pt 2 Flashcards

(524 cards)

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

define gene therapy

A

the delivery of therapeutic genes into the human body to correct conditions created by a faulty gene or genes

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

gene therapy - Cystic Fibrosis

A
  • conventional treatment includes back clapping to drain lung mucus, drugs to thin mucus, antibiotic treatments
  • the CFTR gene was cloned in 1989 and by 1993 scientists had begun gene therapy trials. the challenge, however, was the right amount of CFTR.
    0 in early 1990s, medical researchers placed the normal allele of the gene into the respiratory tissue of mice with the disease. these mice produced a functional CFTR protein
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

gene therapy - sickle cell anaemia

A
  • most widespread inherited blood disorder in North America
  • homozygosity for the HbBs allele
  • current treatment consists of a prescription drug with toxic side effects or bone marrow transplantation
  • in 2001, a Harvard research team successfully treated a sickle-cell mouse model with gene therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

two primary strategies for gene therapy

A

ex vivo gene therapy
in vivo gene therapy

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

ex vivo gene therapy

A

cells are removed from the patient -> gene introduced into the cells -> cells reintroduced into the body

eg bone marrow blood precursor cells

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

in vivo gene therapy

A

introducing genes directly into tissues and organs in the body (injected into retinal cells, inhaled into the lungs)

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

what is the main challenge with in vivo gene therapy?

A

delivering genes only to intended tissues

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

use the liver as an example to compare in vivo gene therapy with ex vivo gene therapy

A

patient with liver cell genetic defect, lacks gene for blood-clotting protein.

ex vivo:
- remove small portion of liver to isolate cells
- grow cells in culture
- introduce normal genes for clotting protein
- transplant liver cells back into patient. genetically altered proteins provide clotting protein

in vivo:
- normal gene for blood-clotting protein is placed into viruses
- these viruses act as vectors for gene delivery
- directly introduce normal gene for clotting protein into patient’s liver cells

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

SCID

A

SCID = severe combined immunodeficiency
- defect in gene called adenosine deaminase (ADA)
- produces an enzyme involved in the metabolism of nucleotide dATP
- accumulation of dATP is toxic to T cells
- without T cells, B cells cannot recognise antigen and make antibodies

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

ex vivo gene therapy - SCID

A

first human gene therapy (1990)
1. remove ADA-deficient T cells from the SCID patient
2. culture cells in the laboratory
3. infect the cells with a retrovirus that contains the normal ADA gene
4. rein fuse the ADA gene containing T cells back into the SCID patient; genetically altered T cells produce ADA

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

in vivo gene therapy - Leber’s congenital amaurosis

A
  • UPenn and Children’s hospital of Philadelphia
  • LCA is a degenerative disease of the retina that affects 1/50000-100,000 infants each year and causes severe blindness
  • young adult patients with defects in the RPE65 gene were given injections of the normal gene
  • complete vision was not restored but 4 children gained enough vision to play sports and several months after a single treatments patients can see more light/navigate an obstacle course
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

in what way do we rely on viruses as vectors for gene delivery?

A

we can use the viral genome to carry a therapeutic gene or genes -> infect human cells, introducing the gene

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

list the various types of viruses we can use for gene delivery

A
  • adenovirus (common cold): immunogenicity
  • Adeno associated virus (AAV): does not integrate into genome
  • retrovirus
  • HIV based (lentiviral vectors): integrates into the genome
  • herpes virus (cold sores, some cause STDs): target CNS
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

when we use viruses as vectors, what must we make sure to do?

A

we must make sure the virus has been genetically engineered so that it can neither produce disease nor spread throughout the body

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

mode of infection of viruses

A
  1. attachment: selective binding to cells
  2. injection: release genetic material into nucleus or cytoplasm
  3. human cell acts as a host to reproduce the viral genome and to produce viral RNA and proteins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

why viruses make good vectors

A
  • they are efficient at infecting many types of human cells
  • retroviruses or lentiviruses (HIV) permanently inserted their DNA into host cell genome (integration)
  • some viruses infect only certain types of cells - good for targeted gene therapy
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

gene therapy with a retroviral vector

A
  1. attachment
  2. injection
  3. RNA -> dsDNA -> insert viral DNA into host genome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

gene therapy with an AAV vector

A
  1. attachment
  2. injection
  3. ssDNA -> dsDNA -> viral DNA not incorporated into host, usually extrachromosomal
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

how to package a gene into retroviral particles

A
  1. make the recombinant retroviral RNA genome:
    - LTRs (Long Terminal Repeats): required for integration and transcription
    - Ψ (psi) packaging signal: ensures RNA is packaged into the viral capsid
    - gene (downstream of promoter)
  2. package recombinant genome into viral particle:
    check video
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

SCID-X1

A

X-linked severe combined immune deficiency

  • LOF mutation in IL2-RG (necessary to promote the growth of several immune system cells)
  • in 2000, several SCID-X1 patients were cured substantially by gene therapy
  • out of the 9 children treated, 8 are still alive
  • however, 4 eventually developed leukaemia due to insertional mutagenesis, 1 succumbed to the cancer
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

potential risks of viruses as vectors

A
  • death of 18 year old Jesse Gelsinger in 1999 due to complications related to adenovirus vector
  • death of two children in France in 2002
  • temporary cessation of a large number of gene therapy trials and FDA stopped most retroviral studies
  • trials eventually resumed with greater patient monitoring
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

alternative gene delivery options (non-viral)

A

liposomes -> lipid nanoparticles with an active pharmaceutical/genetic ingredient

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

how can we tackle dominant negative or toxic alleles?

