• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/154

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

154 Cards in this Set

  • Front
  • Back
Tuberous Sclerosis
TSC1, TSC2 genes
AD, 2/3 de novo
tumor supressor loss of function
prenatal - childhood onset
skin - hypomelanotic macules, facial angiofibromas, shagreen patch, ungula fibroma
CNS - astrocytoma, cortical tubers
renal - angiomyolipomas
heart - rhabdomyoma
eye - retinal hamartomas and achromic patches
Von Hippel Lindau
VHL (3p25)
AD, 20% de novo
null, truncated, missense mtns
tumor suppressor LoF
childhood onset
renal cell ca (40%) - greatest source mortality and morbidity
hemangioblastomas (cerebellum, retina, spinal cord)
pheo (ass'd with missense mutations, not with null mutations)
Hereditary paraganglioma-pheochromoctyoma
SDHD, SDHC, SDHB
new syndrome!
AD, de novo, parent of origin effects
missense, deletions
paragangliomas
pheochromocytomas
tests: epinephrine, norepinephrine, dopamine
Bloom syndrome
BLM
AR
AJ founder mtn - 2281del6ins7 - 1/100
chromosomal instability, hyper-recombination
pre, postnatal growth retardation
microcephaly
skin - photosensitive, butterfly rash
ca predisposition - multiple, esp colon
normal IQ, some learning disab?
tx - BMT, avoid UV, x-ray
Ataxia-telangectasia
ATM (11q22.3)
AR/incomplete dominant (hets have breast and lung ca risk)
Amish founder mtn
mostly null mtns
onset - childhood
ataxia - cerebellar, progressive, 1-4yo onset
ocular apraxia after 3yo (difficulty moving eyes side-to-side, may turn head instead)
conjunctival telangectasia (also - vitiligo, premature greying, cafe-au-lait)
ca predisposition - mostly blood ca
tests: serum AFP, radiation sensitivity, 7;14 translocation, MRI - dystrophic cerebellum
tx - avoid radiation
Fanconi Anemia
13 complementation groups
FancA - 65% of cases
FancC - AJ mtn - IVS4+4A>T (1/89 carrier frequencey)
FancD1 - BRCA2!
AR, except FancB - XLR
childhood onset
congenital anomalies - radial ray, short stature, renal anomalies, skin hypo/hyperpigmentation
Bone marrow failure (90%)
Ca - AML, other blood ca, head and neck, GU, skin, GI
tests: increased sensitivity (chromosome breakage) to crosslinking agents
tx - avoid x-rays, BMT or stem cell transplant
Xeroderma pigmentosum
XPA (25%), XPC (25%), ERCC2 (15%), POLH (21%)
AR
defect in nucleotide excision repair of UV-induced DNA lesions (thymine dimers)
extreme sun sensitivity - sunburn, erythema, freckling, photophobia
ca - 1000x risk of skin and eye cancers; internal neoplasms
neuro (30%) - progressive degeneration - microcephaly, SNHL, cognitive impairment
tests - cellular UV hypersensitivity
Cowden (PTEN hamartoma tumor syndrome)
PTEN (10q23) (85% yield)
AD, de novo
tumor suppressor LoF
allelic - bannayan-riley-ruvalcaba, proteus
macrocephaly
trichilemmomas*, papilomatous papules
ca - breast (25-50%), thyroid (usually follicular*, 10%), endometrial
colon - hamartomatous polyps (40-50%)
Peutz-Jeghers syndrome
STK11 (19p13) (aka LKB1) (50% of cases)
AD, 20-50% de novo
40% have deletions (higher than other tumor supressor syndromes)
distinct polyps - "branching tree," through GI tract, esp. small bowl
pigmented spots on lips, buccal mucosa (dark blue to brown)
ca - breast (54%), colon (39%), pancreas (36%), stomach (29%), uterus, testes,
Lynch (aka HNPCC)
MSH2 and MLH1 (80% of cases), MSH6 (7-10%), PMS1, PMS2 (both rare)
AD
missense, deletions (MLH1, MSH2)
ca - colon (80%), endometrium (20-60%), stomach (11-19%), ovary (9-12%)
dx'c criteria - amsterdam (3 fam members, 2 generations, 1 ca <50yo)
tests - MSI in 90% of lynch tumors (but also 15% of sporadic), IHC identifies which protein/gene
better prognosis than other CRC
2/3 of tumors occur in proximal colon
familial adenomatous polyposis (FAP)
APC 5q21-22
AD, 20-30% de novo
tumor suppressor LoF (gatekeeper)
most mtn truncating (LoF); 10% deletions (so sequencing isn't enough)
dx'c criteria: >100 polyps or 10-100 and +FHx
CRC ca - 100% penetrant, mostly affect recto-sigmoid region
rare other ca - upper GI (5-10%), pancreatic (2%), papillary thyroid (2%), hepatoblastoma (<2%), brain (medulloblastoma)
rare benign - desmoid tumors of abndomen (painful, recur), osteomas of jaw and skull, epidermoid cysts,
CHRPE
100% penetrant
tx - colectomy, upper GI screening
Gardner syndrome
APC 5q21-22
AD
FAP +
osteomas
soft tissue tumors
desmoid tumors of abdominal wall
epidermoid cysts
Turcot syndrome
APC 5q21-22, Lynch genes
FAP +
CNS tumors - medulloblastomas
aFAP
APC 5q21-22, especially 3' and 5'
FAP -
fewer polyps (avg 35 vs >100)
older onset
MYH associated polyposis (MAP)
MYH
AR (vs. related AD syndrome)
Y165C, G382D - 85% of northern european mutations
polyposis, but few polyps (<100)
later onset (20-50yo)
hets have increased colon ca risk
BRCA1
17q21
AJ mtn - 185delAG (1%), 5382insC (0.4%)
Dutch - 3 large del
5-10% (?) del/dup/rearrangement
ca - breast (40-87 ), ovarian (16-63), prostate (25)
90% of hereditary ovarian cancers are due to BRCA1 mutations
breast ca path - triple negative
BRCA2
13q12.3
ovarian ca cluster (exon 11)
AJ mtn - 617delT (1.2%)
iceland mtn - 999del5
5-10% (?) del/dup/rearrangement
path - nothing unique (?)
ca - breast (28-84), ovarian (27), male breast (6-14), prosate (20), melanoma, pancreas, other rare ca
NF1
NF1 (17q11.2)
AD, 50% de novo (v. high mtn rate)
80% of mtn - truncating
tumor suppressor LoF (gatekeeper)
onset - prenatal to childhood
dx'c criteria: >=2 of 6+ CAL, 2+ neurofibromas, 1 plexiform neurofibroma, axilary or inguinal freckling, optic glioma, 2+ lisch nodules, osseous phenotype, affected FDR
up to 50% have MR or LD
optic glioma (50%) (benign)
100% penetrant but highly variable
Li-Fraumeni
p53 (17p13) (70% of cases)
AD, de novo (?%)
tumor suppressor LoF (DNA repair and cell cycle regulation)
dx'c criteria: proband with sarcoma <45yo & FDR w ca <45yo & FDR or SDR w ca <45 or sarcoma at any age
ca (50% by 35yo, 90% lifetime for women, 70% lifetime for men) - SBLA - sarcoma, breast (v. early onset, avg age dx 32yo), brain, leukemia, adrenocortical ca in childhood
retinoblastoma
RB1 (13q14)
AD, 80% de novo (?)
allelic het - missense, deltions, insertions, hypermethylation of promoter
40% of all cases are due to mutation
hereditary cases - more likely to be bilateral (but not always), younger dx (<1y vs. 1-2y)
risk for other ca later in life (70%) - sarcomas, radiation therapy increases ca risk
incomplete penetrance (esp. if residual activity)
Men2a
RET (10q11.2)
AD, <5% de novo
exon 10 & 11 (95%) -
allelic - hirschprung (LoF)
GoF proto-oncogene
ca - medullary thyroid (60%) in early *adulthood* (vs. 2B)
-pheochromocytoma (50%)
-*parathyroid adenoma/hyperplasia* (20%) (vs. 2B)
represents 20-50% of medullary thyroid cancer
screening: prophylactic thyroidectomy by 6yo, pheo and PTH screening annually after 6yo
Men2b
RET (10q11.2)
AD, *50% de novo
exon 15 (95%) - one mtn v. common, so low genetic heterogeneity
allelic - hirschprung (LoF)
GoF proto-oncogene
ca - medullary thyroid (60%) in *childhood* (bilateral, multicentric)
-pheochromocytoma
*muccosal neuromas of tongue and lips
*marfanoid habitus, big lips
*ganglioneuromas of GI tract
*NO parathyroid hyperplasia (vs. 2A)
accounts for <2% of MTC
tx/screening: prophylactic thyroidetomy by 1yo, pheo screening annually after 3yo
congenital adrenal hyperplasia
12-hydroxylase deficiency
CYP1A2 (6p21.3)
AR
1% of mutation are de novo
missense mutations, common dels
most common form of this disorder
1/300 in Yupik Eskimos of Alaska
~1/50 carrier rate in US
defect in biosynth of glucocorticoids and mineralcorticoids, leads to over production of androgens
classic - null alleles, severe, 3/4 salt wasting
nonclassic - one not null allele
sex-dependent pheno
females - most comon cause of pseudohermaph.
