M2M Unit 2 Flashcards
Standard pedigree symbols:
male= square female= circle unknown= diamond black=phenotype positive white= phenotype negative deceased= diagonal line through 3 generations to be "complete"
Patterns of Mendelian Inheritance- generic
genes come in pairs
(note x-linked and mito diseases)
genes’ alleles lead to observed phenotypes
Law of Segregation
Alleles segregate at meiosis into the gametes
Law of Independent Assortment
The segregation of each pair of alleles is independent
exception: genes close together are linked
define hemizygous
a person only has 1 particular gene, not 2
i.e. males have a single copy of each X chromosome gene
also anyone who only has 1 working copy of a gene via deletion (or imprinting)
Horizontal pattern of affected phenotype
tends to be autosomal recessive- more likely affected in siblings and not parents
Rare disease and consanguinity
the rarer the disease/allele, the greater proportion of affected persons will be due to consanguinity (blood related to affected person)
autosomal dominant
autosomal recessive
dom: tends to appear in every generation
phenotypically normal parents tend to not pass it on equally to males and females. (new mutations can occur)
rec: can skip generations; normal parents can pass it on to their children
x-linked recessive
incidence much higher in males.
appears to “skip” through unaffected females
affected males pass on mutations to ALL daughters and NO sons.
Carrier females’ offspring have 50% chance of inheriting
x-linked dominant
disease incidence is much higher in female children
affected males pass it on to all daughters but not sons
carrier females’ children have 50% chance of inheriting phenotype
pedigree AKA useful for... proband consultand consanguineous mating
family history
useful for identifying possible patterns of inheritance and est. genetic risks
starting point of genetic study
person bringing the family to attn
couples w/ >1 known ancestor in common
penetrance
expressivity
pleiotropy
penetrance: fraction who has a genotype and shows the phenotype (can be age dependent, etc.)
expressivity: the extent that the genotype is expressed (severity) (depends on sex, environmental effects, stochastic effects, and modifier genes)
pleiotrophy: a mutation affecting multiple different phenotypes (NOT variable expressivity)
population genetics:
the study of allele frequencies and changes in allele frequencies in populations
Hardy Weinberg principle
and assumptions
p^2 + 2pq + q^2 = 1 p + q = 1 p= common allele freq q= rare allele freq assume: pop is large random matings allele freq's are constant over time because: no mutations equally fit genotypes no sig immigration/emigration
3 events in meiosis that produce genetic variability in offspring
crossing over
assortment of alleles
reduction in genetic material from diploid to haploid
mitotic vs meiotic cell division
Meiosis: paternally and maternally derived homologous chromosomes pair at the onset of meiosis- Prophase 1
Meiosis: reciprocal recombination events between maternal and paternal sister chromatids generate chiasmata between homologs
meiotic recombination vs chromosome nondisjunction
nondisjunction events are related to the positioning of chiasmatas
crossovers occurring too near or far from the centromere increase nondisjunction
centromere-distal exchanges are less effective in ensuring appropriate spindle attachment and sep of paired homologs in meiosis 1
centromere-proximal or excessive exchanges lead to entanglement of paired homologs in meiosis 1 that then undergo reductional division leading what appears to be meiosis 2 errors
nondisjunc events are related to freq of crossover events- the reduction/absence of recombination events increases the likelihood of nondisjunction
*trisomies often result from meiosis 1 nondisjunction
3 common human trisomies
trisomy 13, 18, 21
clinical features of trisomy 13
Patau syndrome
characteristic faces
severe mental retardation
congenital malformations- holoprosencephaly, facial clefts, polydactyly, renal abnormalities
clinical features of trisomy 18
Edwards syndrome
intrauterine growth retardation
characteristic faces, severe mental retardation, characteristic hand positioning
congenital malformations- valvular heart disease, posterior fossa CNS maldevelopment, diaphragmatic hernias, renal abnormalities
clinical features of trisomy 21
Down syndrome
characteristic faces, short stature, hypotonia, moderate mental retardation
congenital malformations- endocardial cushion defects, duodenal atresia and other gastrointestinal anomalies, Hirshprung disease
fundamental principles regarding human genome evolution and organization
- reflects results of different selection pressures that have occurred over evolutionary time and shaped our genome
- genes and genomic features that have been adaptive have been retained
- genotype + environment = phenotype
why is genome variation an essential fuel of evolution and adaptation (and disease)
random variation in a highly ordered structure = almost always deleterious consequences
genetic disease is the price we pay as a species to continue to have a genome that can evolve (adapt to new/changing environments)
organization of the human genome
dynamic; non-random
~30 new mutations per individual
shuffling of regions at each meiosis due to recombination
can produce somatic and germ-line DNA changes
SNP frequencies
average of 1 SNP every 1000 bp between any 2 randomly chosen genomes
99.9% identical and 3,000,000 differences
5 types of variations that occur between genomes:
- insertion-deletion polymorphisms (indels)
- SNPs
- CNVs
- Genome “structural” variation
- others- chromosomal or larger scale variations, rearrangements, translocations, etc.