A

using RNA interference (RNAi) technology

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
how does RNAi work?
uses complementary RNA to silence gene expression
26
how has RNAi proven to be a promising way to turn off disease genes?
used successfully in cell culture but took over a decade to live up to its promise as a treatment for disease
27
example of endongenous RNAi and how we can use it
using gene therapy to stimulate naturally occurring microRNAs (miRNAs)
28
challenges facing gene therapy
- Can therapeutic gene expression be controlled? - Can we safely and efficiently target only cells that require the gene? - How can gene therapy be targeted to specific regions of the genome? - How long will therapy last? - Will immune system reject? - How many cells need to express the therapeutic gene to treat the condition effectively?
29
genome editing: TALENS
transcription activator-like effector nuclease. these are engineered proteins used for precise genome editing which combine: A DNA-binding domain from TALEs (proteins derived from plant pathogens like Xanthomonas) A DNA-cutting domain from the FokI endonuclease
30
CRISPR-cas9 programmable DNA cleavage
Cas9 uses a guide RNA to locate and bind a specific DNA sequence next to a PAM site. It then creates a double-strand break, allowing targeted gene disruption or editing. Has been used successfully in a wide range of organisms
31
type II CRISPR/cas9 system ingredients
nuclease: - Cas9 single-guide RNA: - CRISPR RNA (crRNA) - trans-activating crRNA (tracrRNA) - optional: section of DNA repair template that is utilised in either non-homologous end joining (NHEJ) or homology directed repair (HDR)
32
what two factors does CRISPR/cas9 depend on for specificity?
the target sequence (20bp long) and the PAM
33
main advantage of CRISPR-cas9
specificity - allows us to target and modify particular DNA sequences in the vast expanse of a genome
34
use of CRISPR- cas9 for sickle cell anaemia
Goal: Use CRISPR-Cas9 to cut the mutated HBB gene and provide a corrected DNA template for homology-directed repair (HDR). - Extract patient hematopoietic stem cells (HSCs) from bone marrow or blood. - Use CRISPR-Cas9 + a DNA template to correct the point mutation in HBB ex vivo. - Re-infuse the edited HSCs into the patient → they produce healthy red blood cells.
35
distinguish between a knocking and knockout
A knockout deletes or disrupts a gene using error-prone NHEJ to inactivate it. A knockin inserts or replaces a gene sequence using precise HDR, often to fix or modify a gene.
36
what are the two unifying themes about cancer genetics?
- cancer is a genetic disease - different inheritance pattern than other genetic disorders
37
cancer is a genetic disease caused by
- mutations in genes that regulate cell division, or activate cell death - environmental factors
38
how does cancer have a different inheritance pattern than other genetic disorders?
- inherited mutations can predispose to cancer (germline) - mutations causing cancer occur in somatic cells - mutations accumulate in clonal descendants of a single cell
39
describe two changes in proteins can lead to cancer
1. protein that stimulates cell division starts working more 2. protein that blocks the cell from dividing stops working
40
cancer phenotypes include (4)
- uncontrolled cell growth - genomic and karyotypic instability - potential for immortality - ability to invade and disrupt local and distant tissues
41
molecular changes causing uncontrolled cell growth
- autocrine stimulation - loss of contact inhibition - loss of apoptosis
42
autocrine stimulation
- when autocrine stimulation is absent, the cell receptor responds to the binding of a ligand produced elsewhere - when autocrine stimulation is present, the cell receptor responds to the binding of a ligand produced in the cell
43
loss of contact inhibition
- contact inhibition present: normal cells stop dividing once they cover a surface and touch neighbouring cells. - contact inhibition absent: cancer cells continue to divide even after touching neighboring cells.
44
loss of apoptosis
- following irradiation, most normal cells recognise the DNA damage that has occurred, leading to cell death - following irradiation, most cancer cells continue dividing despite the DNA damage that has occurred
45
how can defects in DNA repair machinery lead to genomic instability?
1. replication occurs 2. mismatch is created by DNA polymerase - in a normal cell, this mismatch is corrected by mismatch repair and the following round of replication results in a normal DNA sequence - in a cancer cell, the mismatch is not correct and the following round of replication results in a mutated DNA sequence
46
karyotypic instability
- increased rate of chromosomal aberrations - this includes gains/losses of chromosomes, chromosomal rearrangements, and formation of abnormal structures (eg dicentric chromosomes)
47
draw and explain the graph for immortality of cancer cells
number of passages (X-Axis): where you take a cell, grow it, move it to another plate, and repeat cumulative cell number (Y-axis) - in normal cells, as number of passages increases after a certain point, cumulative cell number plateaus - in cancer cells, they lose the ability to stop passaging so the line is straight
48
3 steps for the spread of cancer
1. angiogenesis: the formation of blood vessels to distribute nutrients to all the cancer cells 2. metastasis: where cancer cells break through the basement membrane and go to another body part 3. evasion of immune surveillance
49
multi-hit model of carcinogenesis
proposes that cancer arises through the accumulation of multiple genetic and epigenetic alterations in clonal copies of a single cell
50
evidence for the clonal origin of tumours
- scientists explored this through X-inactivation in females - they looked at proteins on the X chromosome - in normal tissue, there was a mixture of cells with proteins from both X chromosomes - in tumour cells, there was a mixture of cells with proteins from only one X chromosome
51
two main pieces of evidence for the multi-hit nature of cancer
1. lung cancer death rates: males started smoking around 1930 and lung cancer death rates peaked around 1990. females started smoking around 1950 and lung cancer death rates peaked around 2000. 2. incidents of cancer increase exponentially between age 0 and age 80
52
give an example of the familial genetic predisposition to certain types of cancer
- retinoblastoma is caused by inherited mutations in the RB gene - individuals who inherit a single RB- mutant allele are more prone to this cancer as loss of heterozygosity through WT mutation is more likely
53
two general types of cancer producing mutations
a) oncogenes b) tumour suppressor genes
54
oncogenes
- members of signal transduction systems that produce proteins promoting cell proliferation - cancer occurs when an oncogene obtains a gain-of-function mutation in ONE allele
55
tumour-suppressor genes
- members of cell cycle checkpoint control and DNA repair mechanisms - produce protein that inhibits cell proliferation or protects the genome - cancer occurs when a tumour-suppressor gene obtains a loss-of-function mutation in BOTH alleles
56
approaches to identifying oncogenes
- tumour-causing viruses - transform normal mouse cells with human tumour DNA - genome wide functional screens
57
role of tumour-causing viruses in identifying oncogenes
- Retroviral DNA integrated near proto-oncogenes in cellular DNA. - The viral promoter overactivated these genes, turning them into activated oncogenes. - Deletion and fusion events also captured proto-oncogenes into the viral genome. - Studying these viral fusion genes revealed which host genes could drive cancer.
58
role of transformation of normal mouse cells with human tumour DNA in identifying oncogenes
- human tumor DNA transformed normal mouse cells, causing them to grow uncontrollably. - By isolating the integrated human DNA, scientists identified specific mutated proto-oncogenes
59
oncogenic changes: constitutive activation - Bcr/c-abl
Bcr: signaling molecule involved in regulating cell growth and cytoskeletal organization. Abl: regulating cell differentiation, division, adhesion, and response to DNA damage. - normal chromosome 9 (containing abl) - normal chromosome 22 (containing bcr) - both chromosomes break and swap segments - changed chromosome 9 now does not contain abl, whilst changes chromosome 22 (Philadelphia Chromosome) contains bcr-abl which is now constitutively active -> cancer
60
oncogenic changes: constitutive activation - oncogenic forms of RAS
RAS is involved in the cell proliferation pathway - in a normal cell, the binding of a growth factor to a receptor activates RAS, causing proliferation - in a cancer cell, proliferation occurs even without the growth factor
61
oncogenic changes: constitutive activation - Her2 gene amplification
Her2 regulates cell growth - in normal cells, each cell has 2 copies of Her2 - in breast cancer cells, each cell has more than 2 copies of Her2, leading to multiple copies of the Her2 receptor on the surface of the cell and the cell being able to activate itself for cell growth
62
how can tumour suppressor genes be identified?
through genetic analysis of families with inherited predisposition to cancer
63
how does inheritance of mutant suppressor alleles lead to increased chance of cancer?
- one normal allele is sufficient for normal cell proliferation in heterozygotes - however, the wild-type allele in somatic cells of the heterozygote can be lost or mutated, leading to abnormal cell proliferation
64
events causing loss of heterozygosity in somatic cells of RB+/RB- individuals
- nondisjunction (loss) - uniparental disomy - mitotic recombination - gene conversion - deletion - point mutation
65
what are the usual functions of tumour suppressor genes whose mutations decrease the accuracy or rate of cell proliferation
1. negative regulators of cell proliferation 2. components of cell cycle checkpoints (before S phase, after G2) 3. involved in DNA damage repair 4. promote apoptosis when DNA damage is excessive
66
how do cell cycle checkpoints ensure genomic stability?
checkpoints monitor the genome and cell-cycle machinery before allowing progression to the next stage of the cell cycle
67
G1 to S checkpoint
DNA synthesis can be delayed to allow time for repair of DNA that was damaged during G1
68
G2 to M checkpoint
mitosis can be delayed to allow time for repair of DNA that was damaged during G2
69
spindle checkpoint
monitors formation of mitotic spindle and engagement of all pairs of sister chromatids
70
describe the role of p53
1. p53 is a transcription factor that is activated by UV or ionising radiation 2. induces expression of CDK inhibitor, p21 3. p21 inhibits the activity of CDK4 - cyclinD complexes 4. if these complexes are inhibited, then Rb is not phosphorylated 5. Rb remains unphosphoryalted so it can inhibit E2F, preventing entry into the S phase of the cell cycle
71
if Rb is phosphorylated
E2F is no longer inhibited, so the genes necessary for DNA synthesis are activated
72
how does loss of P53 function lead to genomic instability?
- inactive p53 means p21 is not induced - p21 cannot inactivated CDK4/cyclin D so this complex phosphorylates Rb - E2F is constitutively phosphorylated, causing the cell to enter the S phase - if mutations in the DNA have occurred, these are not repaired before mitosis
73
how does chemotherapy work?
targets proliferating cells: - disrupts DNA replication - prevents formation of the mitotic spindle
74
problem with chemotherapy
drugs used in chemotherapy target rapidly dividing cells; however, they also affect normal dividing cells, leading to adverse side effects
75
strategies to target the unique molecular properties of the cancer cells
- design drugs that specifically bind and inactivate oncogenic proteins - mobilise the immune system to eradicate cells that overexposes a particular oncogene
76
oncogene protein inactivation: Gleevec
- typically the Bcr/c-Abl enzyme binds to ATP and a target protein, leading to chronic myelogenous leukaemia - Gleevec binds to the ATP active site, inhibiting it
77
monoclonal antibodies to growth factor receptors: Herceptin
- typically, abundant Her2 in cancer cells leads to dimerization, turning the signal transduction pathway on and leading to cell proliferation - Herceptin attracts the immune system cells to attack the cancer cells. this prevents dimerization and recruits killer T cells, turning the signal transduction pathway off and stopping cell proliferation
78
strategies tailored for tumour suppressor genes
- many tumour suppressor gene products are involved in DNA repair/DNA damage checkpoints - in many cells excessive DNA damage induces programmed cell death strategy: utilise the ultra sensitivity of cancer cells to DNA damage to elicit self-destruction by inducing programmed cell death
79
strategies tailored for tumour suppressor genes: LOF-PARP inhibitors
PARP inhibitors block the action of poly(ADP-ribose) polymerase, needed to mend nicks in DNA without PARP, nicks in DNA -> double stranded breaks -> self-destruction by apoptosis
80
how are PARP inhibitors an example of tumour selective cytotoxicity?
in a normal cell with a functional homologous recombination (HR) pathway. there is HR-mediated DNA repair of the double-strand breaks, enabling cell survival. some cancer cells do not have this
81
why are PARP inhibitors not for all cancer cells?
- they increase genomic instability - in cells with dysfunctional apoptotic pathways, this may simply lead to the accumulation of new cancer associated mutations
82
personalised medicine
customising medicine by designing the most effective drug therapy and treatment strategies based on the specific genetic profile of a patient
83
how can we solve the problem of whole-genome sequences of cells derived from different regions of the same tumour having somewhat different genomes (heterogeneity)?
