due to potential risks to the developing fetus.

233
Q

Describe the significance of a history of vaccine-related adverse events

A

Individuals who had severe adverse reactions to a specific vaccine in the past may be advised against further doses

234
Q

Describe the first stage of an immune response.

A

Recognition of Antigen: Immune cells identify foreign substances, known as antigens, which can be parts of pathogens like bacteria or viruses.

235
Q

What is the role of dendritic cells in the immune response?

A

Dendritic cells present antigens to T cells, activating them

236
Q

What are the two types of T cells involved in immune responses?

A

Helper T cells and cytotoxic T cells.

237
Q

What happens when B cells are activated by helper T cells?

A

B cells differentiate into plasma cells that produce antibodies.

238
Q

What is the function of antibodies in the immune response?

A

Antibodies are proteins designed to neutralize or tag pathogens for destruction.

239
Q

What is the purpose of memory cells in the immune response?

A

Memory cells persist in the body to “remember” the specific antigen and provide faster and more robust responses upon future encounters with the same pathogen.

240
Q

Define immunological memory.

A

Immunological memory refers to the ability of the immune system to remember and mount a faster and more effective response upon re-exposure to a specific pathogen.

241
Q

What are the stages of an immune response?

A

Recognition of Antigen, Antigen Presentation, Activation of T Cells, B Cell Activation, Antibody Production, Memory Cells Formation.

242
Q

What is herd immunity?

A

Herd immunity is achieved when a significant portion of a population becomes immune to a disease, either through vaccination or previous infection.

243
Q

How does herd immunity help in controlling the spread of a disease?

A

Herd immunity helps slow or interrupt the spread of a disease, protecting those who are not immune.

244
Q

What are the common vaccinations administered during childhood?

A

Common vaccinations administered during childhood include Hepatitis B, DTP, Hib, IPV, Rotavirus, PCV, MMR, Varicella, Hepatitis A, and Influenza.

245
Q

Describe the life cycle of protozoa.

A

The life cycle of protozoa typically involves a single-celled organism and can have both sexual and asexual stages. Transmission can occur through ingestion of contaminated food water, insect vectors, or direct person-to-person contact. Eg plasmodium - causes malaria

246
Q

Describe the life cycle of helminths (worms).

A

The life cycle of helminths is generally more complex, often involving different stages in the host and external environment. Transmission can occur through ingestion of contaminated food or water, skin penetration, or insect vectors.

247
Q

What are some examples of flatworms (Platyhelminthes)?

A

Some examples of flatworms include Taenia (tapeworms) and Schistosoma.

248
Q

What are some examples of roundworms (Nematodes)?

A

Some examples of roundworms include Ascaris lumbricoides, hookworms, and filarial worms.

249
Q

Describe the life cycle of ectoparasites.

A

Ectoparasites live on the external surface of the host. Transmission often occurs through direct contact.

250
Q

What are some examples of arthropods that are ectoparasites?

A

Some examples of arthropods that are ectoparasites include lice, fleas, ticks, and mites.

251
Q

Describe giardiasis.

A

Giardiasis is an infection caused by the parasite Giardia lamblia, leading to gastrointestinal symptoms like diarrhea, abdominal cramps, and nausea.

252
Q

What is the cause of amoebiasis?

A

Amoebiasis is caused by the parasite Entamoeba histolytica, in severe diarrhea with blood mucus, abdominal pain, and fever.

253
Q

Define malaria.

A

Malaria is a disease transmitted by the Plasmodium parasite through the bite of infected mosquitoes. It causes symptoms such as fever, chills, and flu-like illness

254
Q

How is schistosomiasis transmitted?

A

Schistosomiasis is transmitted when parasitic flatworms called schistosomes penetrate the skin upon contact with contaminated water. Symptoms include fever, abdominal pain, and blood in the urine or stool.

255
Q

What are some measures to prevent enteric and blood-borne parasites?

A

Preventing these diseases involves practicing good hygiene, avoiding contaminated food and water, and taking measures to prevent insect bites, such as using bed nets and insect repellent.

256
Q

Define a parasite.

A

A parasite is an organism that lives in or on another organism (host) and derives its nourishment and other benefits at the expense of the host.

257
Q

Describe the role of clinical history and symptoms in the diagnostic principles of parasitology.

A

Clinical history and symptoms help guide the diagnostic process by providing specific signs and symptoms that may indicate a parasitic infection.

258
Q

What are the different types of clinical samples that can be collected for parasitology diagnostics?

A

Clinical samples for parasitology diagnostics can include blood, stool, urine, tissue biopsies, and other body fluids.

259
Q

What is the purpose of culturing in parasitology?

A

Culturing allows for the observation of developmental stages and facilitates species identification.

260
Q

Describe the identification of Staphylococcus aureus.

A

Gram-positive cocci in clusters.

261
Q

What are the distinguishing features of Staphylococcus aureus?

A

Catalase positive, coagulase positive, and often produces golden-yellow colonies on blood agar.

262
Q

How can Streptococcus pyogenes be identified?

A

Gram-positive cocci in chains.

263
Q

Define the distinguishing features of Streptococcus pyogenes.

A

Beta-hemolytic on blood agar, bacitracin-sensitive, and positive for Group A streptococcal antigen.

264
Q

Describe the identification of Streptococcus pneumoniae.

A

Gram-positive cocci in pairs or lancet-shaped.

265
Q

Describe the identification of Clostridium difficile.

A

Gram-positive, spore-forming bacilli.

266
Q

What are the identifying characteristics of Staphylococcus epidermidis?

A

Gram-positive cocci in clusters, coagulase-negative, novobiocin-resistant, and often part of the skin microbiota.

267
Q

Do Staphylococcus epidermidis bacteria produce coagulase?