majority of variants are silent, but some can have functional effects
Insertion-deletion polymorphisms:
minisatellites
microsatellites
minisatellites: tandemly repeated 10-100 bp DNA blocks
VNTR (variable # of tandem repeats)
microsatellites: di-, tri-, tetra- nucleotide repeats; >5x10^4 per genome
STRPs (short tandem repeat polymorphisms)
SNPs
frequency of 1 in ~1000 bp’s
PCR-detectable markers, easy to score, widely distributed
CNVs
variation in segments of genome to 200bp-20 Mbp’s
can range from one additional copy to many
array comparative genomic hybridization (array CGH)
genome “structural” variation
broadest sense: all changes in genome are not due to single bp substitutions
CNVs: primary type of structural variation
CNV loci may cover 12% of genome
implicated in increasingly larger number of diseases
some CNV regions involved in rapid and recent evo change… such regions are often enriched for:
human specific gene duplications
genome sequence gaps
recurrent human diseases
characteristics of the genome: gene-rich gene-poor stable unstable GC rich AT rich euchromatin heterochromatin
gene-rich: Chr 19 gne-poor: Chr 13, 18, 21 stable- majority of genome unstable- dynamic and often disease-associated (SMA Chr 5q13; DiGeorge Syndrome Chr 22q; 12 diseases 1q21) GC rich: 38% of genome AT rich: 54% of genome euchromatin- relaxed, less repeats heterochromatin- less relaxed, more repeats, generally near centromeres
completely sequenced human genome?
no completely sequenced and assembled human genome
-sequencing focused on euchromatic regions: easiest to access and interpret; repeats are often difficult to decipher
gaps remaining in the euchromatic regions
341 gaps
1% of euchromatic genome
many contain segmental duplications that require more work and new methods
categories of genomic DNA sequences
1.5% translated (protein coding)
20-25% genes (exons, introns, flanking sequences in regulatory gene expression)
50% “single copy” sequences
40-50% classes of “repetitive DNA”
sequences that are repeated 100s to millions of times
2 types of repetitive DNAs
tandem repeats AKA satellite DNAs
dispersed repetitive elements
tandem repeats AKA satellite DNAs
incl micro and minisatellites
some are used in cytogenetic banding
some are found on specific long-arm heterochromatin regions of Chr 1, 9, 16, and Y hotspots for human-specific evo changes
incl alpha satellite repeats- 171 bp repeat unit near centromere (may be important in chromosome segregation)
dispersed repetitive elements
Alu family- (ex. SINEs)
~300bp related members
500K copies in genome
L1 family- (ex. LINEs)
~6K bp related members
100K copies in genome
- both can be sig med relevance
- retrotransposition may cause insertional inactivators of genes
- repeats may facilitate aberrant recomb events between diff copies of dispersed repeats leading to diseases (called Non-Alleltic Homologous Recombination (NAHR)
estimated number and types of human genes
25-30 thousand different genes, comprised of:
protein-encoding genes
RNA-encoding genes
pseudogenes (nonfunc, but homologous copies of existing genes; split into intron-containing and intronless?)
gene families
genes with high sequence similarity;
perform similar functions
arise by gene duplication
gene duplication as an evolutionary mechanism
advantageous
when a gene duplicates, it frees one copy to vary while the other copy continues to perform its function
ALTHOUGH more copies means more chances for errors and negative impacts
2 methods of current genome sequencing and “missing heritability” problem
nextgen DNA sequencing-
- no genome has been completely sequenced/assembled
- relies on short read sequences
- complex, high duplicated areas are often unexamined, but these are implicated in numerous diseases
genome-wide association studies (GWAS)
- many regions are unexamined by “genome wide” screening tech’s;
- “missing heritability” for complex diseases- many large-scale studies implicate loci (SNPs) that account for only a small frac of the expected genetic contribution
genetic variability from meiosis
- meiotic recombination: homologs cross-over (chiasmata); offer steady support for smooth division
- can also occur X-Y in males
- random segregation of chromosomes (2^23 possible combos)
- 1/4 haploid female products becomes an egg
mitosis vs meiosis cell divisions
mitosis- 1 round of chr seg.