males - early virilization
salt wasting -> neonatal death if not treated
tests: on NBS panels
hereditary hemochromatosis
HFE (6p21.3)
AR
Cys282Tyr/Cys282Tyr (90%), Cys282Tyr/His63Asp (10%)
v. high carrier rate in whites (38%)
His63Asp - lower penetrance
incomplete penetrance, variable expressivity, sex-dependent penetrance
GI mucosa absorbs too much iron
bronzing of skin, diabetes, cirrhosis, cardiomyopathy, fatigue (most frequent complaint)
tests: ferritin (want <300ng/ml), liver function
Factor V Leiden thrombophilia
F5
AD, incomplete dominant
Arg506Gln, GoF b/c resistant to deactivation by protein C
incomplete penetrance, genetic & environmental modifiers
2-15% of Europeans
Genetic factors in venous thrombosis
Factor V Leiden (12-14%)
Prothrombin mutations (6-18%)
Antithrombin III or protein C or S (5-15%)
Hemophilia B
F9 (Xq27.1-q27.2)
XLR, but 10% of female carriers affected
hemarthrosis, intracranial bleed
deep muscle hematomas
excessive bruising
severity inversely correlates with activity
"Leyden" - variant - mtn in promoter - severe in childhood but resolves
tets: prolonged PTT (pro-thrombin time)
tx: replace missing factor
Hemophilia A
F8 (Xq28)
XLR, some carriers manifest
allelic hetero - deletions, insertions, inversion, point
common mtn - (50%) intron 22 inversion del of C term due to aberrant homologus recomb - test with PCR or southern
hemarthrosis, intracranial bleed
deep muscle hematomas
prolonged, renewed bleeding after trauma
excessive bruising
severity inversely correlates with activity (<30% is lower limit of nml)
tx: IV replacement of missing factor
Beta-Thalassemia
HBB (11p15.5)
AR
genetic heterogeneity (15 mtn - 90%)
trait - het, asymptomatic, mild anemia
major - homoz, hemolytic anemia postnatally
tests (carriers) - reduced MCV, Hb electrophoresis - elevated Hb A2, Hb F
carrier rates: mediteran'n 1/25, SE asian 1/30
tx - transfusion
Alpha-Thalassemia
HBA1, HBA2 (16pter-p13.3)
AR
90% deletions, 10% point mutations
HB Bart: --/--, hydrops fetalis, perinatal death (SE asia only)
HB H: --/a-, moderate anemia, etc.
trait: --/aa or -a/-a, low MCV, low MCH, mild anemia
carrier: -a/aa - no phenotype
tets (carriers): MCV, MCH - both reduced; HB eletrophoresis - normal
tx: Hb H - transfusion during hemolytic crisis
carrier rates: SE asian, asian 1/20 cis; medit'n, african-am'n, carribean, west african 1/30 trans
Beckwith Wiedeman syndrome
11p15
AD (15% familial, 85% sporadic)
DMR2 (KCNQOT1) loss of meth'n (50%)
paternal UPD 11p15 (10-20%)
DMR1 (H19) gain of meth'n (2-7%)
CDKN1C mtns (40% of familial, 5010% of sporadic)
cytogenetic abnl @ 11p15 (<1%)
11p15 - growth factors, tumor suppressors
9x increase in risk with ART (also for Angelman syn but not for PWS)
key features - omphalocele, macroglossia, macrosomia, embryonal tumor (wilms, hepatoblastoma), neonatal hypoglycemia, ear creases/pits
Pallister-Killian syndrome
tetrasomy 12p, mosaic isochromosome 12p
47,XY i(12)(p10)
tissue-limited mosaicism - NOT in blood, only in fibroblasts (on karyotype. recently detected by aCGH on interphase cells from blood sample -- culturing causes selection bias?)
profound MR
seizures
hypo- or hyper-pigmented streaks
dysmorph: prominent forehead, sparse anterior scalp hair (localized alopecia), flat occiput, short nose, flat nasal bridge, short neck, coarse facies
congenital diaphragmatic hernia
Gaucher
GBA (1q21)
AR
AJ mtn N370S 1/18 carrier (type 1)
LSD
distinct cells named for the disease (macrophages filled with glucosylcerbroside)
hepatosplenomegaly
nose bleeds, reduced platelets (easy bruising, slow clotting)
bone pain ("crises"), bone deterioration, bone infarcts
type 1: most common, bone, organ, blood, lung, no CNS involvement
type 2: + progressive CNS disesase
type 3: + slower progressive CNS disease
also perinatal-lethal and cardiovascular
type - clinically dx'd
tests: enzyme activity
tx: ERT, substrate reduction, BMT for type 3
CHARGE
CHD7 (8q12.1) (60%)
AD (nearly all de novo, germline mosaicism reported, empiric 1-2% recurrence risk)
coloboma (80-90%)
heart defect
choanal atresia (50-60%)
growth and mental retardation
GU malformations
characteristic ear (90-100%)
cranial nerve anomalies (SNHL, swallowing, palsy) (70-90%)
branciootorenal syndrome (BOR)
EYA1 (8q13.3)(40%), SIX1 (14q23)(rare)
AD
point mtn, dup/del (10%)
ear malformations - outer, middle, inner
deafness - conductive, SNHL, mixed
preauricular pits, tags
banchial fistulae, cysts
facial asymmetry
renal malformations - hypoplasia, agenesis
Bardet-Biedl syndrome
12+ genes, M390R in BBS1 (18-32%), 91fsX95 in BBS10 (10%), BBS (8%), BBS6 (6%)
AR, "triallelic" inheritance
cilia-opathy
truncal obesity, normal prenatal growth
cone-rod dystrophy -- blindness
postaxial polydactyly
hypogonadism, complex GU malformations
renal dysfunction
Antley-Bixler syndrome
(aka POR deficiency?)
POR (7q11.2)
AR
highly variable range of allelic disorders of steroidogenesis
ambiguous genitalia in both sexes
maternal virilization during pregnancy w affected fetus
craniosynostosis, frontal bossing, midface hypoplasia, hydrocephalus
choanal stenosis or atresia
stenotic auditory canals
neonatal fractures, bowing of long bones, joint contracture
renal malforamtions
tests: NBS, low uE3 on mat'l serum screen
Aarskog syndrome
FGD1 (Xp11.2) (7-20%)
XLR (some AR, AD reported)
shawl scrotum
cryptorchidism
hypertelorism
brachydactyly, cervical verterbral abnormalities, pectus excavatum
single simian crease
short stature
MR (30%)
hyperextensibility
Leber hereditary optic neuropathy (LHON)
1178A>G in ND4 subunit of complex 1 of eletron transport chain
largely homoplasic (unlike most mtDNA diseaes)
maternal inheritance
rapid onset blindness - young adult
sex bias - penetrance 50% in males vs. 10% in females (so can appear x-linked)
may also have dystonia
NARP
ATPase subunit 6 gene - point mtns
heteroplamic
maternal inheritance
neuropathy, ataxia, RP, dev'l delay, MR, lactic acidemia
Leigh syndrome (aka subacute necrotizing encephalopathy)
ATPase subunit 6 gene - point mtns
heteroplasmic; diverse etiologies - can be mtDNA or nuclear (AR)
maternal inheritance
early-onset progressive neurodeg'n - hypotonia, dev'l delay, optic atrophy, respiratory abnormalities
cranial nerve abnormalities, respiratory dysfunction, ataxia, hyperintense signals on T2-weighed images of head
onset - infancy to early childhood
progressive, lethal
MERRF
tRNAlys point mtns
8344A>G - common mtn
heteroplasmic
maternal inheritance
onset late childhood to adulthood
progressive myoclonic epilepsy with ragged-red muscle fibers, myopathy, ataxia, SNHL, dementia
some pt have symmetrical lipomatosis (large subcutaneous fat masses, usually around neck)
MELAS
tRNAleu(UUR) point mtn
most commonly 3243A>G
heteroplasmic
maternal inheritance
myopathy, mitochondrial encephalomyopathy, lactic acidosis, stroke-like episodes
variable expressivity - may present as only diabetes and deafness
deafness - mtDNA
progressive SNHL, often induced by aminoglycoside antibiotics
12S rRNA gene - two mtns - 1555A>G, 7445A>G
homoplasmic
maternal inheritance
chronic progressive external ophthalmoplegia (CPEO)
mtDNA
the commone MELAS point mtn in tRNAleu(uur) - 3243A>G
large deletions similar to KSS
heteroplasmic
deletions - sporadic
point mutations - maternal inheritance
progressive atrophy of extracoular muscles, ptosis
pearson syndrome
mtDNA
large deletions
heteroplasmic
usually sporadic, somatic mutations
pancreatic insufficiency, pancytopenia, lactic acidosis, KSS in second decade
Kearns-Sayre syndrome (KSS)
mtDNA
~5db large deletion (most cases), heteroplasmic
generally sporadic, due to somatic mutations
other rare causes - mtDNA point mtn, nuclear genes (AR and AD)
progressive myopathy, progressive external ophthalmoplegia of early onset, cardiomyopathy, heart block, ptosis, atypical retinitis pigmentosa, ataxia, diabetes, elevated protein in CSF, cerebellar ataxia
achondroplasia
FGFR3 @ 4p16.3
~100% of cases due to Gly280Arg (98% 1138G>A, 2% 1138G>C), one of the most mutable nucleotides in genome
AD, 80% de novo - only from father, ass'd with APA
GoF constitutive activation --> inhibits chondrocyte proliferation at growth plate
most common form of dwarfism
rhizomelic, macrocephaly, frontal bossing, midface hypoplasia, trident hand, limited elbow extension
onset: prenatal, only detectable by u/s after 24 wks; detectable by x-ray sooner
normal lifespan except 3-7% die in infancy b/c of central or obstructive sleep apnea
normal intelligence except delayted motor dev't b/c macrocephaly, hypotonia
Alzheimer disease
Early-onset mendelian forms
PSEN1 - 20-70% of early onset familial, fully penetrant
APP - 1-2% of early onset familial, fully penetrant
PSEN2 - <5% of early onset familial, reduced penetrance
early onset: <60yo
dementia, confusion, poor judgement, language disturbance, agitation, withdrawal, hallucinations