- possibly most effective treatments would be directed against the common (early_ mutations to target all the cells - help find the driver mutations and 'druggable' targets
84
how can we use immunotherapy to prevent the evasion of immune surveillance?
monoclonal antibody can prevent the interaction between tumour cells and T cells so that the T cell can no longer recognise the tumour cell
85
car-t cell therapies
Car T cells have a chimeric antigen receptor which can recognise the cancer cell
86
challenges with immunotherapy
- easier to implement for hematopoietic cancers (unique antigens) - can induce a cytokine storm (life-threatening)
87
How are all living forms related?
- evolution has conserved basic strategies of development across multicellular eukaryotes - many homologous proteins have highly conserved functions
88
give examples of homologous genes in different organisms
- eyeless (Drosophila), Pax-6 (mouse), and Aniridia (humans) genes are homologous - Pax-6 and Aniridia wild-type genes can direct eye development in Drosophila
89
distinguish between forward and reverse genetics
Forward: Phenotype → Find gene - Mutagenize, screen for trait, map gene - Use when you know the trait but not the gene Reverse: Gene → Find phenotype - Knockout/mutate gene, observe effect - Use when you know the gene but not its function
90
what is an example of mutagenesis screens in organisms?
- motor axon guidance mechanisms in Drosophila. - scientists mutagenise C. elegans, observe the phenotype, then try and identify the gene responsible for each phenotype
91
what is a modifier mutagenesis screen?
type of genetic screen used to find genes that influence the severity or expression of a known phenotype, rather than causing the phenotype on their own.
92
define redundancy
two or more genes perform overlapping or similar functions, so that loss of one gene has little or no effect on phenotype
93
What does the "modifier mutagenesis screen" using sev::Ras^G12V in Drosophila eyes reveal about genetic interactions?
- Wild type: Normal eye structure. - sev::Ras^G12V: Rough eye due to hyperactive Ras signaling. - Enhanced phenotype (E⁻/E⁺; sev::Ras^G12V): worsened mutant phenotype → E is a negative regulator of Ras. - Suppressed phenotype (S⁻/S⁺; sev::Ras^G12V): More normal eye → S is a positive regulator of Ras.
94
Use of Genome wide screens
- we can design RNAs that can target specific genes and inactivate them, similar to a knockout - this produces a phenocopy of a loss of function mutation
95
define a phenocopy
a phenotype that mimics the effects of a gene knockout, caused by methods like RNAi or CRISPR.
96
4 ways in which we can analyse developmental pathways
Nature of the encoded protein: - we can infer amino acid sequence from nucleotide sequence and use computer searches to identify known motifs Location and timing of gene expression: - during development, where and when is the mRNA found? Location of the protein product: - during development, where and when is the protein found? Developmental phenotypes: - what cells or tissues are affected by loss-of-function?
97
two methods to follow the localisation of proteins
- attach antibodies to the protein - tag the proteins with GFP
98
why must the interactions of genes in a developmental pathway be determined?
genes don't work in isolation, and many biological processes are complicated and require the coordinated action of many genes
99
two key steps to ordering genes in a pathway
analysis of effects of one gene on expression of another gene: - does a mutation in one gene affect the level or distribution of mRNA or protein from another gene? analysis of double mutants - epistatic interactions: - do mutations in two different genes define successive steps in a pathway?
100
give an example of the impacts that gene products can have on the expression of other genes
- staining of Drosophila wing imaginal disks for wingless protein (Wg, green) and vestigial protein (Vg, red) - wild-type produces a different staining pattern than wingless mutant - this suggests that the wingless gene product is required for the expression of the vestigial gene
101
2 requirements for an epistasis analysis:
- phenotypes of the two mutants must differ - alleles must either be null or constitutive
102
null allele
A mutant allele that results in complete loss of function of the gene.
103
constitutive allele
A mutant allele that causes the gene to be continuously active, regardless of normal regulatory signals.
104
use the sevenless signalling pathway as an example of epistasis analysis
- the phenotype of the double mutants reveals epistatic interactions - in a double mutant, the phenotypes the same as the Ras^G12V mutant, therefore seven less is upstream of Ras - the downstream gene is the one that is epistatic - thus, Ras is downstream and epistatic to sevenless ('masks' sevenless)
105
what is the programmed cell death pathway and how was it understood?
- eliminates unwanted or damaged cells - was understood by epistatic analysis screens
106
relationship between embryonic and adult body axes in drosophila
each embryonic segment defines a specific structure in the adult: 3 head segments, 3 thoracic segments, 8 abdominal segments
107
who made discoveries concerning the genetic control of early embryo development
Christiane Nusslein-Volhard, Eric Wieschaus, Edward B. Lewis
108
What two mutagenesis screens were conducted to identify genes that control embryonic development?
1. Screening for abnormal embryos in homozygous mutant females - Identified recessive mutations in maternal-effect genes 2. Screening for abnormal homozygous mutant embryos - Identified recessive mutations in three classes of zygotic segmentation genes
109
Four classes of genes responsible for segment formation in drosophila embryos
Function in a hierarchy that progressively subdivides the embryo into successively smaller units - maternal genes - gap genes - pair-rule genes - segment polarity genes
110
maternal genes
- expressed by the mother - mRNAs deposited in egg but not translated until after fertilisation - products of maternal effect genes establish morphogenic gradients in the egg - these gradients ensure that gap genes are expressed only in certain broad regions of the embryo
111
gap genes
- expression is controlled by maternal gene products - expression begins at syncytial blastoderm stage - activate pair-rule genes in a series of seven stripes
112
pair-rule genes
- seven zones of expression are controlled by gap and maternal gene products - levels of pair-rule gene products restrict the expression of segment polarity genes to a series of 14 stripes, one per segment
113
segment polarity genes
- expression in 14 segments is controlled by pair-rule gene products - this divides the embryo into 14 segment-sized units
114
what controls the anterior and posterior patterning in the drosophila embryo?
two maternal-effect gene products which are morphogens: bicoid (bcd) and nanos (nos)
115
define a morphogenetic
a substance that defines cell fate in a concentration-dependent manner
116
how do bcd and nos work?
- bcd and nos are transcribed by the mother and their mRNAs are localized to opposite poles of the oocyte - bcd and nos mRNAs are not translated in the embryo until after fertilisation - each protein forms a gradient in the embryos (diffusion)
117
bcd protein gradient
bcd is highest at anterior and lowest at the posterior
118
nos protein gradient
nos is lowest at anterior and highest at the posterior
119
localisation of bicoid mRNA and protein
- bcd mRNA localizes to the anterior pole of the oocyte - bcd protein diffuses from the anterior pole of the embryo to produce an anterior-to-posterior gradient
120
evidence that bicoid is the anterior morphogenetic
- dosage of maternal bcd gene determines how much of the embryo becomes head structures (the more protein in the anterior, the greater the size of the segment) - bicoid null mothers produce embryos that lack all head and thoracic structures
121
describe and draw the distribution of RNA and protein products of maternal-effect genes within the early drosophila embryo
- bicoid protein activates the expression of hunchback gene and represses translation of caudal mRNA - this causes posterior-to-anterior gradient of caudal protein - nanos protein represses the translation of hunchback mRNA - this causes anterior-to-posterior gradient of hunchback protein
122
how is segment number further specified?
by zygotic genes: - bicoid, hunchback, and caudal proteins are transcription factors that control the spatial expression of zygotic genes - zygotic gene expression begins in the syncytial blastoderm stage
123
three classes of zygotic segmentation genes
- gap genes - pair-rule genes - segment polarity genes
124
look at slide 30
125
two classes of pair-rule genes
primary pair-rule genes and secondary pair-rule genes
126
primary pair-rule genes
- expression is controlled by transcription factors encoded by maternal genes and zygotic gap genes - upstream region of each pair-rule gene has multiple binding sites for transcription activation/repression
127
secondary pair-rule genes
- expression is controlled by transcription factors encoded by other pair-rule genes
128
how are pair-rule genes expressed at the early blastoderm stage?
- in seven stripes - two segment periodicity: each stripe has two segments
129
stripe 2
eve transcription is activated by Bcd and Hb, but repressed by giant (Gt) and Kruppel (Kr) proteins
130
how is individual pair-rule stripe formation controlled?
by combinations of maternal-effect and gap proteins (see slide 32_
131
distribution of engrailed proteins in 14 stripes
- segment polarity genes are expressed in stripes that are repeated with single segment periodicity (one stripe per segment) - interactions between various polarity genes maintains the periodicity
132
many of the Drosophila A-P axis genes that contribute to pattern formation code for
transcription factors
133
genetic hierarchy leading to segmentation in drosophila
in successive levels of the hierarchy, genes are expressed in narrower bands
134
how is segment identity established by homeotic genes?
- transcription of homeotic genes is controlled by gap, pair-rule, and segmentation genes - at the cellular blastoderm stage, each homeotic gene is expressed within a subset of body segments - homeotic genes are master regulators that control transcription of many genes responsible for development of segment-specific structures
135
homeotic transformation
when one body part develops as if it were another, due to mutations in homeotic (Hox) genes, which control body plan identity during development.
136
example of a homeotic transformation in drosophila
Wild type wing vs bithorax mutant vs postbithorax mutant
137
how are hox genes expressed?
in spatially restricted domains
138
mammalian Hox genes are organised into four gene clusters:
each on a different chromosome: HoxA HoxB HoxC HoxD
139
how do we know that Hox genes play an important role in developmental patterning of all animals?
all animal genomes - even those of sponges - contain Hox genes
140
why is it not possible to just mutagenise the wild-type organism in a modifier mutagenesis screen?
if you just mutagenise the wild type, you may have a whole bunch of different mutations, but if there's redundancy you won't see the phenotype you are studying
141
what is the function of the sev gene in Drosophila?
sev is a receptor tyrosine kinase that activates Ras signaling to specify the R7 photoreceptor.
142
what is the function of the Ras gene in Drosophila?
When sev is activated by its ligand, it triggers a cascade that activates Ras, which in turn promotes cell differentiation (the specification of the R7 photoreceptor neuron).
143
what is Ras^G12V?
gain of function mutation - this mimics constant "on" signaling, so R7 cells develop even without the proper upstream signal, or develop abnormally, causing things like a rough eye phenotype.
144
define a maternal-effect gene
genes that are 100% inherited from the maternal side
145
What provides evidence for the importance of epigenetics?
genotype =/= phenotype - monozygotic twins are genotypical identical but not always phenotypically identical, and these differences can't always be explained by environment - mice with genotype A^vy/a genotype are also genetically identical, but some have brown fur and some have yellow fur
146
define epigenetic
the study of mitotically and/or meiotically heritable changes in gene function that cannot be explained by changes in DNA sequence
147
state the 4 molecular mechanisms underlying epigenetics
1. DNA methylation on CpG islands 2. covalent modification of histone tails 3. noncovalent modification of histones 4. non-coding RNAs
148
DNA methylation on CpG islands
CpG = Cytosine-phosphate-guanine nucleotides - some regions have many CpG islands, in which cytosine may be either methylated or unmethylated - typically, unmethylated promoters enable gene expression - typically, methylated promoters result in heterochromatin, repressing gene expression - the effects of methylation depend on the region; sometimes DNAm leads to silencing, sometimes it leads to increased expression
149
covalent modification of histone tails
- acetylation: loosens chromatin by neutralizing lysine charges, promoting gene expression - methylation: can activate or repress gene expression, depending on the site and number of methyl groups. - ubiquitination: tags histones for degradation or signals chromatin remodeling, affecting transcriptional activity
150
non-covalent modification of histones
reposition nucleosomes to make DNA more or less accessible.
151
non-coding RNAs
Transcriptional Silencing: Small RNAs (like siRNAs) guide chromatin modifiers to DNA, causing heterochromatin formation and blocking transcription.
152
how does chromatin structure affect transcription?
