Session 1 Flashcards

1
Q

Outline the stages of the cell cycle

A

Interphase:
G0 - cell cycle arrest
G1 - cellcular components duplicated
S - Chromosome duplication
G2 - Error checking

Mitosis:
Prophase - Chromosome condense and spindle fibres appear
Prometaphase - spindle fibres attach to chromosomes
Metaphase - Chromosomes line up at metaphase plate
Anaphase - Sister chromatids split to opposite poles
Telophase - Nuclear membranes reform and chromosomes decondense. Loss of spindle fibres
Cytokinesis - cytoplasm divides and 2 daughter cells produced.

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

What are the three main cell cycle check points?

A

G1/S - Cell growth enables formation of the CDK4/6-cyclin D. Phosphorylates retinoblastoma protein. Relieves inhibition of E2F transcription factor. Cyclin E now expressed, binds to CDK2

G2/M - CDK1 is activated by phosphorylation and de-phosphorylation of specific amnio acid residues by Cyclin-Activating Kinase (CAK), as well as the inhibition of the wee1 protein (which inhibits CDK1). Enables CDK1-cyclin B formation (aka MPF)

M/Anaphase - Anaphase-promoting complex (APC) activated. Degrades cyclin B = MPF disassembly. Relieves inhibition of ‘separase’ which cuts cohesin. Sister chromatid separation = anaphase entry

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

How is the cell cycle manipulated for metaphase spreads?

A

Mitogens - used to induce division of resting cells:(PHA, pokeweed, concanavilin A)

Synchronisation - Inhibitors block cell cycle during S phase by slowing/stopping DNA synthesis (FudR/uridine, Thymidine)

Block released after 16-22h - cells continue through G2 together

Mitotic arrestants - stop division during mitosis (colchicine/Colcemid®). Prevents spindle fibre apparatus formation. Stops cell at metaphase

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

When does recombination occur?

A

Prophase 1

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

What can occur due to non-allelic homologous recombination?

A

Loss of regions of chromosome - from single gene to multiple genes to entire regions of chromosomes

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

What are the three subtypes of Eukaryotic chromosomes?

A

Metacentric: p and q arms are of roughly equal length (e.g. 1 and 3, 16-18)
Submetacentric: the arms are of unequal length (e.g. 2 and 6-12)
Acrocentric: p arm is very short but still present

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

What are the functions of centromeres?

A

essential for accurate chromosome segregation during cell division as it provides the foundation for assembly of the kinetochore which attaches to the spindle

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

Outline centromere structure

A

Composed of constitutive heterochromatin of families of repetitive satellite DNA - tandem repeats of 171bp AT rich DNA named alpha satellites, that extend for several Mb and make up 3% of the genome.

Around 20 proteins associate to the centromere and exist in two groups on the basis of their spatial positioning throughout the cycle: 1) The first class comprises proteins that are constitutively associated with the centromere such as CENPA, CENPB and CENPC, which are thought to have structural roles in kinetochore formation. 2) The second class known as passenger proteins associate with the centromere transiently during the cell cycle and comprises proteins with diverse roles in cell division such as spindle capture, metaphase to anaphase transition, and sister chromatid cohesion

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

How does CENPA maintain the centromere position?

A

CENP-A is a histone H3 variant and it forms a unique complex with other histones (H2A, H2B, and H4) which is only found at the centromere with specific epigenetic mark

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

Name 3 “diseases” are associated with centromere dysfunction?

A

Premature centromere division (PCD) - age-dependent phenomenon occurring in women, characterised by rod-shaped X chromosome(s) though to be cause of age-dependent increase of X chromosome aneuploidy. no phenotype

Premature chromatid separation (PCS) - consists of separate and splayed chromatids with discernible centromeres and involves all or most chromosomes of a metaphase. It is found in up to 2% of metaphases in cultured lymphocytes from approximately 40% of normal individuals. no phenotype but maybe increased aneuploidy in offspring/reduced fertility

Roberts syndrome - Autosomal recessive breakage syndrome. ESCO2 encodes Acetyltransferase needed for sister chromatid cohesion in S phase. Metaphase spreads show extensive PCS. Phenotype includes pre/post growth retardation, Limb malformations (reduction), craniofacial (microcephally, clefting), intellectual disability and renal and cardiac abnormalities.