beta-amyloid plaques, neurofibrillary tangles, amyloid angiopathy
diagnosis can only be definitively made on autopsy, clinical diagnosis has 80-90% sensitivity, APOE e4 homozygote increases clinical dx sensitivity to 97%
charcot-marie-tooth - most common form
PMP22 duplications or GoF mutations
accounts for 70-80% of all CMT
17p11.2, unequal crossing over of flanking repeats
allelic disorder: HNPP due to deletions, LoF
AD, 1/4-1/3 de novo
abnormal peripneral myelination
onset: <30yo
nearly fully penetrant, but variable expressivity
slowly progressive weakness, atrophy of distal leg muscles, mild sensory impairment
distal weaknes -> foot drop, gait abnormalities, foot deformities (pes cavus, hammer toes)
tests: slow nerve conduction (by 5yo, even if asymptomatic), nerve biopsy, genetic test to confirm dx
Cystic fibrosis
CFTR @ 7q31.2
AR
1/29 carrier rate in caucasians, 1/3200 incidence
deltaF508 (80% of caucasian mutations)
5T - in intron 8, variably penetrant, ass'd with abnl phenotype when cis with R117H, more often when cis with 13TG
carrier panel - 23-25 mtn present in >1/1000 patients, doesn't include polyT, only reflux to that if find R177H
progressive lung disease - chronic infection, progress to end-stage disease
exocrine pancreatic insufficiency (85-95%)
CBAVD, azoospermia (95% of males)
meconium ileus (15-20%)
-20% have rectal prolapse
dx'c criteria: =>1 phenotypic feature AND (2 CFTR mtn OR 2 abnl sweat chloride (>60) OR transepithelial nasal potential difference)
46,XY females
DAX1
duplication of part of Xp that includes DAX1 (Xp21.3)
DAX1 has dosage dependent effect on SRY, excess DAX1 b/c of duplication suppresses normal male-determining function of SRY
DFNB1 - most common AR nonsyndromic prelingual SNHL
GJB2 (connexin26) -98% of cases
common mtn - 35delG (whites), 235delC (asians), 167delT and del35G (AJ)
2% of cases - double het for mtn and GJB6 (connexin30) del
tests: audiometry, dysmorph exam, CT scan to eval for temporal bone abnormalities
genetic test: GJB2 seq'g, including common splice site mtn, test for 2 GJB6 dels
empiric risks - if isolated case to hearing parents and GJB2/6 testing is normal - 14% recurrence risk
Jervell and Lange-Nielson synd
KCNQ1 @11p15.5, KCNE1 @ 21q22.1-q22.2
AR, hets at risk for long QT syndrome (without HL)
congenital bilateral severe-profound SNHL
long QT syndrome
Pendred syndrome
SLC26A4 @ 7q31 (50%)
FOX1I (1%)
AR
bilateral severe SNHL, usually congenital, progressive
temporal bone abnormalities - dilated vestibular aqueducts with or without cochlear hypoplasia (Mondini)
goiter (75%), onset late childhood to early adulthood, normal thyroid
allelic - DFNB4 - no thyroid defects
tests: hearing testing, CT/MRI of temporal bone
genetic testings: SLC26A4 common mtn, sequencing
usher syndrome
11 genes - most common and testing available?
Type I - MY07A (50%), several others
Type II - USH2A (80%), others
AR
Type I - congenital profound bilateral SNHL, congenital balance problems, RP onset pre-puberty
Type II - cogenital mild-severe bilateral SNHL, normal balance, RP onset teens-20s
Type III - progressive later onset HL, progressive balance problems, variable onset RP
tests: hearing tests, electroretinography (ERG), eye exam for pigment changes
Waardenburg synd
PAX 3 @ 2q35
AD, de novo are rare
point mtn (90%), deletions (6%)
highly variable (all features)
type 1 - congenital SNHL, pigmentary abnl (heterochormic irides, white forelock, premature gray, leukoderma), telecanthus, NTD
type 2 - same, no telecanthus
type 3 - 1 + skeletal abnlts (limb hypoplasia or contracture, carpal bone fusion, syndactyly)
type 4 - 1 + hirschprung dis
Jervell and Lange-Nielson synd
KCNQ1 @11p15.5, KCNE1 @ 21q22.1-q22.2
AR, hets at risk for long QT syndrome (without HL)
congenital bilateral severe-profound SNHL
long QT syndrome
Pendred syndrome
SLC26A4 @ 7q31 (50%)
FOX1I (1%)
AR
bilateral severe SNHL, usually congenital, progressive
temporal bone abnormalities - dilated vestibular aqueducts with or without cochlear hypoplasia (Mondini)
goiter (75%), onset late childhood to early adulthood, normal thyroid
allelic - DFNB4 - no thyroid defects
tests: hearing testing, CT/MRI of temporal bone
genetic testings: SLC26A4 common mtn, sequencing
usher syndrome
11 genes - most common and testing available?
Type I - MY07A (50%), several others
Type II - USH2A (80%), others
AR
Type I - congenital profound bilateral SNHL, congenital balance problems, RP onset pre-puberty
Type II - cogenital mild-severe bilateral SNHL, normal balance, RP onset teens-20s
Type III - progressive later onset HL, progressive balance problems, variable onset RP
tests: hearing tests, electroretinography (ERG), eye exam for pigment changes
Waardenburg synd
PAX 3 @ 2q35
AD, de novo are rare
point mtn (90%), deletions (6%)
highly variable (all features)
type 1 - congenital SNHL, pigmentary abnl (heterochormic irides, white forelock, premature gray, leukoderma), telecanthus, NTD
type 2 - same, no telecanthus
type 3 - 1 + skeletal abnlts (limb hypoplasia or contracture, carpal bone fusion, syndactyly)
type 4 - 1 + hirschprung dis
Duchenne muscular dystrophy (DMD)
DMD at Xp21.2
XLR
null mtns - large dels (65%), large dups (5-10%), small dels, insertions, substitutions (25-30%) - nearly all identifiable by molecular testing (multiplex PCR, (formerly southern), MLPA, sequencing)
XLR
onset 3-5yo, proximal weakness then distal, gower maneuver, wheelchair by 12yo, DCM in 100%
cognitive impairment - MR in 1/3
manifesting carrier females
isolated cases - 2/3 from carrier mother, 1/3 de novo (equally from males and females)
germline mosaicism - so if mom not a carrier, 7% recurrence risk (i.e. 14% rate of germline mosaicism)
high mtn rate (10^-4)
carrier testing: for known mtn or linkage (but huge gene so recombination may occur) or by CK (50% penetrant, decreases with age)
-dystrophin recombination rate: 10-12% (v. high, important for linkage)
Becker muscular dystrophy (BMD)
DMD at Xp21.2
XLR, 10% de novo, manifesting carriers (esp DCM)
mtns retain reading frame - large dels (85%), dups (6-10%), point or small mtn (5-10%), unknown (5-10%)
muscle biopsy - some protein present
ambulatory unto 20s, DCM, activity-induced cramping, flexion contractures of elbows, preservation of neck flexor (vs. more severe allelic disorder).
Familial hypercholesterolemia
LDLR @ 19p13.2
AD, incomplete dominant
private mtn, some found mtn
1/500
environmental modifiers - esp. diet
hypercholesterolemia (onset at birth), xanthomas, arcus corneae, atherosclerosis, CAD
homoz - onset in childhood, chol > 600mg/dL
hets - CAD onset in 30s+, chol > 300mg/dL
Fragile X
FMR1 @ Xq27.3
XL, expansion only from mother, expansion risk increases with size and decreases with AGG
nl - 5-40, grey - 41-58, pre - 59-200, full - >200
pre - POF (20%) and FXTAS (?%, <100%)
MR, autistic spectrum, hyperactivity
post-puberty: long face, big ears, prominent chin and forehead, macro-orchidism
connective tissue pheno: hyperflexible, flat feet, MVP, soft velvety skin
testing: PCR (misses full) and/or southern blot (imprecise), methylation testing
Apert synd
FGFR2 @ 10q26
only two mtn - P253R (syndactyly more common), S252W (cleft palate more common)
AD, de novo (APA ass'n)
coronal synostosis, midface hypoplasia
distinct features: bony and soft tissue foot and hand syndactyly (mitten hand), DD/MR (50%)
Crouzon synd
FGFR2 @ 10q26
AD, de novo (APA ass'n)
coronal synostosis,
distinct features: significant proptosis, normal hands, parrot beak nose, mandibular prognathism, progressive hydrocephalus (30%)
variant with acanthosis nigrans Ala391Glu in FGFR3 (not 2!)
Pfeiffer synd
FGFR2 @ 10q26 (most)
FGFR1 (5% of type 1)
AD, de novo (APA assn)
coronal synostosis, proptosis, midface hypoplasia
distinct features: broad, short, medially devaited thumbs and big toes; mild syndactyly, vision and hearing problems
type 1 - rare, less severe, normal IQ
type 2, 3 - more common, more severe - DD/MR, exorbitism, hydrocephalus, seizures, choanal atresia
Thanatophoric dysplasia
FGFR3 @ 4p16.3
de novo, (APA ass'n), germline mosaicism not reported
GoF mtn
Arg248Cys (50% of cases of type I)
Lys650Glu (type II)
micromelia, macrocephaly, perintatal lethal, short ribs and narrow thorax, redundant skin folds along arms, cloverleaf skull (type II, not type I), bowed limbs (type I, not type II),
detectable on u/s (as early as 12-14wk)
Hypochondroplasia
FGFR3 @ 4p16.3
AD, de novo
N540K (70%; C1620A 50%, C1620G 21%), other mtn (10%). i.e. identifiable mtn in 80%
difficult to diagnose
postnatal short stature - childhood presentation (vs. prenatal presentation in allelic disorders)
rhizo- or mesomelic shortening
stocky build, limited elbow extension, brachydactyly, mild joint laxity, macrocephaly, normal facies
mild-mod. MR (rare)
tests: x-ray features
Diastrophic dysplasia
SLC25A2 (DTDST) @ 5q32-q33.1
AR (unlike other similar diseases!)