- promoters can be hidden when wrapped in nucleosomes, leading to lowered gene expression - chromatin remodelling complexes can expose gene promoters, allowing RNA polymerase to bind - nucleosomes in heterochromatin can be tightly packed, generating silenced heterochromatin
153
epigenetic changes causing coat colour differences can be influenced by
the environment
154
regulation of gene expression in A^vy/a mice
- the agouti gene is normally expressed in a regulated pattern, giving banded hairs (black-yellow-black) and brown/gray fur. - the Aᵛʸ allele contains an IAP (Intracisternal A Particle) retrotransposon upstream of the Agouti gene. - the IAP has a promoter that can drive ectopic (abnormal) Agouti expression. - Unmethylated IAP → promoter is active → overexpression of Agouti → yellow coat. - Methylated IAP → promoter silenced → Agouti expressed normally (or not at all) → brown or mottled coat.
155
define a metastable epiallele
an allele whose expression is not fixed but can vary in a stable way between cells or individuals due to epigenetic states established early in development.
156
how is A^vy an example of a metastable epiallele?
its expression is controlled by DNA methylation at a retrotransposon promoter, leading to variable, heritable gene expression without DNA sequence changes.
157
what two main points were derived from experiments involving agouti mice?
1. Maternal diet can directly affect the epigenetic regulation of genes in offspring. 2. These changes can be stable enough to affect not just the immediate offspring, but also subsequent generations.
158
How was the idea that maternal diet can directly affect the epigenetic regulation of genes in offspring understood?
- When pregnant mothers were fed a diet rich in methyl donors, their offspring showed a higher proportion of the pseudoagouti phenotype (brown fur). - This was because methylation silenced the Agouti gene, preventing ectopic expression and resulting in darker coat color. - When mothers were fed a normal diet, their offspring showed more of the yellow or mottled phenotypes. - This was due to lower methylation of the Agouti gene, allowing it to be expressed and leading to lighter coat color.
159
How was the idea of intergenerational epigenetic inheritance understood?
- F0 mothers were supplemented with methyl donors during pregnancy. - Her F1 offspring showed increased methylation at the Agouti locus. - The F2 generation also exhibited more pseudoagouti phenotypes, despite not being directly exposed to the diet. - This suggested that the epigenetic marks were inherited through the germline. - F0 mothers were supplemented with no methyl supplementation. - the F1 and F2 offspring showed more yellow or mottled phenotypes, indicating low methylation.
160
how is epigenetic required for normal development?
specific functions of different cell types are generated through differential gene regulation
161
explain the randomness of X-chromosome inactivation and its consequences
- random X-chromosome inactivation in females occurs early during development for dosage compensation - these X-chromosomes are reactivated in germ cells - inactivation of the paternal/maternal X chromosome is random but persists in the subsequent cells produced - this means that females express X^m in some cells and X^p in others, leading to clonal patches
162
give an example of X-chromosome inactivation and its phenotypic consequences in animals
in the calico cat, X-inactivation leads to a mosaic of fur colours
163
mechanism of X-chromosome inactivation
- process begins at the X-inactivation centre (XIC), which activates the gene Xist. - Xist makes a long non-coding RNA (lncRNA) that spreads across the X chromosome, coating it and starting the silencing process. - Hypoacetylation of a Lys of histones (H3/H4), methylation of histone H3 and underlying DNA shut down gene expression - the inactive X becomes tightly packed into a structure called a Barr body. - most genes on this X are turned off, but a few, called escapees, remain active.
164
how does histone acetylation lead to more loosely packed DNA?
- histone tail usually positively charged, DNA negatively charged - when the tail is acetylated, the charge is neutralised and DNA becomes more loosely packed
165
Hypoacetylation vs Hyperacetylation
Hypoacetylation = regions that are silenced Hyperacetylation = transcriptionally active regions
166
gene therapy approach for Down syndrome
autosome + one copy of Xist -> autosome becomes a heterochromatic Barr body thus, we could add Xist to one of the 3 copies of chromosome 21 in children with Down syndrome
167
what is a synonym for parental imprinting?
genomic imprinting
168
how does parental imprinting come about?
it results from transcriptional silencing
169
define parental imprinting
when the expression of a gene depends upon its parental source (ie whether it is inherited from the maternal or paternal side)
170
imprinted =
silenced
171
paternally imprinted gene
- paternally silenced - only the maternal allele is expressed
172
maternally imprinted gene
- maternally silenced - only the paternal allele is expressed
173
how are imprinted genes usually modified?
- methylated by special methylases - demethylated by demethylases
174
how is epigenetic state maintained across cell generations? draw a diagram
through the action of DNA methyltransferases, slide 22
175
define silencing
long term repression through DNA methylation
176
explain the resetting of genomic imprints during meiosis
- in the early primordial germ cells, existing methylation marks are removed. - later in gametogenesis, new methylation marks are added. - female places maternal imprints on all eggs - male places paternal imprints on all sperm
177
draw the model for the imprinting of the IGF2 and H19 genes
178
describe the model for the imprinting of the IGF2 and H19 genes
- single enhancer downstream of H19 gene controls the expression of both genes on maternal chromosome: genes and regulatory sequences are not methylated, so CTCF binds to insulator blocking activation of IGF2, but allowing for the activation of H19 on paternal chromosome: insulator and promoter region of H19 gene is methylated. CTCF cannot bind to insulator, and activator is able to active transcription of IGF2. - thus, due to mthe methylation of its promoter, the activator cannot active H19
179
draw the mechanism of IGFR2 as an example of noncoding RNA mechanism of imprinting
180
consequences of mutations in imprinted genes
- In regular genes, a mutation in one allele can often be compensated by the other normal allele. - In imprinted genes, since one allele is silenced, a mutation in the active allele acts like a dominant mutation - A mutation in the silenced allele is not expressed despite being present in the genome
181
look over pedigrees of mutations in imprinted genes
182
Prader-Willi syndrome symptoms
obese, small hands and feet, eats uncontrollably, does not mature sexually, short stature, mental retardation
183
Angelman syndrome
developmental delays, severe mental and motor retardation, prominent jaw, happy disposition
184
genetic cause of PWS
A deletion of the paternal copy of 15q11–q13. The same region on the maternal chromosome is silenced by imprinting.
185
genetic cause of AS
A deletion of the maternal copy of 15q11–q13. The paternal allele of a key gene, UBE3A, is silenced in neurons due to imprinting. No active UBE3A in neurons → leads to AS.
186
how does chromosome rearrangement represent a major feature of evolution?
1. rearrangement breakpoint may acquire new patterns of gene expression and create new gene functions by fusion of two separate genes 2. some rearrangements contribute to the process of speciation 3. duplications provide extra gene copies that can acquire new functions
187
state and draw the four classes of chromosomal rearrangements resulting from chromosome breakage and subsequent DNA repair
- deletion - inversion (180 degree rotation of a piece of DNA) - deletion in one chromosome, duplication in another - translocation of a piece of DNA into another chromosome
188
state and draw the four types of chromosomal rearrangements resulting from aberrant crossing over at repeated sequences
- deletion - inversion - deletion in one chromosome, duplication in another - reciprocal translocation of a piece of DNA into another chromosome
189
two types of deletions
1. intragenic: small deletion within a single gene 2. multigenic: many genes deleted
190
Del (Df) homozygotes
- short for deletion (deficiency) homozygote, is an individual that has both copies of the same chromosomal region deleted - usually inviable
191
Del (Df) heterozygotes
- gene imbalance - might result in haploinsufficiency
192
deletion loop
a DNA loop formed during meiosis when one homologous chromosome has a segment deleted. The extra DNA on the normal homolog that has nothing to pair with loops out.
193
pseudodominance
when a recessive allele is expressed in a heterozygous individual because the dominant allele has been deleted or is missing.
194
what is a practical application of deletions?
- deletions may uncover recessive mutations - they can be used to locate genes for mapping
195
deletion mapping: complementation
1. Start with a mutant strain that has a recessive mutation (e.g. mut) causing a known phenotype. 2. Cross it with a strain that carries a known deletion of part of the chromosome (e.g. Df1). 3. Examine the phenotype of the offspring (heterozygotes: mut / Df1): - If the offspring show the mutant phenotype, then the deletion likely removes the same gene as the mutation → No complementation. - If the offspring show the wild-type phenotype, the mutation must lie outside the deleted region → Complementation occurs.
196
two main types of duplications
tandem duplications: the duplicated segment is inserted right next to the original. non tandem (dispersed) duplications: the duplicated segment is inserted elsewhere in the genome, not adjacent to the original.
197
impact of duplications
- less likely to affect phenotype - in some cases causes a dosage effect/genetic imbalance - genes may be placed in a new location that modifies their expression
198
how do duplications arise?
1. X-ray breaks/any other cause of breaks: - X-rays break one chromosome in two places - X-rays break homologous chromosome in one place - during repair, the freed segment from the first chromosome is mistakenly inserted at the break site on the homolog -> non tandem duplication 2. Unequal crossing over: - Homologous chromosomes misalign during meiosis. - Crossing over occurs at these misaligned points. - One chromosome gains extra DNA (duplication), the other loses it (deletion).
199
duplications can result in
unequal crossing over, causing increase and reciprocal decrease in the number of copies (eg Bar-eye in drosophila)
200
potential impacts of inversions
- most inversions to not alter phenotype unless breakpoints occur within genes - but genes may be placed in a new location that modifies their expression (eg Antennapedia)
201
two main types of inversion
1. pericentric inversion - includes the centromere 2. paracentric inversion - does not include the centromere
202
breakpoints between genes
- Genes remain intact. - Order of genes is reversed in the inverted segment. - Usually no gene disruption or loss of function. - May affect gene expression if regulatory regions are affected.
203
breakpoints within ONE gene
- The gene is disrupted (split) and mutated - Usually causes loss of gene function or creates a truncated protein. - Can lead to a nonfunctional or altered gene product.
204
breakpoints within TWO genes
- Both genes are disrupted at the breakpoints. - May create fusion genes by joining parts of two genes. - Can produce novel or dysfunctional proteins. - Often causes loss of function or gain of abnormal function.
205
inversion loops
- form in inversion heterozygotes - enables pairing of homologous regions despite the reversed gene order. - produces abnormal recombinant chromosomes.
206
paracentric inversion loop
Normal chromosome + Inversion chromosome. Inversion loop forms outside the centromere: - if crossing over occurs inside this loop, it produces one dicentric chromosome (with two centromeres) and one acentric fragment (without a centromere) - the acentric fragment is lost - there is a random break in the dicentric bridge of the dicentric fragment Results in: one normal product, two deletion products, and one inversion product with all genes present. Reduced number of viable gametes
207
pericentric inversion loop
Normal chromosome + inversion chromosome. Inversion loop includes the centromere: - if crossing over occurs inside this loop, it results in gene imbalance - one normal product, two different inviable deletion/duplication products, one viable inversion product (all genes present). Reduced number of viable gametes
208
look over how to find the possible gametes arising from a paracentric and pericentric inversion
209
What are balancer chromosomes and why are they useful?
- engineered chromosomes used in genetics that carry multiple inversions and sometimes other rearrangements. - these prevent crossing over from happening during meiosis
210
translocations
- most translocations do not alter phenotype unless breakpoints occur within genes - but genes may be placed in a new location that modifies their expression
211
three types of translocations
a) nonreciprocal intrachromosomal translocation b) nonreciprocal interchromosomal translocation c) reciprocal interchromosomal translocation
212
cause of chronic myelogenous leukaemia
- arises from a specific reciprocal translocation between chromosomes 9 and 22 - this translocation fuses the BCR gene from chromosome 22 with the ABL gene from chromosome 9, creating the BCR-ABL fusion gene. - the BCR-ABL fusion produces a constitutively active tyrosine kinase protein that drives uncontrolled cell division and the malignant proliferation of white blood cells seen in CML.
