Block 1 Exam 4 Part 1 Flashcards

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

Which types of inheritance, males and females, are equally likely to transmit the trait to their offspring, and it shows a vertical transmission?

A

Autosomal Dominant

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

Which of the factors that affect expression of disease-causing genes refers to genes that exert affects on multiple aspects of physiology or anatomy?

A

Pleiotropy

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

The study of chromosomes and their abnormalities is called ___.

A

cytogenetics

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

What techniques are used to arrest dividing somatic cells in metaphase, when chromosomes are easiest to see?

A

Spindle poisons like colchicine and colcemid.

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

What solution causes swelling of cells and better separation of individual chromosomes?

A

Hypotonic (low-salt) solution.

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

What process makes chromosomes easier to identify by producing light and dark bands?

A

Staining materials that are absorbed differently by different parts of chromosomes.

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

In which phase are chromosomes maximally condensed and easiest to observe?

A

Metaphase

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

How long are peripheral lymphocytes usually cultured before chromosome analysis?

A

48 to 72 hours.

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

What substance is added to produce metaphase arrest during chromosome analysis?

A

Colcemid

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

What is used to rupture the cell nucleus during chromosome analysis?

A

Hypotonic saline solution.

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

What is the process called where chromosomes are photographed and arranged according to length?

A

Karyogram (or karyotype).

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

What does the term “karyotype” refer to?

A

The number and type of chromosomes present in an individual.

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

What technology is currently used to display chromosomes?

A

Computerized image analyzers.

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

What term describes a chromosome with its centromere near the middle?

A

Metacentric

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

What is a chromosome called when its centromere is near the tip?

A

Acrocentric

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

What term is used for chromosomes with centromeres located between the middle and the tip?

A

Submetacentric

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

What is the name of the tip of each chromosome?

A

Telomere

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

What label is given to the short arm of a chromosome?

A

p

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

What label is given to the long arm of a chromosome?

A

q

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

In metacentric chromosomes, how are the p and q arms designated?

A

By convention, where the arms are of roughly equal length.

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

How is a normal female karyotype designated?

A

46,XX

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

How is a normal male karyotype designated?

A

46,XY

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

What development in the 1970s improved the detection of chromosomal abnormalities?

A

Staining techniques that produce chromosome bands.

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

How does chromosome banding help in karyotyping?

A

It helps detect deletions, duplications, and other structural abnormalities, and aids in identifying individual chromosomes.

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

How are the major bands on each chromosome designated?

A

They are systematically numbered.

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

What does “14q32” refer to?

A

The second band in the third region of the long arm of chromosome 14.

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

How are subbands designated in chromosome banding?

A

By decimal points following the band number (e.g., 14q32.3).

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

What was the first staining method used to produce specific banding patterns?

A

Quinacrine banding (Q-banding).

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

Which staining method has largely replaced Q-banding?

A

Giemsa banding (G-banding).

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

What is required to produce G-bands?

A

Giemsa stain after partial digestion of chromosomal proteins by trypsin.

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

What does reverse banding (R-banding) do?

A

It reverses the usual white and black pattern seen in G-bands and Q-bands.

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

Which banding technique is helpful for staining the distal ends of chromosomes?

A

Reverse banding (R-banding).

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

What does C-banding specifically stain?

A

Constitutive heterochromatin near the centromere.

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

What areas do nucleolar organizing region stains (NOR stains) highlight?

A

Satellites and stalks of acrocentric chromosomes.

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

Why are C-banding and NOR staining techniques useful?

A

They are useful in identifying unknown chromosomal material.

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

What stages of cell division are chromosomes stained during high-resolution banding?

A

Prophase or early metaphase (prometaphase).

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

How does staining chromosomes during prophase or prometaphase affect the number of observable bands?

A

The number of observable bands increases from about 300 to 450 to as many as 800.

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

What advantage does high-resolution banding provide compared to conventional banding?

A

It allows detection of less obvious abnormalities usually not seen with conventional banding.

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

What does the fluorescence in situ hybridization (FISH) technique use to detect specific DNA sequences?

A

A labeled single-stranded DNA segment (probe).

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

What types of chromosomes can be used in FISH?

A

Denatured metaphase, prophase, or interphase chromosomes.

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

How is the location of a probe visualized in FISH?

A

Under a fluorescence microscope.

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

What does it indicate if a FISH probe hybridizes to only one chromosome in a patient?

A

The patient likely has a deletion on the chromosome that failed to hybridize.

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

How does FISH resolution compare to high-resolution banding approaches?

A

FISH provides considerably better resolution and can detect deletions as small as 1 million base pairs (1 Mb).

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

What are two common deletion syndromes detected by FISH?

A

Prader-Willi syndrome (microdeletion of 15q11.2) and Williams syndrome (microdeletion of 7q11.2).

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

How are extra copies of a chromosome region detected using FISH?

A

The probe hybridizes in three or more places instead of two.

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

What can combinations of FISH probes detect?

A

Chromosome rearrangements such as translocations.

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

What is an advantage of using FISH with interphase chromosomes compared to traditional methods?

A

It allows for faster analyses and diagnoses because it is not necessary to stimulate cells to divide to obtain metaphase chromosomes.

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

What are common applications of FISH analysis of interphase chromosomes?

A

Prenatal detection of fetal chromosome abnormalities and analysis of chromosome rearrangements in tumor cells.

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

How has the FISH technique been extended to detect multiple numerical abnormalities?

A

By using multiple probes, each labeled with a different color, to test several common numerical abnormalities simultaneously.

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

What does spectral karyotyping involve?

A

Using varying combinations of five different fluorescent probes and special cameras to uniquely color each chromosome for identification.

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

What is the advantage of spectral karyotyping?

A

It is useful for identifying small chromosome rearrangements.

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

What technique is used to detect losses or duplications of whole chromosomes or specific chromosome regions?

A

Comparative genomic hybridization (CGH).

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

How is DNA labeled for CGH analysis?

A

DNA from the test source is labeled with one color (e.g., red) and DNA from normal control cells is labeled with a second color (e.g., green).

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

In CGH, what does a red signal indicate on a chromosome?

A

The region is duplicated in the test DNA.

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

In CGH, what does a green signal indicate on a chromosome?

A

The region is deleted in the test DNA.

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

What type of cells is CGH especially useful for analyzing?

A

Cancer cells

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

What is a major limitation of CGH when used with metaphase chromosomes?

A

It cannot detect deletions or duplications smaller than 5 to 10 Mb microscopically.

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

What resolution can array CGH (aCGH) provide?

A

Resolution to 50 to 100 kb or even less.

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

What does cytogenomic microarray (CMA) utilize to detect smaller genomic variations?

A

Microarrays with single nucleotide polymorphisms (SNPs) arranged very closely.

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

What is the resolution capability of cytogenomic microarrays (CMA)?

A

Detection of deletions and duplications of less than 20 kb.

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

What additional features does cytogenomic microarray (CMA) offer compared to array CGH?

A

Detection of loss of heterozygosity and uniparental disomy.

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

Do array CGH (aCGH) and cytogenomic microarrays (CMA) require dividing cells for analysis?

A

No, they do not require dividing cells.

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

How much DNA is needed for analysis using array CGH (aCGH) and cytogenomic microarrays (CMA)?

A

Less than a microgram of DNA is sufficient for analysis of the entire genome.

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

What is a primary disadvantage of array CGH (aCGH) and cytogenomic microarrays (CMA)?

A

They cannot detect balanced rearrangements of chromosomes, such as reciprocal translocations or inversions.

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

What is the term for a cell that contains a complete set of extra chromosomes?

A

Polyploidy

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

What is the karyotype designation for triploidy?

A

69,XXX (or other combinations of sex chromosomes).

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

What is the karyotype designation for tetraploidy?

A

92,XXXX (or other combinations of sex chromosomes).

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

What is the most common cause of triploidy?

A

Fertilization of an egg by two sperm cells (dispermy).

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

What are some effects of having extra chromosomes due to polyploidy?

A

Multiple anomalies such as defects of the heart and central nervous system.

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

Why do most triploid conceptions result in spontaneous abortion?

A

Because of the large amount of surplus gene product leading to severe developmental issues.

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

What is a rare cause of tetraploidy?

A

Mitotic failure in the early embryo or fusion of two diploid zygotes.

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

How long do infants with tetraploidy typically survive?

A

They usually survive for only a short period.

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

What term describes cells with a multiple of 23 chromosomes?

A

Euploid

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

What is the chromosome count for triploidy?

A

69 chromosomes

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

What is the chromosome count for tetraploidy?

A

92 chromosomes

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

Are polyploid conditions compatible with long-term survival?

A

No, all polyploid conditions are incompatible with long-term survival.