5 mtn represent 65% of alleles, rare mtn id'd by sequencing get yield up to >90%
sulfate transporter mtn - biochemical analysis esp. useful if not mtn found
x-rays
limb shortening, normal-sized skull, hitchhiker thumbs, spinal deformities, contractures of large joints, early onset osteoarthritis, clubfoot, normal IQ, ear cysts ("cauliflower ears")
Osteogenesis imperfecta
COL1A1 (17q21.33), COL1A2 (17q21.33)
type 1: null mtn, milder phenotype b/c less normal protein (vs. abnormal protein), blue sclarae, brittle bones, no bone deformity, genetic testing 100% sensitive, hearing loss in 50%
type2: abnl protein/dominant negative, perinatal lethal, severe skeletal abnormalities, fractures, dark sclerae, dentinogenesis imperfecta, de novo mtn, genetic testing 98% sensitive
type 3: abnl protein/dominant negative, progressive deforming, fractures, limited growth, blue sclerae, hearing loss frequent, genetic testing 60-70% sensitive
type4: abnl protein/dominant, moderate severity, normal to grey sclerae, deforming, genetic testing 70-80% sensitive
type 7: AR (vs. others - AD, de novo)
EDS classic type (I and II)
COL5A1 @ 9q34.2, COL5A2 @ 2q31
(50%)
?
AD
skin - strongest skin phenotype of all subtypes - hyperextensible, abnl (slow) wound healing, wide atrophic scars, smooth velvety, easy bruising, molluscoid pseudotumors, subcutaneous spheroids
joints - hypermobile, dislocations, subluxations
hypotonia, hernia, chronic pain, aortic dilatation (rare)
biochem testing - not helpful
genetic testing - 50% sensitivty
EDS hypermobility type (III)
?
TNXB (rare)
AD
least severe subtype
joint hypermobility - primary manifestation - dislocations, subluxations, chronic pain, degenerative joint disease
skin - may be mildly affected (soft, velvety, slightly increased elasticity, easy brusing), but only mildly and no fragility
no testing available
EDS vascular type (IV)
COL3A1 @ 2q31 (~100%)
AD (50%), de novo (50%)
major criteria: arterial rupture, intestinal rupture, uterine rupture during pregnancy, family history of vascular EDS
minor criteria: thin, translucent skin, easy bruising, facies (thin lips and philtrum, small chin, thin nose, large eyes), acrogeria (aged hands), hypermobility of small joints...
testing - collagen III biochem (95%)
genetic tesing - COL3A1 sequencing (98-99%)
EDS kyphoscoliotic type (VI)
PLOD1 (?%)
AR (vs. other subtypes)
enzyme defect - crosslinking
skin - friable, hyperextensible, thin scars, easy bruising
progressive scoliosis, often present at birth
joint laxity, generalized
severe hypotonia at birth
scleral fragility, rupture of globe
rupture of medium-size arteries
tests - urinalysis for cross-linking or enzyme analysis in fibroblasts
Stickler syn.
COL2A1, COL11A1, COL11A2
AD, highly variable
ocular - myopia, cataract, retinal detachment
hearing loss, hypermobile ear systems
craniofacial - cleft palate, bifid uvula, micrognathia, robin sequence
joint - mild spondyloepiphiseal dysplasia, precocious arthritis, hypermobility
genetic testing - <100%
Congenital contractural arachnodactyly (beals synd.)
FBN2 @ 5q23-q31
AD
Marfanoid + major joint contracture, camptodactyly (finger contracture), crumpled ears, muscle hypoplasia, kyphosis/scoliosis (severe), aortic dilatation (rare)
rare severe infantile form
genetic testing - 75%
hereditary multiple osteochondromas (aka multiple exostoses syndrome)
EXT1, EXT2
AD, 10% de novo
96% penetrant
onset-childhood
benign cartilage-capped bone growths/tumors at growth plate of long bones, surface of flat bones
small risk of malignant transformation
short metacarpals, leg length inequity, bowed long bones, compression of nerves or blood vessels
genetic testing - sequencing 70%, deletion analysis 20%
Glucose-6-phosphate dehydrogenase deficiency
G6PD @ Xq28
XL(R) - manifesting female carriers with mild disease
heterozygote advantage - malaria (so prevalent in mediteranean basin, africa, asia - 5-25%)
allelic heterogeneity
african variants milder than medit'n variants
neonatal jaundice
hemolytic anemia
triggers: oxidative stress (b/c leads to glutathione deficiency) - infections, oxidative drugs, toxins, fava beans
testing - enzyme activity in erythrocytes
Hirschsprung disease
RET (isolated - 50% familial, 25% sporadic), EDNRB, EDN3, GDNF, NRTN
genetic heterog. - allelic and locus
AR, AD, multigenic
variable expressivity - intra and interfamilial, isolated vs. syndromic, short vs. long segment
reduced and sex dependent penetrance (males>females, incidence - 4:1 m:f)
empiric recurrence risks dependent on proband's sex and phenotype
congenital intestinal aganglionosis
neurocristopathy (neural crest cell dis.)
constipation, obstruction, failure to pass meconium, abdominal distension, enterocolitis
dx based on histopathology
Holoprosencephaly
chromosomal (50%)
syndromic
nonsyndromic - SHH (30-40% familial AD, 5% overall), TGIF, PTCH, TMEM1, HPE6 (all rare)
diverse etiologies - chromosomal (50%), syndromic, nonsyndromic monogenic, teratogenic (maternal diabetes)
recurrence risk depends on etiology
AD familial nonsyndromic
-variable expressivity
-reduced penetrance (70%)
onset: prental
ventral forebrain maldev't, developmental delay
dysmorphisms - cyclopia, hypotelorism, an/microphthalmia, absent frenulum, single incisor,
males: female (1:2) (vs. most birth defects)
syndromes - SLO, meckel-gruber
tests - MRI/CT, karyotype, prenatal u/s (dx at 16-18wk) sequencing
Huntington disease
HD @ 4p16.3
CAG repeats - polyglutamine - in exon 1
AD
anticipation - expansion in spermatogensis only
nml 10-26; premutation 27-35 (3-10% chance expansion and transmission from a male); reduced penetrant mtn 36-39; fully penetrant mtn >40; juvenile >60
toxic gain of novel property
mvmnt abnormalities (voluntary and involuntary)
cognitive abnormalities (all aspects of cognition)
psychiatric abnormatlities (personality changes, affective psychosis, schizophrenia)
bhvr disturbances (social disinhibition, aggression, outbursts, apathy, sexual deviation, increased apetite)
mean dx: 35-44yo
mean death: 54-55yo
brain atrophy, neuronal dysfunction
tests: PCR-based (<115 repeats), southern only for rare >115 or homozygous
juvenile (5-10% of cases) - more severe presentation, rapid decline, seizures (<10yo)
Miller-Dieker syndrome
17p13.3 deletion
70% cytogenetically detectable, others need FISH or CGH
lissencephaly type 1 or 2
death by 2yo
hypotonia, little-no development, seizures, FTT
microcephaly
dysmorphic - bitemporal narrowing, prominent forehead, vertical furrows in forehead, short nose, upturned nares, protuberant upper lip, thin vermillion border, small jaw
diff'l: isolated lissencephaly due to LIS1 del (35%) or mtn (15%)
SBMA (Kennedy's disease)
AR @ Xq11-12
XLR
CAG (poly-gln) repeats in exon 1
normal <34; (no premutation); reduced penetrance 36-37; full penetrance >=38
progressive neuromuscluar disease - proximal muscle weakness and atrophy, fasciculations
difficulty with swallowing, speaking ('bulbar')
gynecomastia, testicular atrophy, reduced fertility (mild androgen insensitivity)
Myotonic dystrophy (1)
DMPK @ 19q13.2
AD, anticipation - with biggest increases in oogenesis
CTG repeats in 3'UTR
normal: 5-34; premutation: 35-49; full mutation >50
mRNA toxic gain of function
mild (50-150) - mild myotonia, cataract
classic (100-1000) - distal muscle weakness and wasting, myotonia, cataract, balding, arrhythmia
congenital (>1000, ?>2000) - infantile hypotonia, congenital severe generalized weakness, respiratory insufficiency and early death, MR is common
mitotic instability -> variability
tests: EMG, serum CK, muscle biopsy, slit lamp exam
genetic test: PCR for up to 100, then southern for larger
Friedrech's ataxia
FXN @ 9q13
GAA repeat expansion in intron 1
interferes with transcription (LoF)
AR
5% are compound hets for a point mtn and repeat expansion
nomral: 5-33; premutation: 34-65; borderline/reduced penetrance: 44-66; full mtn: 66-1700
involved in iron metabolism and protecting against oxidative stress in mitochondria
onset: 10-15yo
progressive ataxia, depressed tendon reflexes, spasticity of lower limbs, dysarthria, scoliosis, pes cavus, optic nerve atrophy, loss of position and vibration senses, HCM, diabetes
Limb-girdle muscular dystrophy (LGMD)
CAPN3, DKRP, LMNA, SGCA, SGCB, SGCG, DYSF
most AR, some AD
heterogeneous group, phenotype depends on genetic subtype
must distinguish from dystrophinopathies
muscle biopsy - IHC to identify subtype and then targeted genetic testing
progressive, loss of ambulation (20-30y?)