213
3 types of segregation patterns occurring in heterozygotes during meiosis
- alternate - adjacent-1 - adjacent-2
214
draw a diagram and table for alternate segregation pattern
215
draw a diagram and table for adjacent-1 segregation pattern
216
draw a diagram and table for adjacent-2 segregation pattern
217
consequences of different segregation patterns
- semiysterility since <50% of the time there are viable gametes - pseudo linkage since genes can't independently assort - only alternate segregation produces viable progeny
218
robertsonian translocation
reciprocal exchange between acrocentric chromosomes generates a large metacentric chromosome and a small chromosome (which may be lost)
219
robertsonian translocation and Down syndrome
- a parent carries a Robertsonian translocation involving chromosome 21 and another acrocentric chromosome (commonly chromosome 14). - this carrier has 45 chromosomes but is usually healthy because they have all the essential genetic material. - however, during gamete formation, abnormal segregation can lead to a child inheriting two normal chromosome 21s plus the translocated chromosome containing an extra copy of 21 material. - this causes trisomy 21, the genetic cause of Down syndrome. table
220
methods of detection of chromosomal rearrangements
Fluorescent in situ hybridisation (FISH) - FISH Karyotype - Multicolour banding
221
FISH karyotype
- uses fluorescent probes that bind to specific DNA sequences or regions on chromosomes. - detects presence, absence, or location of specific genes or chromosomal regions. - useful for identifying known abnormalities (e.g., deletions, duplications, translocations). - shows bright fluorescent signals on chromosomes under a microscope.
222
Multicolour Banding
- specialized form of FISH that uses multiple probes along a single chromosome to create a unique banding pattern with different colors. - allows high-resolution analysis of structural chromosome rearrangements. - can distinguish small intrachromosomal changes like inversions or complex rearrangements. - provides a detailed “barcode”-like pattern for precise mapping.
223
chromosomes from normal cells vs chromosomes from tumour cells
chromosomes from tumour cells may be present in larger copies
224
detection of chromosomal rearrangements by PCR
- fast - inexpensive - highly sensitive
225
give a real world example of how translocations can contribute to speciation
- house mice in the island of Madeira - different Madeira mouse populations have unique Robertsonian translocations, forming distinct chromosomal races. - reduced fertility of heterozygotes for translocations can contribute to reproductive isolation and promote speciation
226
two main themes underlying the observations on chromosomal changes
1. karyotypes generally remain constant within a species - most genetic imbalances result in a selective disadvantage 2. related species usually have different karyotypes - closely-related species differ by only a few rearrangements - distantly-related species differ by many rearrangements - correlation between karyotypic rearrangements and speciation
227
x =
number of different/unique chromosomes that make up a single complete set eg 23 in humans
228
n =
number of chromosomes in a gamete eg 23 in humans
229
relation between n and x in diploids
n = x - each gamete contains a single complete set of chromosomes
230
relation between n and x in hexaploids
n = 3x
231
euploidy
A condition where a cell has a complete set(s) of chromosomes.
232
aneuploidy
A condition where a cell has an abnormal number of individual chromosomes, not involving whole sets.
233
give 4 examples of euploid
monoploidy (x) diploidy (2x) triploidy (3x) tetraploidy (4x)
234
using diploid species as a basis, explain: - euploidy - nullisomy - monosomy - trisomy
euploidy: 2n nullisomy: 2n-2 monosomy: 2n-1 trisomy: 2n+1
235
monoploidy
- male bees, wasps, and ants - they undergo parthenogenesis - usually lethal in other organisms
236
parthenogenesis
development of unfertilised egg into an embryo (with no fertilisation): - single set of chromosomes - produce gametes by mitosis
237
why is monoploidy lethal in most organisms?
- unmasks recessive lethal alleles (eg X-linked diseases) - if an individual survives to adulthood, this most likely leads to sterility
238
uses of monoploid plants
- visualize recessive traits directly - introduction of mutations
239
how can we create a monoploid plant?
1. take a diploid plant 2. haploid pollen grains are treated and plated onto agar 3. growth of haploid embryoids 4. embryoids treated with plant hormones 4. monoploid plant (usually sterile)
240
what is the issue with having a monoploid plant?
- in meiosis, chromosomes are supposed to pair up and separate. - in monoploids, there's nothing to pair with, so meiosis is disrupted, leading to infertile gametes.
241
how is colchicine useful to scientists?
- colchicine inhibits the formation of the mitotic spindle - thus the plant cells become diploid and meiosis is able to occur normally
242
in what organisms is polyploidy particularly common?
in plants
243
tetraploidy in plants
alfalfa, coffee, peanuts, large apples, pears, grapes
244
octoploidy in plants
large strawberries
245
results of polyploidy in plants
- associated with origin of new species - may positively correlate with size and vigor
246
two types of polyploids
- autopolyploids - allopolyploids
247
autopolyploids
- originate within a species - all polyploids with an odd number of chromosome sets are sterile because they cannot produce balanced gametes, producing aneuploid gametes
248
when can autopolyploids with an odd number of chromosome sets produce balanced gametes?
- if x is small - balanced gametes are only produced if two copies of each chromosome always segregate to the same daughter cell and the third to the other
249
generation of autotetraploids
- when the 2x genome of a diploid is doubled to 4x, with all four sets coming from the same species - could be spontaneous or induced by a drug such as colchicine - often the source of a new species
250
how are balanced gametes generated in autotetraploids?
- each chromosome has three homologs to choose from - in order to form balanced gametes, the four copies of each group of homologues must form two bivalents - successful tetraploids produce balanced 2x gametes and are fertile
251
allopolyploids
- hybrid of two or more closely-related species - partially homologous chromosomes
252
amphidiploid
doubled diploid: contains two different diploid genomes
253
amphipolyploids in agriculture
F1 hybrid of wheat and rye (triticale) is sterile because there are no pairing partners for the rye chromosomes
254
describe the different triticale hybrids that have been generated
- some combine high yield of wheat with ability of rye to grow in unfavourable environments - some combine high level of protein from what with high level of lysine from rye
255
4 examples of aneuploidy in sex chromosomes
XXY, XXX, XO, XYY
256
draw diagrams for nondisjunction during the first and second meiotic division, as well as after fertilisation by a normal gamete
257
what are the potential consequences of mitotic nondisjunction?
can result in a mosaic - am individual has two or more populations of genetically different cells in their body.
258
gynandromorph
an organism that has both male and female physical characteristics — often with distinct regions of male and female tissues
259
two types of mitotic nondisjunction
1. mitotic nondisjunction 2. mitotic chromosome loss
260
why do fertile aneuploids generate aneuploid progeny?
offspring of fertile aneuploids have an extremely high chance of aneuploidy because of production of unbalanced gametes
261
aneuploidy in the human population
- incidence of abnormal phenotypes caused by aberrant chromosome organisation or number is 0.4% - half of spontaneously aborted foetuses have chromosome abnormalities - incidence of abnormal phenotypes caused by single-gene mutations is 0.01%
262
monosomy in humans
2n-1; usually lethal in utero in humans, but there are a few exceptions: - Monosomy 21: born with severe multiple abnormalities but die shortly after birth - Turner Syndrome (XO): 99% of affected foetuses are not born. Those who are born have developmental abnormalities
263
defining characteristics of Turner Syndrome
short stature, sterile due to rudimentary ovaries and lack of menstruation
264
X inactivation occurs in XX individuals, so why are there abnormalities in XO individuals?
- embryogenesis: X inactivation occurs after several rounds of cell division, so it is thought that the 2nd X chromosome has important function within the first hundred divisions - some of the genes on the 'inactivated' X chromosome are expressed - germ-line: X chromosome reactivation
265
trisomy in humans
2n+1: often lethal in animals owing to chromosome imbalance
266
trisomy 21
Down syndrome - females can be fertile - males infertile - average life expectancy is 40-60 years due to congenital heart disease
267
trisomy 18
Edward syndrome - severe physical and mental abnormalities - heart defects, growth retardation, small jaw, kidney abnormalities, narrow pelvis, rocker bottom feet, clenched fists - average life expectancy of a few weeks - 1/6000 to 1/10000 live births
268
trisomy 13
patau syndrome - severe physical and mental abnormalities - major abnormalities of heart kidneys, brain, face and limbs, small or absent eyes, harelip, small malformed head - average life expectancy of 130 days - 1/12500 to 1/21700 live births
269
three types of sex chromosome trisomies
XYY XXX XXY (Klinefelter syndrome)
270
Klinefelter syndrome
1/1000 male births - humans tolerate X chromosome aneuploidy because of X inactivation - sterile
271
why can aneuploidy for X have phenotypic consequences?
some X-linked genes escape inactivation — especially those in the pseudoautosomal regions (PARs). One key gene, SHOX, is essential for bone growth. Loss (like in Turner syndrome) or gain (like in XXX or XXY) of SHOX copy number leads to short or tall stature, respectively.
272
prenatal diagnostic testing
- look for abnormal karyotypes - possible to screen for biochemical and genetic disorders - tests are done in combination with blood tests for certain maternal and fetal proteins, and with ultrasound tests
273
prenatal testing - screening tests
first trimester screening test (11 to 13 weeks) ultrasound (nuchal translucency) and maternal blood test [pregnancy associated plasma protein-A (PAPP-A) and β-human chorionic gonadotrophin (β-hCG)]
274
prenatal testing - diagnostic tests
- chorionic Villi Sampling, CVS (10 to 13 weeks) - amniocentesis (16+ weeks), less invasive
275
which comes first - screening tests or diagnostic tests
screening tests - if there are abnormalities, we then conduct diagnostic tests.
276
define transposition
movement of small segments of DNA called transposable elements from one position to another in the genome
277
who discovered transposition?
Barbara McClintock in the late 1940s, awarded Nobel prize in 1983
278
define a transposable element (TE)
any segment of DNA that evolves the ability to move within a genome
279
how did Rhoades and McClintock infer the existence of TEs?
from genetic studies of corn
280
where are TEs found?
in all organisms
281
how has our understanding of the impact of TEs varied over the years?
- previously considered to be selfish DNA carrying no genetic information useful to the host - now it is known that some TEs have evolved functions that are beneficial to the host sometimes
282
TE length ranges from
50bp to 10kb
283
how many copies of TEs can be present in genomes?
TEs can be present in hundreds of thousands of copies per genome
284
how did McClintock realise the presence of TEs in corn?
- some kernels showed a mottled coloration, rather than a uniform color - the mutant phenotype was colourless, whereas the wild type phenotype was red - this pattern could not be explained by standard Mendelian inheritance, since all cells in a kernel should theoretically have the same genotype and produce the same pigment. - this suggested that gene expression was being turned on and off during development, pointing to the movement of genetic elements within the genome.
285
What is the C mutable (c-m) element in maize?
an unstable version of the C gene responsible for kernel pigmentation. It causes mottled color due to the insertion of a transposable element that disrupts gene function.
286
What phenotype does the C mutable element cause and why?
It causes colorless kernels with red spots, because the pigment gene is inactivated by an inserted Ds element. If Ac is present, it enables Ds to jump out, restoring gene function in some cells and producing spots of pigment.
287
What role does the Ac (Activator) element play in C mutable pigmentation?
Ac is required for Ds to transpose. If Ac is present, it allows the Ds element to move out of the C gene, restoring pigment production in some cells.
288
What is the difference between Ac and Ds elements?
Ac (Activator) is autonomous and can transpose on its own. Ds (Dissociator) is non-autonomous and needs Ac to move.
289
If Ac can move, why doesn't kernel color keep changing throughout the kernel’s life?
Kernel color is set early in development, when cells are still dividing. Ac must be present in the same cell as Ds during this time to make Ds jump out of the C gene and restore pigment. Once the kernel matures, cells stop dividing and pigment production is fixed, so even if Ac moves later, it can’t change the color anymore.
290
TEs in bacteria
several types, inserted several times
291
TEs in drosophila
approximately 12.5% of the genome
292
TEs in humans
44% of the genome
293
two main classes of TEs in eukaryotes
DNA transposons Retrotransposons
294
DNA transposons
move directly without being transcribed into RNA (eg TEs studied by McClintock in corn, P elements in Drosophila)