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

What is the kinetochore?

A

Large protein complex which assembles on centromere and acts as point of attachment for spindle fibres

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

What are telomeres?

A

Highly conserved gene-poor, DNA-protein complexes at end of chromosomes

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

What is the function of telomerase?

A

Complex made of TERT and TERC. TERC is RNA subunit used by TERT as template to extend telomeric sequence and maintain telomeres

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

What is the nucleolus? And what regions organise it? How can it be visualised?

A

Site of ribosomal RNA (rRNA) transcription, pre-rRNA processing and ribosome subunit assembly

nucleolar organising regions (NOR) located on the short arms of the acrocentric chromosomes

Silver nitrate staining if active

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

What are the three phases of DNA replication?

A

Initiation - DNA replication initiates at the origins of replication recognised by the origins recognition complex (ORC).. Topoisomerases create a nick in a single DNA strand to be unwound by Helicases. This releases the tension holding the DNA helix in coils and supercoils and allows the double helix 3’ end of the RNA primers followed by the Polymerase.

Elongation - primers are removed and replaced with new DNA nucleotides and the backbone is sealed by DNA ligase. The two replication forks are created in opposite directions

Termination - two replication forks meet each other and they are dismantled and the two ends are joined by DNA ligase

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

How do the leading and lagging strands differ?

A

Leading strand - polymerase moves 5’ to 3’ continuously

Lagging strand - Cannot be continuous due to 5’ to 3’ rule. Polymerase elongates a short stretch of DNA and then moves to a new primer while the helicase moves along the DNA. Synthesis is discontinuous. The short fragments are called Okazaki fragments. Replicative Polymerase is replaced by a different polymerase which has an exonuclease activity to degrade the RNA primer and synthetize DNA. A DNA Ligase then joins the two sections of DNA.

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

What is the overall error rate per cell division? How is it improved?

A

1 in 10^3
Proofreading by polymerase and MMR reduces to 1 in 10^9

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

Name a genetic disease which arises due to variants in aspects of DNA replication?

A

Origin of replication defect - Meier-Gorlin syndrome
Helicase defect - Bloom Syndrome
Polymerase defect - Hutchinson-Gilford progeria syndrome
Telomerase defect: Dyskeratosis congenita

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

What families of DNA polymerase exist?

A

Family B = high-fidelity, DNA polymerases for nuclear DNA
Family A = DNA polymerase γ (gamma) which replicates mitochondrial DNA

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

What is the structure of DNA?

A

Linear sugar and phosphate backbone with Carbon 5’ bound to Carbon 3’ of next sugar by phosphdiester bond.

Bases bound to Carbon 1 of sugar molecular

Two strands run anti-parallel to each other with base pairing between­ Adenine and Thymine (2 hydrogen bonds) and Guanine and Cytosine (3 hydrogen bonds) to form double helix.

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

What is the structure of RNA?

A

Single stranded molecule with additional ­hydroxyl group at the 2’ position which makes it more unstable

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

What is a histone?

A

Lysine and arginine rich +vely charged proteins which an affinity for -vely charged DNA

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

What is a nucleosome?

A

­147bp of 2nm DNA helix coiled in less than two turns around a central core of 8 histone proteins (x2 each of H2A, H2B, H3 & H4)

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

How is DNA packaged further?

A

Nucleosomes are separated by stretches of linker DNA which bind H1.

H1 facilitates further packaging into a 30nm thick chromatin fibre made of nucleosome spiral with 6-8 nucleosome per turn

The chromatin forms loops and condenses further to form chromosome

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

What are the two main levels of chromatin compaction observed in interphase?