A

No, Staphylococcus epidermidis is coagulase-negative.

268
Q

Describe how Escherichia coli (E. coli) can be identified.

A

Escherichia coli can be identified by its ability to ferment lactose on selective media like MacConkey agar. Biochemical tests, such as the indole test, can also be used.

269
Q

What is the identification method for Neisseria gonorrhoeae?

A

Neisseria gonorrhoeae can be identified by the presence of gram-negative diplococci seen on microscopy from clinical samples. Culture on selective media like Thayer-Martin agar can confirm its presence.

270
Q

How can Neisseria meningitidis be identified?

A

Neisseria meningitidis can be identified by the presence of gram-negative diplococci seen on microscopy, similar to N. gonorrhoeae.

271
Q

Describe how Helicobacter pylori is often identified.

A

Helicobacter pylori is often identified through biopsy samples from the stomach lining. Special staining methods like the urease test and PCR can confirm its presence.

272
Q

Describe the role of serotyping in the identification of bacteria.

A

Serotyping involves the classification of bacteria based on their specific O and H antigens, which can help differentiate between different strains or types of bacteria.

273
Q

Describe why some clinically significant bacteria are not readily identified by conventional Gram staining.

A

Some clinically significant bacteria are not readily identified by conventional Gram staining due to atypical cell wall structures or other characteristics.

274
Q

Define Treponema pallidum.

A

Treponema pallidum is the causative agent of syphilis and is thin and tightly coiled, making it difficult to visualize with standard Gram staining.

275
Q

Describe the shape of spirochetes.

A

Spirochetes have a helical shape.

276
Q

Describe hyphae.

A

Hyphae are thread-like structures that form the basic body of a fungus. They can be septate or non-septate.

277
Q

What is mycelium?

A

Mycelium is a mass of hyphae that constitutes the body of the fungus. It serves as the main vegetative structure involved in nutrient absorption.

278
Q

Define spores.

A

Spores are reproductive cells that can give rise to new fungi. They are produced through both sexual and asexual reproduction.

279
Q

What are fruiting bodies?

A

Fruiting bodies are specialized structures that house and produce spores. Examples include mushrooms in Basidiomycota and ascocarps in Ascomycota

280
Q

Describe the cell wall of fungi.

A

Fungal cell walls contain chitin, a complex polysaccharide, providing structural support.

281
Q

How many nuclei do fungal cells typically have?

A

Fungal cells typically have multiple nuclei.

282
Q

What are rhizoids?

A

Rhizoids are root-like structures in some fungi that anchor the fungus and aid in nutrient absorption.

283
Q

What are sporangia?

A

Sporangia are structures that produce and release spores in certain fungi, such as Rhizopus in Zygomycota.

284
Q

Describe the difference between septate and non-septate hyphae.

A

Septate hyphae are divided by cross-walls called septa, while non-septate hyphae are coenocytic and do not have these cross-walls.

285
Q

Describe Candidiasis.

A

Candidiasis is a fungal infection caused by Candida species, most commonly Candida albicans. It can affect various organs, leading to mucocutaneous candidiasis, systemic candidiasis, and invasive candidiasis. It is commonly seen in immunocompromised individuals.

286
Q

What is the causative agent of Aspergillosis?

A

Aspergillosis is caused by Aspergillus species, primarily Aspergillus fumigatus.

287
Q

Define Cryptococcosis.

A

Cryptococcosis is a fungal infection caused by Cryptococcus neoformans, commonly associated with HIV/AIDS patients. It primarily affects the lungs and central nervous system, causing pneumonia and meningitis.

288
Q

Describe Histoplasmosis.

A

Histoplasmosis is a fungal infection caused by Histoplasma capsulatum. It primarily affects the respiratory system and can lead to pulmonary histoplasmosis and disseminated histoplasmosis. It is common in endemic areas.

289
Q

What are the manifestations of Coccidioidomycosis?

A

Coccidioidomycosis causes respiratory infections, ranging from mild flu-like symptoms to severe pneumonia. It is caused by Coccidioides immitis and Coccidioides posadasii.

290
Q

Describe Tinea Corporis (Ringworm)

A

Circular, red, and scaly skin lesions on non-hairy areas of the body caused by various dermatophyte fungi.

291
Q

Do Tinea Pedis (Athlete’s Foot) cause

A

Fungal infection of the feet, causing itching, peeling, and redness, commonly caused by Trichophyton rubrum and Trichophyton mentagrophytes.

292
Q

Do Tinea Pedis (Athlete’s Foot) cause

A

Fungal infection of the feet, causing itching, peeling, and redness, commonly caused by Trichophyton rubrum and Trichophyton mentagrophytes.

293
Q

Describe Tinea Capitis

A

Fungal infection of the scalp, often seen in children, leading to hair loss and scaly patches, caused mainly by Microsporum and Trichophyton species.

294
Q

What is Tinea Unguium (Onychomycosis)?

A

Fungal infection of the nails, causing discoloration, thickening, and brittleness, caused by various dermatophytes including Trichophyton rubrum.

295
Q

Describe the immune status of individuals who are more susceptible to fungal infections.

A

Immunocompromised individuals, such as those with HIV/AIDS, undergoing chemotherapy, or receiving immunosuppressive therapy, are more susceptible to fungal infections.

296
Q

How does age affect susceptibility to fungal infections?

A

Age plays a role in susceptibility to fungal infections, with neonates and the elderly being more vulnerable due to the immaturity or decline of the immune system, respectively.

297
Q

Describe how hormonal fluctuations can impact the immune response.

A

Hormonal fluctuations, such as those occurring during pregnancy, menstruation, or hormonal therapy, can impact the immune response and create an environment favorable for fungal growth.

298
Q

What are some occupational or environmental exposures that can increase the risk of fungal infections?