2 identical daughter cells
DNA replication preceeds each round of chr segregation
no homolog pairing
infrequent recomb
sister chromatids separate
occurs in somatic cells (and pre-meiosis germ cells)
meiosis- 2 rounds of chr seg 4 unique haploid daughter cells homolog pairing crossing over homologs THEN sister chromatids separate occurs only in germ line cells
banding of karyotypes
giemsa- dye used to create banding patterns based on selective binding (G-banding)
ideogram- banding pattern depiction, w/ bands numbered prox-> distal from centromere
p and q chromosome types: metacentric submetacentric acrocentric
p- short arm
q- long arm
m- central centromere
s-offset centromere; longer and shorter arm
a- centromere is near the end, with “stalk” rRNA- producing DNA and “satellite” region in the nub
noting chromosome abnormality
general format:
total # of chromosomes,
gender chromosomes,
type of mut (loc of mut)
\+ additional chromosome del inv dup ins r (ring)
aneuploidy
loss/gain of selected chromosomes usually fatal)
often due to selective meiotic disjunction
specifically maternal meiosis 1
polyploidy
extra copies of all chromosomes (triploidy);
almost always fatal
complete meiotic disjunction
2 sperm + 1 egg, or a diploid sperm/egg
mosaicism
when a zygote contains 2 cell lines differing in chromosome number
- post-zygotic mitotic event results in chromosomal abnormality
- affects various tissues, depending on nature of abnormality
- can be poly or aneu-ploidy mosaic, but generally less severe than a complete poly/aneu
3 common trisomies
trisomy 13, 18, 21
Patau Syndrome
AKA Trisomy 13
most clinically severe of trisomies polydactylity CNS abnormalities omphalocele (GI organ herniation outside abdomen) renal dysplasia congenital heart disease
Edwards Syndrome
AKA Trisomy 18
SGA
rocker bottom feet
clenched fists
congenital heart disease
hypertonicity (clenched hands, narrow hips)
severe CNS abnormalities, severe retardataion
Down Syndrome
AKA Trisomy 21
most common survivable trisomy congenital heart disease hypotonia GI abnormalities early-onset Alzheimer's
2 common mech’s of chromosomal structural rearrangements
1- dsDNA break and repair by NHEJ (info lost)
2- crossing over between repetitive DNA sequences. this can delete segments of a stretch, can delete on one and duplicate on another, invert, reciprocally translocate, etc.
balanced vs unbalanced structural rearrangements
balanced- normal, but rearranged, complement of chromosomal material. often phenotypically neutral
no gain or loss
unbalanced- abnormal chromosome content. often phenotypically abnormal
3 types of balanced rearrangement
alternate segregation
inversion- ds segment flipped paracentric- excl centromere potential to have dicentric and acentric outcomes pericentric- incl centromere chromosome has to loop in meiosis ex. Rec8 infants- term births; wide face
reciprocal translocation-
break/reform create recombination of 2 non-homologous chromosomes
observed in ~1/500
creates “quadravalance”- 4 homologs align instead of 2
disease states- chronic myelogenous leukemia
lethal risk 5-10%
Robertsonian translocation-
2 acrocentric long arms fuse; you lose the p arms
chr count goes down by 1, and can give you DS w/o Trisomy
21 (ex. Chr 14 stuck w/ Chr 21)
(13, 14, 15, 21, 22 are acrocentric)
leads to potentially giving your children a trisomy
4 types of unbalanced rearrangement
adjacent segregation
deletion-
1- del seg on 1 chromsome arm- terminal deletion
2- del seg contains centromere- interstitial deletion
duplication- generally less harmful than deletion
isochromosomes-
1 missing arm; other has mirrored itself to replace the missing arm
most common on X chr, sometimes 21
(100% of viable offspring are abnormal, since it’s either three or a single Chr 21)
marker (ring) chr’s-
an interstitial deletion frag becomes circ and is stably transmissible to offspring, due to its containment of the centromere
family risks with balanced translocations leading to unbalanced progeny
most chromosomal abnormalities aren’t likely to recur, but if the mother has the translocation, it’s more likely the child will be unbalanced
some stable rearrangements are transmissible
ranges from 0-30%
risk of unbalanced progeny is low due to: size of exchanged material whether genes are involved (DGAP) tolerated mono/trisomies sex of carrier
