Block 1 Exam 4 Part 2 Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

What happens to anti-apoptotic proteins like BCL-2 and BCL-X_L as the T cell response declines?

A

Their levels drop, leading to increased cell death.

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

Which protein is activated due to growth factor deprivation and triggers apoptosis?

A

BH3-only protein BIM.

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

What role do phosphatases such as SHP-1 and SHP-2 play in T cell activation?

A

They dephosphorylate signaling substrates to limit the duration or magnitude of T cell activation.

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

What is the role of PD-1 in T cell regulation?

A

It restrains previously activated CD8+ T cells in peripheral tissues, limiting their activity.

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

How do regulatory T cells help in the decline of T cell responses?

A

By engaging CTLA-4 with B7 on APCs and suppressing effector T cell responses, thereby limiting immune responses and preventing autoimmunity and allergy.

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

What is the ultimate source of genetic variation?

A

Mutation

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

What is the probability that a parent will transmit a specific allele at a locus?

A

1/2, as each allele has an equal chance of being transmitted.

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

What is the multiplication rule in probability?

A

The probability of independent events happening together is the product of their individual probabilities.

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

What is the addition rule in probability?

A

The probability of either one outcome or another is the sum of their individual probabilities.

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

If a couple wants to know the probability that all three of their children will be girls, what is the answer?

A

1/2 _ 1/2 _ 1/2 = 1/8.

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

What is the probability of getting either two heads or two tails in two coin tosses?

A

1/4 + 1/4 = 1/2.

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

What is the probability that a couple planning three children will have all of the same sex (either all boys or all girls)?

A

1/8 + 1/8 = 1/4.

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

What does genotype frequency represent?

A

The proportion of each genotype in a population.

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

do you calculate the frequency of a genotype in a population?

A

Divide the number of individuals with that genotype by the total number of individuals in the population.

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

If 64 out of 200 people have the MM genotype, what is the frequency of MM?

A

0.32 (64/200)

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

What does gene frequency represent?

A

The proportion of each allele at a specific locus in a population.

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

How do you calculate the frequency of an allele in a population?

A

Divide the number of copies of the allele by the total number of alleles at that locus.

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

How do you calculate the frequency of allele M if there are 248 M alleles out of 400 total alleles?

A

0.62 (248/400).

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

What is the relationship between the sum of all genotype frequencies and the sum of all gene frequencies in a population?

A

Both sums must equal 1.

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

What does the Hardy-Weinberg principle describe?

A

The relationship between gene frequencies and genotype frequencies in a population under random mating conditions.

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

What are the allele frequencies labeled as in the Hardy-Weinberg principle?

A

p for allele A and q for allele a.

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

How do you calculate the frequency of the AA genotype using allele frequencies?

A

p_ (p multiplied by p).

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

How do you calculate the frequency of the aa genotype?

A

q_ (q multiplied by q).

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

What is the formula for the frequency of the heterozygote genotype (Aa)?

A

2pq.

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

In a population, if the frequency of allele A (p) is 0.7, what is the frequency of allele a (q)?

A

0.3 (since p + q = 1).

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

What does q_ represent in the context of recessive diseases?

A

The frequency of affected individuals (homozygotes aa) in the population.

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

If q_ for a recessive disease like cystic fibrosis is 1/2500, what is the value of q?

A

0.02 (q = Ã(1/2500)).

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

How do you estimate the frequency of heterozygous carriers (Aa) if q is 0.02?

A

2pq, which simplifies to 2q because p Å 1, so 2 _ 0.02 = 1/25.

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

Why are recessive disease alleles often hidden in populations?

A

Because they are commonly carried by heterozygotes (Aa) who do not show symptoms of the disease.

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

What is the source of all genetic variation?

A

Mutation

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

How does natural selection affect genetic variation?

A

It increases the frequency of favorable mutations and decreases the frequency of unfavorable ones.

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

What typically happens to disease-causing mutations in a population?

A

They are continually introduced through errors and removed by natural selection.

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

How can a disease mutation provide a selective advantage?

A

In certain environments, like areas with malaria, a mutation can provide survival benefits, as seen with sickle cell heterozygotes.

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

Why do sickle cell heterozygotes have an advantage in malaria-endemic regions?

A

The malaria parasite does not survive well in the erythrocytes of sickle cell heterozygotes, reducing their risk of dying from malaria.

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

What happens to the frequency of the sickle cell mutation in environments without malaria?

A

Natural selection acts against it, reducing its frequency.

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

Why does the sickle cell mutation persist at a high frequency in certain populations?

A

Because it provides a survival advantage in malaria-endemic regions, despite being harmful in homozygotes.

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

What does the example of sickle cell disease illustrate about genetic variation?

A

It shows how natural selection can cause variation in the incidence of genetic diseases among different populations based on environmental factors.

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

What is natural selection?

A

It is the process where alleles that provide survival or reproductive advantages increase in frequency, while those with disadvantages decrease.

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

How does natural selection affect skin pigmentation as humans moved to higher latitudes?

A

It increased the frequency of variants that adapted skin pigmentation to the new environments.

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

What is an example of natural selection related to lactase persistence?

A

In populations that began drinking cow’s milk through adulthood, natural selection increased the frequency of hereditary lactase persistence.

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

How has natural selection acted on populations living in high-altitude environments?

A

It has increased the frequency of adaptations in the hypoxia-inducing factor pathway, allowing survival in oxygen-deficient areas like the Tibetan plateau.

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

What is genetic drift?

A

A random evolutionary process that causes larger changes in gene frequencies in smaller populations.

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

What is the founder effect?

A

It is a special case of genetic drift where a small founder population experiences large changes in gene frequency due to its small size.

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

How did genetic drift affect the Old Order Amish population in Pennsylvania?

A

Genetic drift increased the frequency of Ellis-van Creveld syndrome among the Amish, a population founded by about 50 couples.

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

What is gene flow?

A

The exchange of genes between populations, which tends to make them genetically more similar over time.

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

What happens to sickle cell disease frequency in African Americans compared to African populations due to gene flow?

A

Gene flow between African Americans and European Americans decreased the frequency of sickle cell disease in African Americans.

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

What is mutation-selection balance?

A

It predicts a relatively constant gene frequency when new mutations introduce harmful alleles, while natural selection removes them.

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

Why are recessive disease-causing alleles generally more common than dominant ones?

A

Because recessive alleles are often hidden in heterozygotes, protecting them from natural selection.

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

What does the selection coefficient (s) represent?

A

It represents the reduction in offspring for those carrying a specific allele, used to predict gene frequency in mutation-selection balance.

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

What is non-Mendelian inheritance?

A

It refers to patterns of inheritance that do not follow Gregor Mendel’s principles, including sex-linked mutations, mitochondrial inheritance, anticipation, and imprinting.

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

What are sex-linked mutations?

A

Mutations that occur on the sex chromosomes (X and Y). Diseases caused by these mutations are referred to as X-linked or Y-linked.

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

What is significant about the X chromosome in relation to disease?

A

The X chromosome is large (155 Mb) and contains nearly 1300 genes, many of which are associated with X-linked diseases.

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

How does the Y chromosome compare to the X chromosome?

A

The Y chromosome is much smaller (60 Mb) and contains only a few dozen genes.

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

What is mitochondrial inheritance?

A

It refers to the inheritance of the mitochondrial genome, which is passed down only from the mother.

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

Why do mitochondrial diseases have a unique pattern of inheritance?

A

Because the mitochondrial genome is inherited exclusively from the mother, mitochondrial diseases show a maternal inheritance pattern.