A

Certain occupations or environmental exposures, such as working in agriculture or construction, can increase the risk of exposure to fungal spores, leading to respiratory infections.

299
Q

How can lifestyle and behaviors affect susceptibility to fungal infections?

A

Lifestyle and behaviors, such as smoking and substance abuse, can weaken the immune system and increase susceptibility to fungal infections.

300
Q

Explain how previous infections or colonization can increase the risk of recurrent fungal infections.

A

Individuals who have previously had fungal infections or are colonized by fungi may be more susceptible to recurrent infections, especially if the underlying risk factors persist.

301
Q

Describe the process of clinical evaluation for fungal infections.

A

Common clinical presentations of fungal infections include skin rashes, respiratory symptoms, or systemic manifestations.

302
Q

What are the techniques used in microscopic examination of clinical samples for identifying fungi?

A

The techniques used in microscopic examination include KOH Mount and Calcofluor White Stain.

303
Q

Describe the Enzyme-Linked Immunosorbent Assay (ELISA)

A

ELISA is a diagnostic test used to detect fungal antigens in blood or other body fluids.

304
Q

How do molecular methods contribute to fungal identification?

A

Molecular methods, such as Polymerase Chain Reaction (PCR) and DNA sequencing, are used to accurately and rapidly identify fungal species.

305
Q

Describe the mechanism of action of polyenes like Amphotericin B.

A

Polyenes bind to ergosterol in fungal cell membranes, leading to membrane disruption and cell death.

306
Q

What is the mechanism of action of Echinocandins like Caspofungin?

A

Echinocandins inhibit the synthesis of beta-glucan, a component of the fungal cell wall.

307
Q

What is the mechanism of action of 5-Flucytosine as an antifungal drug?

A

5-Flucytosine is converted to 5-fluorouracil inside the fungal cell, disrupting RNA and protein synthesis.

308
Q

Describe how Griseofulvin works as an antifungal drug.

A

Griseofulvin interferes with microtubule function, inhibiting mitosis and fungal growth.

309
Q

Define oral administration as a route of administration for antifungal drugs.

A

Oral administration refers to the administration of drugs through the mouth, commonly done with antifungal drugs like azoles and 5-flucytosine.

310
Q

What is the common route of administration for drugs like Amphotericin B and Caspofungin?

A

Amphotericin B and Caspofungin are often given intravenously (IV), especially in severe systemic infections.

311
Q

Describe topical administration of antifungal agents.

A

Topical administration involves applying antifungal agents directly to the skin or mucous membranes the treatment of fungal infections.

312
Q

What are some issues associated with antifungal agents?

A

Some issues associated with antifungal agents include resistance, drug interactions, toxicity, and limited spectrum of activity.

313
Q

Define fungal resistance to antifungal drugs.

A

Fungal resistance refers to the ability of fungi to survive and grow in the presence of antifungal drugs, reducing their effectiveness in treating fungal infections.

314
Q

What are some potential toxic effects of Amphotericin B?

A

Amphotericin B may cause toxic effects, including nephrotoxicity (kidney damage) and infusion-related reactions.

315
Q

What is hepatotoxicity and which antifungal agents may cause it?

A

Hepatotoxicity refers to liver damage. Some antifungal agents, such as azoles, may cause liver enzyme abnormalities and hepatotoxicity.

316
Q

Describe viral pathogenesis.

A

Viral pathogenesis refers to the process by which viruses cause disease in their host organisms, involving events from initial infection to clinical symptoms manifestation.

317
Q

What is tissue tropism in viral pathogenesis?

A

Tissue tropism refers to the preference of different viruses for specific tissues or cell types, influencing the organs affected during infection.

318
Q

What happens during primary replication in viral pathogenesis?

A

During primary replication, viruses undergo the synthesis of viral components and the production of progeny virions after entering host cells.

319
Q

How do viruses spread within the host?

A

Viruses can spread within the host through mechanisms such as cell-to-cell transmission, viremia (spread through the bloodstream), and neural spread (traveling along nerves).

320
Q

Describe the innate immune system’s response to infection.

A

The innate immune system responds rapidly to infection.

321
Q

Define cytopathic effects (CPE) caused by viruses.

A

Cytopathic effects refer to the damage caused to host cells by viruses.

322
Q

How do viruses evade recognition by the immune system?

A

Many viruses exhibit antigenic variation, where they change their surface proteins to evade recognition by the immune system.

323
Q

How can viral infections predispose individuals to secondary infections?

A

Viral infections can compromise the host’s immune defenses, allowing opportunistic pathogens to cause secondary bacterial or fungal infections.

324
Q

Define viral pathogenesis.

A

Viral pathogenesis refers to the complex interplay between the virus and the host, influenced by viral and host factors that contribute to the outcome of infection.

325
Q

Give examples of pathogens.

A

Examples of pathogens include influenza virus, Salmonella bacteria, and Plasmodium parasites.

326
Q

Define a commensal.

A

A commensal is an organism, typically a microorganism, that lives in or on another organism without causing harm or benefiting it.

327
Q

What is the symbiotic relationship between a comensal and its host?

A

Commensals have a symbiotic relationship with the host, deriving benefits from the association while neither significantly harming nor benefiting the host.

328
Q

How do commensal bacteria in the human gut influence the host’s immune system?

A

Commensal bacteria in the human gut can influence the host’s immune system without causing disease.

329
Q

Define pathogen.

A

A pathogen is a microorganism or other agent that causes disease in its host organism.

330
Q

Describe the role of skin and mucous membranes in innate immunity.

A

They form the first line of defense, preventing the entry of pathogens. Mucous membranes produce mucus, which traps and expels microbes.

331
Q

What are Pattern Recognition Receptors (PRRs) and what do they recognize?