weakness and wasting in limb musculature, proximal > distal
heart and bulbar spared in most subtypes
SMA (spinal muscular atrophy)
SMN1 (aka telSMN), SMN2 @ 5q12.2-13.3
AR, 2% have one de novo mtn
95-98% homozygous del of exons 7 and 8 in gene 1
2-5% compound hets for 7/8 del and a point mtn in gene 1
multiple copies of gene 2 can make phenotype milder
variable spectrum, no definitive genotype-phenotype corr'n
arthrogryposis, progressive muscle weakness (degneration and loss of anterior horn cells), peripheral nerve hypomyelination
type O - prenatal onset, type I - <6mo onset, type II - onset >6mo, type III - onset 2-3yo, type IV - adult onset
carrier testing: recommended by ACMG, PCR dosage assay, complicated by extra copies of gene 1 and point mtns
SCA (spinocerebellar ataxias)
many genes
AD, AR, XL
group of disorders, each characterized by causative gene and mode of inheritance
slowly progressive incoordination of gait, wide-based gait/stance
poor coordination of hands, speech, eye mvmt
atrophy of cerebellum
CAG/polyQ - 1, 2, 3, 6, 7 (expand in spermatogenesis)
CAG CTG - 8 (expands in oogenesis)
CAA/CAG - 17
others caused by point mtn, del, etc.
must rule out non-genetic cause of ataxia
cri du chat
5p- (5p15.2 and distal?)
contiguous gene syndrome/partial aneusomy
most dels are visible on karyotype
85% de novo deletions (most paternal in origin)
12% due to parental translocation or inversion
distinct cry, decreases with age
slow growth
mental retardation
microcephaly
hypotonia
dysmorphisms - round face, hyperterlorism, micrognathia, epichanthal folds, low-set ears
wolf-hirschhorn
4p-
partial aneusomy/microdeletion/contiguous gene syndrome
deletions vary in size, 156kb critical region, interstitial usualy p12-16
75% have de novo del (mostly from father)
13% have imbalance from parent's balanced rearrangement
12% have unusual cytogenetic abnormality (ex. ring 4)
distinct facies - 'greek warrior helmet' - broad nasal bridge that continues up to forehead, hypertelorism, epicanthus, high arched eyebrows, short philtrum, downturned mouth,micrognathia, ptosis
pre and postnatal growth deficiency
hypotonia
MR in all (variable degree)
seizures, structural brain abnlty (33%)
skeletal anomalies (60-70%)
cleft lip/palate, heart defect (50%), hearing loss (>40%), urinary malformation (25%)
tests: karyotype detects 60-70% of deletions, FISH/array CGH detects >95%
Williams syndrome
7q11.23 deletions
contiguous gene del/microdel syndrome - submicroscopic, requires FISH or array
one gene (ELN) can cause the cardiac phenotype
AD, but most de novo
heart - aortic and other artery stenosis, MVP
connective tissue - joint limitation or laxity, hoarse voice
hypotonia
MR - mostly mild
distinct facies - broad brow, bitemporal narrowness, periorbital fullness, stellate/lacy iris pattern, strabismus, short nose, full nasal tip, malar hypoplasia, long philltrum, full lips, wide mouth, prominent ear lobes
kids - epicanthal folds, full cheeks, widely spaced teeth
adults - long face and neck, sloping shoulder, appear 'gaunt'
endocrine disorders - hypercalcemia/uria, hypothyroidism
FTT in infancy
hernia, rectal prolapse
bhvr/perosnality - overly friendly, highly empathic, anxiety, ADD
Rubinstein-Taybi
CREBBP (del 10%, mtn 40-60%) @ 16p13.3
EP300 (mtn 3%)
AD, mostly de novo
MR
postnatal growth delay (prenatal usually normal)
microcephaly, short statur
dysmorphism - beaked nose, ptosis, downslanting palpebral fissures, columella extending below the nares, highly arched palate, 'grimicing' smile, talon cusps (on teeth)
broad thumbs, toes, often angulated
Noonan syndrome
PTPN11 @ 12q24.1 (50%)
SOS1 @ 2p22-p21 (13%)
KRAS @ 12p12.1 (<5%)
RAF1 @ (3-17%)
AD, 25-70% de novo
cardiac - HCM (20-30%), pulmonic valve stenosis (20-50%), other
feeding problems
short stature, scoliosis, pectus
learning disabilities (25%), mild MR (33%)
bleeding problems
facies - short, broad/webbed neck, tall forehead, hypertelorism with downslanting palpebral fissures, deeply grooved philtrum, low posterior hairline, thickened or ptotic eye lids, myopathic facies in childhood, triangular face as teenager, nasolabial folds as adult
sequencing of known genes - ~75% yield
Phenylketonuria
PAH
BH4
AR, significant allelic heterogeneity (and sort of locus heterogeneity)
first step of PHE metabolism: PHE-> TYR
severe MR, microcephaly, seizures
psych-autistic, hyperactive, anger, anxiety, depression
hypopigment'n (hair, skin)
odor (esp. ear wax) - musty, sweet
maternal: aim for <300umol/L during pregancy; pheno: microcephaly, MR, CHD, IUGR, postnatal growth retard'n
normal: 30-90umol/L; benign hyper: 120-600: hyper: 600-100; abnormal >1000
NBS: ~100% sensitive, also picks up benign hyper, need to confirm ASAP to start tx by 4 wk, assay substrate to product ratio and substrate levels
tx: nearly curative (executive fxn defects, hyperactivity, depression, anxiety), low protein, special food w/o that AA, large neutral AA to compete across blood brain barrier, cofoactor, aim for 40-350umol/L
must distinguish between cofactor and enzyme etiology b/c treatment differes
Tyrosinemia type I
FAH
AR
AA disorder, in TYR/PHE pathway
fumarylacetoacetase deficiency
presents in infancy, death by 10yo if untreated
liver - liver failure, cirrhosis, ascites, coagulopathy, fasting intolerance, hepatocellula carcinoma
renal - enlargement, dysfunction, renal fanconi/renal tubular acidosis (AA in urine)
skeletal - rickets (secondary to renal disease)
growth failure
odor - boiled cabbage
NBS - tyrosine (other causes of elevated tyrosine: liver disease, transiently elevated in newborns, prematurity....?)
tests: plasma - high tyr, met, phe; serum - high AFP
urine - high tyr metabolites, succinylacetate
genetic testing: 4 common mtn, sequencing - total yield 95%
tx - phe and tyr free formula, low protein diet, NTBC - inhibits upstream enzyme, converts phenotype to tyrosinemia II (better growth, improved liver fxn, kidney fxn, add skin and eye pheno of II)
tests:
Homocystinuria
CBS
cystationine beta-synthase deficiency
AR
AA disorder
myopia, ectopia lentis (early, by 8yo) (downward vs. upward in Marfan)
marfanoid habitus, scoliosis, pectus
osteoperosis (vs. Marfan)
thromboemoblism (vs. Marfan)
MR (vs. Marfan)
high arches (vs. marfan)
no aortic dissection (vs. Marfan)
Plasma AA: high homocysteine, methionine
tx: low protein diet, Met-free formula, cofactor supplementation - B6 (30% responsive), cofactor to augment alternative pathway (B12/folate)
glycine encephalopathy
GLDC (80%), AMT (10-15%), GCSH
AR
AA disorder
usually immediate and devastating neonatal presentation
lethargy, hypotonia (right after birth)
myoclonic jerks, seizures, pathoneumonic EEG pattern (burst suppression)
decreased respiration
MR
tests: urina AA: high glycine, CSF: high glycine, high CSF:plasma glycine ratio, enzyme activity on liver biopsy
genetic testing (90-95%)
tx: sodium benzoate (binds gly, forms hippurate, excreted), destromethorphan (NMDA antagonist), not very treatable
glutaric aciduria type 1
glutaryl-CoA dehydrogenase deficiency
AR
OA disorder
macrocephaly (unlike other metabolics)
normal and then trigger event (catabolic) -> encephalopathy/metabolic stroke -> hypotonia, profound dystonia and or choreoathetosis
motor >> cognitive dysfunction
MRI abnormalities
tests: metabolic acidosis, hyperammonemia during episodes, UOA - glutaric acid, MRI - fluid and atorphy of frontal lobes, basal ganglia changes
tx: fluids, glucose, carnitine in episode; riboflavin trial, lysin-restricted diet
isovaleric acidemia
isovaleryl CoA dehydrogenase
AR
OA disorder, defect in leucine degradation
pt normal and then sick/not eating for some reason -> catabolic -> excess leucine -> metabolic acidosis, respiratory alkalosis, ketosis, hyperammonemia
lethargy, vomiting, dehydration
thrombocytopenia, neutropenia
encephalopathy, seizures, coma, death
odor: sweaty feet
tests: metabolic acidosis, high anion gap, hyperammonemia
UOA: isovaleric and OH isovaleric acid, isovaleric-glycine, ketones
prognosis better than PA, MMA, and good if treatment started early
tx - low protein diet, Leu free formula
carnitine and glycine to help clear excesses
emergency attn in crises
methylmalonic acidemia
MUT, MMAA, MMAB
AR
cbl C - combined MMA, homocystinuria (relatively common)
OA disorder
final step in breakdown of multiple AA (isoleu, val) and some FA, right before kreb cycle
methylmalonate builds up
more severe than isovaleric acidemia
acute: severe metabolic acidosis, ketosis, hyperammonemia (secondary), lethargy, vomiting, dehydration, hypotonia, encephalopathy
hepatomegaly (vs. IVA), thrombocytopenia, neutropenia, coma, death
other: cardiomyopathy, basal ganglia infarcts/metabolic stroke, pancreatitis, bone marrow suppression, progressive renal disease
tests: metabolic acidosis, high anion gap, high ammonia
paa - high gly
uoa - methylmalonate, ketones
tx: sodium benzoate for hyperammonemia, carnitine (metabolic sponge), vit b12 b/c cofactor (some responsive, others not), metronidazole b/c gut flora produce a lot of proprionate, kidney liver transplant (not curative), restrict protein and precursors (VOMIT), emergency attn when sick
NBS: B12 deficiency can cause false positive
propionic acidemia
PCCA, PCCB
propionylCoA carboxylase deficiency
AR
OA disorder
second last step in breakdown of isoleucine and valine (and others)
similar phenotype to MMA
PFVLCSz - vomiting prominent
severe metabolic acidosis, ketosis, hyperammonemia
FTT
lethargy, vomiting, dehydration
hypotonia, encephalopathy
hepatomegaly (vs. IVA)
thrombocytopenia, neutropenia
coma, death
other: MR, basal ganglia infarct, CM, pancreatitis
tests: metabolic acidosis, high anion gap, high ammonia
paa: high glycine
uoa: propionate metabolites and ketones
tx: sodium benzoate for hyperammonemia, carnitine, biotin (enzyme cofactor), metronidazole b/c gut flora produce a lot of proprionate, restrict precursors (VOMIT), emergency attn when sick, kidney/liver transplant (not curative)
poor prognosis, even with tx
galactosemia
GALT
AR
classic - <5% activity, Q188R common mtn, G allele on isoelectric focusing
duarte - 5-20%, N314D, D allele, treat??