the most common contiguous gene syndroms in humans
deletion or duplication of 22q11.2
a disorder due to overexpression or deletion of multiple gene loci that are adjacent to e/o
ex. velocardiofacial syndrome and DiGeorge Syndrome
define epigenetics and how modifications may affect gene expression
epigenetics- heritable changes in gene expression that occur without a change in DNA sequence
ex. patterns of reversible post-translational modifications of histones and pattern of DNA methylation
genetic imprinting and its molecular basis
small subset of genes that are inherited in a transcriptionally active state from one parent and transcriptionally inactive state from the other parent
clinical interpretation- we’re normally hemizygous for all imprinted genes, so we’re particularly vulnerable at all of those genes since there’s no backup
act/inactivation seems to depend on methylating CpG islands in promoter regions of particular genes
1st- meiosis
2nd- erasure
3- sex specific gene silencing (myelination)
4- fertilization
3 rules for epigenetic DNA methylation
1- modification must be est during gamete genesis (all maternal (and paternal) must be imprinted the same way
2- modification must be stably maintained in somatic cells (which will contain half paternal/maternal
3- modifications must be reversible so that they can be reset during gametogenesis to transmit the appropriate sex-specific imprint to progeny (ex. if they’re female, maternally methylated; males are paternally methylated)
genetic imprinting with Prader Willi Syndrome and Angelman Syndrome
both have a deletion on chromosome 15
PW= 15q11-q13
-maternally activated region on maternally inherited 15 (inactive= Angelman)
-paternally activated region on paternally inherited 15 (inactive = PW)
imprinting pattern is determined by your parents, NOT your gender
uniparental disomy with PW and Angelman Syndrome
one gamete has 2 copies of a chromosome
if this gamete fuses w/ another normal gamete, the zygote will be trisomy for that chromosome
if zygote has nondisjunction in an early mitosis, it may continue w/ normal chr # but has both chromsomes from 1 parent….
15 maternal disomy= PW
15 paternal disomy= Angelman
PW= 70% deletion; 25% disomy
Angelman- 70% deletion;
2 of the most common leukemia translocations
CML and APML
chronic myeloid leukemia CML- night sweats, fatigue, weight loss, anemia, large/lobulated cells
translocation w/ 9 and 22 (BCR/ABL rearrangement)
treat w/ Gleevec- tyrosine kinase Inhibitor
acute pro-myeloid leukemia APML- auer rods, excess bleeding (teeth), incr blood blasts
translocation w/ 15 and 17 (PML/RARA rearrangement)
treat w/ Vit A for immediate remission
childhood B-cell leukemia
AKA ALL
high hyper-diploidy revealed by chromosome and FISH analyses (hypodiploidy doesn’t have good prognosis)
pain in extremities;
abdominal distention;
high blast count in peripheral blood but none in CSF;
fever;
irritability;
scattered bruising
6 types of FISH probes
centromere- “cen”
(enumeration- prenatal trisomy, etc) ALL, p53 cancer
locus-specific- “LSI”
gene deletion/duplication
dual fusion/fusion- “DF/F”
translocations
MCL and APL leukemias
break apart- “BAP”
rearrangement + translocation
MLL cancer
Whole chromosome paint- “WCP”
FISH basic definition
fluorescence in situ hybridization
method to examine subtle deletions or changes in chromosomes that may not be picked up by banding patterns alone:
small deletions
test host vs donor marrow cells after transplant
can look at large # of cells at once
usually done after prelim chromosome dye banding
chromosomal microarray analysis CMA roles and limits
can detect genomic deletions (200kb) or duplications (400kb), but NOT translocations
uses DNA oligomer probes to interrogate for SNPs
reveals info on intensity and runs of homozygosity > 5Mbs (reporting threshold 10Mbs), possibly revealing autosomal recessive conditions
investigates whole genome simultaneously via DNA amplicfication and labeling
deletions/dups w/o a phenotypic consequence can’t be detected at the chromosomal level
translocation can’t be detected- whole genome is interrogated, not sensitive to location; only detects gains and losses
lab test algorithm for children w/ learning disorders, developmental delays, autism, dysmorphic features, failure to thrive
run microarray to see whole genome and chromosomes (high-res w/ chr banding)