A

PRRs are expressed by innate immune cells and recognize conserved patterns on pathogens (PAMPs).

332
Q

What is the process of phagocytosis and which cells are involved?

A

Phagocytosis is the engulfing and digestion of pathogens by neutrophils and macrophages. Dendritic cells also play a role in antigen presentation.

333
Q

How does the inflammatory response contribute to innate immunity?

A

Innate immune cells release cytokines, chemokines, and other mediators to initiate inflammation, recruit immune cells, and enhance blood flow to the site of infection.

334
Q

Explain the role of the complement system in innate immunity.

A

Complement proteins can opsonize pathogens, facilitating phagocytosis, and induce the lysis of microbial cells.

335
Q

What is the function of Natural Killer (NK) cells in innate immunity?

A

NK cells recognize and destroy infected or abnormal cells, providing early defense against viruses and tumor cells.

336
Q

Define PAMPs and give an example.

A

PAMPs (Pathogen-Associated Molecular Patterns) are conserved patterns on pathogens that are recognized by Pattern Recognition Receptors (PRRs). An example of a PAMP is lipopolysaccharide (LPS) found on the outer membrane of Gram-negative bacteria.

337
Q

Describe the role of dendritic cells in innate immunity.

A

Dendritic cells play a role in antigen presentation to the adaptive immune system, helping to initiate an adaptive immune response.

338
Q

What are acute-phase proteins and where are they produced?

A

Acute-phase proteins are produced by the liver and contribute to the systemic response and modulation of immune and inflammatory processes.

339
Q

Define antigen presentation.

A

Antigen-presenting cells (APCs), such as dendritic cells, present antigens derived from pathogens to T cells and B cells.

340
Q

How are T cells activated in response to antigens?

A

T cells, specifically CD4+ helper T cells and CD8+ cytotoxic T cells, are activated in response to antigens. CD4+ T cells help coordinate immune responses, while CD8+ T cells directly kill infected cells.

341
Q

Describe the activation of B cells in the immune response.

A

B cells are activated and differentiate into plasma cells, producing antibodies specific to the encountered pathogen.

342
Q

What are antibodies and what is their function?

A

Antibodies, also known as immunoglobulins, circulate in the blood and bind to specific pathogens or their toxins, neutralizing them, promoting phagocytosis, or activating complement.

343
Q

Describe immunological memory.

A

Immunological memory refers to the ability of T cells and B cells to develop memory cells that remember specific pathogens, allowing for a quicker and more effective immune response upon re-exposure.

344
Q

What is the role of regulatory T cells in the immune system?

A

Regulatory T cells help control the immune response by preventing excessive inflammation and maintaining tolerance to self-antigens.

345
Q

Describe colonization.

A

Colonization refers to the presence and multiplication of microorganisms on or within a host organism without causing disease.

346
Q

Define latent infection.

A

Latent infection occurs when a microorganism, typically a virus, persists within the host without causing active disease.

347
Q

What is an asymptomatic infection?

A

An asymptomatic infection is one in which the individual is infected with a microorganism, but no clinical symptoms or signs of illness are present.

348
Q

Describe infection.

A

Infection refers to the invasion and multiplication of microorganisms within a host organism, which can result in a range of outcomes from asymptomatic to severe illness.

349
Q

Describe clinical infection.

A

Clinical infection refers to an active and symptomatic manifestation of a microbial infection within a host organism.

350
Q

What are some common symptoms of clinical infection?

A

Common symptoms of clinical infection include fever, fatigue, pain, inflammation, respiratory symptoms, gastrointestinal symptoms, and other signs of illness.

351
Q

Define signs of inflammation in clinical infections.

A

Signs of inflammation in clinical infections can include redness, swelling, heat, and pain at the site of infection. Systemic inflammation can lead to symptoms such as fever.

352
Q

Describe pathogenicity.

A

Pathogenicity refers to the ability of a microorganism to cause disease in a host organism.

353
Q

Define infectivity.

A

Infectivity is a measure of the ability of a pathogen to establish an infection in a susceptible host and spread within the host organism.

354
Q

What is virulence?

A

Virulence is a measure of the severity of disease caused by a pathogen.

355
Q

What are some factors that can influence infectivity?

A

Factors may include the pathogen’s ability to evade host defenses, enter host cells, and replicate within the host.

356
Q

Describe the role of virulence factors.

A

Virulence factors, such as toxins, enzymes, or mechanisms for evading the immune system, contribute to the pathogen’s ability to cause harm.

357
Q

Describe pathogenic toxins.

A

Pathogenic toxins are substances produced by certain microorganisms that have the ability to cause harm to the host organism.

358
Q

What are the two types of pathogenic toxins?

A

The two types of pathogenic toxins are exotoxins and endotoxins.

359
Q

Give an example of an exotoxin and the bacteria that produces it.

A

An example of an exotoxin is botulinum toxin, which is produced by Clostridium botulinum.

360
Q

What is an endotoxin?

A

An endotoxin is part of the outer membrane of Gram-negative bacteria and is released when the bacteria die

361
Q

How do pathogenic toxins cause cellular damage?

A

Pathogenic toxins often target specific cells or tissues, leading to damage or dysfunction.

362
Q

What are the effects of neurotoxins and enterotoxins?

A

Neurotoxins affect nerve cells, while enterotoxins affect the digestive system.

363
Q

What cellular processes can toxins interfere with?

A

Toxins can interfere with essential cellular processes, such as protein synthesis, by disrupting ribosomes or blocking key enzymes.

364
Q

How are exotoxins and endotoxins released?

A

Exotoxins are often released during bacterial growth and are actively produced by the bacterium. Endotoxins are released when the bacterial cell wall is disrupted, such as during cell death.

365
Q

Describe botulism.