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

What typically happens to most polyploid conceptions?

A

They are spontaneously aborted.

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

What term describes cells with missing or additional individual chromosomes?

A

Aneuploid

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

What are the two main types of autosomal aneuploidy?

A

Monosomy (one copy of a chromosome) and trisomy (three copies of a chromosome).

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

Are autosomal monosomies typically compatible with survival to term?

A

No, autosomal monosomies are nearly always incompatible with survival to term.

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

Why do some trisomies produce less-severe consequences than monosomies?

A

The body can tolerate excess genetic material more readily than it can tolerate a deficit of genetic material.

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

What is the most common cause of aneuploidy?

A

Nondisjunction during meiosis.

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

What can result from nondisjunction during meiosis?

A

A gamete that either lacks a chromosome or has two copies of it, leading to a monosomic or trisomic zygote, respectively.

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

In which stages of meiosis can nondisjunction occur?

A

Meiosis I or Meiosis II.

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

What is the karyotype for Trisomy 21?

A

47,XY,+21 or 47,XX,+21.

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

What is the most common autosomal aneuploid condition compatible with survival to term?

A

Trisomy 21 (Down syndrome).

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

What is a highly consistent feature of Down syndrome that aids in diagnosis?

A

Decreased muscle tone (hypotonia).

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

Are males with Down syndrome typically fertile?

A

No, males with Down syndrome are nearly always sterile, with very few reported cases of reproduction.

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

What percentage of females with Down syndrome fail to ovulate?

A

Approximately 40%.

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

What is the risk for a female with Down syndrome to produce a gamete with two copies of chromosome 21?

A

50%

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

Why is the risk of producing affected live-born offspring lower than 50% in women with Down syndrome?

A

Because about 75% of trisomy 21 conceptions are spontaneously aborted.

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

What can be said about the majority of cases of trisomy 21 in relation to new mutations?

A

Nearly all cases of trisomy 21 can be regarded as new mutations.

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

What is tissue-specific mosaicism?

A

Mosaicism confined to certain tissues, which can complicate diagnosis due to cytogenetic analysis being based on a single tissue type.

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

How can mosaicism affecting the germline of a parent influence the recurrence of Down syndrome?

A

It can lead to multiple recurrences of Down syndrome in the offspring.

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

What is the recurrence risk for Down syndrome among mothers younger than 30 years?

A

About 1%, which is approximately 10 times higher than the population risk for this age group.

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

What is the karyotype for Trisomy 18?

A

47,XY,+18.

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

What is another name for Trisomy 18?

A

Edwards syndrome

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

What is the most common congenital heart defect seen in Trisomy 18?

A

Ventricular septal defects (VSDs) with dysplasia of multiple heart valves.

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

What is the karyotype for Trisomy 13?

A

47,XY,+13

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

What is another name for Trisomy 13?

A

Patau syndrome.

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

What percentage of individuals with Trisomy 13 have heart defects?

A

80%

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

What is cutis aplasia, and how can it be useful in diagnosing Trisomy 13?

A

Cutis aplasia is a defect of the skin on the scalp, particularly on the posterior occiput, and can be a diagnostic clue for Trisomy 13.

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

What is the karyotype for Turner Syndrome?

A

45,X

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

What type of kidney defects are seen in Turner Syndrome, and how do they impact health?

A

Structural kidney defects are present in about 50% of individuals, but they usually do not cause medical problems.

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

How does Turner Syndrome affect spatial perceptual ability and intelligence?

A

There is typically some diminution in spatial perceptual ability, but intelligence is usually normal.

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

What is the karyotype for Klinefelter Syndrome?

A

47,XXY.

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

What reproductive issues are associated with Klinefelter Syndrome?

A

Most males with Klinefelter Syndrome are sterile due to atrophy of the seminiferous tubules.

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

What is a common secondary sexual characteristic observed in Klinefelter Syndrome?

A

Gynecomastia (breast development).

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

How does Klinefelter Syndrome affect testosterone levels and muscle mass?

A

Testosterone levels are low, and muscle mass tends to be reduced.

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

What cognitive effects are associated with Klinefelter Syndrome?

A

There is a predisposition for learning disabilities and a reduction in verbal IQ, typically 10 to 15 points lower than that of the affected person’s siblings.

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

Why is Klinefelter Syndrome often diagnosed later in life?

A

Due to its subtlety, it is often not diagnosed until after puberty and is sometimes first discovered in fertility clinics.

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

What is the most common known cause of pregnancy loss?

A

Chromosome abnormalities

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

What percentage of conceptions with chromosome abnormalities are lost before term?

A

At least 95%

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

What chromosome abnormality is thought to be the most common at conception but is rarely seen in live births?

A

Trisomy 16

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

What structural abnormalities are seen in approximately 1% of oocytes and 5% of sperm cells?

A

Structural chromosome abnormalities

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

What are structural chromosome abnormalities?

A

Alterations in chromosome structure that can involve the loss or gain of chromosome parts or changes in chromosome arrangement.

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

What can cause structural chromosome abnormalities?

A

Structural abnormalities can occur due to improper alignment of homologous chromosomes during meiosis, unequal crossover, or chromosome breakage during meiosis or mitosis.

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

What are clastogens?

A

Harmful agents that can increase the likelihood of chromosome breakage.

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

What is the term for the interchange of genetic material between nonhomologous chromosomes?

A

Translocation

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

What type of translocation involves a reciprocal exchange of segments between two nonhomologous chromosomes?

A

Reciprocal translocation

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

What type of translocation involves the fusion of two acrocentric chromosomes at their centromeres, resulting in a single chromosome?

A

Robertsonian translocation

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

Which type of translocation is most commonly associated with an increased risk of chromosomal abnormalities in offspring?

A

Robertsonian translocation

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

What happens during a reciprocal translocation?

A

Breaks occur in two different chromosomes, and the material is mutually exchanged.

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

What are the chromosomes called that result from a reciprocal translocation?

A

Derivative chromosomes

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

Why are carriers of reciprocal translocations usually unaffected?

A

They have a normal complement of genetic material.

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

What types of genetic material can the offspring of a carrier of a reciprocal translocation inherit?

A

The offspring can inherit normal genetic material, carry the translocation, or have duplications and deletions of genetic material.

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

What can result in the offspring of a person with a reciprocal translocation?

A

The offspring may have partial trisomy or partial monosomy and an abnormal phenotype.

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

What happens to the short arms of chromosomes during a Robertsonian translocation?

A

The short arms of two nonhomologous chromosomes are lost.

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

What occurs to the long arms of chromosomes in Robertsonian translocations?

A

The long arms fuse at the centromere to form a single chromosome.

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

Which chromosomes are involved in Robertsonian translocations?

A

Acrocentric chromosomes (13, 14, 15, 21, and 22).

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

Why are carriers of Robertsonian translocations phenotypically unaffected?

A

The short arms of these chromosomes are very small and contain no essential genetic material.

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

What is the karyotype designation for a male carrier of a Robertsonian translocation involving chromosomes 14 and 21?

A

45,XY,der(14;21)(q10;q10).

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

What can result from adjacent segregation during meiosis in a carrier of a Robertsonian translocation?

A

Gametes may be unbalanced, leading to trisomy or monosomy of the long arms of the involved chromosomes.

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

What is the expected percentage of live-born offspring with Down syndrome from a carrier of a Robertsonian translocation? In mothers? In fathers?

A

About 10%-15% for mothers and 1%-2% for fathers.

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

What is a terminal deletion?

A

A loss of genetic material including the chromosome’s tip after a single break.

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

What is an interstitial deletion?

A

A loss of genetic material between two breaks in a chromosome.

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

What happens when a gamete with a deletion unites with a normal gamete?

A

The zygote has one normal chromosome and one chromosome with the deletion.

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

What is cri-du-chat syndrome caused by?

A

A deletion of the distal short arm of chromosome 5.

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

What is the characteristic cry associated with cri-du-chat syndrome?

A

A distinctive cry that resembles a cat’s cry.

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

What chromosome deletion causes Wolf-Hirschhorn syndrome?

A

chromosome 4.

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

What is the role of high-resolution banding in detecting microdeletions?

A

It allows for the identification of smaller deletions that were previously undetectable.

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

What microdeletion of a chromosome is Prader-Willi syndrome associated with?

A

Chromosome 15q11-q13

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

Which chromosome microdeletion is associated with Angelman syndrome?

A

chromosome 15

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

What chromosome deletion is associated with Williams syndrome?

A

chromosome 7

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

What gene is crucial in the Williams syndrome critical region and what is its role?

A

The ELN gene, which encodes elastin and is important for the aortic wall.

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

How does the deletion size in Williams syndrome affect the phenotype?