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

What is anticipation in genetics?

A

Anticipation is the phenomenon where some genetic diseases manifest at an earlier age in more recent generations of a family.

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

What is genetic imprinting?

A

Imprinting is when certain genes are expressed only from the chromosome inherited from one parent, either maternal or paternal.

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

What is X inactivation?

A

It is the process by which one of the two X chromosomes in each somatic cell of a female is randomly inactivated to achieve dosage compensation.

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

Why is X inactivation necessary?

A

To equalize the amount of X-linked gene products between males (who have one X chromosome) and females (who have two X chromosomes).

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

When does X inactivation occur?

A

Early in female embryonic development.

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

How is the X chromosome chosen for inactivation?

A

The X chromosome to be inactivated is chosen randomly in each cell, resulting in roughly half of the cells having the maternal X chromosome active and the other half having the paternal X chromosome active.

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

What happens to the X chromosome after it is inactivated?

A

It remains inactive in all descendants of that cell.

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

Why are females considered mosaics in terms of X chromosome activity?

A

Because they have two populations of cells, one with an active maternal X chromosome and the other with an active paternal X chromosome.

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

Are males mosaics for the X chromosome?

A

No, males are hemizygous for the X chromosome, meaning they have only one X chromosome and do not undergo X inactivation.

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

What are sex-linked genes?

A

Genes located on either the X or the Y chromosome.

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

Why are X-linked diseases more common than Y-linked diseases?

A

The X chromosome is larger and contains more genes compared to the Y chromosome.

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

Why are X-linked recessive diseases more common in males than females?

A

Males have only one X chromosome, so if they inherit the disease-causing allele, they will express the disease.

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

Can X-linked recessive diseases be passed from father to son?

A

No, fathers pass their Y chromosome to sons, not the X chromosome.

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

How can X-linked recessive diseases appear to skip generations?

A

The disease can be passed through phenotypically normal heterozygous females (carriers), who may pass it on to their sons.

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

What is a manifesting heterozygote?

A

A female carrier of an X-linked recessive disease who shows mild symptoms due to random X inactivation favoring the diseased allele.

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

Why might a female with Turner syndrome be affected by an X-linked recessive disease?

A

Because she has only one X chromosome, any recessive disease allele on that chromosome will be expressed.

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

What is the probability of a female being affected by an X-linked recessive disease?

A

It is much lower than in males because females need two copies of the recessive allele to express the disease.

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

What is an example of an X-linked dominant disease?

A

Hypophosphatemic rickets, which impairs the kidneys’ ability to reabsorb phosphate.

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

Why are X-linked dominant diseases less common than X-linked recessive diseases?

A

X-linked dominant diseases are generally fewer and often less prevalent due to their inheritance patterns.

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

Why are X-linked dominant diseases more common in females than in males?

A

Females have two X chromosomes, so they are about twice as likely to inherit the disease-causing gene.

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

Can X-linked dominant diseases be passed from father to son?

A

No, because fathers pass the Y chromosome to their sons, not the X chromosome.

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

What happens to males with X-linked dominant diseases that are lethal?

A

They often do not survive to term, as seen in disorders like incontinentia pigmenti type 1 and Rett syndrome.

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

What is a key feature of Rett syndrome?

A

It is a neurodevelopmental disorder characterized by autistic behavior, intellectual disability, seizures, and gait ataxia.

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

How does random X inactivation affect the severity of X-linked dominant diseases in females?

A

It can lead to milder symptoms if a larger percentage of X chromosomes with the disease mutation are inactivated.

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

How are Y-linked traits inherited?

A

They are passed strictly from father to son.

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

What is an example of a Y-linked trait?

A

The gene that initiates differentiation of the embryo into a male.

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

What is a sex-limited trait?

A

A trait that occurs only in one of the sexes, such as uterine defects in females or testicular defects in males.

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

What is a sex-influenced trait?

A

A trait that occurs in both sexes but is more common in one sex, like male-pattern baldness.

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

Is male-pattern baldness strictly X-linked?

A

No, it is influenced by both X-linked and autosomal genes, which explains father-to-son transmission.

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

What is mitochondrial DNA (mtDNA)?

A

It is a small, circular DNA molecule found in mitochondria that encodes rRNAs, tRNAs, and polypeptides involved in oxidative phosphorylation.

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

How is mtDNA inherited?

A

Exclusively through the maternal line, as only eggs contribute mitochondria to the developing embryo.

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

What is heteroplasmy?

A

It is the presence of a mixture of normal and mutated mtDNA within a cell, leading to variable expression of mitochondrial diseases.

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

Why do mitochondrial diseases often affect the central nervous system?

A

The central nervous system requires large amounts of ATP, making it highly vulnerable to mtDNA mutations that impair energy production.

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

What is Leber hereditary optic neuropathy (LHON)?

A

A mitochondrial disease caused by missense mutations in mtDNA that leads to rapid loss of central vision due to optic nerve death

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

What is the role of single-base mutations in mitochondrial diseases?

A

They can cause disorders like myoclonic epilepsy with ragged-red fibers (MERRF) and mitochondrial encephalomyopathy with stroke-like episodes (MELAS), both of which are heteroplasmic and highly variable in expression.

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

What types of mutations cause Kearns-Sayre disease and Pearson syndrome?

A

Duplications and deletions in mtDNA.

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

How common are mitochondrial diseases?

A

Approximately 1 in 4000 individuals is affected by a mitochondrial disease, with most cases due to mitochondrial mutations

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

Can mitochondrial mutations contribute to aging?

A

Yes, it is suggested that mtDNA mutations accumulating through life due to free radical formation could contribute to the aging process.

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

What is the significance of mitochondrial mutations in common diseases?

A

Mitochondrial mutations are linked to late-onset deafness, some cases of type 2 diabetes, and possibly Alzheimer’s disease.

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

What is genomic imprinting?

A

It is a process where the expression of an allele depends on whether it is inherited from the mother or the father, leading to gene silencing of one allele.

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

How does genomic imprinting affect gene expression?

A

One allele is transcriptionally inactive due to imprinting, resulting in only one active copy of the gene.

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

What are imprinted genes typically associated with?

A

Heavily methylated DNA and chromatin condensation that inhibit transcription.

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

What is Prader-Willi syndrome (PWS) and how is it caused?

A

PWS is a disorder caused by a deletion on chromosome 15 inherited from the father, affecting genes that are normally active only on the paternal chromosome.

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

What is Angelman syndrome (AS) and how is it caused?

A

AS is a disorder caused by a deletion on chromosome 15 inherited from the mother, affecting genes that are normally active only on the maternal chromosome.

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

What is the gene associated with Angelman syndrome?

A

UBE3A, which is active only on the maternal chromosome in brain tissue.

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

What is Beckwith-Wiedemann syndrome and how is it caused?

A

It is an overgrowth disorder caused by imprinting errors on chromosome 11, leading to overexpression of genes like IGF2 or loss of expression of growth-regulating genes.

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

How does uniparental disomy contribute to Beckwith-Wiedemann syndrome?

A

By inheriting two copies of the paternal chromosome 11 or losing imprinting on the maternal copy, leading to overexpression of IGF2.

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

What causes Silver-Russell syndrome?

A

A loss of methylation of DMR1 on chromosome 11p15.5, leading to underexpression of IGF2 and diminished growth.

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

What distinguishes Silver-Russell syndrome from Beckwith-Wiedemann syndrome in terms of IGF2 expression?