individuals who are compound hets for duarte/gal alleles can appear normal
hypoglycemia, feeding problems, FTT, vomit'g, diarrhea
catracts**, e coli sepsis**
jaundice**, hepatomegaly, hepatocellular damage, bleeding
language delays**, POF**

tests: NBS detects ~100%, high substrate and reduced enzyme activity
confirmation: enzyme activity, then molecular testing
tx: lactose-restriced diet, start within first 10 days, calcium supplementation; residual phenotype: cataracts, speech delay, DD, poor growth, cognitive deficits (exec fxn), POF
glycogen storage disease type I
G6PC (80%) - G6Pase
SLC37A4 (20%) - G6Pase translocase
AR
defect in glycogen metabolism, glycogen builds up in liver, kidneys, muscles
doll like faces**, thin extremities, short stature, protuberant abdomen
hepatomegaly, hepatocellular ca
renomegaly, renal dysfxn
hypoglycemia (3-4hr post feed), lactic acidosis, neutropenia
osteoperosis, delayed puberty, polycystic ovaries
tests: no response to glucagons, metabolic acidosis, high lactate, high uric acid, high chol, TG
dx by enzyme activity on liver sample or by genetic testing
tx - avoid fasting (frequent feeds, glucose drip at night, cornstarch), kidney/liver transplant
VLCAD
ACADVL
AR
FAO disorder - most severe form
can't breakdown fatty acids C14-20
LCHAD and TFP have similar phenotypes
HCM/DCM (vs. MCAD, SCAD; reversible with tx), sudden death, DD, seizures, hepatomegaly, fatty infiltration of liver, hypoketotic hypoglycemia
severe early-onset multiorgan form - present in infancy w HCM/DCM, preicardial effusion, arrhythmias, hypotonia, hepatomegaly, intermittent hypoglycemia
hepatic or hypoketotic hypoglycemic form - present early chidlhood w hypoketotic hypoglycemia and hepatomegaly, no CM
later onset episodic myopathic form - intermittent rhabdomyolysis, muscle cramps and/or pain, and/or exercise intolerance, usually w/o hypoglycemia
tests: acylcarnitine analysis of plasma during crisis/stress, enzyme activity, analysis of FAO in fibroblasts, molecular testing
tx - avoid fasting, very low fat diet, supplement MCFA, emergency attn if sick, not eating
MCAD
ACADM
AR, 50% homozygous for K304E, 40% K304E/another allele
FAO disorder, can't break down C8, C10, C12
sudden death, ~5% of SIDS
present 3-24months, sometimes later
fatal in 40% of cases
hypoketotic hypoglycemia
lethargy, vomiting
DD, seizures
hepatomegaly, fatty infiltration of liver
muscle and heart normal (vs. L, VL)
tests: plasma acylcarnitines (elevated C8), plasma fatty acid profile, UOA, urine acylglycines, octenoic acid (C8:1) is isgnature metabolite, enzyme activity in fibroblasts, molecular testing for common mtn (A985G) and sequencing
-treatable w early diagnosis
tx: avoid fasting (good prognosis), don't need to overly restrict fats ('heart healthy diet'), carnitine, emergency attn if sick, lethargic, not eating
SCAD
ACADS
AR
FAOD - mildest form (some think shoudln't be on NBS)
can't breakdown C4
hypoketotic hypoglycemia
DD, seizures
FTT
muscle weakness (vs. M)
NBS
tx - avoid fasting, emergency attn if sick, not eating
OTC - ornithine transcarbamylase deficiency
OTC @ Xp21.1
XL**
90% from carrier mother, 10% de novo (vs. usually 2/3 carrier mother for genetic lethal XL - difference b/c mutation rate high in spermatogenesis so many moms carry mtn de novo from MGF) -- important to determine mother's carrier status
most common UCD
v. high ammonia (>2000), high glutamine (with ammonia), high orotic acid** (only UCD with this)
low - citrulline, arginine; normal UOA (vs. UOA disorders)
usually neonatal and severe presentation - first 48-72 hours - vomiting, lethargy, coma
late (esp. girls) - FTT, DD/MR, episodic ataxia, vomiting, protein avoidance, neuropsych, postpartum hyperammonemia
acute tx - 10% dextrose, eliminate protein intake, IV ammonul (sodium benzoate and sodium phenylacetate), hemodialysis if resistent to tx
chronic tx - protein restriction, avoid catabolism, oral phenylbuterate, close monitoring, liver tx
prenatal testing must be done by molecular testing b/c enzme not expressed in chorionic vili or amniotic cells
CPSI, NAGS deficiency
CPSI, NAGS
AR
UCD disorder - most severe (similar severity to OTC)
classical neonatal presentation, milder later onset forms
high - ammonia, glutamine
low - orototic acid* (vs. OTC), citrulline, arginine
tx - protein restriction, arginine supplementation, augment alternate pathways
Arginase deficiency
ARG1
AR
UCD, last step in UC
distinct from other UCDs - chronic not acute, later presentation, ammonia not elevated
high - arginine
normal until 1-3yo -- growth retardation, spasticity, seizures, DD or regression, loss of ambulation, bowel, bladder control, severe MR
tx - low protein diet, avoid fasting, urgent care if sick, ammonia scavenging agents, liver transplant
Tay-Sachs
HEXA @ 15q23-24
AR
AJ - 1/30 carrier - 1278insTATC (82%), two others
LSD - GM2 gangliosides accumulate in lysosomes
enzyme activity inversely correlates iwth severity (infantile vs. juvenile vs. adult)
infantile form: normal until ~6mo - slowing dev't then regression (loss of motor skills) by 8-10mo
hyperacusis*, apathy, cherry red macula/spot*
progresses to seizures, blindess, spasticity
death by 2-5yo
no hepatosplenomegaly* (vs. other storage disorders)
juvenile - muscle coordination problems, seizures, vision problems (start 1st year), survival into late childhood, adolescence
juvenile form - onset 2-4y w neuro deterior'n, ataxia, incoordination --> seizures, spasticity, +/- cherry red spot
chronic adult form - rare, slower progression, bipolar psychosis, progressive dystonia, spinocerebellar degeneration, motor neuron disease, muscle weakness and fasiculations, quite variable
enzyme testing: dx based on absent HEXA activity with normal HEXB activity; false positives due to pseudodeficiency alleles (f/u with DNA testing) (2% of AJ with abnl activity have pseudodef., 35% of non-AJ do)
genetic testing: 6 common mutations - 98% yield in AJ, 59% yield in non-jews
Sandhoff
HEXB
activator protein
LSD disease, similar toTay-Sachs - distinguish with enzyme testing, AR, deficiency in HEXA activity and HEXB activity b/c of mutations in HEXB or in activator protein.
MPS I
alpha-L-iduronidase
(aka Hurler, Hurler-Scheie, Scheie)
AR
MPS, spectrum of phenotype - now called MPS I and attenuated MPS I
severe - onset 6-12mo, die by 10yo
coarse facies
corneal clouding*
hydrocephalus, macrocephaly
skeletal dysplasia - dysostosis multiplex** (x-ray finding, bullet shape metacarsals)
hepatomegaly
umbilical or inguinal hernias
short stature - linear growth stopes by 3yo)
cardiomyopathy
mitral and aortic valve regurg
hearing loss, DD, MR, decreased ROM
tx - BMT, ERT
MPS II - Hunter syndrome
iduronate sulfatase
XLR*
LSD
a lot like similar AR disorder but no corneal clouding**
coarse facies, hydrocephalus, CM, heart failure, dysostosis multiplex, short stature, MR, decreased ROM in joints, carpal tunnel syndrome
tx - ERT, BMT less successful than other similar disorder
Pompe disease/GSD II
GAA
AR
GSD and LSD
usually infantile onset (sometimes prenatal, sometimes childhood)
hypotonia, muscle weakness**, HCM**, respiratory distress
FTT, feeding difficulties
hearing loss, fractures
enzyme testing is diagnostic, DNA testing for GC'ing
tx - ERT (die by 1-2yo without it)
RT/OT/PT
Fabry
alpha galactosidase a deficiency
GLA
XL**, rarely de novo
LSD - GL-3 builds up - esp. in blood vessels, kidneys, heart
onset - adolescence for males, adulthood for females
vascular occlusion, ischemia, infract -> TIA, stroke
corneal verticillata/whorls**
angiokeratomas (groin, umbilicus, back)
acroparasthesias (provoked by exercise, temp changes)
hypohidrosis, anhydrosis
renal disease
CM, arrhythmia
heat and cold intolerance
vomiting, diarrhea, cramping
depression, anxiety, fatigue
proteinuria
tests: enzyme activity (but not good for females)
genetic testing: ~100% sensitive
tx: ERT
MNGIE
thymidine phosphorylase gene
AR
one of nuclear-encoded diseases affecting mtDNA - mtn in thymidine phosphorylase gene cause multiple mtDNA deletions
manifestations: mitochondrial myopathy, peripheral neuropathy, GI and encephalopathy disease
mtDNA depletion syndrome
AR
mtn in nuclear-encoded genes needed for mtDNA replication (gamma polymerase), maintenance of nucleotide pools (TK2, dGK)
heterogeneous group of disorders
infantile onset
myopathic, hepatic
dx made in muscle (not blood) by compairing mtDNA: nDNA reations in patients vs. controls
MERRF - myoclonic epilepsy with ragged-red fibers
mtDNA tRNAlys
maternal inheritance
heteroplasmic
common mutations: 80% 8344G>A, 10% 8356T>C or 8363G>A
highly variable (including ragged red fibers) - severity increases with somatic mtDNA mutation rate, i.e. with age
complexes I and IV most severely affected b/c have most components transcribed in mitochon.
