• 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

1/95

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;

95 Cards in this Set

  • Front
  • Back

marfan syndrome: what are the common features?

cardio: aortic root dilation, dissection of ascending aorta, mitral valve prolapse, dilation of pulm artery or descending aorta


skeletal: tall stature, long arms and legs compared to trunks, long thin digits, flat feet


other features: dural ectasia, hernia, stetch marks, spontaneous pneumothorax, high arched palate, dental crowding

marfan syndrome: inheritance pattern

AD with 25% new mutations

marfan syndrome: mutations


what protein is involved?

allelic heterogeneity, various mutations within the FBN1 gene on chromosome 15q21



protein fibrillin

marfan syndrome: lab tests

diagnosis based on FH and clinical features with highest weight given to arotic root dilation and to ectopia lentis (lens displacement)



genetic testing by mutation scanning and sequence analysis



FBN1 mutation itself does not confer dx of marfans

how do you manage marfans?

life time follow up for cardiac issues


obtain ECHO-- assess for surgical or medical management (beta blockers)


monitor skeletal abnormalities


annual optho exams

what do you counsel marfans patients to do?

avoid contact sports, strenuous exercise


pregnant women with marfan need high risk OB and cardiac management bc of aortic dissection/rupture

turner syndrome: clinical features

prenatally: 45, X karyotype associated with cystic hygroma, severe lymphedema, hydrops fetalis with high risk of fetal demise



newborn: lymphedema, webbed neck, congenital heart defects most commonly coarctation of the aorta, usually normal linear growth until 2-3



childhood: short stature, low posterior hairline, cubitus valgus, short 4th metacarpals, disproportionately short legs, broad chest, prominant ears, pigmented neci, scoliosis micrognathia



adolescence: short stature and delayed puberty, delayed eiphyseal fusion


adults: intertility/anovulation



other features: inc risk for chronic otitis, hearing loss, hip dysplasia, strabismus, scoliosis, liver deisease, HTN, osteoporosis, diabetes, dyslipidemia



incresaed risk for aorticaortic dilation


learning disabilities, but not intellectual disability


vast majority are infertile, most will not develop secondary sexual characteristics without hormone replacement


inheritance pattern of turners syndrome

chromosomal


incidence is 1 in 2500 femal live births, 1-2% of all conceptions

mutations of turner syndrome

half have 45, X karyotype (cusaed by paternal meiotic non disjunction)


also structural abnormalities

what lab tests do you do for turners syndrome?

karyotyping- peripheral blood


if karyotyping reveals non-mosaic 45, X karyotype, FISH for X/Y should be completed to rule out sex chromsome mosaicism



prenatal dx using CVS or amniocentesis

management issues with turners syndrome

medical: peds card evalutaion with ECHO and EKG, renal ultrasound, audiology, thryoid function test, celiac disease monitoring ,formal optho exam, evaluate for scoliosis



increased risk for gonadoblastoma if karyotype includes a Y chromsome cell line

counseling issues for turners syndrome

review recurrence of less htan 1% for future pregnancy


most girls with turner syndrome are infertile


can use GH and sex hormone replacement

down syndrome: clinical features

dysmorphic features include upslanting palpebral fissues, epicanthal folds, flat nasal bridege, small, dysplastic ears, large tongue/small mouth, excess nuchal skin, short fingers, fifth finger clinodactylly, wide space between first and 2nd toes, single transverse palmar crease


microcephalybrachycephaly


intellectual disability with most individuals with a mild to moderate disability


hypotonia, short stature


congenital heart defects (commonly endocardial cushion defects, ASD, VSD)


hearing problems


vision problems (cataracts, refractive errors, nystagmus, strabismus)


obstructive sleep apena


GI atresias, hypothyroidism, seizures, hematologic problems, celiac disease

down syndrome: incidence


inheritance pattern


mutation

incidence: 1/800


inheritance: chromosomal


mutations: 95% have trisomy 21 caused by maternal meiotic nondisjunction


3-4% 14, 21 translocation is most common

lab tests for DS

karyotyping


peripheral blood karyotype detects almost all cases


prenatal dx available using CVS or amniocentesis



management issues for DS

medical: echo w/ cardio referral


thyroid screening


hearing screeing


vision


cervical spine x rays



counsel about recurrence risk

What deletion is involved in DiGeorge syndrome?

variable expressivisity



common features:


facial features: prominent nose with squared nasal root, small eyes, small ears


cognitive-- intellectual disability, mean


palatal abnormalities: velopharyngeal incompetence, submucosal cleft palate


hypoparathyroidism: hypocalcemia, may lead to seizures


congenital heart disease-- tetralogy of fallot, interrupted aortic arch, truncus arteriosus


immune defiency: imparied T cell production resulting from thymic hypoplasia, recurrent infection, autoimmune disease


can also have ADHD, autism, anxiety, depression, schizophrenia


endocrine abnormalities-- hypo/er thyroidism, GH, vsion concerns, hearing loss, vertebral, GU tract

incidence of digeroge


inheritance pattern


mutations

1/4000


chromosomal (michrodeletion)-- 10% inherited from parent


mutations: 85% have 3 MB deletion, 15% have nested deletion within 22q11.2

lab testing for digeorge

FISH for 22q11.2 deletion or microarray (if phenotype is non specific)