A

Beckwith-Wiedemann syndrome is caused by overexpression of IGF2, while Silver-Russell syndrome results from underexpression of IGF2.

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

What is humoral immunity mediated by?

A

B lymphocytes; secreted antibodies

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

What is the main function of humoral immunity?

A

Defense against extracellular microbes and microbial toxins.

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

What is the key difference between humoral immunity and cell-mediated immunity?

A

Humoral immunity uses antibodies, while cell-mediated immunity is mediated by T lymphocytes.

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

What types of microorganisms does humoral immunity primarily combat?

A

Extracellular bacteria, fungi, and viruses before they infect cells or after being released from infected cells.

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

What happens if there are defects in antibody production?

A

Increased susceptibility to infections by bacteria, fungi, and viruses.

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

How do most current vaccines provide protection?

A

By stimulating the production of antibodies.

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

In what harmful conditions can antibodies mediate tissue injury?

A

In allergies, autoimmune diseases, blood transfusion reactions, and transplant rejection.

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

What does antibody-mediated elimination of antigens involve?

A

Effector mechanisms involving phagocytes and complement proteins.

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

Where are antibodies produced?

A

Plasma cells in secondary lymphoid organs, inflamed tissues, and bone marrow.

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

What is the significance of antibodies being transported across the placenta?

A

They protect the developing fetus by entering the fetal circulation.

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

Where do long-lived plasma cells reside?

A

Mainly in the bone marrow.

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

Why are antibodies the primary defense mechanism in mucosal organs and the fetus?

A

T lymphocytes are not transported into mucosal secretions or across the placenta.

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

What region of immunoglobulin (Ig) molecules mediates many of the effector functions of antibodies?

A

The Fc region.

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

Which immunoglobulin isotypes activate the complement system?

A

IgM and some IgG subclasses (IgG1, IgG2 to a limited extent, IgG3).

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

What is the only antibody function mediated entirely by antigen binding?

A

Neutralization.

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

How is complement activation triggered by antibodies?

A

By binding to a multivalent antigen, clustering antibody molecules close together.

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

How do antibodies neutralize microbes and microbial toxins?

A

By blocking the binding of microbes and toxins to cellular receptors.

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

What is an example of a microbe that uses surface molecules to infect host cells?

A

Influenza virus uses hemagglutinin to infect respiratory epithelial cells.

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

How do antibodies prevent microbes from interacting with cellular receptors?

A

Through steric hindrance, preventing the microbes from binding to receptors.

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

How do microbial toxins like tetanus toxin and diphtheria toxin cause injury?

A

By binding to specific cellular receptors and interfering with normal cell functions.

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

How do antibodies protect against microbial toxins like tetanus and diphtheria?

A

By preventing toxins from binding to their target receptors and causing tissue injury.

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

What is the role of IgA antibodies in the lumen of the gut?

A

They agglutinate microbes, reducing infectivity and facilitating clearance by peristalsis.

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

What effect can antibodies have on microbes besides steric hindrance?

A

They can induce conformational changes in microbial surface molecules, preventing receptor interaction.

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

What are the two primary antibody isotypes involved in neutralization?

A

IgG in the blood and IgA at mucosal sites.

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

What makes antibodies most effective at neutralizing microbes?

A

High affinity for their antigens, produced by affinity maturation.

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

How do some microbes evade antibody-mediated neutralization?

A

By mutating the genes encoding their surface antigens.

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

How do IgG antibodies promote the phagocytosis of microbes?

A

By coating (opsonizing) the microbes and binding to Fc receptors on phagocytes.

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

What immune cells express Fc receptors that promote phagocytosis of opsonized particles?

A

Mononuclear phagocytes and neutrophils.

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

What is the function of the Fc_ receptors on phagocytes?

A

To bind IgG-coated particles and promote phagocytosis.

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

Which complement activation product can coat microbes to promote their phagocytosis?

A

C3b

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

What is the process of coating particles to promote phagocytosis called?

A

Opsonization

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

Which Fc_ receptor has the highest affinity for IgG antibodies?

A

Fc_RI (CD64).

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

Which immune cells express the high-affinity Fc_RI receptor?

A

Macrophages and neutrophils.

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

What is the function of the Fc_RIIB receptor?

A

It is an inhibitory receptor that regulates immune responses.

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

What happens when multiple Fc receptors are cross-linked by IgG-coated particles?

A

It leads to phagocyte activation and engulfment of the particles.

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

What are the primary microbicidal substances produced by activated phagocytes?

A

Reactive oxygen species, nitric oxide, and hydrolytic enzymes.

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

What is ADCC (Antibody-Dependent Cell-Mediated Cytotoxicity)?

A

Process where NK cells and other leukocytes destroy antibody-coated cells.

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

Which receptor do NK cells use in ADCC?

A

Fc_RIIIA (CD16).

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

What triggers NK cells to perform ADCC?

A

Antibody-coated target cells.

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

Why doesn’t ADCC occur with free IgG in the plasma?

A

Fc_RIII binds only to clustered IgG on cell surfaces, not to free IgG.

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

What do NK cells release during ADCC?

A

Cytokines like IFN-_ and granule contents to kill the target cells.

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

Can ADCC be mediated by other cells?

A

Yes, macrophages can also mediate ADCC.

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

What is the complement system?

A

A set of serum and cell surface proteins involved in eliminating microbes and complementing antibody function.

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

How does complement activation enhance immune response?

A

It focuses immune attack on microbial surfaces and amplifies the immune response through sequential proteolysis of proteins.

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

What are zymogens in the context of complement activation?

A

Inactivated proteins that gain proteolytic activity through sequential activation, leading to a proteolytic enzyme cascade.

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

How are complement proteins activated?

A

By binding to microbial surfaces, antibodies, and other antigens, leading to their stable activation.

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

What role do regulatory proteins play in complement activation?

A

They inhibit complement activation on normal host cells to prevent damage, while microbes lack these regulatory proteins and are susceptible to complement activation.

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

What are the three pathways of complement activation?

A

Classical, alternative, and lectin pathways.

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

What activates the classical pathway of complement activation?

A

IgM and IgG antibodies bound to antigens.

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

What activates the alternative pathway of complement activation?

A

Microbial cell surfaces in the absence of antibody.

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

What activates the lectin pathway of complement activation?

A

Plasma lectins binding to surface carbohydrates on microbes.

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

What is the central event in all complement activation pathways?

A

Proteolysis of the complement protein C3 to generate C3a and C3b.

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

What is the role of C3b in complement activation?

A

C3b covalently attaches to microbial surfaces or antibody-bound antigens and promotes phagocytosis.

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

What enzyme complex cleaves C5 into C5a and C5b?

A

The C5 convertase.

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

What are the functions of C5a and C5b in complement activation?

A

C5a stimulates inflammation, and C5b contributes to pore formation in microbial membranes.

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

What initiates the alternative pathway of complement activation?

A

The stable attachment of C3b to microbial surfaces without the need for antibodies.

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

What is C3 tickover?

A

The continuous low-rate hydrolysis and cleavage of C3 to generate C3b.

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

What causes C3b to become covalently attached to cell surfaces?

A

The exposure of the reactive thioester bond in C3b after cleavage.

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

What happens to C3b if it does not form covalent bonds with cell surfaces?

A

It remains in the fluid phase and is quickly hydrolyzed, becoming inactive.

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

What is the role of Factor B in the alternative pathway?

A

Factor B binds to C3b on cell surfaces and is cleaved by Factor D to form the alternative pathway C3 convertase.

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

What is the C3bBb complex?

A

The alternative pathway C3 convertase that cleaves more C3 molecules.