A

Larger deletions encompass more genes and produce the full Williams syndrome phenotype, while smaller deletions may result in isolated features like SVAS.

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

What is a contiguous gene syndrome?

A

A condition caused by the deletion or duplication of a series of adjacent genes on a chromosome, such as in WAGR syndrome.

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

What chromosome deletion is WAGR syndrome associated with?

A

chromosome 11p

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

What causes the consistent size of deletions in microdeletion syndromes like Prader-Willi syndrome?

A

The presence of multiple low-copy repeats at the deletion boundaries promotes unequal crossing over.

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

What role do low-copy repeats play in microdeletion syndromes?

A

They promote unequal crossing over, leading to duplications and deletions of the chromosomal region bounded by these repeats.

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

What is the size of the deletion critical region in Prader-Willi syndrome?

A

Approximately 4 Mb.

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

What is a common consequence of rearrangements near chromosome telomeres?

A

They often result in genetic disease due to the high density of genes in these regions.

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

What is the most common subtelomeric rearrangement?

A

chromosome 1p36.

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

What is uniparental disomy?

A

A condition where one parent contributes two copies of a chromosome and the other parent contributes no copies.

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

What is the term for uniparental disomy when one parent contributes two copies of one homolog?

A

Isodisomy

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

What is the term for uniparental disomy when one parent contributes one copy of each homolog?

A

Heterodisomy

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

How can uniparental disomy of an imprinted chromosome cause diseases such as Prader-Willi syndrome?

A

It can cause the absence of active paternal genes in the imprinted region if two copies are inherited from the mother and none from the father.

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

What is a potential result of isodisomy in the offspring of a heterozygous parent?

A

It can result in autosomal recessive disease if the parent contributes two copies of the chromosome with a disease-causing mutation.

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

What is an example of a disease caused by uniparental disomy where the parent transmits two copies of the chromosome containing a mutation?

A

Cystic fibrosis

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

How can uniparental disomy arise in a trisomic conception?

A

It can arise if the embryo loses one of the extra chromosomes, resulting in two copies of the chromosome from one parent.

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

What is a mechanism by which uniparental disomy can occur in the early embryo?

A

It can occur due to mitotic errors, such as chromosome loss followed by duplication of the homologous chromosome.

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

What is a partial trisomy or duplication of genetic material?

A

It is a condition where extra genetic material is present, often seen in offspring of individuals with a reciprocal translocation or caused by unequal crossover during meiosis.

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

Why are duplications generally less severe than deletions?

A

Because a loss of genetic material usually has more serious consequences than an excess of genetic material.

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

What forms when deletions occur at both tips of a chromosome and the remaining ends fuse?

A

A ring chromosome.

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

What is the karyotype notation for a female with a ring X chromosome?

A

46,X,r(X).

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

What can happen to ring chromosomes during cell division?

A

They can often be lost, resulting in monosomy for the chromosome in some cells, leading to mosaicism.

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

What are two of the most common ring chromosomes described in humans?

A

Rings of chromosomes 14 and 22.

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

What happens during an inversion on a chromosome?

A

Two breaks occur on the chromosome, and the intervening fragment is reinserted at the original site but in inverted order.

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

What is the difference between a pericentric and a paracentric inversion?

A

A pericentric inversion includes the centromere, while a paracentric inversion does not involve the centromere.

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

Why do inversions rarely produce disease in the inversion carrier?

A

Because inversions are typically balanced structural rearrangements and do not usually result in a loss or gain of genetic material.

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

How can inversions affect meiosis and offspring?

A

Inversions can interfere with meiosis, leading to the formation of a loop during prophase I. Crossing over within this loop can result in duplications or deletions in the chromosomes of daughter cells.

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

What is the estimated frequency of people carrying an inversion?

A

About 1 in 1000 people.

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

What is an isochromosome?

A

A chromosome with two copies of one arm and no copies of the other.

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

What typically happens to isochromosomes of most autosomes?

A

They are usually lethal due to substantial alteration of genetic material.

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

Which isochromosomes are most commonly observed in live births?

A

Isochromosomes involving the X chromosome.

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

What condition is often associated with isochromosome Xq?

A

Turner syndrome

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

What phenotype can be observed with isochromosome 18q?

A

Edwards syndrome phenotype.

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

How can isochromosomes also be created aside from faulty division?

A

By Robertsonian translocations of homologous acrocentric chromosomes.

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

What chromosome alteration is consistently seen in patients with chronic myelogenous leukemia (CML)?

A

A reciprocal translocation between chromosomes 9 and 22, known as the Philadelphia chromosome.

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

What is the Philadelphia chromosome characterized by?

A

A translocation of most of chromosome 22 onto the long arm of chromosome 9 and a small distal portion of 9q translocated to chromosome 22.

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

What gene is altered in CML due to the Philadelphia chromosome?

A

ABL proto-oncogene

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

What type of cancer is associated with a reciprocal translocation between chromosomes 8 and 14?

A

Burkitt lymphoma.

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

How many different chromosome rearrangements have been observed in cancer, and how does this help in treatment?

A

More than 100 different rearrangements in over 40 types of cancer. Identifying these rearrangements leads to more accurate prognosis and better therapy.

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

What additional factor can increase chromosome breakage in patients with Fanconi anemia?

A

Exposure to certain alkylating agents.

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

What is a notable cytogenetic feature of Bloom syndrome?

A

A high incidence of somatic cell sister chromatid exchange.

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

What common underlying issue is thought to contribute to chromosome instability syndromes?

A

Faulty DNA replication or repair.

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

What is the relationship between chromosome instability syndromes and cancer risk?

A

All chromosome instability syndromes are associated with a significant increase in cancer risk.

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

Which type of inheritance, males and females, are equally likely to transmit the trait to their offspring, and it shows a vertical transmission?

A

Autosomal Dominant

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

Which factors that affects expression of disease-causing genes refers to genes that exert affects on multiple aspects of physiology or anatomy?

A

Pleiotropy

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

In autosomal dominant diseases frequently affected offspring are produced by the union of an unaffected parent with an affected heterozygote. In this scenario, what will be the probability for children to be heterozygote?

A

0.5

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

In which type of inheritance do affected fathers cannot transmit the trait to their sons but can transmit the trait to all their daughters and it shows a horizontal transmission?

A

X linked recessive

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

Asthma is a multifactorial and polygenic disease characterized by variable and recurring symptoms, reversible airflow obstruction, and easily triggered bronchospasms. Studies of twins and of families of asthmatic individuals demonstrate a range of heritability of asthma from 25 to 80 percent. How will be the recurrence risk of asthma?

A

Affected by the severity of asthma in the family members

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

What is inflammation?

A

A protective response that delivers leukocytes and molecules of host defense to sites of infection and cell damage.

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

Why is inflammation important?

A

It helps rid the body of the cause of cell injury and the consequences of injury, such as necrotic cells and tissues.

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

What are the key components involved in inflammation?

A

Phagocytic leukocytes, antibodies, and complement proteins.

196
Q

Where do the mediators of inflammation usually reside?

A

In the blood, in a resting state, or in tissues as sentinels.

197
Q

What does the suffix “-itis” indicate?

A

Inflammation in a specific part of the body (e.g., appendicitis, conjunctivitis).

198
Q

What is the first step in a typical inflammatory reaction?

A

Recognition of the noxious agent by cells equipped with receptors.

199
Q

What happens after the noxious agent is recognized in inflammation?

A

Recruitment of leukocytes and plasma proteins into the tissues.

200
Q

Which leukocytes are first to be recruited to the site of inflammation?

A

Mainly neutrophils, followed by monocytes and lymphocytes.

201
Q

What is the main function of phagocytic cells during inflammation?

A

To ingest and destroy microbes and dead cells, removing the stimulus for inflammation.

202
Q

Why is regulation of the inflammatory response important?

A

To terminate the reaction once it has accomplished its purpose.

203
Q

What occurs during the repair phase of inflammation?

A

Damaged tissue is healed through regeneration of surviving cells and filling of defects with connective tissue (scarring).

204
Q

What are the major participants in the inflammatory reaction in tissues?

A

Blood vessels and leukocytes.

205
Q

How do blood vessels respond to inflammatory stimuli?

A

By dilating and increasing their permeability, allowing proteins to enter the site of infection or tissue damage.

206
Q

What changes occur in the endothelium of blood vessels during inflammation?

A

It changes so that circulating leukocytes can adhere and migrate into the tissues.

207
Q

What is the role of leukocytes once they are recruited to the site of inflammation?

A

To ingest and destroy microbes, dead cells, foreign bodies, and other unwanted materials.

208
Q

What are the potential harmful consequences of inflammation?