A

Euchromatin - “open” with weak binding of H1 histones and acetylation of the 4 nucleosomal histones

Heterochromatin - highly condensed throughout cell cycle associated with tight H1 histone binding. Can be Constitutive (generally inactive - largely of repetitive DNA) or Facultative (varies)

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

What are the three main histone post translational modifications?

A

methylation, acetylation, phosphorylation

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

What is the effect of histone methylation?

A

Methylation - commonly on lysines and arginine residues of N-terminal tails which protrude from nucleosomes. Effect depends on which residue is modified due to effect on basicity/hydrophobicity of histones and their affinity with certain proteins . 1. Lysine methylation can be involved in both transcription repression, 2. Arginine methylation has similarly been implicated in both transcription repression

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

What is the effect of histone acetylation?

A

­Occurs on lysines and almost always associated with activation of transcription. Acetyl group on a lysine neutralises its positive charge and weakens interactions between histones and DNA, de-stablising chromatin architecture. Mediates by HATs and HDACs

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

What is the effect of histone phosphorylation?

A

Occurs on Serines, Threonines and Tyrosines.­ Addition of phosphate to amino acid adds significant negative charge to the histone, influencing chromatin structure. Phosphorylation of H3S10 during mitosis occurs genome-wide - associated with chromatin condensing

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

What is the mechanism of disease in Rett syndrome?

A

LoF variants in MECP2

MECP2 binds methylated DNA and recruits HDACS and HMT to respress expression through histone modification. LoF variants lead to activation of genes normally repressed. (mainly in verve cells for phenotype)

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

What are the three main mechanisms of epigenetic gene regulation?

A

DNA methylation
Histone modification
RNA-associated silencing

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

How are genes methylated?

A

Addition of methyl group to the C5 position of cytosine to form 5-methylcytosine (5MeC). Almost exclusively at CpG dinucleotides - two methylated cytosines diagonal to each other on opposing DNA strands.

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

What enzymes are involved in DNA methlation?

A

DNMT1 - maintains methylation of new strand during DNA replication
DNMT3A and DNMT3B - mainly embryonic and add initial methylation

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

What are short Non-coding RNAs ?

A

MicroRNAs small strands of RNA ~22 nucleotides long, interfere with gene expression at the level of translation. Bind to the 3′-untranslated regions of their target mRNAs, thereby inducing enzymatic degradation and preventing translation

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

What are long Non-coding RNAs ?

A

~200bp long and are thought to form ribonucleoprotein complexes that interact with chromatin, regulating histone modifications and the structural transformations that distinguish heterochromatin from euchromatin.

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

What is rRNA?

A

Component of cytoplasmic and mitochondrial ribosomes

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

What is snRNA?

A

9 in total and are components of spliceosome

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

What is tRNA?

A

cytoplasmic tRNA (encoded by nuclear DNA) and mitochondrial tRNA (mt-tRNA) (encoded by mitochondrial DNA) transfer the correct amino acid to the ribosome during translation

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

What is the core promoter?

A

most proximal pmromter of a gene - contains RNA polymerase binding site - the TATA box

40
Q

What are Proximal promoter elements?

A

located ~250 bp upstream from TSS; contains many primary regulatory elements; site where general transcription factors bind

41
Q

What is encoded by the mitochondrial genome?

A

37 genes coding:
two ribosomal rNAs (rRNAs)
22 transfer RNAs (tRNAs)
13 polypeptides subunits of the oxidative phosphorylation system

42
Q

What are LCRs?

A

chromosome-specific low copy repeats of around 1-400Kb which are the substrate for NAHR

43
Q

What are some “common” LCR/NAHR mediated deletions/duplications?