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

How does the alternative pathway C3 convertase amplify complement activation?

A

By forming a complex with C3b and Bb, which cleaves additional C3 molecules.

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

What role does properdin play in the alternative pathway?

A

Properdin binds to and stabilizes the C3bBb complex, especially on microbial surfaces.

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

What forms the alternative pathway C5 convertase?

A

A complex containing one Bb moiety and two molecules of C3b.

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

What initiates the classical pathway of complement activation?

A

Binding of C1 to the CH2 domain of IgG or the CH3 domain of IgM bound to antigen.

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

Which IgG subclasses are the most efficient at activating the classical pathway?

A

IgG1 and IgG3.

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

Why can only antibodies bound to antigens initiate classical pathway activation?

A

Because C1q needs to bind to at least two Ig heavy chains to be activated, which requires antibodies to be bound to antigens.

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

What role does C1q play in the classical pathway?

A

C1q binds to the Fc regions of IgG or IgM, initiating the activation of C1r and C1s.

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

How does C1s contribute to complement activation in the classical pathway?

A

C1s cleaves C4 into C4b and C4a, with C4b forming covalent bonds on the cell surface.

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

What is the function of the C4b2a complex?

A

It is the classical pathway C3 convertase that cleaves C3 into C3a and C3b.

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

How does the C3b generated by the classical pathway affect the complement system?

A

C3b can bind Factor B, generating more C3 convertase and amplifying complement activation.

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

What forms the classical pathway C5 convertase?

A

The C4b2a3b complex, which cleaves C5 and initiates the late steps of complement activation.

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

Why is IgM a more efficient complement-fixing antibody than IgG?

A

Because a single IgM molecule can bind multiple C1q molecules, while IgG requires multiple molecules to bind C1q.

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

What triggers the lectin pathway of complement activation?

A

Binding of microbial polysaccharides to circulating lectins such as mannose-binding lectin (MBL) or ficolins.

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

What are the key components of MBL and ficolins involved in the lectin pathway?

A

MBL has a collagen-like domain and a carbohydrate recognition domain; ficolins have a collagen-like domain and a fibrinogen-like domain.

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

Which proteins are associated with MBL in the lectin pathway?

A

MBL-associated serine proteases (MASPs) including MASP1, MASP2, and MASP3.

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

What role do MASPs play in the lectin pathway?

A

MASPs cleave C4 and C2 to activate the complement pathway, similar to the function of C1r and C1s in the classical pathway.

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

MASPs cleave C4 and C2 to activate the complement pathway, similar to the function of C1r and C1s in the classical pathway.

A

Formation of the membrane attack complex (MAC) that can lyse cells.

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

What are the components that form the membrane attack complex (MAC)?

A

C5b, C6, C7, C8, and multiple C9 molecules.

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

What is the function of the C9 protein in the MAC?

A

C9 polymerizes to form pores in plasma membranes, allowing the movement of water and ions, leading to cell lysis.

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

How does C5b-8 contribute to the formation of pores in the membrane?

A

C5b-8 complex inserts into the lipid bilayer and forms unstable pores, which are then stabilized by C9.

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

What structural similarity does C9 have with perforin?

A

C9 is structurally homologous to perforin and forms similar membrane pores.

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

What is the main function of the type 1 complement receptor (CR1, CD35)?

A

Promotes phagocytosis of C3b- and C4b-coated particles and clearance of immune complexes from the circulation.

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

Where is CR1 primarily expressed?

A

On bone marrow-derived cells such as erythrocytes, neutrophils, monocytes, macrophages, eosinophils, T and B lymphocytes, and follicular dendritic cells.

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

What type of complement fragments does CR2 (CD21) bind?

A

C3d, C3dg, and iC3b.

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

What is the role of CR2 on B cells and follicular dendritic cells (FDCs)?

A

On B cells, CR2 enhances activation and response to antigens; on FDCs, it traps antigen-antibody complexes in germinal centers.

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

What is the primary function of the type 3 complement receptor (MAC-1, CR3)?

A

Promotes phagocytosis of microbes opsonized with iC3b and binds to ICAM-1 to aid in leukocyte attachment to endothelial cells.

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

What distinguishes CR4 (p150,95) from MAC-1 (CR3)?

A

CR4 has a different _ chain (CD11c) but the same _ chain (CD18) as MAC-1 and likely has similar functions.

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

Where is the complement receptor CRIg found and what is its role?

A

CRIg is found on macrophages in the liver (Kupffer cells) and is involved in the clearance of opsonized bacteria and blood-borne pathogens.

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

What is the role of C5aR1 and C3aR?

A

Both are G protein-coupled receptors that mediate the proinflammatory effects of C5a and C3a, respectively.

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

What is the function of the C1q receptor?

A

Assists in the clearance of apoptotic bodies and protein fibers such as amyloid fibrils by binding C1q.

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

What is the main role of C1 inhibitor (C1 INH) in complement regulation?

A

Inhibits the proteolytic activity of C1r, C1s, and MASP2 to prevent excessive activation of the classical and lectin pathways.

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

What disease is caused by a deficiency of C1 INH, and what are its symptoms?

A

Hereditary angioedema; symptoms include intermittent edema in the skin and mucosa, abdominal pain, vomiting, diarrhea, and airway obstruction.

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

How do regulatory proteins like MCP, CR1, and DAF inhibit complement activation on normal cells?

A

By binding to C3b and C4b on cell surfaces, preventing the assembly of C3 and C5 convertases.

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

What is the role of Factor H in complement regulation?

A

Inhibits the binding of Bb to C3b, regulating the alternative pathway of complement activation.

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

What disease is characterized by recurrent hemolysis due to unregulated complement activation, and what causes it?

A

Paroxysmal nocturnal hemoglobinuria; caused by a deficiency of GPI-linked proteins such as DAF and CD59 due to an acquired somatic mutation.

201
Q

What is the function of CD59 in complement regulation?

A

Inhibits the formation of the membrane attack complex (MAC) by preventing the addition of C9 molecules to the C5b-8 complex.

202
Q

How do plasma carboxypeptidases regulate inflammation induced by C3a and C5a?

A

By cleaving their C-terminal arginine residues to form C3a des-Arg and C5a des-Arg, which have reduced activity.

203
Q

What is the function of S protein in complement regulation?

A

Binds to soluble C5b-6-7 complexes to prevent their insertion into cell membranes and the formation of MACs.

204
Q

What is the hierarchy of importance for inhibiting complement activation based on the abundance of regulatory proteins on cell surfaces?

A

CD59 > DAF > MCP

205
Q

What is the role of complement fragments C3b and iC3b in phagocytosis?

A

They act as opsonins by binding to specific receptors on macrophages and neutrophils, enhancing the phagocytosis of coated microbes.

206
Q

How does the complement system contribute to the defense against bacteria with polysaccharide-rich capsules?

A

IgM antibodies against these capsules activate the classical pathway, leading to complement-mediated phagocytosis of the bacteria.

207
Q

Which complement fragment is the most potent mediator of mast cell degranulation?

A

C5a.

208
Q

What is the primary function of the membrane attack complex (MAC) in the complement system?

A

To mediate the lysis of foreign organisms by creating pores in their cell membranes.

209
Q

To mediate the lysis of foreign organisms by creating pores in their cell membranes.

A

By binding to antigen-antibody complexes, complement proteins help dissolve these complexes and facilitate their clearance by phagocytes.

210
Q

What is the function of C3d in relation to B cell activation?