A

Local tissue damage, pain, and functional impairment, which may be self-limited or may lead to chronic diseases if misdirected or uncontrolled.

209
Q

How can inflammation contribute to diseases like rheumatoid arthritis and atherosclerosis?

A

By being misdirected or inadequately controlled, leading to tissue damage as a dominant feature of the disease.

210
Q

Why are anti-inflammatory drugs important in clinical medicine?

A

They help control the harmful effects of inflammation without interfering with its beneficial aspects.

211
Q

What is the difference between local and systemic inflammation?

A

Local inflammation is confined to the site of infection or damage, while systemic inflammation, such as sepsis, affects the entire body.

212
Q

What is sepsis?

A

A serious form of systemic inflammation caused by a widespread bacterial infection, leading to pathologic abnormalities.

213
Q

What triggers the vascular and cellular reactions in inflammation?

A

Soluble factors called mediators of inflammation, produced by various cells or derived from plasma proteins.

214
Q

What can trigger the elaboration of inflammatory mediators?

A

crobes, necrotic cells, and hypoxia.

215
Q

What is acute inflammation?

A

A rapid, short-term response to easily eliminated offending agents, characterized by edema and the emigration of neutrophils.

216
Q

How does chronic inflammation differ from acute inflammation?

A

Chronic inflammation lasts longer and may involve different cells and mediators, often due to persistent offending agents or repeated exposure.

217
Q

What characterizes the cellular response in acute inflammation?

A

The emigration of leukocytes, primarily neutrophils, to the site of inflammation.

218
Q

What happens if the offending stimulus in acute inflammation is eliminated?

A

The inflammatory reaction subsides, and any residual injury is repaired.

219
Q

What is chronic inflammation?

A

A longer-lasting inflammatory response to agents that are difficult to eradicate

220
Q

What are the potential consequences of chronic inflammation?

A

Increased tissue destruction and scarring (fibrosis).

221
Q

Can chronic inflammation coexist with acute inflammation?

A

Yes, chronic inflammation may coexist with unresolved acute inflammation, such as in peptic ulcers.

222
Q

How does tissue necrosis trigger inflammation?

A

Inflammation is triggered by molecules released from necrotic cells, regardless of how the cells died (e.g., ischemia, trauma, chemical injury).

223
Q

How can foreign bodies cause inflammation?

A

By causing tissue injury or carrying microbes.

224
Q

Can endogenous substances trigger inflammation?

A

Yes, substances like urate crystals (in gout), cholesterol crystals (in atherosclerosis), and lipids (in metabolic syndrome) can cause inflammation when they deposit in tissues.

225
Q

What role do cytokines play in inflammation caused by immune reactions?

A

They induce and sustain inflammation in autoimmune and allergic reactions.

226
Q

What is the initiating step in inflammatory reactions?

A

Recognition of microbial components or substances released from damaged cells.

227
Q

Which family of receptors is best defined for recognizing microbes?

A

Toll-like receptors (TLRs).

228
Q

What do cytosolic receptors like NOD-like receptors (NLRs) recognize?

A

Molecules released or altered due to cell damage

229
Q

What is the inflammasome and its role in inflammation?

A

A multiprotein cytosolic complex that induces the production of interleukin-1 (IL-1), which recruits leukocytes and triggers inflammation.

230
Q

What is the function of receptors for the Fc tails of antibodies and complement proteins on leukocytes?

A

They recognize microbes coated with antibodies and complement (opsonization), promoting their ingestion, destruction, and inflammation.

231
Q

What is the role of the complement system in inflammation?

A

It reacts against microbes and produces mediators of inflammation.

232
Q

What does mannose-binding lectin do in the immune response?

A

It recognizes microbial sugars, promotes ingestion of microbes, and activates the complement system.

233
Q

What are collectins and their function?

A

Proteins that bind to and combat microbes, aiding in the immune response.

234
Q

What triggers phagocytes and other sentinel cells to release mediators of inflammation?

A

The recognition of foreign or abnormal substances, such as microbes or dead cells.

235
Q

What is exudation in the context of acute inflammation?

A

The escape of fluid, proteins, and blood cells from the vascular system into the interstitial tissue or body cavities.

236
Q

What is an exudate?

A

Extravascular fluid with a high protein concentration and cellular debris, indicating increased permeability of small blood vessels due to inflammation.

237
Q

How does a transudate differ from an exudate?

A

A transudate is a fluid with low protein content, little or no cellular material, and low specific gravity, produced due to osmotic or hydrostatic imbalance without increased vascular permeability.

238
Q

What is edema?

A

An excess of fluid in the interstitial tissue or serous cavities, which can be either an exudate or a transudate.

239
Q

What is pus (purulent exudate)?

A

An inflammatory exudate rich in leukocytes (mostly neutrophils), dead cell debris, and often microbes.

240
Q

What is the first vascular change that occurs during acute inflammation?

A

Vasodilation, induced by mediators like histamine on vascular smooth muscle.

241
Q

What effect does vasodilation have on blood flow at the site of inflammation?

A

It increases blood flow, leading to heat and redness (erythema) at the site of inflammation.

242
Q

What follows vasodilation in the acute inflammatory response?

A

Increased permeability of the microvasculature, allowing protein-rich fluid to enter the extravascular tissues.

243
Q

What is the most common mechanism of vascular leakage in acute inflammation?

A

Contraction of endothelial cells, leading to the opening of interendothelial gaps.

244
Q

What mediators are known to cause endothelial cell contraction and increased vascular permeability?

A

Histamine, bradykinin, leukotrienes, and other chemical mediators.

245
Q

What is the immediate transient response in the context of vascular permeability?

A

It is the rapid and usually short-lived opening of interendothelial gaps occurring 15 to 30 minutes after mediator exposure.

246
Q

What is delayed prolonged leakage, and what might cause it?

A

It is vascular leakage that starts 2 to 12 hours after exposure and lasts for hours or days, often due to mild endothelial damage or contraction, such as from sunburn or certain bacterial toxins.

247
Q

What causes endothelial injury and detachment in severe acute inflammation?

A

Severe physical injuries (e.g., thermal burns), microbial actions, and microbial toxins that damage endothelial cells.

248
Q

How does the presence of neutrophils affect endothelial cells during inflammation?

A

Neutrophils adhering to the endothelium can injure endothelial cells, amplifying the inflammatory reaction.

249
Q

When does leakage typically start and how long does it last in cases of endothelial injury?

A

Leakage starts immediately after endothelial injury and can last for several hours until the damaged vessels are repaired or thrombosed.

250
Q

How do lymphatic vessels participate in acute inflammation?

A

They help drain edema fluid that accumulates due to increased vascular permeability, as well as leukocytes, cell debris, and microbes.

251
Q

What happens to lymphatic vessels during inflammation?

A

They proliferate to handle the increased load of fluid and cellular debris.

252
Q

What is lymphangitis?

A

Inflammation of the lymphatic vessels.

253
Q

What is lymphadenitis?

A

Inflammation of the lymph nodes, often characterized by hyperplasia of lymphoid follicles and increased numbers of lymphocytes and macrophages.

254
Q

What is reactive lymphadenitis?

A

A constellation of pathologic changes in lymph nodes due to inflammation, including enlargement and hyperplasia.

255
Q

What might be seen alongside lymphangitis as a sign of bacterial infection?

A

Painful enlargement of the draining lymph nodes, indicating lymphadenitis.

256
Q

What is the primary function of leukocytes in inflammation?

A

To eliminate offending agents by ingesting and destroying bacteria, microbes, necrotic tissue, and foreign substances.

257
Q

Which leukocytes are most important in typical inflammatory reactions and why?

A

Neutrophils and macrophages, because they are capable of phagocytosis and can ingest and destroy pathogens and debris.

258
Q

What additional role do macrophages play beyond phagocytosis during inflammation?

A

They produce growth factors that aid in tissue repair.

259
Q

What is a downside of the defensive potency of activated leukocytes?

A

They can induce tissue damage and prolong the inflammatory reaction by harming normal bystander host tissues.

260
Q

What mediates the journey of leukocytes from the vessel lumen to the tissue?

A

Adhesion molecules and chemokines (cytokines that attract cells).

261
Q

What happens during the margination and rolling phase of leukocyte migration?

A

Leukocytes move to the periphery of the blood vessel, rolling along the activated endothelium before adhering.

262
Q

What occurs during the migration phase across the endothelium and vessel wall?

A

Leukocytes pass through the endothelial cells and the vessel wall to enter the tissue.

263
Q

What guides leukocytes to the site of infection or injury once they are in the tissue?

A

Chemotactic stimuli, which are signals that attract leukocytes to the area.

264
Q

What is margination in the context of inflammation?