A

Williams-Beuren Syndrome - chromsome 7
Prader-Willi Syndrome/Angelman syndrome - chromosome 15
Charcot-Marie-Tooth (dup) / HNPP (del) - chromosome 17
Smith-Magenis (del) / Potocki-Lupski (dup) - chromosome 17
DiGeorge syndrome and reciprocal duplication - chromsome 22

44
Q

What are the three mRNA surveillance mechanisms?

A

Nonsense Mediated mRNA decay pathway (NMD)
Nonstop Mediated mRNA decay pathways (NSD)
No-go Mediated mRNA decay pathway (NGD)

45
Q

What therapies have been trialled to avoid NMD on PTC transcripts causing disease?

A

Increase PTC read through (happens 0.1-1% of time anyway)
Chimeric tRNAs, that specifically recognise the termination codon triplets and introduce an amino acid instead of termination
antisense oligonucleotides to remove PTC portion of a mutated transcript
NMD inhibitors

46
Q

What are two example common pericentric inversion variants which class as normal variation?

A

inv(3)(p13p25) and inv(11)(q21q23)

47
Q

What are the four common constitutive heterochromatic normal variants?

A

1qh, 9qh, 16qh and YqhWh

48
Q

What are transposable elements?

A

units of DNA that move within the genome - either “copy and paste” or “cut and paste”

They account for 45% of genome. Most commonly Long Interspersed Nuclear Elements (LINES ~500,000 per genome) and Short Interspersed Nuclear Elements (SINES ~1,500,000 per genome).

49
Q

How can transposable elements cause disease?

A

Insertion within a gene may disrupt a coding region and homologous recombination and/or mismatch repair between these elements may induce structural chromosomal rearrangements

50
Q

What are some examples of Satellites?

A

Satellite DNA, Minisatellites and Microsatellites

51
Q

What are fragile sites?

A

Segments of uncoiled chromatin that is liable to show gaps and breaks in chromosomes which are exposed by our efforts to control passage through the cell cycle and are normal variation

52
Q

Give an example fragile site which is not classed as normal variation

A

Xq27.3 (FRAXA) and Xq28 (FRAXE) due to hypermethylation of FMR1

53
Q

What sex chromosome aneuploidy is thought to always be mosaic? What types of mosaicism are observed?

A

Turner syndrome
Numerical - e.g. 45,X/46,XY or 45,X/46,XX/47,XXX
Structural - e.g. 45,X/46,X,i(X)(q10) or 45,X/46,X,+mar, 45,X/46,X,r(X)

54
Q

What autosomal chromosome aneuploidy is always mosaic?

A

Pallister Killian syndromeW

55
Q

What is Pallister Killian syndrome?

A

i(12p) - tetrasomy for 12p

Demonstrates tissue specific mosaicism with normal diploid complement in PB due to instability i(12)p at mitosis so is lost through rounds of mitosis. But significant levels in fibroblast, AF, CVS and BM

Buccal smears may be of use using interphase FISH or qPCR to detect the additional i(12)p

56
Q

What is MMEJ ?

A

microhomology-mediated end joining

double stranded repair mechanism that results in deletions and insertions that flank the break sites -requires short regions of homology (5-25bp) at either side of a double strand break allowing repair to occur through annealing and ligation

57
Q

What is the result of Breakage-fusion-bridge cycle ?

A

Dicentric chromosomes which are then pulled apart and break in anaphase. This will occur multiple times until a chromosome acquires a telomere

58
Q

What is FoSTeS?

A

Fork stalling and template switching

3’ end of the lagging DNA strand disengage from the original template and anneal, by virtue of microhomology, to a single-stranded section of DNA in a nearby replication fork. Can result in deletions, inversion, duplication, translocations

59
Q

What is Chromothripsis?

A

hundreds of genomic rearrangements occur in single one-off event to produce highly complex derivative chromosomes seen in some cancers

60
Q

What is the most common reciprocal translocation?

A

t(11;22)(q23.3;q11.2)

Risk of der(22) offspring (Emmanuel syndrome) as result of 3:1 segregation

61
Q

What are the two most common robertsonian translocations? And how do they arise?