A

C3d binds to CR2 on B cells, enhancing B cell activation and the development of humoral immune responses.

211
Q

What role do follicular dendritic cells (FDCs) play in complement-mediated humoral immunity?

A

They trap opsonized antigens in germinal centers, displaying them to B cells and aiding in the selection of high-affinity B cells.

212
Q

How does complement contribute to the clearance of apoptotic cells?

A

Complement proteins and natural antibodies help clear apoptotic cells and their fragments by promoting their recognition and phagocytosis.

213
Q

How can complement activation lead to ischemic tissue injury?

A

Complement activation can result in intravascular thrombosis and damage to endothelial cells, which favors coagulation and can lead to ischemic injury.

214
Q

What are immune complex-mediated diseases, and how are they related to complement activation?

A

These diseases are caused by the deposition of antigen-antibody complexes in blood vessels and kidney glomeruli, leading to complement activation, inflammation, and tissue damage.

215
Q

These diseases are caused by the deposition of antigen-antibody complexes in blood vessels and kidney glomeruli, leading to complement activation, inflammation, and tissue damage.

A

Complement activation by deposited immune complexes leads to inflammation, destruction of vessel walls or glomeruli, thrombosis, and ischemic damage.

216
Q

Complement activation by deposited immune complexes leads to inflammation, destruction of vessel walls or glomeruli, thrombosis, and ischemic damage.

A

Antibodies against C5 are used to treat paroxysmal nocturnal hemoglobinuria, complement-mediated hemolytic uremic syndrome, and neuromyelitis optica. Recombinant human C1 INH is used to treat hereditary angioedema.

217
Q

How do some pathogens use sialic acids to evade complement activation?

A

Pathogens recruit sialic acids to their surfaces, which attract Factor H, displacing C3b and inhibiting the alternative pathway of complement.

218
Q

Which pathogen incorporates host complement regulatory proteins into its envelope to evade complement?

A

Human immunodeficiency virus (HIV) incorporates GPI-anchored complement regulatory proteins DAF and CD59 into its envelope.

219
Q

What protein does Staphylococcus aureus produce to inhibit complement activation?

A

Staphylococcus aureus produces staphylococcal complement inhibitor, which binds to and inhibits both the classical and alternative pathway C3 convertases.

220
Q

How does the complement inhibitory protein of the vaccinia virus affect complement pathways?

A

The vaccinia virus complement inhibitory protein resembles human C4BP, binding to both C4b and C3b, and accelerates the decay of C3 and C5 convertases.

221
Q

What role does the protein chemokine inhibitory protein of staphylococci (CHIPS) play in complement evasion?

A

CHIPS acts as an antagonist of the C5a anaphylatoxin, inhibiting complement-mediated inflammation.

222
Q

Which cancer is curable?

A

Hodgkin lymphoma

223
Q

Which cancer is virtually always fatal?

A

Pancreatic adenocarcinoma

224
Q

What is necessary for controlling cancer?

A

Learning more about its causes and pathogenesis

225
Q

What happened to cancer mortality rates in the United States during the last decades?

A

Declined

226
Q

What helps to distinguish benign and malignant tumors?

A

Morphologic characteristics

227
Q

What provides a measure of the impact of cancer on human populations?

A

Epidemiology

228
Q

What plays a critical role in the development of neoplasia?

A

Genetic alterations

229
Q

What is the focus of new technologies in cancer diagnosis?

A

Targeting particular molecular lesions

230
Q

What does neoplasia mean?

A

New growth

231
Q

What does the term tumor originally describe?

A

Swelling caused by inflammation

232
Q

What is the study of tumors or neoplasms called?

A

Oncology

233
Q

What is a modern definition of neoplasm?

A

A genetic disorder of cell growth

234
Q

What gives neoplastic cells a survival and growth advantage?

A

Mutations affecting genes

235
Q

What are the two components of all tumors?

A

Neoplastic cells and reactive stroma

236
Q

What determines the classification and biologic behavior of tumors?

A

Parenchymal component

237
Q

What term describes tumors with abundant collagenous stroma?

A

Desmoplasia

238
Q

What are stony hard desmoplastic tumors called?

A

Scirrhous

239
Q

Where do benign tumors remain localized?

A

At their site of origin

240
Q

What is the general outcome for patients with benign tumors?

A

Survival

241
Q

What type of benign tumor occurs in the brain and may be fatal?

A

Benign brain tumors

242
Q

What suffix is used to name benign tumors of mesenchymal cells?

A

NAME?

243
Q

What is a benign tumor of fibroblast-like cells called?

A

Fibroma

244
Q

What is a benign cartilaginous tumor called?

A

Chondroma

245
Q

What is a benign epithelial neoplasm derived from glandular tissues called?

A

Adenoma

246
Q

What are benign epithelial neoplasms with fingerlike projections called?

A

Papillomas

247
Q

What is a benign epithelial tumor forming large cystic masses called?

A

Cystadenoma

248
Q

What is a tumor producing papillary projections into cystic spaces called?

A

Papillary cystadenoma

249
Q

What is a neoplasm producing a projection above a mucosal surface called?

A

Polyp

250
Q

What is a polyp with glandular tissue called?

A

Adenomatous polyp

251
Q

What can malignant tumors do that benign tumors cannot?

A

Invade and metastasize

252
Q

What are malignant tumors collectively referred to as?

A

Cancers

253
Q

What is the origin of the word “cancer”?

A

Latin for crab

254
Q

What is the outcome for some cancers discovered at early stages?

A

Surgical excision or cure

255
Q

What is the term for malignant tumors arising from solid mesenchymal tissues?

A

Sarcomas

256
Q

What are malignant tumors of blood-forming cells called?

A

Leukemias

257
Q

What are tumors of lymphocytes or their precursors called?

A

Lymphomas

258
Q

What are malignant neoplasms of epithelial origin called?

A

Carcinomas

259
Q

What type of carcinoma has tumor cells resembling stratified squamous epithelium?

A

Squamous cell carcinoma

260
Q

What is the term for malignant epithelial cells growing in a glandular pattern?

A

Adenocarcinoma

261
Q

What is the term for cancers with cells of unknown origin?

A

Undifferentiated malignant tumors

262
Q

What is an example of a mixed tumor?

A

Salivary gland mixed tumor

263
Q

What does pleomorphic adenoma refer to?

A

A neoplasm with both epithelial and mesenchymal components

264
Q

From how many germ layers are most neoplasms composed?

A

One

265
Q

What is a tumor containing cells from more than one germ layer called?

A

Teratoma

266
Q

Where do teratomas originate from?

A

Totipotential germ cells

267
Q

What type of tumor contains bone

A

epithelium, muscle, fat, and nerve tissue?,Teratoma

268
Q

What is a common pattern seen in ovarian cystic teratoma?

A

Differentiation along ectodermal lines

269
Q

What is an ovarian cystic teratoma also known as?

A

Dermoid cyst

270
Q

What is the key distinction between benign and malignant tumors?

A

Histologic and anatomic features

271
Q

What does differentiation refer to in neoplasms?

A

Extent of resemblance to normal cells

272
Q

What is the term for lack of differentiation in tumors?

A

Anaplasia

273
Q

What type of tumor is usually well differentiated?

A

Benign tumors

274
Q

What type of tumor is highly predictive of malignant behavior?

A

Highly anaplastic tumors

275
Q

What is pleomorphism?

A

Variation in cell size and shape

276
Q

What is the typical nuclear-to-cytoplasm ratio in cancer cells?

A

1:01

277
Q

What do undifferentiated cancers often contain many of?