A

Botulism is a toxin-mediated disease caused by botulinum toxin, which leads to muscle paralysis.

366
Q

What are the symptoms of diphtheria?

A

Diphtheria is a toxin-mediated disease that results in respiratory and systemic symptoms.

367
Q

Define cholera.

A

Cholera is a toxin-mediated disease caused by cholera toxin, which leads to severe diarrhea and dehydration.

368
Q

What are the effects of tetanus toxin?

A

Tetanus is a toxin-mediated disease caused by tetanus toxin, which causes muscle stiffness and spasms.

369
Q

How do antitoxins work?

A

Antitoxins neutralize the effects of toxins and can be used to treat diseases caused by toxins.

370
Q

Describe how respiratory viruses can enter the body.

A

Respiratory viruses can enter the body through inhalation of respiratory droplets containing the virus or by touching surfaces contaminated with respiratory secretions and then touching the nose or mouth.

371
Q

What are some common sites of viral entry in the gastrointestinal tract?

A

The oral cavity is a common site of viral entry in the gastrointestinal tract. Contaminated food, water, or surfaces can serve as sources of viral entry.

372
Q

How can sexually transmitted viruses enter the body?

A

Sexually transmitted viruses can enter the body through the genital mucosa during sexual activity.

373
Q

What is the ocular surface and how can viruses enter through it?

A

The ocular surface refers to the eyes. Certain viruses, such as adenoviruses, can enter through the eyes by touching them with contaminated hands or exposure to respiratory secretions.

374
Q

Describe how bloodborne viruses can enter body through breaks in the skin.

A

Bloodborne viruses, such as HBV, HCV, and HIV, can enter the body through cuts, abrasions, or needlestick injuries.

375
Q

What is vertical transmission?

A

Vertical transmission is when certain viruses cross the placental barrier and infect the developing fetus during pregnancy.

376
Q

Define vector in the context of viral transmission.

A

In the context of viral transmission, a vector is an organism, such as a mosquito, that carries and transmits the virus to a host through a bite.

377
Q

Describe the characteristics of acute viral infections.

A

Acute viral infections are characterized by a sudden onset of symptoms, a relatively short duration, and an active viral replication phase within the host.

378
Q

What is the role of viral replication in acute viral infections?

A

Acute infections involve active viral replication within the host, leading to a high viral load in the early stages of infection.

379
Q

Name two specific viruses that cause acute respiratory symptoms.

A

Influenza virus and rhinovirus are two examples of viruses that cause acute respiratory symptoms.

380
Q

What are some examples of acute viral infections?

A

Examples of acute viral infections include influenza, the common cold, norovirus infection, chickenpox, and measles.

381
Q

Describe enteroviruses.

A

Enteroviruses are a group of viruses that primarily infect the gastrointestinal tract but can also affect other systems, including the respiratory and nervous systems.

382
Q

Name some common enteroviruses.

A

Common enteroviruses include coxsackieviruses, echoviruses, and enteroviruses.

383
Q

How are enteroviruses typically transmitted?

A

Enteroviruses are typically transmitted through the fecal-oral route, often via contaminated food, water, or surfaces.

384
Q

When do enterovirus infections often peak?

A

Enterovirus infections often exhibit a seasonal pattern, with higher incidence during the summer months.

385
Q

Describe Hand, Foot, and Mouth Disease (HFMD).

A

HFMD is caused by coxsackieviruses and is characterized by sores in the mouth and a rash on the hands and feet. It is common in children.

386
Q

Define Herpangina.

A

Herpangina is an illness caused by coxsackieviruses and is characterized by ulcers in the throat and mouth.

387
Q

What are the cardiac complications associated with enteroviruses?

A

Enteroviruses can cause inflammation of the heart (myocarditis) or the lining around the heart (pericarditis), leading to chest pain and cardiac symptoms.

388
Q

Describe why certain viruses are known as oncoviruses.

A

Certain viruses are known as oncoviruses because they have the potential to contribute to the development of cancer.

389
Q

What is insertional mutagenesis and how does it relate to oncoviruses?

A

Insertional mutagenesis is when oncoviruses integrate their genetic material into the host cell’s DNA, disrupting the normal regulation of cellular genes and leading to uncontrolled cell growth.

390
Q

Provide an example of an oncovirus that can integrate its DNA into the host genome.

A

Human Papillomavirus (HPV) can integrate its DNA into the host genome, contributing to the development of cervical and other cancers.

391
Q

Describe how Epstein-Barr virus (EBV) contributes to the development of lymphomas.

A

EBV can inhibit the function of the tumor suppressor gene p53, which promotes tumor development.

392
Q

Define genomic instability and how it relates to tumor development.

A

Genomic instability refers to the increased likelihood of genetic mutations within host cells, which can promote tumor development.

393
Q

Describe humoral immunity.

A

Humoral immunity is a branch of the immune system that involves B cells and the production of antibodies. It takes place in the blood and lymph and functions to neutralize pathogens, activate the complement system, enhance phagocytosis, and eliminate toxins.

394
Q

Define cell-mediated immunity.

A

Cell-mediated immunity is a branch of the immune system that involves T cells, specifically cytotoxic T cells and helper T cells. It primarily takes place in infected cells and tissues and functions to directly kill infected cells.

395
Q

How does humoral immunity activate the complement system?

A

Humoral immunity activates the complement system, which is a group of proteins that enhance the immune response. Antibodies produced by B cells bind to pathogens, triggering the complement system to eliminate the pathogens.

396
Q

Describe the mechanism of Type 1 Hypersensitivity.

A

Type 1 Hypersensitivity involves the activation of effector cells- mast cells and basophils by antigen-specific IgE antibodies.

397
Q

Give an example of Type 2 Hypersensitivity.

A

An example of Type 2 Hypersensitivity is hemolytic transfusion reactions.