A

The process where white blood cells move to the periphery of blood vessels and assume a position along the endothelial surface due to slowed blood flow.

265
Q

What are the two major families of adhesion molecules involved in leukocyte adhesion and migration?

A

Selectins and integrins.

266
Q

What role do selectins play in leukocyte adhesion?

A

They mediate the initial rolling interactions of leukocytes on the endothelial surface.

267
Q

What are the three types of selectins and where are they found?

A

L-selectin (on leukocytes), E-selectin (on endothelium), and P-selectin (on platelets and endothelium).

268
Q

How do integrins contribute to leukocyte adhesion?

A

They mediate firm adhesion of leukocytes to the endothelium after initial rolling interactions.

269
Q

What induces the expression of selectins and their ligands on endothelial cells?

A

Cytokines like TNF and IL-1, produced in response to infection or injury.

270
Q

What triggers the redistribution of P-selectin to the endothelial surface?

A

Mediators like histamine and thrombin.

271
Q

What is the role of chemokines in leukocyte adhesion?

A

They activate rolling leukocytes and increase the affinity of integrins, leading to firm adhesion.

272
Q

What is the consequence of integrin activation on leukocytes?

A

It converts integrins to a high-affinity state, promoting firm adhesion to the endothelium.

273
Q

What are the ligands for integrins expressed on the endothelium?

A

VCAM-1 (for VLA-4 integrins) and ICAM-1 (for LFA-1 and MAC-1 integrins).

274
Q

What is the process of leukocyte migration through the endothelium called?

A

Transmigration or diapedesis.

275
Q

What is the role of chemokines in leukocyte migration?

A

Chemokines stimulate adherent leukocytes to migrate through interendothelial gaps toward the site of injury or infection.

276
Q

What adhesion molecule is involved in the migration of leukocytes through interendothelial gaps?

A

CD31 or PECAM-1 (platelet endothelial cell adhesion molecule).

277
Q

How do leukocytes penetrate the basement membrane during transmigration?

A

By secreting collagenases.

278
Q

What is chemotaxis in the context of inflammation?

A

The movement of leukocytes along a chemical gradient toward the site of injury or infection.

279
Q

How do chemoattractants affect leukocyte movement?

A

They bind to seven-transmembrane G protein-coupled receptors on leukocytes, leading to cytoskeletal reorganization that drives migration.

280
Q

What is the result of cytoskeletal reorganization in leukocytes during chemotaxis?

A

Extension of filopodia at the leading edge and pulling of the back of the cell in the direction of the chemical gradient.

281
Q

What is the significance of leukocyte adhesion molecule deficiencies?

A

They result in increased susceptibility to bacterial infections.

282
Q

In most acute inflammatory responses, which leukocytes predominate during the first 6 to 24 hours?

A

Neutrophils

283
Q

After the initial neutrophil response in acute inflammation, which leukocytes typically replace them within 24 to 48 hours?

A

Monocytes

284
Q

Why do neutrophils predominate early in acute inflammation?

A

Neutrophils are more numerous, respond rapidly to chemokines, and adhere more firmly to early adhesion molecules like P-selectin and E-selectin.

285
Q

How long do neutrophils typically survive in extravascular tissues?

A

A few days, after which they undergo apoptosis.

286
Q

What allows monocytes to become the dominant leukocyte population in prolonged inflammation?

A

Monocytes survive longer and can proliferate in tissues.

287
Q

In what type of infection might neutrophils be continuously recruited for several days?

A

Infections caused by Pseudomonas bacteria.

288
Q

Which cells are typically the first to arrive in viral infections?

A

Lymphocytes

289
Q

In hypersensitivity reactions, which cells are often predominant?

A

Activated lymphocytes, macrophages, and plasma cells.

290
Q

What types of infections or reactions are eosinophils mainly associated with?

A

Helminthic infections and allergic reactions.

291
Q

What is the collective term for the responses induced in leukocytes by the recognition of microbes or dead cells?

A

Leukocyte activation.

292
Q

What are the key signaling pathways triggered during leukocyte activation

A

Increases in cytosolic Ca__, activation of protein kinase C, and phospholipase A_.

293
Q

What are the primary functional responses of leukocytes important for destroying microbes?

A

Phagocytosis and intracellular killing.

294
Q

Name two enzymes activated during leukocyte activation.

A

Protein kinase C and phospholipase A_.

295
Q

What role does an increase in cytosolic Ca__ play in leukocyte activation?

A

It is part of the signaling pathways that enhance leukocyte functions.

296
Q

How do activated leukocytes destroy microbes?

A

Through phagocytosis and intracellular killing mechanisms.

297
Q

What types of receptors on phagocytes help in binding and ingesting microbes?

A

Mannose receptors, scavenger receptors, and receptors for various opsonins.

298
Q

What does the macrophage mannose receptor recognize?

A

Terminal mannose and fucose residues on microbial glycoproteins and glycolipids.

299
Q

How do scavenger receptors function in phagocytosis?

A

They bind and mediate endocytosis of oxidized or acetylated LDL particles and a variety of microbes.

300
Q

What enhances the efficiency of phagocytosis?

A

The coating of microbes with opsonins like IgG antibodies, C3b, mannose-binding lectin, and collectins.

301
Q

What happens during the engulfment process of phagocytosis?

A

Extensions of the cytoplasm flow around the particle, forming a phagosome, which then fuses with a lysosomal granule to form a phagolysosome.

302
Q

What is phagocytosis dependent on?

A

Polymerization of actin filaments.

303
Q

What are the primary mechanisms of intracellular destruction of microbes?

A

Reactive oxygen species (ROS), reactive nitrogen species (NO), and lysosomal enzymes.

304
Q

Under what circumstances can leukocytes cause injury to normal cells and tissues?

A

When they cause collateral damage during infection, in autoimmune diseases, or in allergic reactions.

305
Q

What types of substances do activated leukocytes produce that can damage host tissues?

A

Reactive oxygen species (ROS), reactive nitrogen species (NO), and lysosomal enzymes.

306
Q

What is frustrated phagocytosis and what does it trigger?

A

Occurs when leukocytes cannot easily ingest materials like immune complexes, leading to strong activation and release of lysosomal enzymes into the extracellular environment.

307
Q

What roles do activated leukocytes play in host defense beyond eliminating microbes and dead cells?

A

They produce cytokines that modulate inflammation, growth factors for cell proliferation and collagen synthesis, and enzymes for connective tissue remodeling.

308
Q

How do activated macrophages contribute to tissue repair?

A

By producing growth factors that stimulate endothelial cell and fibroblast proliferation and collagen synthesis.

309
Q

What is the role of Th17 cells in acute inflammation?

A

They produce IL-17, which induces the secretion of chemokines to recruit other leukocytes.

310
Q

What happens in the absence of effective Th17 responses?

A

Individuals are more susceptible to fungal and bacterial infections and develop “cold abscesses” that lack classic signs of acute inflammation.

311
Q

What is the source and action of histamine in inflammation?

A

Source: Mast cells, basophils, platelets; Action: Vasodilation, increased vascular permeability, endothelial activation.

312
Q

What effects do prostaglandins have during inflammation?

A

Vasodilation, pain, fever.

313
Q

Which cells produce leukotrienes, and what are their effects?

A

Produced by mast cells and leukocytes; Effects: Increased vascular permeability, chemotaxis, leukocyte adhesion and activation.

314
Q

What are the local and systemic actions of cytokines such as TNF, IL-1, and IL-6?

A

Local: Endothelial activation (expression of adhesion molecules); Systemic: Fever, metabolic abnormalities, hypotension (shock).

315
Q

What role do chemokines play in inflammation?

A

Chemotaxis, leukocyte activation.

316
Q

What are the functions of platelet-activating factor in inflammation?

A

Vasodilation, increased vascular permeability, leukocyte adhesion, chemotaxis, degranulation, oxidative burst.

317
Q

What are the roles of complement in inflammation?

A

Leukocyte chemotaxis and activation, direct target killing (membrane attack complex), vasodilation (mast cell stimulation).

318
Q

What effects do kinins have in the inflammatory response?

A

Increased vascular permeability, smooth muscle contraction, vasodilation, pain.

319
Q

What cells are the primary source of histamine?

A

Mast cells, basophils, platelets.

320
Q

What are the main effects of histamine in inflammation?

A

Dilation of arterioles, increased permeability of venules, contraction of some smooth muscles.

321
Q

What is serotonin’s primary function and its role in inflammation?

A

Neurotransmitter in the gastrointestinal tract and central nervous system; Vasoconstriction (importance unclear).

322
Q

What is arachidonic acid (AA), and how is it produced?