aCGH to detect deletions/dups
FISH w/ specific probes
then compare to gene report
Down Syndrome- 3 chromosomal abnormalities
Trisomy 21- 95%;
nondisjunction or error in maternal meiosis
unbalanced translocation between Chr 21 and another acrocentric Chr- 3-4%; important to check parents’ karyotypes
mosaic Tri 21- 1-2%
mix of normal and Tri 21 cells
typically more mild phenotype
testing for Down Syndrome
genetic testing- timing of results for karyotype and FISH (more important for other trisomies)
FISH- usually looking for trisomy; results within hours
Karyotypic analyses to confirm suspicions: amniocentesis to look at prenatal chromosomes (1.5-2 weeks); early 2nd trimester
chorionic villous sampling- fetal tissue attached to uterine wall; (quicker results); after 1st trimester
fetal ultrasound- look for webbed neck; short femurs; nuchal fold translucency
blood screens- look for fetal blood markers
Down Syndrome Phenotype
flattened occiput- bracheocephaly midface hypoplasia (incomplete midface dev) epicanthal folds (corners of eyes) ears small and set low in head bilateral transverse palmar creases accentuated space between 1st and 2nd toes hypotonia (low muscle tone) abnormal tooth development GI tract problems normal tongue, but small oral cavity
Down Syndrome medical problems
Congenital heart disease
commonly- AV canal (hole between chambers- surgery)
GI- esophageal atresia (immediate surgery) dudodenal atresia Hirshprung's constipation feeding problems GERD Celiac disease
endocrine problems- autoimmune disorders thyroid disease (hypthyroidism) insulin dependent diabetes alopecia areata reduced fertility (normal puberty)
ophthalmological- blocked tear ducts myopia lazy eye nystagmus (giggly eyes) cataracts
hematologic issues
inc risk of leukemia
iron deficiency anemia
ENT problems ear infections deafness nasal congestion enlarged tonsils and adenoids (obstructive sleep apnea)
orthopedic problmes
hips
joint sublaxation
atlantoaxial sublaxation
Down Syndrome developmental and behavioral phenotype
developmental issues-
hypotonia affects gross motor development
intellectual disability spectrum
speech problems (sign language)
psychiatric issues-
depression
early Alzheimer’s
Autism- 1/10 patients
neurologic problems-
hypotonia spectrum
seizures, esp infantile spasms
Prader Willi Syndrome chromosome abnormalities
DEL on a paternal 15q11-q13 maternal disomy (gamete has 2 copies of a chr), leading to zygotic trisomy (could have early mitotic nondisjunction and have normal chr #, but 2 from same parent- mom)
70% from paternal gene del
25% from maternal disomy
Diagnose PW
made with FISH or microarray
methylationg tests on maternal and paternal alleles
imprinting regions and disorders on Chr 15
PW and Angelman syndromes both have deletions on Chr 15
2 imprinted regions on each of your 2 chromosomes- one is maternally and other is paternally activated
deactivating paternal region on paternal chr= PW
deactivating maternal region= Angelman
imprinting pattern is dependent on your parents, not your gender
Prader Willi phenotype
infancy-
hypotonia and dysmorphic features, almond eyes, undescended testicles, light pigmentation, feeding issues
toddler- feeding problems reverse; persistent hunger
PW medical problems
early failure to thrive and feeding problems- reverse to hyperphagia and weight gain; growth hormone treatments to promote height and hinder obesity
ophthalmologic problems common, esp nystagmus (jiggly) and strabismus- (lazy) eyes
ortho- scoliosis
resp- obstructive sleep apnea
PW developmental and behavioral phenotype
mild to mod dev delay leading to intellectual disabilities as adults
behavioral issues are common
other abnormalities associated with Chromosome 15 abnormalities
Angelman- associated w/ deletion on maternal chromsome 15
mildly dysmorphic facial features which evolve w/ age;
hypotonia in infancy, progressing to spasticity
intellectual disabilities
seizures
autism
marker chromosmes- inverted dup (autism, NOT dysmorphic; often hypotonic; seizures common)
interstitial duplications- dup of part of chr (partial trisomy)
phenotype only if derived from mother (autism, NOT dysmorphic, seizures common, hypotonia common in infancy)
linkage disequilibrium between patients w/ autism and polymorphisms in the GABAa-b3 locus (2 15’s put together?)