A

rob(13;14) and rob(14;21)

centric fusion

62
Q

How can UPD cause disease?

A

1) Genes within the UPD region are subject to genomic imprinting
2) Homozygosity for recessive mutations: two copies of a recessive mutation transmitted from a heterozygous carrier parent (‘isozygosity’)
3) UPD resulting from a somatic recombination can cause loss of heterozygosity (LOH) or loss of imprinting (LOI)

63
Q

What are the different types of UPD?

When does each arise?

A

Uniparental isodisomy (UPID) - Meiosis II nondisjunction or mitotic error

Uniparental heterodisomy (UPHD) - Meiosis I nondisjunction

64
Q

What occurs due to UPD for the entire chromosome complement?

A

Complete hydatidiform mole - paternal UPD (usually 46,XX)

Benign cystic ovarian teratoma - Maternal UPD

Partial hydatidiform mole - triploidy with extra set of either mat or pat chromosomes

65
Q

How can complete chromosome UPD occur?

A

Trisomy rescue
Monosomic rescue
Mitotic error

66
Q

What is trisomy rescue?

A

Meiotic nondisjunction in one parent results in a disomic gamete
Fertilisation of this disomic gamete with a normal haploid gamete results in a trisomic conceptus
Rescue’ through loss of one homologue (perhaps through anaphase lag) at a very early postzygotic stage (possibly even in the zygote), results in UPD in 1/3 cases (usually mat)

67
Q

What is monosomic rescue?

A

Meiotic nondisjunction in one of the parents results in a nullisomic gamete.- Fertilisation of this nullisomic gamete with a normal haploid (monosomic) gamete results in a monosomic conceptus.
‘Rescue’ of the remaining homologue results in specifically isodisomy (usually pat)

68
Q

What is mitotic error?

A

Conceptus and subsequent cell line are initially normal. Mitotic error leading to either trisomy or monosomy is then ‘rescued’ to result in isodisomy

69
Q

What is the function of sodium bisulphite?

A

converts unmethylated cytosine nucleotides to uracil so can be used to differentiate between methylated and unmethylated DNA

70
Q

What techniques for UPD detection use sodium bisulphite?

A

Methylation Sensitive (MS) PCR

Bisulphite restriction analysis and PCR

Methylation Sensitive (MS) melting curve analysis

Pyrosequencing

71
Q

What techniques can detect UPD without sodium bisulphite?

A

Microsatellite Analysis

MS- MLPA

Southern Blotting with MS restriction enzymes

SNP array

WES/WGS

72
Q

What is most common cause of 47,XXY offspring?

A

Paternal XY non-disjunction at MI accounts for 50% of XXY cases

73
Q

What are the three major classes of eukaryotic pseudogenes?

A

Non-processed (duplicated) pseudogenes

Processed (retrotransposed) pseudogenes

Unitary (solitary) pseudogenes

74
Q

Give an example a pseudogene can have at a DNA level

A

gene conversion/recombination with their parental homolog - e.g. between PSM2 and PMS2CL

75
Q

Give an example a pseudogene can have at a RNA level

A

post-transcriptional regulation. e.g. PTENP1 (a pseudogene of PTEN) acts as a ‘miRNA decoy’ by binding to miRNA and allowing PTEN to escape miRNA-mediated silencing

76
Q

What is the resolution of G banding?

A

3-5Mb

77
Q

How does G banding work?

What underlies the pattern?

A

Treating aged/dried metaphase preparations with a protease (dilute trypsin) then staining with Giemsa stain

Heterochromatin stains darker whereas euchromatin stains light

78
Q

What is R banding? What is main benefit?

A

Almost mirror of G - dark and light swap.

Allows visualisation of telomeres

79
Q

What is Q banding?

A

Fuorescent technique using Quinacrine dihydrochloride which stains A-T rich sites

80
Q

What is C banding?