A

AA is a 20-carbon polyunsaturated fatty acid derived from dietary sources or synthesized from linoleic acid. Produced from membrane phospholipids by phospholipase A2.

323
Q

What are the two major classes of enzymes involved in synthesizing arachidonic acid metabolites, and what do they produce?

A

Cyclooxygenases (produce prostaglandins), lipoxygenases (produce leukotrienes and lipoxins).

324
Q

What are eicosanoids, and how do they affect inflammation?

A

Eicosanoids are AA-derived mediators synthesized from 20-carbon fatty acids. They bind to G protein-coupled receptors and mediate virtually every step of inflammation.

325
Q

What is the complement system, and what is its primary function?

A

The complement system is a collection of plasma proteins that function in host defense against microbes and in pathologic inflammatory reactions.

326
Q

How many complement proteins are there, and how are they numbered?

A

There are more than 20 complement proteins, some of which are numbered C1 through C9.

327
Q

What are the main effects of complement protein cleavage products in inflammation?

A

Increased vascular permeability, chemotaxis, and opsonization.

328
Q

How does the complement system contribute to both innate and adaptive immunity?

A

By defending against microbial pathogens and participating in inflammatory responses.

329
Q

What defines chronic inflammation?

A

Chronic inflammation is a prolonged response (weeks or months) where inflammation, tissue injury, and repair coexist in varying combinations.

330
Q

What are some causes of chronic inflammation?

A

Persistent infections, hypersensitivity diseases, and prolonged exposure to toxic agents.

331
Q

How can persistent infections lead to chronic inflammation?

A

By causing an immune reaction that is difficult to eradicate, such as with mycobacteria, certain viruses, fungi, and parasites.

332
Q

What is a granulomatous reaction, and when might it occur?

A

It is a specific pattern of chronic inflammation that can develop in response to persistent infections or irritants that cannot be easily cleared.

333
Q

What role does chronic inflammation play in hypersensitivity diseases?

A

It results from excessive and inappropriate immune system activation, leading to autoimmune diseases or allergic diseases.

334
Q

How does prolonged exposure to toxic agents cause chronic inflammation?

A

It results from continued irritation or damage by substances such as particulate silica or excess cholesterol, leading to diseases like silicosis or atherosclerosis.

335
Q

Can acute and chronic inflammation occur simultaneously?

A

Yes, acute and chronic inflammation can coexist, as seen in conditions like peptic ulcers.

336
Q

What are the dominant cells in chronic inflammatory reactions?

A

Macrophages

337
Q

How do macrophages contribute to chronic inflammation?

A

By secreting cytokines and growth factors, destroying foreign invaders and tissues, activating T lymphocytes, and promoting tissue repair.

338
Q

Where do macrophages originate from?

A

From hematopoietic stem cells in the bone marrow, and from progenitors in the embryonic yolk sac and fetal liver.

339
Q

What is the difference in lifespan between blood monocytes and tissue macrophages?

A

Blood monocytes have a half-life of about 1 day, while tissue macrophages can live for several months or years.

340
Q

What are the two major pathways of macrophage activation?

A

Classical (M1) and Alternative (M2).

341
Q

What characterizes classical macrophage activation (M1)?

A

Production of NO and lysosomal enzymes, and secretion of cytokines that stimulate inflammation.

342
Q

What is the primary role of alternatively activated macrophages (M2)?

A

To terminate inflammation and promote tissue repair.

343
Q

What are the main functions of lymphocytes in chronic inflammation?

A

Amplifying and propagating chronic inflammation, with CD4+ T lymphocytes promoting inflammation through cytokine secretion.

344
Q

What cytokines do Th1 cells produce and what is their role?

A

Th1 cells produce IFN-_, which activates macrophages via the classical pathway.

345
Q

What cytokines are secreted by Th2 cells and what is their function?

A

Th2 cells secrete IL-4, IL-5, and IL-13, which recruit and activate eosinophils and support alternative macrophage activation.

346
Q

What is the role of Th17 cells in chronic inflammation?

A

They secrete IL-17, which induces chemokine secretion and recruits neutrophils and monocytes.

347
Q

How do macrophages and lymphocytes interact in chronic inflammation?

A

Macrophages display antigens to T cells and produce cytokines that stimulate T cells, which in turn produce cytokines that recruit and activate macrophages.

348
Q

What are tertiary lymphoid organs and where are they commonly found?

A

Organized clusters of lymphocytes resembling lymph nodes, often found in chronic conditions like rheumatoid arthritis and Hashimoto thyroiditis.

349
Q

What role do eosinophils play in chronic inflammation?

A

They are involved in IgE-mediated immune reactions and parasitic infections but can also cause tissue damage.

350
Q

How do mast cells contribute to chronic inflammation?

A

By releasing mediators like histamine and cytokines, which can promote inflammatory reactions.

351
Q

What role do neutrophils play in chronic inflammation?

A

They may persist in chronic inflammation, particularly in cases of persistent microbes or irritant stimuli.

352
Q

What characterizes granulomatous inflammation?

A

Collections of activated macrophages, often with T lymphocytes, sometimes associated with necrosis.

353
Q

What is the primary goal of granuloma formation?

A

To contain an offending agent that is difficult to eradicate.

354
Q

What are epithelioid cells?

A

Activated macrophages with abundant cytoplasm that resemble epithelial cells.

355
Q

What are multinucleate giant cells?

A

Macrophages that have fused together, forming cells with multiple nuclei.

356
Q

What induces foreign body granulomas?

A

Inert foreign bodies that induce inflammation without a T cell-mediated immune response.

357
Q

What materials commonly cause foreign body granulomas?

A

Talc, sutures, or other fibers that are too large to be phagocytosed and are not immunogenic.

358
Q

What characterizes immune granulomas?

A

They are caused by agents that induce a persistent T cell-mediated immune response, such as persistent microbes.

359
Q

What role do Th1 cells play in immune granulomas?

A

They produce cytokines like IFN-_ that activate macrophages.

360
Q

In which infections are immune granulomas associated with Th2 responses and eosinophils?

A

Some parasitic infections, such as schistosomiasis.

361
Q

Name two notable conditions associated with granulomatous inflammation.

A

Crohn disease and sarcoidosis.

362
Q

Why is it important to recognize granulomatous inflammation?

A

Because it can be caused by a limited number of conditions, some of which can be life-threatening.

363
Q

How can specific granulomatous diseases be diagnosed?

A

Through special stains for organisms, culture methods, molecular techniques, and serologic studies.

364
Q

What stimulates the production of prostaglandins in the hypothalamus during inflammation?

A

Cytokines TNF and IL-1.

365
Q

What are acute-phase proteins, and what stimulates their production?

A

Proteins like C-reactive protein; their production is stimulated by cytokines (e.g., IL-6) acting on liver cells.

366
Q

What causes leukocytosis during inflammation?

A

Cytokines (CSFs) stimulate the production of leukocytes from bone marrow precursors.

367
Q

What is septic shock, and what induces it?

A

Severe infection leading to a fall in blood pressure, disseminated intravascular coagulation, and metabolic abnormalities, induced by high levels of TNF and other cytokines.

368
Q

What is the term for the phenomenon where genetic diseases appear earlier and/or more severely in successive generations?

A

Anticipation

369
Q

What genetic mutation causes myotonic dystrophy type 1?

A

An expanded CTG trinucleotide repeat in the DMPK gene.

370
Q

How does an expanded CTG repeat in myotonic dystrophy type 1 affect the disease severity and age of onset?

A

Increasing repeat number leads to earlier onset and more severe disease.

371
Q

hat type of mutation is responsible for myotonic dystrophy type 2?

A

A 4-bp (CCTG) repeat expansion in an untranslated region of a gene on chromosome 3.

372
Q

How do expanded repeats in untranslated regions of genes cause disease in myotonic dystrophy?

A

The abnormal mRNA interacts with RNA-binding proteins, disrupting normal protein function and causing disease symptoms.

373
Q

What is a common feature of repeat expansions in neurological diseases like Huntington disease?

A

CAG or CTG repeat expansions in protein-coding regions, with disease-causing ranges generally from 50 to 100 repeats.

374
Q

How do repeat expansions in diseases of the third category (e.g., fragile X syndrome, myotonic dystrophy) typically differ from those in neurological diseases?

A

They are usually much larger (100 to several thousand repeats), located outside protein-coding regions, and often produce harmful RNA products.

375
Q

In which category of repeat expansion diseases is anticipation commonly observed?

A

Both neurological diseases and disorders with very large repeat expansions.

376
Q

What is the primary cause of Fragile X Syndrome?

A

A CGG repeat expansion in the FMR1 gene.

377
Q

How many CGG repeats are typically found in unaffected individuals?