onset: childhood through adulthood; slowly progressive or rapid decline
myoclinic epilepsy
myopathy (with or without ragged red fibers)
other (variable) features: abnl brainstem provoked responses, SNHL, ataxia, renal dysfunction, diabetes, CM, dementia
prenatal and predictive molecular testing are not useful b/c mtn load in blood/amnio/etc. doesn't reflect other tissues and b/c can't predict replicative segregation, tissue selection, somatic mtDNA accumulation
ADPKD (polcystic kidney disease)
PKD1 @ 16p13.1 (85%), PKD2 @ 4q21 (15%)
AD, 10% de novo
common! 1/300-1/1000
variable expressivity (inter and intrafamilial), two-hit hypothesis for cyst formation, age-dep't penetrance
renal - progressive failure, recurrent UTI, hematuria, nocturia, secondary htn
intracranial saccular aneurysms, dilatation of aortic root, dissection of root, MVP
sequencing - 85% yield
also - deletions of PKD1 and TSC2 - tuberous sclerosis and PKD

cysts - renal, hepatic, ovarian, pnacreat
Prader-Willi syndrome (PWS)
15q11-q13
etiology:
70% pat del (<1% recurrence risk)
15-25% mat UPD (<1% recurrence risk)
5% imprinting defect (50% recurrence risk)
infancy: feeding difficulties, severe hypotonia, hypogonadism with cryptorchidism
childhood: hyperhagia, obesity, delayed motor and language dev't, bhvr pheno (temper tantrums, stubbornness, manipulative, OCD), delayed puberty (b/c of hypogonadism)
sterility, scoliosis, sleep apnea, short stature, osteoperosis
dysmorphism: narrow bifrontal diametere, almond-shaped eyes, trangular mouth, small hands and feet, hypopigmentation of eyes, hair, skin
testing: Meth-sensitive PCR or meth-sensitive restriction enzyme - 99% yield! then need further testing to determine etiology (for recurrence risk)
tx: symptomatic, food management, growth hormone
Rett syndrome
MECP2 @ Xq28
(CDKL5 mtn in aytpical presentation w early onset seizures)
XLD, almost exclusively females (rare males with mosaicism or XXY)
mtn (85%), del by MLPA, qPCR (15%)
loss of fxn
99% de novo (70% from dad), rare unaffected carrier mother with skewed x inactivation
germline mosaicism reported
sex-dependnet phenotype
males - lethal
females - classic to atypical
classic: normal dev't to 6-18 months, then develop'l slowing, stagnation, decelerating head growth (-> acquired microcephaly), neurodev'l regression (esp. speech and purposeful hand use, also motor skills), stereotyped repetitive hand movements (wringing); several years of stabilization, then progress to severe MR, progressive spasticity, rigidity, scoliosis
ataxia, seizures, breathing irregularities, bruxism (teeth clenching)
bhvr/psych - fits of screaming, inconsolable crying, autistic features, panic-like attacks
long QT, reduced lifespan due to sudden death
mtn in this gene show wide variability of phenotype with poor geno-pheno correlations
dx confirmed by molecular testing, must rule-out angelman synd.
Sickle Cell disease
HBB
AR
60-70% homozygous Hb S (Glu6Val), rest are compound hets - Hb S/Hb C, Hb S/beta-thal, etc.
het advantage - malaria resistance - african-americans 1/14 carriers, non-hispanic caribbean w. indian 1/14, west african 1/6
Hb S reduced solubility when deoxygenated, form polymers, sickle shape of RBC, occlude capillaries (ischemia), get cleared --> hemoyltic anemia
present first 2y of life - anemia, FTT, splenomegaly, repeat infections/sepsis, dactylitis (painful swelling of hands or feet)
painful crises - bone vasoocclusions
infarct --> organ damage - most significant - spleen, brain, lungs, kidneys
NBS (to allow for antibiotic prophylaxis)
test for Hb S - HPLC, IEF, or Hb electrophoresis
DNA testing for Glu6Val
tx - antibiotic prophylasix, vaccination, oral hydration, folate supplements, avoid hypoxia/exhaustion/temp extremes
chronic RBC transfusions
analgesics for pain crises
Turner syndrome
45,X
sporadic - 80% of cases due to sex chromosome missing from father
1/2000-1/5000 births
etiology:
-50% 45,X
-25% structural abnormality of X
-25% mosaic 45,X
second sex chromosome important for intrauterine survival? (1-2% of all conceptions are 45,X but only 1% of those survive, yet postnatal phenotype is mild)
second X needed for maintenance (but not develop't) of oocytes and ovaries --> fibrous streak gonads --> 90% don't spontaneously enter puberty, hormones needed for secondary sex characteristics
short stature (100%) (SHOX!)
broad chest, wide spaced nipples, webbed neck, low nuchal hairline, cardiac anomalies, renal anomalies, SNHL, edema of hands and feet, dyspalstic nails
shy, withdrawn
osteoperotic fractures, thyroiditis, diabetes, IBD, heart disease
tx - growth hormone for linear growth, progest./est. for secondary sex charact., echo, renal u/s, diabetes screening
Russel-Silver syndrome
matUPD7 (10% of cases)
11p15.5 imprinting changes (?%)
recurrence risk - low, most are sporadic
pre and postnatal growth retardation - 2+ standard deviations below the mean; proportionate short stature with normal head circumfrence (so relative macrocephaly)
traingular facies
areas of hyper and hypopigmentation
hemihypertrophy
excessive sweating
delayed bone maturation
NF2
NF2 @22q12.2
AD, 50% de novo
75% sequencing variants
15% deletions
dx'c criteria:
-bilateral vestibular schwanommas
OR
-a FDR with NF2 and
-unilateral vestibular schwanomma
-two of the following: meningioma, glioma, schwannoma, juvenile posterior subcapsular lenticular opacity/juvenile cortical cataract
some have cafe-au-lait
ca - acoustic neuromas, meningiomas, gliomas, ependymomas, mesotheliomas
denys-drash
WT1 @ 11p13
AD, most de novo
-urogenital anomalies - XY males with ambiguous or female genitalia
-wilms tumor (>90%)
-mesangial sclerosis of kidneys
WAGR
11p13 contiguous gene deletion (including WT1 and PAX6)
true contiguous gene syndrome
Wilms tumor (WT1) (40-50% risk)
aniridia (PAX6)
genital anomalies
retardation
testing: FISH detects 30%
Emery-Dreifuss muscular dystrophy
EMD @ Xq28, LMNA @ 1q21.2
XL - EMD - most female carriers unaffected, but can have cardiac or even full blown disease
AD, AR (rare) - LMNA (75% de novo)
joint contracturs (early childhood onset)
slowly progressive muscle weakenss and wasting (starts humero-peroneal, extends to scapular and pelvic girlde)
cardiac involvemnt - palpitations, presyncope, syncope, CHF
inter and intrafamilial variability
dx based on muscle biopsy - path, emerin, lamins A/C and genetic testing (LMNA)
Acute intermittent porphyria
HMBS @ 11q23.3
AD
onset - after puberty
reduced penetrance (10-50%), women>men
potentially severe and debilitating
intermittent acute attacks: abdominal pain(most common symptom), nausea, vomitting, constipation, diarrhea,
peripheral neuropathy - muscle weakness, neuropathy,
psych - hysteria, anxiety,
hepatocellular carcinoma, *no cutaneous findings
tests: high urine delta-amonolevulinic acid (ALA) and porphobilinogen (PBG) during attack
genetic tests: sequencing (>98%)
tx: stop or treat precipitant (medication, infection, alcohol, dehydration, smoking, etc.); intubate if needed; IV dextrose: IV hemin; pain control
Alagille syndrome
JAG1 (95%), NOTCH2 (<1%)
AD, 60% de novo, possible germline mosaicism
seq JAG1 (88%), FISH 20p12 (7%, includes JAG1)
liver, heart, eyes, face, skeleton
highly variable
cholestatis (96%) (bile duct paucity on liver biopsy) (85%)
congenital heart defects - pulmonary arteries
posterior embryotoxon of eye (78%)
DD (16%), growth failure
dysmorph - prom't forehead, deep-set eyes, moderate hypertelorism, pointed chin, saddle or straight nose w bulbous tip (triangular face)
butterfly vertebrae (51%)
HHT - Hereditary Hemoorrhagic Telangectasia
ENG, ACVRL1(ALK1), SMAD4 (total - 85%)
AD, marked intrafamilial variability
arteriovenous malformations - multiple - in skin (telangectasias), nose, brain, liver, lungs
age-dependent, usually not dx'd until adolescence or later
easy bleeding after slight trauma
spontaneous and recurrent nosebleeds (most common feature, onset 12yo)
GI bleeding (25%, after 50yo)
genetic testing - seq'g all 3 genes: 85%, dup/del (10%)
annual eval: for anemia, pulmonary AVM by pulse oximetry or echo (?head MRI, u/s for hepatic AVM)
Holt-Oram syndrome
TBX5 (70%)
AD - 85% de novo
upper-extremity malformations (radial, thenar, carpal bones) - carpal (100%, may require hand x-ray), symmetric or asymmetric
CHD - ASD, VSD - (75%)