295
Retrotransposons
move via reverse transcription of an RNA intermediate (eg copra elements in Drosophila, L1 and Alu in humans)
296
further classification of retrotransposons
LINEs, SINEs, HERVs
297
LINEs
encode their own reverse transcriptase and endonuclease, allowing them to copy and insert themselves into new genomic locations.
298
SINEs
non-autonomous retrotransposons that rely on LINE enzymes to move - they do not encode any proteins
299
HERVs
ancient LTR retrotransposons that originally encoded enzymatic components but are mostly now inactive due to mutations
300
what do DNA transposons encode?
an enzyme called transposase that helps them cut and move to different places in the DNA.
301
how was it experimentally demonstrated that retrotransposons move through an RNA intermediate?
An intron was added to a yeast Ty1 retrotransposon (located on a plasmid and flanked by LTRs). After splicing and reverse transcription, the DNA inserted into the yeast's genomic DNA lacked the intron, proving the RNA (not DNA) was the template.
302
How do LTR retrotransposons move within the genome?
Target-site duplication: - they are transcribed into RNA, which is reverse-transcribed into cDNA by reverse transcriptase. This cDNA is inserted into a new genomic site via endonuclease cleavage, generating target-site duplications flanking the new insertion.
303
transposon structure
most transposons contain two components: - inverted repeats (IRs) of 10-200 bp long at each end - gene encoding transposase, which catalyses the transposition through recognition of the IRs (cuts at border between the IR and genomic DNA)
304
how do P element transposons move?
P element excised from its original location and is transposed to new location by transposase enzyme
305
what are the two ways in which the original location where the P element was is repaired?
in both cases, exonucleases first widen the gap. Then there is: - repair of gap using a sister chromatid or homologous chromosome containing a P element, resulting in the transposon remaining in its original position OR - repair of gap using a homologous chromosome lacking a P element, resulting in the transposon no longer being at the original position
306
how do animals and plants survive with so many mobile elements?
- these are usually inserted into introns - often defective and unable to transpose again due to mutations (ie lack of repeats or active transposase) - epigenetic changes from heterochromatin, resulting in no transcription/transposition - inserted into safe havens (non-essential/repetitive regions)
307
cellular mechanisms that inhibit TE activity
- production of transposon repressors through alternate splicing - piRNAs (kiwi-interacting RNAs) that block TE transcription and translation (both DNA transposons and retrotransposons)
308
consequences of transposable elements on organisation and function of genes and chromosomes
- unequal crossing over can occur between TEs found in slightly different locations on homologous chromosomes due to misalignment - recombination between them can lead to duplications, deletions, or inversions of the DNA between them.
309
Bar eye
- bar gene product limits eye growth - its enhancers increase transcription of the gene Bar (one duplication of gene + enhancer): reduced expression leading to bar eye Double Bar (two duplications of gene + enhancer): even more reduced expression
310
how can transposition relocate genes?
- two transposons can form a large, composite transposon - composite transposon can move to new location - the level of expression of the gene between them may be altered
311
3 types of transposons
1. insertion sequence (IS) element: IR - transposase - IR 2. complex transposon (IS + genes): IR - transposase - gene - IR 3. composite transposon (two transposons) - IR - transposase - IR - gene A - gene B - IR - transposase - IR
312
give examples of gene mutations caused by TEs
- wrinkled pea mutation likely resulted from insertion of a TE near the sbe1 gene (starch branching enzyme) - ~100 human genetic diseases i.e. forms of Hemophilia A, Hemophilia B, cystic fibrosis, neurifibromatosis, muscular dystrophy - many spontaneous mutations in the white eye gene (Drosophila)
313
define a mutation
change genes from one allelic form to another, sometimes leading to the creation of entirely new alleles
314
when do genes mutate?
genes mutate randomly, at any time and in any cell of an organism
315
how often do genes mutate?
very rarely; on average 1.2x10^-8 mutations/gene/gamete in humans
316
what causes a gene to mutate?
spontaneous: - arise in absence of known mutagen (polymerase errors, reactive oxygen species etc) - provide 'background rate' of mutation induced (by geneticist -> mutagenesis) - action of mutagen alters nucleotide sequence
317
what types of mutations can and can't be passed onto progeny?
only mutations in germline cells can be transmitted to progeny; somatic mutations cannot be transmitted
318
inherited mutations appear as ---- in populations
alleles
319
state 3 observations of mutation rates
- mutations affect phenotype rarely - different genes mutate at different rates (mutation rates range from less than 10^-9 to more than 10^-3/gene/gamete) - mutation rate can increase after exposure to a mutagen (eg UV light, certain chemicals)
320
define a substitution
a base is replaced by one of the other 3 bases
321
indels
deletions - block of one or more nucleotide (base) pairs is lost insertion - block of one or more nucleotide (base pairs is added)
322
what are the two types of base substitutions?
transition: purine to purine (A<->G) or pyrimidine to pyrimidine (C<->T) transversioin: purine to pyrimidine or pyrimidine to purine
323
purines
guanine, adenine
324
pyrimidines
thymine, cytosine
325
why do we study mutations?
- they act as markers for genes - mutations can disrupt gene function. this allows for the study of how the wild-type gene works
326
wild-type allele
the form found in nature (or in a standard laboratory stock); an allele whose frequency is 1% or more of the population
327
mutant allele
the form that has changed due to a mutation; an allele whose frequency is less than 1% of the population
328
forward mutation
changes wild-type allele to a different allele
329
reverse mutation (reversion)
causes novel mutation to revert back to wild-type allele
330
which is higher - rate of forward or rate of reverse mutation
rate of forward mutation is almost always higher than rate of reverse mutation (except TEs)
331
depurination
the hydrolysis of a purine base from the deoxyribose-phosphate backbone, leading to an apurinic site
332
how often does depurination occur?
100 times/hour in every human cell
333
deamination
the removal of an amino group from a cytosine, leading to the conversion of cytosine to uracil
334
how is deamination fixed?
by uracil-DNA glycosylase - most of the time, it is easy for cells to recognise deamination as uracil is only meant to be in RNA, not DNA
335
how does deamination affect DNA strands resulting from replication?
- after replication, one of the strands will be normal (CG) and the other will be mutant (UA) as adenine base pairs with uracil - after another round of replication, one of the strands will be (TA) and the other will be (UA) overall, there is a C-G to T-A transition mutation
336
give examples of radiation that may cause mutations
naturally occurring radiation such as cosmic rays and x rays often lead to mutations like deletions
337
what kind of mutation does UV light cause and how?
- UV light (especially UV-B and UV-C) primarily causes pyrimidine dimers - induces covalent bonds between adjacent pyrimidines (especially thymine-thymine dimers, but also C-T and C-C).
338
describe how oxidation leads to mutation
- oxidative damage can occur to any of the 4 base pairs - ROS oxidize guanine into 8-oxoG. 8-oxoG mispairs with adenine during replication. This causes a G:C → T:A transversion in the next round of replication.
339
describe how DNA polymerases may lead to mutation
- DNA polymerases may lead to DNA replication mistakes - this is caused by the incorporation of an incorrect during replication
340
how often do DNA replication mistakes occur?
DNA polymerase has very high fidelity, so such errors are exceedingly rare
341
where do indel mutations tend to occur?
in regions of repeated bases during replication or crossing over in meiosis
342
trinucleotide repeat expansion
DNA polymerase can slip or misalign on the template strand, forming a loop of extra repeats on the new strand, leading to expansion
343
trinucleotide repeat contraction
DNA polymerase can slip or misalign on the template strand, forming a loop of extra repeats on the template strand, leading to contraction
344
give examples of diseases caused by intel mutations of regions with repeated bases
Fragile X-syndrome, Huntington and other disorders of the nervous system
345
define genetic code
- dictionary used to translate nucleic acids to a amino acids - codon language - redundant (multiple codons per amino acid)
346
why is redundancy of the genetic code important?
this protects against the negative impacts of mutations
347
silent (synonymous) mutation
altered codon resulting from the mutation specifies the same amino acid
348
missense mutation
altered codon resulting from the mutation specifies a different amino acid
349
southern blot
identifies specific DNA sequences
350
northern blot
identifies (m)RNA sequences
351
western blot
identifies protein sequences
352
two types of missense mutations
conservative: - substitutes chemically similar amino acid - less likely to alter function or structure of protein non-conservative: - substitutes chemically different amino acid - more likely to alter function or structure of protein
353
nonsense mutation
introduction of a (early) stop codon (TAG, TAA, TGA)
354
frameshift mutation
- the insertion or deletion of nucleotides in a DNA sequence not in multiples of three - shifts the reading frame used during translation, drastically altering the resulting protein
355
intragenic suppressor mutation
a second mutation that occurs within the same gene as a previous mutation and restores, at least partially, the original function that was lost or disrupted by the first mutation
356
two types of mutations outside the coding sequence
- mutations in promoter or termination signal sites - splice donor/acceptor site mutations
357
splice donor/acceptor site mutations
- disrupts splice donor/acceptor site, resulting in incorrect retention/excision of intron - often leads to large additions or deletions that may cause frameshift
358
loss-of-function mutation
result in reduced or abolished protein activity
359
what is the typical inheritance mode of loss of function mutations?
loss-of-function mutations are usually recessive
360
two types of loss-of-function mutations
- null (amorphic) mutations - completely block function of gene product (eg deletion of an entire gene) - hypomorphic mutations - gene product has weak, but detectable, activity
361
haplosufficiency
- when one WT allele reaches threshold for the WT phenotype, the WT allele is haplosufficient - even with one mutant allele, we still see the WT phenotype - recessive mutations
362
haploinsufficiency
- when one WT allele does not reach the threshold for the WT phenotype, the WT allele is haploinsufficient - heterozygotes are affected - dominant mutations
363
give an example of haploinsufficiency
polydactyly - phenotype is seen in individuals with only one mutant copy
364
incomplete dominance
phenotype varies with the amount of functional gene product
365
gain of function mutations
enhance a function or confer a new activity
366
what mode of inheritance do gain of function mutations typically have?
they are typically dominant
367
two types of gain-of function mutations
hypermorphic mutations: - generate excessive gene product or more potent gene product neomorphic mutations: - generate gene product with new functions or ectopically expressed at inappropriate time or place, generating a novel phenotype
368
give an example of a hypermorphic mutation
achondroplasia - caused by a hypermorphic allele of the FGFR3 gene, which leads to increased receptor activity. - this overactive FGFR3 inhibits bone growth, resulting in the characteristic short stature.
369
give an example of a neomorphic allele
mutation in Antennapedia gene of drosophila causes ectopic expression of a leg-determining gene in structures that normally produce antennae
370
antimorphic/dominant negative mutations
- usually occur in genes that encode multimeric proteins - produce a mutant gene product that interferes with the function of the normal (wild-type) protein
371
How did Garrod use mutations to understand gene function in phenylketonuria (PKU)?
- proposed that genes control metabolism by encoding specific enzymes. - In PKU, a mutation in the gene for phenylalanine hydroxylase leads to a loss of enzyme function, causing a buildup of phenylalanine. - This showed that mutations can block metabolic pathways, linking gene defects to biochemical function
372
compare a normal pathway to alkaptonuria
normal pathway: phenylalanine -> tyrosine -> p-hydroxyphenylpyruvate -> homogentisic aid (HA) -> maleylacetoacetic acid -> CO + H2O in alkaptonuria: - HA oxidase nonfunctional - HA accumulates (toxic) - turns urine black in air - pathway stops
373
how can we analyse biosynthetic pathways?
- compounds that are used latest in the pathway will support the growth of the most mutants - compounds that are used earliest in the pathway will support the growth of the fewest mutants
374
define a genome
the total digital information contained within the DNA sequences of an organism's chromosomes
375
the human genome is ----- nucleotides
3 billion
376
define genomics
the branch of biology dedicated to the study of the whole genomes (both human and not human)
377
define bioinformatics