398
Q

What is the timing of Type 3 Hypersensitivity?

A

Type 3 Hypersensitivity can be acute or chronic, depending on the immune response.

399
Q

Define Type 3 Hypersensitivity.

A

Type 3 Hypersensitivity is immune complex-mediated hypersensitivity where antigen-antibody complexes deposit in tissues, activating complement and attracting inflammatory cells.

400
Q

What are the effector cells in Type 3 Hypersensitivity?

A

The effector cells in Type 3 Hypersensitivity are neutrophils and macrophages.

401
Q

Describe the mechanism of Type 2 Hypersensitivity.

A

Type 2 Hypersensitivity involves antibodies (IgG or IgM) binding to antigens on host cells, leading to cell destruction or dysfunction.

402
Q

Give an example of Type 1 Hypersensitivity.

A

An example of Type 1 Hypersensitivity is hay fever.

403
Q

Describe the mechanism of Type 4 hypersensitivity.

A

Type 4 hypersensitivity involves sensitized T cells recognizing antigens, leading to the recruitment of inflammatory cells.

404
Q

What are the effector cells in Type 4 hypersensitivity?

A

T cells and macrophages cause tissue damage in Type 4 hypersensitivity.

405
Q

Define delayed-type hypersensitivity (DTH).

A

Delayed-type hypersensitivity is a type 4 hypersensitivity reaction that has a delayed onset, typically appearing 24 to 72 hours after exposure.

406
Q

Describe the process of sensitization.

A

Sensitization is the initial exposure to an allergen, which leads to the activation and differentiation of CD4+ T helper cells into Th2 cells.

407
Q

What is the role of antigen-presenting cells (APCs) in sensitization?

A

APCs process and present allergenic peptides to CD4+ T helper cells during sensitization.

408
Q

How do Th2 cells contribute to sensitization?

A

Th2 cells release cytokines, particularly interleukin-4 (IL-4) and interleukin-13 (IL-13), which promote B cell class switching to produce IgE antibodies.

409
Q

Define class switching in the context of sensitization.

A

Class switching refers to the process in which B cells produce IgE antibodies specific to the allergen during sensitization.

410
Q

Describe the process of re-exposure to an allergen after sensitization.

A

Upon subsequent exposure to the same allergen, it binds to multiple IgE antibodies on the surface of sensitized mast cells and basophils, leading to the release of inflammatory mediators.

411
Q

What is degranulation and what triggers it?

A

Degranulation is the release of pre-formed granules containing inflammatory mediators, such as histamine, leukotrienes, and prostaglandins. It is triggered by the activation of mast cells and basophils.

412
Q

What are the effects of histamine release during an allergic response?

A

Histamine causes vasodilation, increased vascular permeability, and smooth muscle contraction.

413
Q

How do leukotrienes and prostaglandins contribute to an allergic response?

A

Leukotrienes and prostaglandins contribute to inflammation, bronchoconstriction, and increased mucus production.

414
Q

What is the early phase of the allergic response?

A

The early phase of the allergic response refers to the immediate immune responses that occur within minutes of allergen exposure.

415
Q

Define Fc receptors.

A

F receptors are the high-affinity receptors for IgE found on the surface of mast cells, basophils, and a subset of antigen-presenting cells (APCs) like dendritic cells.

416
Q

What is the sensitisation function of the IgE-Fc interaction?

A

The binding of IgE to Fc sensitizes mast cells and basophils to specific antigens.

417
Q

What is the function of IgG antibodies?

A

IgG antibodies are involved in various immune functions, including opsonization and neutralization.

418
Q

Define opsonization.

A

Opsonization is the process in which IgG antibodies bind to pathogens, marking them for phagocytosis by immune cells that express Fc receptors.

419
Q

How do IgG antibodies contribute to immune complex formation?

A

IgG antibodies can form immune complexes with antigens, which are recognized and cleared by cells expressing Fcγ receptors.

420
Q

Describe the role of Fcγ receptors.

A

Fcγ receptors are receptors for the Fc region of IgG antibodies and are expressed on various immune cells, including macrophages, neutrophils, monocytes, and B cells.

421
Q

What is the most abundant class of antibodies in the circulation?

A

Immunoglobulin G (IgG) is the most abundant class of antibodies in the circulation.

422
Q

What is the function of Fcγ receptors on phagocytes?

A

Fcγ receptors on phagocytes, such as macrophages and neutrophils, bind to the Fc region of IgG antibodies attached to pathogens, facilitating phagocytosis.

423
Q

Describe the role of Fcγ receptors in immune complex clearance.

A

Fcγ receptors on various immune cells are involved in the clearance of immune complexes, preventing their deposition in tissues.

424
Q

What is the function of opsonization in immune complex clearance?

A

Opsonization of bacteria by IgG antibodies enhances phagocytosis by macrophages and neutrophils.

425
Q

What immune responses are triggered by the binding of antibodies to Fc receptors?

A

The binding of antibodies to Fc receptors triggers various immune responses, such as degranulation, phagocytosis, and immune complex clearance.

426
Q

Define atopy.

A

Atopy refers to a genetic predisposition to develop allergic reactions.

427
Q

Summarise Type 2 hypersensitivity

A

Type 2: Involves direct cell damage, cytotoxic T cells, and macrophages. Rapid onset.

428
Q

Summarise Type 3 hypersensitivity

A

Type 3: Involves immune complex formation, neutrophils, and macrophages. Delayed onset.

429
Q

What are autoimmune diseases?

A

Autoimmune diseases result from an immune response against the body’s own tissues.

430
Q

Define central tolerance in T cell development.

A

Central tolerance is the process in the thymus where T cells with high affinity for self-antigens are eliminated through negative selection.