pharmacogenetics vs
pharmacogenomics
genes vs genome
pharmacogenetics- study of how variance in a single gene influences variability in drug response, usually based on prior knowledge of drug action pathways
pharmacogenomics- study of how variance across multiple genes influences variability in drug response, usually not based on prior knowledge of drug action pathways
pharmacodynamics vs pharmacokinetics
2 major physiologic responses to drugs
pharmacodynamics- response of drug binding to its targets and downstream targets (receptors, enzymes, metabolic pathways); ACTION of a drug once it reaches target
pharmacokinetics- rate at which the body absorbs, transports, metabolizes, or excretes drugs on their metabolites
Phase 1 and Phase 2 in drug metabolism
Phase 1- “first pass” metabolism; hydroxylates drug, usually by cytochrome P450 enzymes in the liver
Phase 2- conjunction rxns; glycosylation or acetylation to deactivate drug, make it more soluble, and excrete it faster
central role of CYP450 enzyme sys in drug metabolism
3 families that break down 90% of all drugs
while most CYP genes are important in the rate of inactivation of a drug, in some cases the CYP gene(s) is required to activate a drug
classic example- CYP2D6 activty needed to convert inactive codeine to active morphine
diff combo’s of 2 chromosomes give you normal, poor, and ultrarapid/ultrafast phenotypes
substrates, inhibitors, and inducer of
CYP450 gene CYP3A
sub- felodipine and cyclosporine
inhibitors- ketoconazole, grapefruit juice
inhibitor- rifampin
substrates and inhibitors of
CYP450 gene CYP2D6
sub- tricyclic antidepressants and codeine
inhibitors- quinidine, fluxotine, paroxetine
substrate and dosing of CYP450 gene CYP2C9
sub- Warfarin
overdose- clotting
underdose- bleed out
start w/ 5mg/day and adjust
NAT gene substrate
isoniazid for tuberculosis
TMPT gene substrates and comments
sub- 6-mercaptopurine and 6-thioguanine
can kill a child w/ ALL
classic pharm mech that can be fatal if ignored
G6PD gene substrates, mech, and comments
sub- sulfonamide, dapsone
mech- x-linked enzyme
deficient individuals subject to hemolytic anemia after drug exposure
VKORC1 gene substrate, and comments
sub- Warfarin
blood thinner
prescribed to >20 mil patients annually
population genetics and relevance of “population field”
the study of allele frequencies and changes in allele frequencies in populations
nuclear DNA >99% similar in humans
“polymorphism” refers to any common genetic variant of an allele- it occurs in greater than or equal to 1% of pop
mutation rate for autosomal dominant- direct and indirect methods
direct- assuming 100% penetrance, count number of new cases with no family history and divide by pop TIMES 2 (for alleles)
indirect- reproductive fitness is 0, so all cases represent new mutations; incidence rate is 2x the mutation rate
autosomal recessive less likely to be affected by fitness and selective criteria
how physicians managing genetic diseases could affect prevalence of genetic diseases
theoretically improve fitness and health; alter chances for reproductive success, so mutation rates for disease X may increase, depending on inheritance and severity
recessive- mutant allele increases are slow
dominant/x-linked- rates could be higher
biological advantages of sexual reproduction
diploidy- protects against effects of some mutations (still 1 working copy)
recombination- creates new combos of haploid genes in germ line
sex- allows random chrosomal assortment by combo of haploid cells
sex- gender-dependent epigenetic imprinting
permits rapid evolution and increases survival via genetic variability
sexual dimorphism allows for division of labor and cooperation
x chromosome inactivation and implications
all diploid somatic cells have a single active X chromosome
normal females- one is inactivated in every cell, and the other is a barr body (mosaicism)
inactivated via methylation and histone modification
XIST region on inactive X transcribes an RNA that coats X to attract methylators and HDACs
(10-15% Barr body genes are still transcribed)
non-random X inactivation
occurs when an X chromosome is abnormal, so abnormal X is preferntially inactivated; almost all cells have same abnormal X inactivation due to non-viability of cells with abnormal X
genetic regulation of sexual differentiation-
WT gene, SRY, MIF
WT gene directs the differentiation of embryological genital ridge (for gonads)