A

Mitoses

278
Q

What is a key feature of malignant tumors regarding cell polarity?

A

Loss of polarity

279
Q

What causes areas of ischemic necrosis in rapidly growing cancers?

A

Insufficient blood supply

280
Q

What is metaplasia?

A

Replacement of one cell type with another

281
Q

What is dysplasia?

A

Disordered growth of cells

282
Q

What is carcinoma in situ?

A

Severe dysplasia involving the full thickness of epithelium without invasion

283
Q

Where are dysplastic changes often found?

A

Adjacent to foci of invasive carcinoma

284
Q

What frequently antedates the appearance of cancer in the cervix?

A

Severe epithelial dysplasia or carcinoma in situ

285
Q

Can mild dysplasias contain mutations associated with cancer?

A

Yes

286
Q

Does dysplasia always progress to cancer?

A

No

287
Q

What can happen to dysplasias with removal of inciting causes?

A

They may be completely reversible

288
Q

How long can carcinoma in situ persist before becoming invasive?

A

Years

289
Q

What is needed for the evolution of full-blown cancers from in situ lesions?

A

Accumulation of mutations

290
Q

Does dysplasia always occur in metaplastic epithelium?

A

No

291
Q

What is the growth of cancers accompanied by?

A

Progressive invasion and destruction of surrounding tissue

292
Q

What separates benign tumors from surrounding normal tissue?

A

A capsule

293
Q

What are benign neoplasms composed of tangled blood vessels called?

A

Hemangiomas

294
Q

What pattern of growth is typical of malignant tumors?

A

Crablike pattern

295
Q

What is the most reliable discriminator of malignant and benign tumors?

A

Invasiveness

296
Q

What defines metastasis?

A

Spread of a tumor to discontinuous sites

297
Q

What two malignant neoplasms rarely metastasize?

A

Gliomas and basal cell carcinomas

298
Q

What percentage of solid tumors present as metastatic disease?

A

30%

299
Q

What are the three pathways of cancer spread?

A

Direct seeding, lymphatic spread, hematogenous spread

300
Q

What type of cancer often spreads by seeding in the peritoneal cavity?

A

Ovarian carcinomas

301
Q

What is the term for a gelatinous neoplastic mass filling the peritoneal cavity?

A

Pseudomyxoma peritonei

302
Q

What is the most common pathway for carcinoma dissemination?

A

Lymphatic spread

303
Q

What is the first site breast cancer usually spreads to?

A

Axillary lymph nodes

304
Q

What is the term for bypassing local lymph nodes in cancer spread?

A

Skip metastasis

305
Q

What is a sentinel lymph node?

A

The first node to receive lymph flow from a primary tumor

306
Q

How is sentinel node mapping performed?

A

Injection of radiolabeled tracers or colored dyes

307
Q

What can cause enlarged lymph nodes besides metastasis?

A

Immune response or hyperplasia

308
Q

What type of tumors typically spread through hematogenous routes?

A

Sarcomas

309
Q

Which organs are most frequently involved in hematogenous metastasis?

A

Liver and lungs

310
Q

Which cancers often metastasize to the vertebrae via the paravertebral plexus?

A

Thyroid and prostate carcinomas

311
Q

Which cancer can invade the renal vein and grow into the inferior vena cava?

A

Renal cell carcinoma

312
Q

What are the main characteristics that distinguish benign from malignant tumors?

A

Differentiation, invasiveness, and spread

313
Q

How do benign tumors compare to malignant tumors in differentiation?

A

Benign tumors are well differentiated

314
Q

What type of tumor is more likely to retain functions of its cells of origin?

A

Benign tumors

315
Q

How do benign tumors compare to malignant tumors in growth rate?

A

Benign tumors grow slower

316
Q

How are benign tumors physically characterized?

A

Circumscribed with a capsule

317
Q

How do malignant tumors invade surrounding tissues?

A

They are poorly circumscribed and invasive

318
Q

Where do benign tumors remain?

A

Localized at the site of origin

319
Q

Which tumors prefer to spread via lymphatics?

A

Carcinomas

320
Q

Which tumors prefer the hematogenous route for spreading?

A

Sarcomas

321
Q

What has epidemiology linked smoking to?

A

Lung cancer

322
Q

What dietary pattern is linked to colon cancer?

A

High fat and low fiber diet

323
Q

What percentage of global deaths were caused by cancer in 2018?

A

Nearly 1 in 6

324
Q

Which cancers are the most common in men?

A

Prostate, lung, and colon/rectum

325
Q

Which cancers are the most common in women?

A

Breast, lung, and colon/rectum

326
Q

What test led to a decline in cervical cancer deaths?

A

Papanicolaou (Pap) smear

327
Q

What is the main cause of the decline in lung cancer deaths?

A

Decreased tobacco use

328
Q

Which infectious agent is associated with cervical cancer?

A

Human papillomavirus (HPV)

329
Q

Which cancer is linked to alcohol abuse?

A

Hepatocellular carcinoma

330
Q

What percentage of cancers worldwide are caused by infectious agents?

A

About 15%

331
Q

What is a sentinel lymph node?

A

The first node that receives lymph from the primary tumor

332
Q

Which cancers are more common in obese individuals?

A

Breast, colorectal, and endometrial cancers

333
Q

What is a major cancer risk factor in older adults?

A

Accumulation of somatic mutations

334
Q

What is the usual cause of cancer in families with inherited traits?

A

Germline mutations in tumor suppressor genes

335
Q

What percentage of cancers in the U.S. are sporadic?

A

Roughly 95%

336
Q

Can hereditary factors still play a role in sporadic cancers?

A

Yes

337
Q

What is an example of a gene mutation that increases breast cancer risk?

A

BRCA1 or BRCA2

338
Q

What environmental factor has been linked to an increased breast cancer risk in women with BRCA mutations?

A

Changes in reproductive history

339
Q

What genetic system influences the activation of procarcinogens?

A

Cytochrome P-450

340
Q

What gene polymorphism increases susceptibility to smoking-induced lung cancer?

A

P-450

341
Q

What factors influence the incidence of cancer?

A

Geography, age, race, and genetic background

342
Q

What age group is cancer most common in?

A

Adults older than 55

343
Q

What is the primary cause of geographic variation in cancer incidence?

A

Different environmental exposures

344
Q

What are important environmental factors implicated in carcinogenesis?

A

Infectious agents, smoking, alcohol, diet, obesity, reproductive history, and carcinogen exposure

345
Q

What increases cancer risk related to chronic inflammation or injury?

A

Reparative proliferations

346
Q

What other conditions increase the risk of cancer?

A

Certain forms of hyperplasia and immunodeficiency

347
Q

What may be important determinants of cancer risk?

A

Interactions between environmental and genetic factors

348
Q

What lies at the heart of carcinogenesis?

A

Nonlethal genetic damage

349
Q

What forms a tumor?

A

Clonal expansion of a single precursor cell with genetic damage

350
Q

What are the four principal gene targets of cancer-causing mutations?

A

Proto-oncogenes, tumor suppressor genes, apoptosis-regulating genes, DNA repair genes

351
Q

What do proto-oncogene mutations cause?

A

Gain of function

352
Q

What do tumor suppressor gene mutations cause?

A

Loss of function

353
Q

What is a mutator phenotype?

A

A state marked by genomic instability

354
Q

What is the result of carcinogenesis?

A

Accumulation of mutations over time

355
Q

What are driver mutations?

A

Mutations that contribute to cancer hallmarks

356
Q

What is an initiating mutation?