A

C-banding stains constitutive heterochromatin

81
Q

What is Cd staining?

A

pair of dots at each centromere, one on each chromatid

82
Q

What is Replication banding?

How is it useful?

A

5’-Bromo-2’deoxyuridine (BrdU) is a thymidine analogue that is readily incorporated into chromosomes

Can be used to look for sister chromatid exchange that occurs during mitosis. Normally 5-8 SCEs per mitosis. In Bloom syndrome will be 10X more than this

83
Q

What usually informs on whether adjacent-1 or adjacent-2 is likely to happen?

A

Adjacent-1 = the translocated segments are shorter than the centric ones

Adjacent-2 = the centric segments are shorter than the translocated ones

84
Q

What are the two types of 2 break inversions?

A

Pericentric includes centromere

Paracentric does not include the centromere

85
Q

What is the spliceosome?

A

Complex of snRNAs and their associated proteins

86
Q

Outline the activity of the spliceosome

A

U1 snRNA binds to donor splice site
U2 snRNA binds to branch site (Adenine)
U1 and U2 come together to form a loop
U4, U5 & U6 bind and GT donor “attacks” branch point and AG is cleaved
Exons joined

87
Q

How is splicing regulated?

A

trans-acting elements

cis-acting elements:
Exonic splicing silencer (ESS) - short region (usually 4-18 nucleotides) of an exon which recruit proteins that will negatively affect the core splicing machinery and involved in exon skipping
Exonic splicing enhancer (ESE) - DNA sequence motif consisting of 6 bases within an exon which enhance splicing

88
Q

What are the three “box” promoters of transcription?

A

TATA box - 25-35bp from transcription start and indicates direction for transcription and strand to be read
GC box - ~110 bases upstream from the transcriptional start site
CAAT box - ~80bps upstream and strongest determination of promoter efficiency

89
Q

What are enhancers?

A

cis-acting short sequence elements
bind gene regulatory proteins which loop DNA between promotor and enhancer to allow proteins bound to enhancer to interact with promoter bound TFs or RNA polymerase

90
Q

What is 5’ capping?

A

Methylated nucleoside, 7-methylguanosine (m7G) is linked to the 5’ end of the RNA via a 5’-5’ phosphodiester bond

91
Q

When does X inactivation happen?

A

Random process likely in late blastula stage and the inactivated X is inherited

92
Q

How is X inactivated?

A

Xist is encoded by X inactivation centre (XIC) on Xq13 and codes for long non-coding (lnc) RNA molecule

Xist RNA molecule spreads along the chromatin outwards from the XIC. Resulting heterochromatin modifications such as histone deacetylation and methylation of CpG islands are epigenetic processes which organise the chromatin into a closed confirmation.

93
Q

What regions escape X inactivation?

A

Genes which require bi-allelic expression in the pseudo-autosomal regions (coded for by both X in females and X and Y in males)

94
Q

How can X inactivation effect other chromosomes?

A

X-autosome translocation. Expression of Xist on derivative chromosome will inactivate autosomal regions

95
Q

What is skewed X-inactivation?

A

In theory should be totally random but can be skewed - either benefit (no phenotype from X-linked condition) or drawback (increased phenotype from X-linked condition)

96
Q

What is most common method for X-Inactivation studies?

A

HUMARA Assay

PCR-based technique which amplifies a polymorphic CAG repeat region located within the first exon of the androgen receptor gene

Genomic DNA is first digested using HpaII, a methylation-specific enzyme that will only cut the unmethylated (i.e. active) allele therefore allowing us to distinguish between the active and inactive X chromosomes.

PCR primers are specific to regions flanking HpaII sites, such that digestion of the unmethylated allele disrupts PCR amplification. As a result, only the methylated (i.e. inactive) allele will give rise to a PCR product.

PCR products are then separated by capillary electrophoresis and amplification patterns between the undigested and digested alleles can be compared