A

6 to 50 copies

378
Q

What is the repeat range for a premutation in Fragile X Syndrome?

A

50 to 200 copies of CGG repeats.

379
Q

What is the repeat range for a full mutation in Fragile X Syndrome?

A

200 to 1000 or more copies of CGG repeats.

380
Q

Why do males with Fragile X Syndrome often have a more severe presentation than females?

A

Due to higher penetrance and expression in males.

381
Q

What is the Sherman paradox in the context of Fragile X Syndrome?

A

The observation that daughters of normal transmitting males are never affected, but their sons can be, despite both being carriers.

382
Q

How does the premutation in Fragile X Syndrome affect females differently than males?

A

Premutations can expand to full mutations in females, leading to Fragile X Syndrome in their offspring, whereas males do not pass on full mutations.

383
Q

What are the common clinical features of Fragile X Syndrome?

A

Intellectual disability, distinctive facial appearance (large ears, long face), hypermobile joints, and macroorchidism in postpubertal males.

384
Q

What effect does the full mutation have on FMR1 mRNA production?

A

No FMR1 mRNA is produced due to heavy methylation of the gene.

385
Q

What health issues are associated with FMR1 premutations?

A

Neurological disorders such as ataxia and tremors in males, and premature ovarian insufficiency in females.

386
Q

How does Fragile X Syndrome demonstrate anticipation?

A

The number of CGG repeats often increases in successive generations, leading to earlier onset and more severe symptoms in descendants.

387
Q

What is the function of the FMRP protein, and why is it important?

A

FMRP binds to RNA and regulates its translation, playing a role in mRNA transport from the nucleus to the cytoplasm.

388
Q

What diagnostic method is preferred for Fragile X Syndrome over cytogenetic analysis?

A

DNA testing for the length of the CGG repeat sequence and degree of methylation of the FMR1 gene.

389
Q

What is the first step in T lymphocyte activation?

A

The recognition of the same antigen that the T lymphocyte will later target to eliminate.

390
Q

Where does the initial activation of naive T lymphocytes primarily occur?

A

In secondary (peripheral) lymphoid organs.

391
Q

What type of cells present antigens to naive T lymphocytes to initiate activation?

A

Mature dendritic cells (DCs).

392
Q

What are the two outcomes after the initial activation of naive T lymphocytes?

A

Proliferation and differentiation into effector and memory cells.

393
Q

What is the role of effector T cells in immune responses?

A

To eliminate the source of the antigen, such as infected cells or tumors.

394
Q

Where do naive T lymphocytes circulate before they are activated?

A

Throughout the body in a resting state.

395
Q

What specialized regions are involved in the activation of naive T lymphocytes?

A

Lymph nodes, spleen, and mucosal lymphoid tissues.

396
Q

How do naive T lymphocytes acquire their powerful functional capabilities?

A

By being activated after recognizing antigens presented by dendritic cells.

397
Q

What are the three major biologic responses in T cells after antigen recognition and other activating stimuli?

A

Cytokine secretion, increased cytokine receptor expression, and clonal expansion.

398
Q

What is clonal expansion in T cells?

A

The proliferation of T cells, leading to an increase in the numbers of cells in antigen-specific clones.

399
Q

What do naive T cells differentiate into after activation?

A

Effector and memory lymphocytes.

400
Q

How does T cell activation affect surface molecule expression?

A

It changes the expression of surface molecules, which are involved in T cell trafficking and regulating T cell responses.

401
Q

What additional roles do antigen-presenting cells (APCs) play besides displaying antigens?

A

APCs express surface molecules and secrete cytokines that influence the magnitude and nature of the T cell response.

402
Q

How do activated T cells regulate APCs?

A

Activated T cells deliver signals back to APCs, increasing their ability to activate T cells in a positive feedback loop.

403
Q

What role do surface molecules and cytokines from activated T cells play in the immune response?

A

They inhibit further activation to establish safe limits to the response.

404
Q

Where do naive T cells and effector T cells mainly get activated?

A

Naive T cells are activated in secondary lymphoid organs, while effector T cells can be activated in any tissue.

405
Q

What is the main function of effector CD4+ T cells, also known as helper T cells?

A

They activate macrophages or B cells to kill microbes or help B cells differentiate into antibody-secreting plasma cells and memory cells.

406
Q

What do cytotoxic T lymphocytes (CTLs) do?

A

They kill infected cells and tumor cells and secrete cytokines that activate macrophages and induce inflammation.

407
Q

Where are memory T cells most abundant, and what is their main feature?

A

Memory T cells are abundant in mucosal tissues, the skin, and lymphoid organs, and they have an enhanced ability to rapidly react against the antigen.

408
Q

Why do T cell responses decline after antigen elimination?

A

Because most antigen-activated effector T cells die by apoptosis due to the deprivation of survival stimuli and the activation of inhibitory mechanisms.

409
Q

What is the first signal necessary for T lymphocyte activation?

A

Recognition of antigen.

410
Q

What do CD4+ and CD8+ T lymphocytes recognize on APCs?

A

Peptide-MHC complexes.

411
Q

Where are protein antigens captured and transported to after crossing epithelial barriers?

A

Lymph nodes.

412
Q

How are naive T cells and mature DCs drawn to T cell zones of secondary lymphoid organs?

A

By chemokines engaging the CCR7 chemokine receptor.

413
Q

What happens when naive T cells recognize the specific antigen displayed by DCs?

A

They stop moving and initiate the activation program.

414
Q

Which cells can present antigens to effector T cells besides dendritic cells (DCs)?

A

B cells, macrophages, and virtually any nucleated cell.

415
Q

What is required in addition to antigen-induced signals for the proliferation and differentiation of naive T cells?

A

Costimulatory signals from APCs.

416
Q

What happens to T cells that encounter antigens without costimulation?

A

They fail to respond, become unresponsive, or die.

417
Q

What is the best-characterized costimulatory pathway in T cell activation?

A

The CD28 receptor binding to B7-1 (CD80) and B7-2 (CD86) on APCs.

418
Q

Which cells express B7-1 (CD80) and B7-2 (CD86)?

A

Activated APCs, including dendritic cells (DCs), macrophages, and B lymphocytes.

419
Q

What is the structure of the CD28 receptor?

A

A disulfide-linked homodimer with a single extracellular Ig domain on each subunit.

420
Q

What stimulates the expression of B7 molecules on APCs?

A

Microbial products, Toll-like receptor engagement, and cytokines like IFN-_.

421
Q

Why do mature dendritic cells express high levels of costimulators?

A

They are the most potent stimulators of naive T cells.

422
Q

What role do adjuvants play in T cell activation?

A

They stimulate the expression of B7 costimulators on APCs.

423
Q

What happens to potentially self-reactive T cells when they encounter self antigens presented by resting APCs?

A

They are not activated and may become permanently unresponsive.

424
Q

How do CD28 signals contribute to T cell activation?

A

They promote survival, proliferation, and differentiation of antigen-specific T cells.

425
Q

What protein kinase is recruited by CD28 signaling?

A

PI3-kinase, which activates AKT.

426
Q

What is the outcome of the signaling pathways activated by CD28 in T cells?

A

Increased expression of anti-apoptotic proteins, metabolic activity, proliferation, cytokine production, and differentiation into effector and memory cells.

427
Q

What is a key difference between naive T cells and effector/memory T cells regarding costimulation?

A

Effector and memory T cells are less dependent on the B7

428
Q

Why can effector CTLs kill cells that do not express costimulators?

A

Because once differentiated, effector CTLs can respond to antigens without needing costimulation.

429
Q

What is ICOS and its role in T cell responses?

A

A costimulator important for T cell-dependent antibody responses, especially in the germinal center reaction.

430
Q

Which ligand does ICOS bind to?

A

Binds to ICOS-L (CD275), which is expressed on DCs, B cells, and other cells.

431
Q

What is the role of T follicular helper cells that are activated by ICOS?

A

They are essential for the formation of germinal centers and the generation of B cells that produce high-affinity antibodies.

432
Q

How do other receptors related to CD28 and B7 influence T cell responses?

A

They can either positively or negatively regulate T cell responses.

433
Q

What is the role of CTLA-4 and PD-1 in T cell activation?

A

They act as inhibitory receptors, limiting immune responses by counterbalancing the activating signals from receptors like CD28.

434
Q

How does CTLA-4 inhibit T cell activation?

A

It competes with CD28 for binding to B7 molecules, binding more strongly and thus reducing T cell activation.

435
Q

What is the main function of PD-1 in T cell inhibition?

A

PD-1 recruits a tyrosine phosphatase that blocks TCR and CD28 signaling, inhibiting T cell activation.

436
Q

What is the significance of the CD28 interaction in T cell responses?