A

The first driver mutation that starts malignancy

357
Q

What increases the frequency of both driver and passenger mutations?

A

Genomic instability

358
Q

What are passenger mutations?

A

Mutations with no phenotypic consequence

359
Q

How do tumors evolve during their growth?

A

Under Darwinian selection (survival of the fittest)

360
Q

What happens to tumor cells during expansion?

A

They acquire additional mutations at random

361
Q

What is the term for tumors becoming more aggressive over time?

A

Tumor progression

362
Q

What is a common feature of tumors by the time they become clinically evident?

A

Genetic heterogeneity

363
Q

What explains changes in tumor behavior following therapy?

A

Selection of subclones resistant to treatment

364
Q

What epigenetic modification tends to silence gene expression?

A

DNA methylation

365
Q

What role do histone modifications play in gene expression?

A

They can enhance or dampen gene expression

366
Q

How are epigenetic changes different from DNA mutations in cancer treatment?

A

Epigenetic changes are potentially reversible by drugs

367
Q

What is responsible for silencing some tumor suppressor genes in cancer cells?

A

Aberrant DNA methylation

368
Q

What enables tumors to proliferate without external stimuli?

A

Oncogene activation

369
Q

Why do tumors not respond to growth-inhibitory signals?

A

Inactivation of tumor suppressor genes

370
Q

What metabolic switch do tumor cells undergo?

A

Aerobic glycolysis (Warburg effect)

371
Q

How do tumors evade programmed cell death?

A

By resisting apoptosis

372
Q

What allows tumors to have limitless replicative potential?

A

Avoidance of cellular senescence and mitotic catastrophe

373
Q

Why do tumors need to induce angiogenesis?

A

To supply nutrients and oxygen for growth

374
Q

What causes the majority of cancer deaths?

A

Tumor metastases

375
Q

How do tumors evade the host immune response?

A

By altering the immune recognition of abnormal antigens

376
Q

What two enabling characteristics accelerate tumor progression?

A

Genomic instability and cancer-promoting inflammation

377
Q

What are oncogenes?

A

Mutated genes that cause excessive cell growth

378
Q

What are normal cellular genes that can become oncogenes called?

A

Proto-oncogenes

379
Q

What do oncoproteins do?

A

Promote uncontrolled cell proliferation

380
Q

What is the first step in normal growth factor signaling?

A

Binding of a growth factor to its specific receptor

381
Q

What happens after growth factor receptor activation?

A

Activation of cytoplasmic signal-transducing proteins

382
Q

What is the result of signal transduction in normal cells?

A

Induction of transcription factors and gene expression

383
Q

What do oncoproteins disrupt in cancer cells?

A

Normal checkpoints that regulate cell proliferation

384
Q

Which signaling pathways are often implicated in cancer?

A

Receptor tyrosine kinase, JAK/STAT, WNT, Notch, Hedgehog, TGF-_/SMAD, and NF-_B pathways

385
Q

What does cell growth require besides DNA replication?

A

Biosynthesis of membrane, proteins, and organelles

386
Q

What is a new target for cancer therapies?

A

Oncogenic pro-growth signaling and altered cellular metabolism

387
Q

What do oncogenes encode?

A

Constitutively active oncoproteins

388
Q

How do cancer cells create an autocrine loop?

A

By synthesizing growth factors to which they are responsive

389
Q

What are receptor tyrosine kinases activated by?

A

Mutations that cause constitutive tyrosine kinase activity

390
Q

What gene is amplified in certain breast carcinomas?

A

ERBB2 (HER2)

391
Q

What fusion gene is found in a subset of lung adenocarcinomas?

A

EML4-ALK

392
Q

What is the most common type of proto-oncogene mutation in human tumors?

A

RAS mutations

393
Q

What causes RAS to remain in an active GTP-bound form?

A

Point mutations that reduce GTPase activity

394
Q

What role does NF1 play in RAS signaling?

A

It acts as a tumor suppressor by negatively regulating RAS

395
Q

What protein kinases are involved in downstream signaling from RAS?

A

BRAF and PI3K

396
Q

What phenomenon occurs when tumors are dependent on a specific oncogene for survival?

A

Oncogene addiction

397
Q

What is the BCR-ABL fusion gene associated with?

A

Chronic myeloid leukemia (CML)

398
Q

What has revolutionized the treatment of CML?

A

BCR-ABL kinase inhibitors

399
Q

What is an example of oncogene addiction in CML?

A

Dependence on BCR-ABL signaling

400
Q

Why is inhibition of BCR-ABL effective in CML treatment?

A

CML tumor cells require BCR-ABL signaling for proliferation and survival

401
Q

Why doesn’t treatment with BCR-ABL inhibitors cure CML?

A

Rare CML stem cells persist without requiring BCR-ABL signals

402
Q

What pathway is JAK2 involved in?

A

JAK/STAT signaling pathway

403
Q

What do activating mutations in JAK2 cause?

A

Independence from hematopoietic growth factors

404
Q

Where do all signal transduction pathways ultimately converge?

A

The nucleus

405
Q

What is the ultimate consequence of deregulated mitogenic signaling pathways?

A

Continuous stimulation of nuclear transcription factors

406
Q

What can cause growth autonomy in cancer cells?

A

Mutations affecting transcription factors that regulate pro-growth genes

407
Q

Which transcription factor is most commonly affected in cancer?

A

MYC

408
Q

What are some other transcription factors involved in cancer?

A

MYB, JUN, FOS, and REL

409
Q

What type of gene is MYC?

A

Proto-oncogene

410
Q

How is MYC normally regulated?

A

Through transcription, translation, and protein stability

411
Q

What is one way MYC contributes to cancer?

A

Activates expression of genes involved in cell growth

412
Q

Which genes involved in cell cycle progression are targets of MYC?

A

D cyclins

413
Q

How does MYC enhance protein synthesis?

A

By upregulating ribosomal RNA (rRNA) genes and processing

414
Q

How does MYC contribute to the Warburg effect?

A

Upregulates genes involved in glycolysis and glutamine metabolism

415
Q

Which type of cancer is associated with MYC translocation?

A

Burkitt lymphoma

416
Q

What role does MYC play in cell immortality?

A

Upregulates telomerase expression

417
Q

In which cancers is MYC often amplified?

A

Breast, colon, lung, neuroblastomas, and small cell lung cancers

418
Q

How do upstream signaling pathways elevate MYC levels?

A

By increasing MYC transcription, mRNA translation, and stabilizing MYC protein

419
Q

What do CDK-cyclin complexes do?

A

Phosphorylate target proteins to drive cell cycle progression

420
Q

What silences CDKs to exert negative control over the cell cycle?

A

CDK inhibitors

421
Q

Where are the two main cell cycle checkpoints located?

A

G1/S transition and G2/M transition

422
Q

What happens if DNA damage sensors are activated?

A

Cell cycle progression is arrested or apoptosis is initiated

423
Q

What type of mutations promote unregulated G1/S progression?

A

Gain-of-function mutations in D cyclin genes and CDK4

424
Q

In which cancers is CDK4 gene amplification commonly found?

A

Melanomas, sarcomas, and glioblastomas

425
Q

What effect do CDK4 inhibitors have in cancer treatment?

A

They are effective in treating advanced breast cancers with excessive CDK4 activity

426
Q

What type of mutations inhibit G1/S progression?

A

Loss-of-function mutations in tumor suppressor genes

427
Q

What gene mutation is common in melanoma-prone families?

A

Germline mutations of p16 (CDKN2A)

428
Q

Which tumor suppressor genes inhibit G1/S progression?