gonadal differentiation is dependent largely on whether or not genes promoting testes are present (SRY gene on Y chromosome)
SOX9 gene (also interacts w/ SRY), SF1, and DAX1 other important genes
Mullerian inhibiting factor (MIF)- allows form. of ductus deferens, etc from mesonephric ducts
true hermaphroditism vs. pseudohermaphroditism
true- 46XX /46XY
show ovaries, testes, partial uterus
pseudo- ambiguous external genitalia but normal ovaries or testes (NOT both)
sex reversals
XX males in which the Y chromosome has translocated autosomally
XY females in which regions of the Y chromosome have been deleted or certain sex-developing genes on the X chromosome have been duplicated
45XO Turner Syndrome
normal early female gonadal development in utero, but degerneration of developing ovaries later in fetal life
clinical features- short height; perceptual disorders; coarction of the aorta; fused kidneys
mostly driven by meiotic nondisjunction events
47XXY Kleinfelter Syndrome
develop as anatomic males, but have degeneration of gonads.
infertile, low levels of testosterone development
clinical- tall stature, gynecomastia (breasts)
mostly driven by meiotic nondisjunction events
Androgen insensitivity
46XY
presents as non-menstruating females
does not result in a uterus (testes still produce working MIF even if testosterone isn’t able to affect development)
clinical apporach to disorders of sexual differentiation
1st day of life:
- obtain FISH for sex chromosomes and a karyotype (or CMA)
- order hormone studies (LH, FSH, testosterone, dihydrotestosterone, +/- AMH)
- consider US study (gondads and uterus)
- consider consult w/ specialized team (endocrine, genetics, urology, psych)
issues to be considered: underlying genetics family cultural/social perspective medical and surgical outcomes risks for tumor development fetal brain development in context of hormone exposure and future gender identity future sexuality future fertility
multifactorial inheritance
combo of genetic variants and nongenetic factors
spectrum of disease- simple Mendelian to extremely complex multifactorial; tend to aggregate in families but don’t follow simple inheritance modes
diseases w/ characteristics not explained by the genotype at the causative locus; and diff alleles at the same gene can result in diff levels of severity
complex traits characteristics- incomplete penetrance variable expressivity phenocopies heterogeneity
incomplete penetrance- not everyone w/ genetic variance develops the disease (type 1 diabetes)
variable expressivity- people w/ same genetic variant have different disease characteristics (age of diagnosis)
phenocopies- people w/ same clinical presentation, but for reasons that aren’t primarily genetic (ex. thalidomide-induced limb malformations vs genetically induced)
heterogeneity- same or similar diseases caused by different alleles at 1 location or alleles at alleles at different locations in one gene or among many genes
heterogeneity
allelic vs locus
allelic heterogeneity- different alleles in same gene result in same OR different traits
(cystic fibrosis- lots of alleles lead to CF, variable severity)
locus heterogeneity- variants in different genes result in very similar clinical presentation
(Alzheimer’s- mutations in 1, 14, 21 all lead to same presentation of Alzheimer’s)
multifactorial inheritance disease examples
cystic fibrosis Alzheimer's some cancers diabetes 1 and 2 inflammatory bowel disease asthma Schizophrenia hypertension cleft lip/palate rheumatoid arthritis
strategies to determine importance of genetic vs non-genetic factors contributing to variations in complex traits
epidemiologic twin, adoption, and immigration studies
twins- mono vs dizygotic
adoptive vs biological siblings, or biological siblings raised apart
examine disease freq and risk patterns in relatives
(lambdaS= risk of disease in siblings of affected/risk of disease in general pop)
heritability and difficulties w/ quantifying role of genetics in populations and individuals
heritability- proportion of total variance in a trait that is due to genetic variation
high h^2= differences are more due to genetics (low= environ)
divide it up based on what you think is genetic and what is environmental
roles of these 2 factors vary so much that it’s hard to lay down strict guidelines for genetic markers and disease predispositions