the science of using computational methods to analyse biological information
378
define bioprospecting
harnessing the function of newly discovered genes/identifying more targets for therapy
379
why do we study genomes?
- allows genes and mutations of interest to be identified much more rapidly and easily - opens up prospects for larger-scale, more complete understanding of how genes interact for biological functions (systems biology)
380
how was the human genome sequenced?
through the publicly funded human genome project (HGP)
381
what were the two people and methods that were used to sequence the human genome?
- Francis Collins 'Hierarchical Shotgun' - J. Craig Venter Celera Genomics Parallel (whole-genome shotgun)
382
hierarchical shotgun
- first determine the physical location of large pieces of DNA (large-insert clones) - sequence these pieces
383
Whole-genome shotgun
- Fragment the genome into many small pieces. - Sequence all fragments to get short reads. - Assemble overlapping reads into contigs. - Use paired-end reads to link contigs across gaps. - Build scaffolds from connected contigs. - Use paired reads to help resolve repeats and place contigs in the correct order and orientation.
384
what were the achievements that resulted from the human genome project?
1. sequencing the human genome 2. advancements to sequencing technologies
385
Moore's Law
computing power doubles every 18 months
386
how did the HGP relate to Moore's law?
advances in sequencing technology (resulting in lowered cost) outpaced Moore's Law
387
what 8 things did we learn from the HGP?
- the human genome consists of approximately 3.1 billion base pairs - the genome is approximately 99.9% the same between individuals of all nationalities - single nucleotide polymorphisms (SNPs) and copy number variations (CNVs) account for much of the genome diversity identified between humans - less than 2% of the genome codes for genes - the vast majority of our DNA is non-protein coding, and repetitive DNA sequences account for at least 50% of the noncoding DNA - the genome contains ~20,000 protein-coding genes - nearly 50% genes do not yet have a function - high degree of conservation between species
388
why is the species relatedness and genome conservation between humans and other vertebrates important?
because it means we can use other animals as model organisms
389
how can we overcome the limitations associated with relying on pedigrees?
one can do association studies without paying attention to pedigrees, just by treating groups of individuals that have a trait as though they are related for that trait
390
define a genome-wide association study, or GWAS
the application of SNP genotyping to large populations of people for the purpose of discovering genetic associations between particular SNPs and traits
391
what has GWAS led to?
SNPs being identified as tightly linked to, or playing causative roles in, a range of common diseases such as breast cancer, diabetes, Crohn's etc
392
what are the advantages of GWAS studies as opposed to pedigree analysis?
- GWAS studies are more broadly applicable and provide greater power and resolution than traditional pedigree analysis - GWAS studies do not depend on the analysis of closely related family members. there is no limit to the number of humans that can be included in a GWAS test population - direct comparative studies between affected and unaffected can be performed. we can also map and identify traits' associated genes that follow any pattern of inheritance, simple or complex
393
describe the gene for body size in dogs found by GWAS
IGF1 encodes a hormone involved in juvenile growth in mammals and is the major contributor to the difference in size between small and large breeds of dogs
394
GWAS catalogue contains:
- 7255 studies - 808, 580 unique SNP-trait associations
395
HGP -> SNPs -> Ancestry
fragments of genomes carried by our distant ancestors can be observed as blocks of DNA called haplotypes that are shared between many 'unrelated' people who are in fact distant relatives
396
what is an application of the HGP?
we have the power to develop personalised drugs for someone with a particular genetic disease or drugs that alter the genome
397
describe how a GWAS would be carried out
1. take a group of patients with a disease, and a group of non-patients who don't have the disease 2. take their DNA and detect disease-specific SNPs by comparing the differences within the two groups 3. use linkage disequilibrium (higher frequency of co-segregation of marker and trait to identify locus of interest)
398
list the challenges in mapping complex traits
- incomplete penetrance - phenocopy - locus heterogeneity - polygenic determination
399
incomplete penetrance
an individual carrying the disease genotype may not express the disease phenotype
400
give an example of incomplete penetrance
only 66% of individuals with a mutant BRCA1 allele will develop cancer by the age of 55
401
phenocopy
disease phenotype can be expressed by an individual who does not have the disease genotype, caused by environmental conditions
402
give an example of phenocopy
3% of women still develop breast cancer by age 55 from somatic mutations in the breast cells themselves
403
locus heterogeneity
mutations in more than one locus cause the same phenotype in different families: different disease genotypes are responsible for the same phenotype
404
give an example of locus heterogeneity
BRCA1 (chromosome 17) and BRCA2 (chromosome 13) both predispose to breast cancer
405
polygenic determiantion
mutant alleles at more than one locus influence expression of the disease phenotype in a single individual
406
define Quantitative Trait Loci (QTL)
loci that influence quantitative traits
407
a quantitative effect can be attributed to
2 or more important genes
408
how are QTLs identified?
- using genetic mapping and association of molecular markers with the trait - statistical methods allow the identification of more than one QTL at a time
409
state the 5 steps that are carried out in order to dissect QTLs
1. make an informative cross between individuals which differ at the trait(s) of interest. This step creates a population with a range of phenotypes that can be studied. 2. determine the frequency distribution in the F2. This step shows how variable the trait is and suggests how many genes may be involved (more genes = smoother distribution). 3. use molecular markers to genotype the individuals, attempting to find markers that co-segregate within the trait. 4. use a statistical method to determine if markers are co-segregating (associating) with the trait or not. 5. plot the degree of association (LOD score) on a linkage map. This tells you where in the genome the important genes affecting the trait are located.
410
RFLP as a molecular marker
RFLP - Restriction Fragment Length Polymorphism - a type of molecular marker based on SNPs - if one allele has a restriction site, the enzyme cuts there - if the other allele lacks the site, it produces fragments of different lengths. - these fragment patterns are detected using gel electrophoresis and probes.
411
STR analysis as a molecular marker
STRs - Short Tandem Repeats - alleles vary in the number of STRs that they contain - electrophoresis can be used to separate different alleles based on size
412
describe how different alleles will move through an electrophoresis gel
- longest/heaviest alleles move the least, whereas shorter/lighter alleles move the most - when a sequence of DNA is cut by a restriction enzyme, the resulting fragments will result in different bands based on their size
413
if there is no association between a particular genotype and the phenotype of interest, what will we notice in the experimental results?
we will notice that the phenotype does not change with different genotypes - different sized parents overlap in terms of the alleles being studied
414
if there is an association between a particular genotype and the phenotype of interest, what will we notice in the experimental results?
we will notice that the phenotype does change with different genotypes - different sized parents have different (combinations of) alleles
415
odds ratio =
probability of linkage/probability of no linkage
416
LOD =
log (OR)
417
what is the LOD?
the log of the probability/likelihood of co-segregation of trait and alleles at a locus being due to chance
418
if the LOD = 3,
p = 0.001 likelihood of association due to chance (ie there is a 99.9% chance that the association is REAL)
419
in terms of LOD, what does the threshold value represent?
The threshold value represents the minimum LOD score required to declare a statistically significant association between the marker and the trait.
420
what do we do once the QTL have been identified?
molecular techniques - fine mapping - can be employed to narrow the QTL down to candidate genes
421
RILs
Recombinant Inbred Lines: - Generated by crossing two genetically distinct parents (that differ in the trait), then selfing or sibling-mating for many generations. - Each line becomes homozygous but carries a unique combination of parental segments. - This allows for mapping of QTLs with increased precision by comparing many recombinant genotypes with their phenotype.
422
NILs
Near-Isogenic Lines: - Backcrossing a trait donor (carrying the QTL) into a recurrent parent (without the QTL) for many generations, selecting for the QTL region each time. - Until you get an organism that is differs from the recurrent pattern in only the QTL trait
423
what can we do once we have a candidate gene?
genetic engineering can be used to test the function of the candidate gene, proving its role in determining the genotype
424
use tomatoes as an example of dissecting phenotypes
- there are candidate genes in the fw2.2 QTL interval - these are thought to determine tomato size - the predicted fw2.2 is related to the human proto-oncogene RAS, which is involved in cell cycle control - genetic engineering: take a large fruit NIL. take another, same large fruit NIL with the small fruit allele introduced as a trans gene by genetic engineering. If there is a fruit size decrease in the second tomato, then the gene is involved in determining this trait; if not, then it is not
425
limitations of linkage mapping
- requires either extensive pedigree data or crosses: this is difficult in some taxa like humans - only maps effect in single cross/pedigree - no guarantee of widespread importance in population
426
To prove that two genes are linked, the LOD score (Logarithm of the Odds) must be at least
3.0
427
look over how to calculate LOD score with neurofibromatosis
428
why must one parent be a double heterozygote when we conduct positional cloning?
otherwise we will not know which allele came from which parent in the offspring
429
positional cloning
Positional cloning is a technique used to identify the specific gene responsible for a trait based on its location on the chromosome
430
how can we determine how much variation is attributable to genetic or environmental variation?
we can separate the genetic effects from the environmental effects by quantifying one variable, while controlling the other
431
draw a graph of stem length vs number of plants, and label the mean and variation
432
equation for mean
433
equation for variance
434
Ve
environmental variance - the portion of phenotypic variation in a population that is due to environmental factors, rather than genetic factors
435
Vg
genetic variance - a measure of the variation in a trait due to differences in genotype within a population
436
Vp =
Vg + Ve
437
Broad sense heritability of a trait
the extent of phenotypic variation that is attributable to genetic variation H^2 = Vg/Vp
438
describe what different values of H^2 could mean
H^2 = 1 - all the phenotypic variation is attributable to genetic variation H^2 = 0 - all the phenotypic variation is attributable to environmental effects
439
how and why is the heritability of a trait defined?
- heritability of a trait is always defined for a specific population or specific family in a specific set of environmental conditions - this is because the amounts of genetic, environmental, and phenotypic variation may differ among traits, families, populations and among different environments
440
what does H^2 tell us? what does H^2 NOT tell us?
Tells Us: If H^2 is high, the phenotype of an individual is likely to be attributable to its genotype IN THAT FAMILY Does not tell us: 1. What phenotype an individual will have based on their parents' phenotypes. Even if H^2 is high, an individual's precise phenotype cannot be predicted based on its parents' phenotypes (parents pass on their alleles, not their genotype) 2. What is going on in other families. H^2 is family specific and varies among different families, populations, or environments
441
why is H^2 not predictive?
Vp = Vg + Ve however, Vg = Va + Vd + Vi where - Va is variation due to additive effects - Vd is variation due to dominance effects - Vi is variation due to epistatic effects
442
Vd
dominance variance, which is the portion of genetic variance attributable to interactions between alleles at the same locus
443
is Vi predictive?
epistatic interaction variation is not transmitted from parents to offspring -> new genotypes and thus new epistatic relationships are formed with each generation
444
if a trait is due to additive effects, is it predictive?
yes
445
narrow-sense heritability
the extent of phenotypic variation that is attributable to additive genetic variation
446
describe different h^2 values and what they can mean
if all the phenotypic variation is attributable to additive variation, h^2 = 1 (the maximum it can be) if all the phenotypic variation is attributable to other genetic and environmental effects, h^2 approaches 0
447
h^2 =
Va/Vp
448
use bill depth in Darwin's finches as an example of narrow sense heritability