A

RB and TP53

429
Q

What are proto-oncogenes?

A

Normal genes that promote cell proliferation

430
Q

What are oncogenes?

A

Mutated or overexpressed proto-oncogenes that function autonomously

431
Q

What is an oncoprotein?

A

A protein encoded by an oncogene that drives increased cancer cell proliferation

432
Q

What creates an autocrine signaling loop in cancer cells?

A

Constitutive expression of growth factors and receptors

433
Q

Which mutation activates the EGF receptor in lung cancer?

A

Point mutations

434
Q

How is the HER2 receptor activated in breast cancer?

A

Gene amplification

435
Q

Which gene is involved in the BCR-ABL fusion in CML?

A

ABL nonreceptor tyrosine kinase

436
Q

What is the most common mutation in RAS activation?

A

Point mutations

437
Q

Which kinases are activated by mutations in many cancers?

A

PI3K and BRAF serine/threonine kinases

438
Q

How is MYC deregulated in Burkitt lymphoma?

A

Chromosomal translocations

439
Q

What promotes cell cycle progression in cancer cells?

A

Increased activity of CDK4/D cyclin complexes

440
Q

What do tumor suppressor genes do?

A

Apply brakes to cell proliferation

441
Q

What happens when tumor suppressor genes are mutated?

A

Failure of growth inhibition

442
Q

What regulatory network do many tumor suppressors belong to?

A

One that recognizes genotoxic stress and shuts down proliferation

443
Q

What happens when oncogenes are expressed in cells with intact tumor suppressors?

A

Quiescence or permanent cell cycle arrest (oncogene-induced senescence)

444
Q

What is the “two-hit” hypothesis for retinoblastoma?

A

Both RB alleles must be mutated for cancer to occur

445
Q

How is retinoblastoma inherited in familial cases?

A

As an autosomal dominant trait

446
Q

What happens in sporadic cases of retinoblastoma?

A

Both RB alleles undergo somatic mutation

447
Q

How does RB function at the cellular level?

A

As a recessive trait when lost in individual cells

448
Q

What is the role of many tumor suppressors?

A

Inhibiting cell cycle progression and ensuring genomic stability

449
Q

How can tumor suppressors prevent cellular transformation?

A

By altering cell metabolism or ensuring genomic stability

450
Q

What is RB’s role in the cell cycle?

A

Key negative regulator of the G1/S cell cycle transition

451
Q

In what state is RB active?

A

Hypophosphorylated state

452
Q

How can RB be inactivated?

A

Loss-of-function mutations or hyperphosphorylation by CDK/cyclin complexes

453
Q

What drives the expression of genes needed for S phase progression?

A

E2F transcription factors

454
Q

How does growth factor signaling affect RB?

A

Upregulates CDK/cyclin activity, leading to RB hyperphosphorylation

455
Q

Why do patients with RB mutations mainly develop retinoblastoma?

A

The reason is unclear, but other RB-like proteins may compensate in other tissues

456
Q

What mutations mimic RB loss in cancer?

A

Mutations in cyclin D, CDK4, or CDK inhibitors

457
Q

How do DNA viruses inactivate RB?

A

By binding to RB and releasing E2F transcription factors

458
Q

What does RB do when hypophosphorylated?

A

Binds and inhibits E2F transcription factors

459
Q

What leads to RB hyperphosphorylation and inactivation?

A

Normal growth factor signaling

460
Q

What is required for cells to pass the G1/S phase checkpoint?

A

E2F transcription factors

461
Q

How is the antiproliferative effect of RB abrogated in cancer?

A

Through loss-of-function RB mutations, CDK4/cyclin D amplification, or loss of CDK inhibitors

462
Q

Which viral oncoprotein inhibits RB?

A

E7 protein of HPV

463
Q

What does the TP53 gene encode?

A

The protein p53

464
Q

What is the role of p53?

A

Regulates cell cycle, DNA repair, senescence, and apoptosis

465
Q

What percentage of human cancers have TP53 mutations?

A

More than 50%

466
Q

What syndrome is associated with inherited TP53 mutations?

A

Li-Fraumeni syndrome

467
Q

How is p53 kept in check in normal cells?

A

By association with MDM2, which leads to p53 degradation

468
Q

What activates p53 in stressed cells?

A

DNA damage, hypoxia, or oncogenic stress

469
Q

What are the three possible outcomes of p53 activation?

A

Transient cell cycle arrest, senescence, or apoptosis

470
Q

What gene does p53 activate for cell cycle arrest?

A

CDKN1A, which encodes the CDK inhibitor p21

471
Q

How does p53 promote apoptosis?

A

By activating pro-apoptotic genes like BAX and PUMA

472
Q

What happens when p53 function is lost?

A

DNA damage goes unrepaired, leading to malignant transformation

473
Q

How does p53 status affect cancer treatment response?

A

Tumors with wild-type TP53 are more responsive to chemotherapy and radiation

474
Q

What does p53 monitor in the cell?

A

Stress from anoxia, mutated oncoproteins, or DNA damage

475
Q

What proteins does p53 control?

A

Proteins involved in cell cycle arrest, DNA repair, senescence, and apoptosis

476
Q

Which kinases phosphorylate p53 in response to DNA damage?

A

ATM/ATR family kinases

477
Q

What effect does p53 activation have on the cell cycle?

A

It upregulates p21, causing cell-cycle arrest at the G1/S checkpoint

478
Q

What happens if DNA damage cannot be repaired?

A

p53 induces senescence or apoptosis

479
Q

What mutation is found in the majority of human cancers?

A

Biallelic loss-of-function mutations in TP53

480
Q

What syndrome is associated with inherited TP53 mutations?

A

Li-Fraumeni syndrome

481
Q

How is p53 inactivated by viruses?

A

Viral oncoproteins like HPV E6 protein

482
Q

How are tumor suppressor genes often disabled?

A

Through recurrent chromosomal deletions

483
Q

What role does the APC gene play in colonic neoplasia?

A

It downregulates growth-promoting signaling pathways

484
Q

What disorder is associated with germline APC mutations?

A

Familial adenomatous polyposis

485
Q

What percentage of nonfamilial colorectal carcinomas show APC defects?

A

70% to 80%

486
Q

What pathway does APC regulate?

A

The WNT signaling pathway

487
Q

What does APC normally do to _-catenin?

A

It helps form a destruction complex that degrades _-catenin

488
Q

What happens when APC is lost in cells?

A

Beta-catenin is stabilized, leading to continuous growth signaling

489
Q

What genes are upregulated by _-catenin?

A

MYC, cyclin D1, and other growth-promoting genes

490
Q

What percentage of hepatocellular carcinomas have _-catenin gain-of-function mutations?

A

About 20%

491
Q

What is the role of E-cadherin?

A

Maintains intercellular adhesiveness and regulates _-catenin

492
Q

What happens when E-cadherin is lost?

A

Cells disaggregate, promoting invasion and metastasis

493
Q

Which cancers show reduced E-cadherin expression?

A

Esophagus, colon, breast, ovary, and prostate

494
Q

What syndrome is associated with germline E-cadherin mutations?

A

Familial gastric carcinoma

495
Q

What proteins are encoded by the CDKN2A gene?

A

p16/INK4a and p14/ARF

496
Q

What is the role of p16/INK4a?

A

Blocks CDK4/cyclin D-mediated phosphorylation of RB

497
Q

What cancers are associated with CDKN2A mutations?

A

Melanoma, bladder, head and neck, and cholangiocarcinoma

498
Q
A