A

It is crucial for initiating T cell responses by activating naive T cells.

437
Q

Which costimulatory pathway is critical for helper T cell-dependent antibody responses?

A

The ICOS pathway is essential for helper T cell-dependent antibody responses.

438
Q

How does the CD40L interaction enhance T cell responses?

A

It activates APCs, making them more potent by enhancing the expression of B7 molecules and cytokines, indirectly amplifying T cell responses.

439
Q

What is the role of OX40 in T cell responses?

A

It maintains cell survival and sustained T cell responses.

440
Q

Which superfamily do OX40 and 4-1BB belong to, and what is their general function?

A

They belong to the TNFR superfamily and are involved in regulating immune responses, including maintaining T cell activation and survival.

441
Q

What is the significance of CD40L on activated T cells?

A

CD40L engages CD40 on APCs, enhancing their ability to stimulate T cells by increasing B7 expression and cytokine production.

442
Q

How do TIM-3 and LAG-3 contribute to T cell regulation?

A

They are inhibitory receptors on T cells, with roles in regulating immune responses, although their exact physiological functions are still being studied.

443
Q

What is costimulatory blockade?

A

It is a therapeutic approach that inhibits costimulation to control injurious immune responses.

444
Q

How does CTLA-4-Ig work in costimulatory blockade?

A

CTLA-4-Ig binds to B7-1 and B7-2, blocking the B7

445
Q

Why is CTLA-4 used in CTLA-4-Ig instead of CD28?

A

CTLA-4 has a higher affinity for B7 molecules than CD28, making it more effective in blocking the interaction.

446
Q

What are the clinical uses of CTLA-4-Ig?

A

CTLA-4-Ig is used to treat rheumatoid arthritis and to prevent transplant rejection.

447
Q

What is checkpoint blockade in cancer immunotherapy?

A

It involves using antibodies to block inhibitory receptors like CTLA-4 and PD-1, reducing inhibition and enhancing T cell activation against tumors.

448
Q

What is a potential side effect of checkpoint blockade therapy?

A

It can induce autoimmune reactions due to the role of inhibitory receptors in maintaining self-tolerance.

449
Q

What is the role of the Fc portion of IgG in CTLA-4-Ig?

A

It increases the in vivo half-life of the CTLA-4-Ig fusion protein.

450
Q

Which pathway inhibitors are in clinical trials for transplant rejection and autoimmune diseases?

A

Inhibitors of the CD40L pathway are in clinical trials for these conditions.

451
Q

What is the role of CD69 in T cell activation?

A

CD69 binds to and reduces the expression of S1PR1, retaining activated T cells in lymphoid organs for proliferation and differentiation.

452
Q

Why is CD25 (IL-2R_) important in T cell activation?

A

CD25 is a component of the IL-2 receptor, allowing activated T cells to respond to the growth factor IL-2.

453
Q

What does CD40 ligand (CD40L) do in T cell activation?

A

CD40L enables CD4+ T cells to help macrophages and B cells and activates DCs to enhance T cell responses.

454
Q

What is the function of CTLA-4 and PD-1 in T cells?

A

CTLA-4 and PD-1 act as inhibitors to limit immune responses after T cell activation.

455
Q

What changes occur in adhesion molecules and chemokine receptors during T cell activation?

A

Activated T cells decrease molecules for lymphoid organ migration (e.g., CD62L, CCR7) and increase molecules for migration to infection sites (e.g., LFA-1, VLA-4, CD44).

456
Q

What is the role of IL-2 in T cell activation?

A

IL-2 promotes survival, proliferation, and differentiation of antigen-activated T cells.

457
Q

How is IL-2 produced and regulated in T cells?

A

IL-2 is produced mainly by CD4+ T cells after antigen recognition, with production peaking at 8-12 hours and declining by 24 hours.

458
Q

What are the components of the high-affinity IL-2 receptor (IL-2R)?

A

IL-2R consists of IL-2R_ (CD25), IL-2/15R_ (CD122), and _c (CD132).

459
Q

What happens to IL-2R expression after T cell activation?

A

IL-2R_ is transiently expressed on activated T cells, forming a high-affinity IL-2R complex with IL-2R_ and _c.

460
Q

How does IL-2 affect regulatory T cells?

A

IL-2 is required for the survival and function of regulatory T cells, which are more sensitive to IL-2 and do not produce it themselves.

461
Q

What effect does IL-2 have on cell cycle progression in T cells?

A

IL-2 activates mTOR, inducing cyclin synthesis and degrading the cell cycle inhibitor p27, thus promoting cell cycle progression.

462
Q

What mediates T cell proliferation in response to antigen recognition?

A

T cell proliferation is mediated by signals from the antigen receptor, costimulators, and autocrine growth factors, primarily IL-2.

463
Q

What is the frequency of naive T cells specific for any antigen before exposure?

A

Before exposure, the frequency is 1 in 10^5 to 10^6 lymphocytes or fewer.

464
Q

To what extent can the frequency of antigen-specific CD8+ T cells increase after microbial antigen exposure?

A

The frequency can increase to as many as 1 in 3 CD8+ T lymphocytes, representing a greater than 50,000-fold expansion.

465
Q

What is the increase in frequency of antigen-specific CD4+ T cells after exposure to an antigen?

A

The frequency can increase to up to 1 in 100 CD4+ lymphocytes, representing a 1000-fold expansion.

466
Q

How quickly can a massive expansion of antigen-specific T cells occur after infection?

A

This expansion can occur within as little as 1 week after infection.

467
Q

Do bystander T cells proliferate during the expansion of antigen-specific T cells?

A

No, bystander T cells that are not specific for the virus do not proliferate during this expansion.

468
Q

What do T cell-mediated immune responses usually result in the generation of?

A

Memory T cells specific for the antigen.

469
Q

How long may memory T cells persist?

A

Memory T cells may persist for years, even a lifetime.

470
Q

What are the two possible fates of lymphocytes activated by antigen and other stimuli?

A

They can either become short-lived effector cells or enter the long-lived memory cell pool.

471
Q

What is the small pool of cells called from which memory populations are mainly generated?

A

Memory precursor effector cells (MPECs).

472
Q

What factors may influence the development of memory T cells?

A

Factors may include the strength of TCR stimulation, the level of costimulation, and the cytokine environment.

473
Q

Is there a single transcription factor that determines whether a T cell becomes a terminal effector cell or a memory cell?

A

No, the choice is controlled by quantitative differences in numerous transcription factors and epigenetic reprogramming.

474
Q

What allows memory T cells to survive for prolonged periods?

A

Increased levels of anti-apoptotic proteins such as BCL-2 and BCL-X_L.

475
Q

How do memory T cells respond to antigen stimulation compared to naive T cells?

A

Memory T cells respond more rapidly and effectively.

476
Q

How does the number of memory T cells specific for an antigen compare to the number of naive cells before antigen exposure?

A

Memory T cells are 10- to 100-fold more numerous than naive cells.

477
Q

What allows memory T cells to migrate to various tissues and respond to antigens?

A

Differences in the expression of adhesion molecules and chemokine receptors.

478
Q

What is the primary cytokine required for the maintenance of memory T cells?

A

IL-7. (Memory CD8+ T cells also depend on IL-15.)

479
Q

Which surface markers are commonly associated with memory T cells?

A

High expression of IL-7 receptor (CD127) and CD27, and absence of markers of naive and recently activated T cells.

480
Q

What are the three major subsets of memory T cells?

A

Central memory T cells (T_CM), Effector memory T cells (T_EM), and Tissue-resident memory T cells (T_RM).

481
Q

Where do central memory T cells (T_CM) primarily home to and what is their main function?

A

They home mainly to lymph nodes and generate many effector cells on antigen challenge.

482
Q

Where do effector memory T cells (T_EM) home to and what is their primary function?

A

They home to peripheral sites and mucosal tissues and rapidly produce effector cytokines or become cytotoxic.

483
Q

What is a characteristic feature of tissue-resident memory T cells (T_RM)?

A

They express high levels of CD69, reducing S1PR1 expression, which helps them stay in tissues.

484
Q

What is the function of the TEMRA subset of memory T cells?

A

TEMRA cells express the CD45RA isoform and are thought to have properties similar to naive T cells, but their unique functional properties are not well-defined.

485
Q

How are memory T cells heterogeneous in terms of cytokine profiles?

A

They can be derived from activated T cells of various subsets (Th1, Th2, Th17) and retain or differentiate into these cytokine profiles upon reactivation.

486
Q

What triggers the decline of T cell responses after antigen elimination?

A

The reduction of costimulation and growth factors like IL-2, which decreases anti-apoptotic proteins and increases pro-apoptotic signals.