~ 18% of variation is attributed to the environment and or dominant or epistatic genetic variation slope = 0.82 ~ 82% of variation is attributed to additive genetic variation
449
what does h^2 tell us vs not tell us?
tells us: if h^2 is high, the phenotype of an individual is predictable based on the phenotype of its parent in that family does not tell us: what is happening in other families
450
how can twins be used to study heritability of polygenic traits?
1. monozygotic twins: single ovulated egg fertilised by one sperm, then the embryo splits into two. 100% of alleles are shared, so we can study the effect of different environments on the same genotype 2. dizygotic twins: two ovulated eggs fertilised by different sperm, so 50% of alleles are shared, so we can study the effect of different genotypes on the same environment
451
define a discontinuous trait and give an example
clear-cut, 'either-or' phenotypes between alternative alleles eg. all of the traits Mendel studied in peas
452
how does the phenotype vary in incomplete dominance?
the phenotype varies continuously with levels of protein function/the amount of functional gene product
453
molecular mechanisms of incomplete dominance in snapdragon colour
A1: functional enzyme A2: nonfunctional enzyme A1A1: red (100% pigment production) A1A2: pink (50% pigment production) A2:A2: white (0% pigment production)
454
how is familial hypercholesteremia an example of incomplete dominance?
the heterozygous phenotype (250-500mg/dl) is distinct from and intermediate compared to either homozygous phenotype (<250mg/dl and >500mg/dl)
455
qualitative trait: - examples - mode of inheritance - environmental effects
- blood and serum types, enzyme defects - simple Mendelian (monogene) - little environmental effect
456
quantitative trait - examples - mode of inheritance - environmental effects
- stature height, intelligence, milk yield - complex (polygene) - moderate to great environmental effect
457
threshold trait - examples - mode of inheritance - environmental effects
- hare lib, many diseases - complex (polygene) - moderate to great environmental effect
458
what kind of distribution does continuous variation typically have?
a normal distribution, which is bell-shaped
459
what is the Mendelian explanation of continuous variation?
alleles are incompletely dominant and have additive effects. the more genes or alleles, the more possible phenotypic classes, the greater the similarity to continuous variation
460
what interaction results in the normal distribution of a quantitative trait?
genetic and environmental influences
461
draw 3 graphs for no of individuals vs trait value: - two genes, two incompletely dominant alleles - environmental influences - normal distribution
462
what could be the result of more than two alleles of the same gene?
additional variation might arise
463
additive alleles
contribute to traits in a cumulative manner. the more of a particular allele an individual has, the stronger the expression of that trait
464
give an example of the impact of additive alleles on human traits
- a handful of ultra rare inherited mutations likely shave years off a person's life - each of these DNA variants can reduce lifespan by as much as 6 months - and different combinations dictate how early age-related diseases such as cancer, diabetes, and dementia will develop - thus, each variant is an additive allele
465
define continuous traits and give examples in humans
are determined by segregating alleles of many genes that interact together and with the environment eg height, weight, skin colour
466
what is another name for continuous traits?
quantitative traits
467
continuous traits are usually
polygenic
468
three categories of quantitative traits
meristic (counting traits): takes on a range of discrete values. these traits are quantitative, but restricted to certain discrete values (eg salamander spots) continuous traits: takes on a potentially infinite number of states or values over a continuous range (eg height) threshold trait: individuals who have a certain number of risk factors will exceed a threshold and develop the disease.
469
do threshold traits follow Mendelian rules?
no, they do not follow simple Mendelian rules or produce Mendelian ratios in pedigrees
470
give an example of environmental modification of certain mutations
siamese cats
471
quantitative trait locus (QTL)
region of the genome that correlates with the quantitative trait and may contain the genes affecting the quantitative trait
472
how can quantitative traits be described by a frequency distribution?
frequency distributions associated with each genotype at the QTL overlap. we cannot determine genotype by simply looking at an individual's phenotype as we can with genes that segregate in Mendelian ratios
473
how does our predictive power of genotype from phenotype vary with trait value? draw a graph
at either end (low or high trait value) we can guess the genotype from the phenotype. however, at intermediate trait values, this is very hard
474
phenotype =
genotype + environment
475
population genetics
the study of the genetics of a population and how the alleles vary with time
476
population
an interbreeding group of the same species within a given geographical area
477
gene pool
the collection of all alleles in the members of the population
478
gene flow
alleles can move between populations when individuals migrate and mate
479
give an example of how phenotype frequencies vary in different populations
phenylketonuria (PKU), a heritable metabolic disorder, autosomal recessive trait
480
what does the Hardy-Weinberg Law clarify?
the relation between genotype and allele frequency within a generation and from one generation to the next
481
five assumptions that must be met for a population to be at hardy-weinberg equilibrium
- infinitely large population - individuals mate at random - no new mutations appear in gene pool - no migration into or out of population - no genotype-dependent differences in ability to survive and reproduce
482
does any population follow all assumptions of Hardy-Weinberg law?
no; all populations violate one or more assumptions of Hardy-Weinberg law
483
why can HW law be used despite its limitations? 1
- equations derived based on assumptions are remarkably robust - HW law can be used as a null model
484
p =
allele frequency of one allele
485
q =
allele frequency of a second allele
486
p + q =
1 all of the allele frequencies together equals 1 or the whole collection of alleles
487
HW law
p^2 + 2pq + q^2 = 1 all of the genotype frequencies together equals 1 p^2 and q^2 = genotype frequencies for each homozygote 2pq = genotype frequency for heterozygotes
488
allelic frequency=
of particular allele/total # of alleles in the population - count both chromosomes of each individual - allele frequencies affect the genotype frequencies
489
how does HW law relate from generation to generation?
- allele frequencies do not change from generation to generation in a population at HW equilibrium - a HW population achieves the genotype frequencies of p2, 2pq, q2 in just one generation and once at equilibrium maintains them in subsequent generations - only 1 generation is required to reach HW equilibrium
490
why do all natural populations violate one or more assumptions of the HW law?
- natural populations are undergoing microevolution: genetic change due to changing allelic frequencies in populations - allelic frequencies can change with: nonrandom mating, gene flow, genetic drift, natural selection (unequal reproductive success)
491
impact of genetic drift on evolutionary equilibrium
- chance random fluctuations in allele frequency that have a neutral effect on fitness - random allele frequency fluctuations might make certain disease alleles more common in particular populations
492
are smaller or larger populations more affected by genetic drift?
smaller populations are more affected than larger populations
493
sampling biases are ---- pronounced in small populations
more
494
impact of natural selection
acts on differences in fitness to alter allele frequencies
495
fitness
individual's relative ability to survive and transmit genes to the next generation (viability and reproductive success)
496
natural selection
individuals with higher fitness strive and reproductive more than individuals with lower fitness
497
why might there be a decrease in the frequency of a recessive allele over time?
- homozygous recessive genotype (rr) has decreased fitness - fitness of RR and Rr is the same
498
why is selection unable to reduce the frequency of recessive lethal alleles to zero?
- when q is small, the frequency of homozygous recessive individuals is low - most copies of the q allele are in heterozygotes, who do not have reduced fitness
499
heterozygote advantage
although allele frequency of lethal alleles should continue to decline, some stay in population at stable frequencies
500
heterozygote advantage conferred by the sickle-cell allele
- In heterozygotes, the presence of some sickled red blood cells interferes with the life cycle of the malaria parasite. - As a result, HbA/HbS individuals are more resistant to severe malaria compared to HbA/HbA individuals.
501
how does the time of onset of a recessive, negative-impact trait influence frequency?
- if onset is in middle or late age, after reproduction, then the trait will not affect fitness - sustain little or no negative selection
502
define evolutionary equilibrium:
a balance between mutation to a new allele and selection against the allele
503
what is the ultimate source of new variation?
mutations: - when new allele has effect on fitness, selection will drive frequency towards an equilibrium with wild-type allele - if new allele has no effect on fitness, genetic drift will determine its frequency
504
define a haplotype
short for haploid genotype - a group of alleles or DNA variations that are inherited together from a single parent because they are located close to each other on the same chromosome.
505
what is a possible application of Hardy-Weinberg equilibrium?
1. DNA collected from evidence at crime scene 2. DNA collected from suspected 3. five specific DNA sequences from different chromosomes are labeled and separated by size - blood (victim), blood (suspect), skin (evidence), blood (evidence) 4. use HW to calculate the probability that another person in the suspect's population group has the same pattern of these alleles
506
what is the purpose of haplotype analysis?
enables the correlation of occurrence of trait with occurrence of a particular allele
507
use of SNP analysis
genotype many individuals and correlate with phenotype
508
crossover frequency is a function of the
distance between two loci
509
how can we mathematically prove if a test cross result is due to linkage or not?
perform a chi-square test, which assesses the likelihood that a deviation from expectation is due to chance
510
why is the chi-square test necessary to prove linkage?
- deviations from 1:1:1:1 ratios can represent chance events OR linkage - the chi-square test accounts for sample size
511
chi-square test: framing a hypothesis
null hypothesis - observed values are not different from the expected values. for linkage studies, this is no linkage where you expect a 1:1:1:1 ratio of gametes alternative hypothesis - observed values are different from expected values. for linkage studies, this is that the genes are linked and you expect a significant deviation from 1:1:1:1 ratio
512
4 steps to conducting a chi-square test
1. formulate null hypothesis: genes are not linked (1:1:1:1 ratio of phenotypes 2. compute: x^2: Σ (O-E)^2/E 3. determine degrees of freedom (number of independent measurements) 4. consult x^2 chart of critical values
513
how do we know whether to accept or reject the null hypothesis?
if p<0.05, then we reject the null hypothesis and accept the hypothesis that genes are linked to
514
what does p<0.05 mean?
less than 5% of the time, we are incorrectly rejecting the null
515
what did Alfred Sturtevant do?
he postulated that the frequency of crossovers between two genes is a function of their distance apart on the chromosome; he also created the first genetic linkage map
516
recombination frequency (RF) =
number of recombinants/total number of progeny
517
one map unit (m.u.)=
one centiMorgan (cM) = 1% recombination - 1 product of meiosis out of 100 is recombinant
518
in any given cross, the recombination frequency (RF) cannot be greater than
50% (=unlinked)
519
steps in analysing a three-point test cross
1. anticipate and identify 8 kinds of meiotic products 2. identify pairs of reciprocal products 3. identify parental types as the most frequent pair of products 4. identify double crossover products as least frequent pair of products 5. compare parental and double crossover to deduce locus order 6. compute map distances (distances between loci) by breaking down the results for each interval - RF = SXO+DXO/total
520
define interference
crossover in one region interferes with simultaneous crossing over in adjacent regions
521
how can we calculate interference?
1. expected frequency of DCO = product of frequency crossovers in two regions 2. coefficient of coincidence = observed DCO/expected DCO 3. interference = 1- coefficient of coincidence
522
genetic vs physical distances on a chromosome
- recombination and physical maps are co-linear (syntenic) but are frequently misaligned as recombination distances (cM) are frequently not the same as physical distances (bp of DNA) - that is, one cannot say that X bp of DNA sequence always equals Y cM on a linkage map - this is because recombination varies across the length of a chromosome
523
do traits need to be phenotypic for linkage mapping?
no - we can also use molecular markers - any polymorphism can be mapped
524
why can only female drosophila be used in linkage experiments?
In male Drosophila melanogaster, meiosis does not involve recombination, a phenomenon known as achiasmy