cardiac cnduction disease
genetic testing - 70%
LEOPARD syndrome
PTPN11 (80%), RAF1 (3%)
AD, de novo (?%)
LoF mtn (vs. GoF in Noonan)
L-letigines - onset 405yo, increase to thousands by puberty, flat black-brown macules on face, neck, upper trunk
E-ECG conduction abnlts
O-ocular hypertelorism
Pulmonic stenosis (+ HCM) - 85%
R - retardation of growth, postnatal, short stature - 50%
D - SNHL
MR - mild, 30%
genetic test - 83-93% yield
Greig cephalopolysyndactyly syndrome (GCPS)
GLI3 @ 7p13
AD, de novo (esp. cytogenetic)
preaxial polydactyly or mixed pre and postaxial polydactyly, also syndactyly of fingers 3-4 and toes 1-3
microcephaly, ocular hypertelorism, subtle craniofacial findings
seizures, hydrocephalus, MR
karyotype for gross cytogenetic abnormalities involving 7p13
FISH (array?) - 5-10%
sequencing - 70%
Joubert syndrome
MPHP1, CEP290, AHI1, TMEM67 (total - 40%)
AR, M:F 2:1
hypotonia in infancy
DD - severe MR to normal
apnea - episodic - improves with age
abnormal eye movements
MRI - *molar tooth sign
VACTERL (VATER) association
gene unknown
sporadic, dx of exclusion, dx requires 3/7 features
V - vertebral anomalies
A - anal atresia
C - cardiac defect (VSD, PDA, TOF, TOV)
T - treacheoesophageal fistula
E - esophageal atresia
R - renal anomalies
L - limb anomalies (polydactyly, humeral hypoplasia, radial aplasia, proximally placed thumb)
variant - VACTERL - with hydrocephalus (AR or XL)
-differential dx: fanconi anemia, townes-brocks, infant of diabetic embryopathy, feingold syndrome
X-linked adrenoleukodystrophy
ABCD1 @ Xq28
XLR, 7% de novo
peroxismal disorder, defect in peroxismal membrane protein that may be a transporter, accumulation of saturated VLCFA
tests:
childhood cerebral presentation: ADHD -> total disability within 2 years, mild adrenal dysfxn, decreasing intellectual performance, MRI white matter changes
adrenomyeloneuropathy: late 20s, progressive paraparesis, sphincter disturbance, adrenocortical dysfunction
adrenocortical insufficiency (only) - majority by 8yo (20% of carrier females)
tests: abnl plasma very long chain fatty acids, MRI white matter changes
genetic tests: ABCD1 sequencing (92%), del/dup (6%)
CADASIL
NOTCH3 @ 19p13.2
AD
C - cerebral
AD - autosomal dominant
A - arteriopathy
S - with subcortical
I - infarcts and
L - leukoencephalopathy
stroke-like episodes before 60yo, cognitive disturbances, bhvrl abnlts, migraine and aura
tests: skin bx EM, brain MRI
genetic tests: NOTCH2 sequencing (>90%)
mean age to walk with asst: 60yo, bedridden by 64y, med. age of death 68y
Hereditary diffuse gastric cancer (HDGC)
CDH1
AD
diffuse gastric ca (aka signet ring type ca or isolate cell ca)
adult onset (avg age 38yo)
lifetime risk of ca - 67% for men, 83% for women
lobular breast ca risk - 39%
dx - 2 cases that are FDR or SDR and one <50yo OR 3 cases in FDR or SDR
Canavan disease
ASPA
AR - enzyme deficiency
AJ carrier rate - 1/40
3 common mtn account for 99% of mutations in AJ; 50-55% of mutn in non-AJ
leukodystrophy; accumulation of n-acetyl-aspartic acid
macrocephaly, lack of head control
DD by 3-5 mos - never sit, walk, speak
severe hypotonia, evolves to spasticity
life expectancy - teens
demyelination
tests: high urine n-acetyl aspartic acid (NAA) on UOA
Familial dysautonomia
IKBKAP @ 9q31
AR
AJ carrier rate: 1/35, 2 common mtn - 99% of alleles
disease affects dev'l and survival of sensory, sympathetic, parasympathetic neurons
onset: birth, progressive
GI dysfunction, vomiting crises, recurrent pneumonia, CV instability
altered sensitivity to pain and temp
autonomic crises, hyptonia, in adulthood broad base degenerative gait, decreased life expectancy
absence of overflow tears with crying
Krabbe disease
GALC @ 14q31
AR, enzyme deficiency
infantile onset (most cases) - normal at birth, then irritability, spasticity, DD. psychomotor regression -> no voluntary mvmt. progressive. death by 2yo.
later onset (6mos-5th decade): weakness, vision loss, intellectual regression
abnl CT, MRI, EEG
enzyme testing: 0-5% of normal activity
targeted mtn analysis: 30kb del (35-45%), 809G>A (50% of late onset), sequencing (100%)
Wilson disease
ATP7B
AR, defect in copper-transporting ATPase 2
present 3-50you, variable
liver disease: jaundice, self-limited hepatitis-like illness, autoimmune hepatitis, chronic liver disease
neuro/psych: mvmt disorder, rigid dystonia, depression, neurotic bhvr, disorganization of personality
tests (for dx): kayser-fleisher rings on cornlea exam (Cu deposition), low serum cu and ceruloplasmin, increased urinary copper, increased copper storage on liver bx
genetic tests: sequencing (98%), common mutations
tx: chelating agents, liver transplant
FMF - familial mediterranean fever
MEFV
AR (but in prevalent areas may be pseudo-dominant) - (armenian, turkish, arab, north african jewish, iraqi jewish, AJ)
type 1: recurrent febrile episodes with peritonitis, synovitis, or pleurotis, also erythema - presents as fever and pain at site of inflamation; amyloidosis (AA type, can lead to renal failure)
type 2: amyloidosis as first presentation
tests: increased erythrocyte sedimentation rate (ERS); leukocytosis; increases serum fibrinogen; proteinuria
genetic testing: targted mtn analsyis = 70-90%, sequencing - 90%
tx: colchicine
Hermansky-Pudlak syndrome
HPS1, AP3B1,HPS4,5,6,7,8
AR
oculocutaneous albinism - skin, hair and eye pigment decreased
bleeding diathesis - platelet storage pool deficiency - easy bruising, frequent epistaxis, gingival bleeding, postpartum hemorrhage, colonic bleeding, prolonged bleeding with menses, surgery, etc.
decreased visual acuity, nystagmus, foveal hypoplasia, increased crossing of optic nerve fibers
may have skin ca, pulmonary fibrosis, colitis
tests: prolonged bleeding time; absent platelet dense bodies (sine qua non) on platelet EM
genetic tests: depends on ancestry
Hutchinson-Gilford Progeria syndrome
LMNA
100% G608G
AD, all de novo, paternal age effect
normal at birth, profound FTT in 1st year
short stature, wt
Androgen insensitivity syndrome
AR @ Xq11-q12
XLR
46,XY - feminization of external genitalia, abnl secondary sexual dev't, infertility
spectrum of severity: complete (female genitalia), partial (ambiguous genitalia) mild (nl male genitalia)
tests: normal or increased testosterone; normal or increased LH; deficient androgen binding activity of genital skin fibroblasts
genetic tests: sequencing (>95% yield in complete, <50% yield in partial)
treatment: removal of testes after pregnancy (ca prevention), estrogen replacement therapy
Kallman syndrome - type 1 and 2
KAL @ Xp22.3, FGFR1 @ 8p11.1-11.2
XLR, AD
hypogonadotropic hypogonadism, delayed pubertal dev't
anosmia
type1: mirror hand movements, ataxia, GU anomaly, high palate, pes cavus
type2: MR, CL/P, cryptorchidism, choanal atresia, CHD, SNHL
tests: low FSH and LH, low testosterone in males, low estradiol in females, MRI: hypo/aplasia of olfactory bulbs and tracts
genetic tests: sequencing - 13-26% yield (total
tx: normalize gonadal steroid levels
Klinefelter syndrome
47XXY
tall stature,
slightly delayed motor and language skills, learning problems
testosterone plateaus at age 14, small fibrosed testes, azoospermia and infertility
gynecomastia
increased cholesterol, increased risk autoimmune disorders and mediastinal germ cell tumors (1%)
maternal > paternal origin, AMA affect
tx: testosterone in mid-late adolescence (bone density, secondary sex dev't, sucle mass, chol, increased libido, improved energy).
McCune-Albright syndrome
GNAS @ 20q13.2
sporadic
polyostotic fibrous dysplasia, pathologic fracture, cranial foramina thickening --> deafness and blindness
large irregular cafe aut lait ('coast of maine')
precocious puberty, hyperthyroidism, increased growth hormone, PRL (?) or PTH
ovarian cysts
tests: x-ray, pevlic u/s vision and hearing testing, pituitary hormone analysis
genetic testing: targeted - GoF mtn
Biotinidase deficiency
MRD
AR
neuro - seizures, hypotonia, ataxia, developmental delay, vision problems, hearing loss
cutaneous - alopecia, skin rash, candidiases
older children/teens - motor limb weakness, spastic paresis, decreases visual acuity
treatable with biotin BUT DD, HL and vision problems are irreversible
testing: <10% biotinidase enzyme activity
NBS; genetic test - targeted (60%), sequencing (99%)
2nd week
2nd week of conditions