431
Q

How can autoreactive T cells become activated in peripheral tissues?

A

Autoreactive T cells may become activated in peripheral tissues due to exposure to self-antigens that are not encountered in the thymus.

432
Q

Describe the role of proinflammatory cytokines in autoimmune diseases.

A

Proinflammatory cytokines contribute to inflammation and tissue damage, especially in organs targeted by autoimmune responses.

433
Q

Define molecular mimicry in the context of T cell responses.

A

Molecular mimicry refers to the ability of T cells to respond to both foreign antigens and self-antigens due to molecular similarities.

434
Q

Describe chronic stimulation in the context of autoimmune diseases.

A

Chronic stimulation refers to persistent antigen exposure and continuous inflammation, which can sustain T cell activation and contribute to tissue damage in autoimmune diseases.

435
Q

What are some examples of autoimmune diseases where T cells play a significant role in tissue damage?

A

Some examples of autoimmune diseases where T cells play a significant role in tissue damage include rheumatoid arthritis, multiple sclerosis, type 1 diabetes, and systemic lupus erythematosus.

436
Q

What is immune tolerance?

A

Immune tolerance refers to the immune system’s ability to recognize and tolerate the body’s own tissues and molecules, preventing harmful immune responses against self-antigens.

437
Q

Describe systemic autoimmune disease.

A

Systemic autoimmune disease affects multiple organs and presents with diverse and often systemic symptoms.

438
Q

Describe organ-specific autoimmune disease.

A

Organ-specific autoimmune disease targets a specific organ or tissue, leading to symptoms related to that organ.

439
Q

What does MIC stand for?

A

MIC stands for Minimum Inhibitory Concentration.

440
Q

How is MIC determined?

A

MIC is determined through laboratory testing, often using techniques like broth dilution or agar dilution.

441
Q

What does a lower MIC value indicate?

A

A lower MIC value indicates greater potency, as it means that a lower concentration of the antimicrobial agent is needed to inhibit bacterial growth.

442
Q

What does MBC stand for?

A

MBC stands for Minimum Bactericidal Concentration.

443
Q

What does MBC represent?

A

MBC represents the lowest concentration of an antimicrobial agent that kills a specified percentage (commonly 99.9% or more) of the bacterial population.

444
Q

Describe the difference between MIC and MBC.

A

MIC is the concentration that inhibits visible growth, while MBC is the concentration that kills a specified percentage of the bacterial population.

445
Q

What is the purpose of MBC testing?

A

To assess the bactericidal activity of an antimicrobial agent.

446
Q

Describe the clinical relevance of MBC testing.

A

MBC helps assess the ability of an antibiotic to kill bacteria. Some antibiotics are considered bacteriostatic at concentrations near the MIC and bactericidal at concentrations significantly above the MIC.

447
Q

Describe the term ‘sensitive’ in the context of antimicrobial susceptibility testing.

A

A microorganism is considered sensitive or susceptible to an antimicrobial agent if it can be inhibited or killed by concentrations of the agent that are achievable with normal dosages.

448
Q

What is the clinical implication of a microorganism being sensitive to an antibiotic?

A

When a microorganism is sensitive to an antibiotic, it means that the antibiotic is likely to be effective in treating an infection caused by that particular microorganism.

449
Q

How is sensitivity often denoted in susceptibility reports?

A

In susceptibility reports, the interpretation of sensitivity is often denoted as ‘S’ or ‘Susceptible.’

450
Q

Define the term ‘resistant’ in the context of antimicrobial susceptibility testing.

A

A microorganism is considered resistant to an antimicrobial agent if it cannot be inhibited or killed by concentrations of the agent that are achievable with normal dosages.

451
Q

What is the clinical implication of a microorganism being resistant to an antibiotic?

A

Resistance implies that the microorganism is not responsive to the antibiotic in question, and an alternative treatment may be necessary.

452
Q

Describe the interpretation of resistance in susceptibility reports.

A

Resistance is often denoted as ‘R’ or ‘Resistant’ in susceptibility reports.

453
Q

Define intermediate susceptibility.

A

Intermediate susceptibility indicates that the microorganism may be inhibited by higher-than-normal doses of the antibiotic.

454
Q

What is the clinical implication of intermediate susceptibility?

A

Intermediate susceptibility suggests that the effectiveness of the antibiotic may be limited, and its use should be approached with caution.

455
Q

Describe synergy.

A

Synergy occurs when the combined effect of two or more substances is greater than the sum of their individual effects.

456
Q

Define antagonism.

A

Antagonism occurs when the combined effect of two or more substances is less than the sum of their individual effects.

457
Q

Describe the mechanism of action of beta-lactam antibiotics.

A

Beta-lactam antibiotics inhibit bacterial cell wall synthesis by binding to penicillin-binding proteins (PBPs).Penicillins, Cephalosporins, Carbapenems:

458
Q

What is the effect of quinolones on bacterial cells?

A

Quinolones prevent DNA unwinding and replication, leading to bacterial cell death.Ciprofloxacin, Levofloxacin:

459
Q

Define the mechanism of action of macrolides.

A

Macrolides inhibit bacterial protein synthesis by binding to the 50S ribosomal subunit.Erythromycin, Azithromycin:

460
Q

How do tetracyclines disrupt bacterial protein synthesis?

A

Tetracyclines prevent aminoacyl-tRNA binding by binding to the 30S ribosomal subunit.Doxycycline, Minocycline:

461
Q

Describe the mechanism of action of aminoglycosides.

A

Aminoglycosides inhibit bacterial protein synthesis by causing misreading of mRNA.Gentamicin, Amikacin:

462
Q

What is the mechanism of action of azoles?

A

Azoles inhibit fungal cytochrome P450 enzymes, disrupting ergosterol synthesis.

463
Q

What effect do azoles have on fungal cell membranes?

A

Azoles alter fungal cell membrane integrity, leading to cell death.

464
Q

Do beta-lactam antibiotics weaken or strengthen the bacterial cell wall?

A

Beta-lactam antibiotics weaken the bacterial cell wall, leading to cell lysis.

465
Q

Beta-lactam antibiotics weaken the bacterial cell wall, leading to cell lysis.

A

Binds to ergosterol in fungal cell membranes.

466
Q

What is the effect of Amphotericin B on fungal cells?

A

Forms pores, causing leakage of cellular contents and fungal cell death.

467
Q

How do Echinocandins work?

A

They inhibit fungal cell wall synthesis by targeting beta-glucan synthesis.

468
Q

What is the effect of Echinocandins on fungal cells?

A

They weaken the fungal cell wall, leading to cell lysis.

469
Q

Define Nucleoside/Nucleotide Analog Reverse Transcriptase Inhibitors (NRTIs).

A

They are antiviral drugs that incorporate into viral DNA, terminating chain elongation.

470
Q

Describe the mechanism of action of Protease Inhibitors.

A

They inhibit viral protease, preventing maturation of viral proteins.

471
Q

How do Neuraminidase Inhibitors work?

A

They inhibit the neuraminidase enzyme, preventing the release of viral progeny.

472
Q

Describe the spectrum of penicillins.

A

Penicillins are effective against a broad range of Gram-positive bacteria, including Streptococcus and Staphylococcus species.

473
Q

What is the spectrum of azoles, such as Fluconazole?

A

Broad-spectrum antifungal activity.

474
Q

What viruses are Neuraminidase Inhibitors, such as Oseltamivir, effective against?

A

Influenza viruses (Type A and B).

475
Q

Describe the resistance mechanism of beta-lactam antibiotics known as beta-lactamase production.

A

Bacteria produce enzymes (beta-lactamases) that hydrolyze the beta-lactam ring, inactivating the antibiotic.

476
Q

What is the resistance mechanism of quinolones?

A

Changes in target enzymes reduce the binding affinity of quinolones.

477
Q

Define the resistance mechanism of macrolides called ribosomal modification.

A

Modification of the bacterial ribosome prevents binding of macrolides.

478
Q

How do bacteria develop resistance to beta-lactam antibiotics through altered Penicillin-Binding Proteins (PBPs)?

A

Bacteria modify PBPs, reducing the binding affinity of beta-lactam antibiotics.

479
Q

What is the resistance mechanism of beta-lactam antibiotics involving efflux pumps?

A

Bacteria pump out antibiotics, reducing intracellular drug concentrations.

480
Q

What is the resistance mechanism of tetracyclines in bacteria?

A

Bacteria pump out tetracyclines using efflux pumps and proteins bind to the bacterial ribosome, preventing tetracycline binding.

481
Q

How does the site of infection influence the choice of antimicrobial treatment?

A

Different body sites may require different antimicrobials due to variations in tissue penetration and bioavailability.

482
Q

Do host factors play a role in selecting the appropriate antimicrobial treatment?

A

Yes, patient-related factors such as age, weight, renal and hepatic function, and immune status should be considered when choosing antimicrobial dosages.

483
Q

Describe the importance of assessing the severity of an infection when selecting antimicrobial treatment.

A

Assessing the severity helps determine the appropriate choice of antimicrobial and dosage, based on the clinical presentation and potential complications.

484
Q

What should be checked for when considering allergies to specific antimicrobials or drug classes?

A

Known allergies to specific antimicrobials or drug classes should be checked.

485
Q

Do microbial sensitivity testing help guide antibiotic selection?

A

Yes, microbial sensitivity testing can help guide antibiotic selection.

486
Q

Describe the importance of avoiding unnecessary prolonged courses of antimicrobial treatment.

A

To minimize the risk of resistance.

487
Q

How should the route of administration be determined for antimicrobial treatment?

A

Based on the patient’s condition and the severity of the infection.

488
Q

How can patient compliance be enhanced in antimicrobial treatment?

A

By choosing antimicrobials with convenient dosing regimens and educating patients on the importance of completing the prescribed course of treatment.

489
Q

Describe the common side effects of beta-lactam antibiotics.

A

Allergic reactions and gastrointestinal upset are common side effects of beta-lactam antibiotics.

490
Q

What are the common side effects of quinolones?

A

Gastrointestinal upset and central nervous system effects, such as headache and dizziness, are common side effects of quinolones.

491
Q

Define macrolides and list their common side effects.

A

Macrolides are a class of antibiotics, and their common side effects include gastrointestinal upset, liver enzyme abnormalities, and QT interval prolongation.

492
Q

What are the common side effects of tetracyclines?

A

Gastrointestinal upset and increased sensitivity to sunlight (photosensitivity) are common side effects of tetracyclines.

493
Q

Describe the common side effects of aminoglycosides.

A

Nephrotoxicity (kidney damage) and ototoxicity (hearing loss) are common side effects of aminoglycosides.

494
Q

What are the common side effects of azoles?

A

Gastrointestinal upset, liver enzyme abnormalities, and QT interval prolongation are common side effects of azoles.

495
Q

Define polyenes and list their common side effects.

A

Polyenes are a class of antifungal drugs, and their common side effects include infusion-related reactions (fever, chills) and nephrotoxicity.

496
Q

Describe the role of laboratories in pathogen identification.

A

Laboratories identify the causative pathogens in clinical samples.

497
Q

How does knowing the specific pathogen impact antimicrobial usage?

A

Knowing the specific pathogen allows for targeted and appropriate antimicrobial therapy. It helps avoid broad-spectrum empiric treatments, minimizing the risk of resistance and reducing unnecessary drug exposure.

498
Q
A
499
Q
A