prenatal dx using cells obtained by CVS or amnio

management issues for digeorge

at dx:


serum ionized Ca conc


PTH, TSH, CBC w/ dif, immune eval


otpho, audiology, renal ultrasound, cardiac exam, chest x ray for thoracid vertebral anomalies, cervical spine films, evaluate palate

fragile x: clinical features

phenotype is variable, becomes more pronounced with age


physical features (more evident in late childhood/early adolescence): prominent forehead, long thin face with prominent jaw, large protuberant ears, post pubertal macroochidsm


cog: mild to severe dvlp delay, males more severly affected


behavior: hyperactivity, lack of eye contact, repetitive behaviors, perserverative speech, autism, social shyness


increased risk for: seizures, ocular problems, recurrent otitis media, orthopedic problems, mitral valve prolapse

incidence of fragile x


inheritance pattern


mutation

1/4000 for males and 1/8000 for females


inheritance: x linked with ancipation due to CGG trinucleotide repeat expansion



mutation: expansion of CGG trinucleotide repeat in first exon of FMR1 gene on Xq27



repeat sizes: normal-- 5-44


intermediate: 45-54


premutation:55-200


full mutation: >200, highly methylated


premutations ONLY expand when inherited from a carrier female


daughts of males with a premutation are obligate carriers, but intellectually normal



shows anticipatioN!

lab testing for fragile x

PCR assays specifically designed to detect long CGG repeats


methylation specific PCR assays or southern blotting are then used to determine methylation status

management issues for fragile x

medical: optho (strabismus, hyperopia, astigmatism)


audiology (recurrent serous otitis)


orthopedic (flat feet, scoliosis)


neuro (seizures)



20% of premutation carrier females develop premature ovarian failure/insufficiency, with highest risk at repeat size 80-99



40% of premutation males and 8-16% females develop fragile X associated tremor/ataxia syndrome-- cerebellar gait ataxia, intention tremor, parkinsonism, execuitve dysfunction, onset >50 yrs

huntingtons: clinical features

progressive disorder of cognition, motor, and psych disturbance. 2/3 of affected individuals initially present with neuro features (incoordination, involuntary movements) while 1/3 present with psych changes (depression, irritability)


motor symptoms include gait disturbance, progressive chorea, dysarthia


global decline in cognitive abilities


significant personality change


mean age of onset is 35-44, survival time after onset is 15018 yrs

inheritance pattern of huntingtons

AD


most will have affected parent, new mutations are rare


disorder shows anticipation


de novo cases of HD are more likely due to intermediate sized or reduced penetrance alleles in parent

mutations in huntingtons

expansion of a CAG triplet repeat within HD gene of chromosome 4p-- more repeats, earlier onset


normal: <26


intermediate: 27-35-- no risk to individual, but can expand to offspring


reduced penetrance alleles (36-39)


adult onset: 40-59


juvenile onset: above 60


expansions more likely to occur through paternal gametogenesis-- juvenile onset HD almost exclusively inherited through fathers

lab tests for huntingtons


management issues

lab: PCR detects repeat number



management issues: symp tx (neuroleptics, antipsychotics)



genetic testing of asymptomatic individuals is predictive rather than diagnostic


dont usually test asymptomatic children

whats the difference between a deletrious mutation and a common polymorphism?

polymorphism: common alteration in DNA sequence seen in >1% of popultaion (most are benign, but they may account to diff in drug response or disease risk)



mutation: any alteration in DNA sequence (deleterious mutation is associated with disease)

which CYP450 variants....



increase affect warfarin by inactivating ability to break down metabolites


decrease clopidogrel efficacy by not metabolizing it to its active form


decrease tamoxifen activity


increase codeine toxicity by rapidly metabolizing it to morphine

warfarin increased: CYP2C9



clopidogrel decreaesed: CYP2C19



Tamoxifen increased: CYP2D6



codeine increased: CYP2D6

what does warfarin sensitivity genotyping test show?

CYP2C9*3/*3-- VKORC1


reduced enzyme activity and negative variants in VKORC1 assoc with warfarin sensitivity



CYP2C9 is enzyme responsible for inactivation of warfarin, and CYP2C9*2, *3 are reduced metabolism variant alleles that give you inc sens to warfarin


seen in 40% cauc, <10% asians, af am



(VKOR is vitamin K oxidase reductase which is important for replenishing vit K-- warfarin blocks this so that clotting factors are not formed)


VKORC1*2 is associated with inc sensitivity to warfarin


seen in 40-50% caucasians, 90% asians, 10-15% af am



so they have increased sensitivity to warfarin compared to pt with norml CYP2C9 genotype

what is clopidogrel? what enzyme activates it? what alleles have loss of activity?

platelet inhibitor used in a-fib, coronary artery stents


progdrug activated by CYP2C19


CYP2C19*2 and *3 alleles are most common variants with loss of enzymatic activity-- inc death from CV events

what is one enzyme that is recommended to genotype? in what population

genotype HLA-B*5701 for HIV rx to aviod abacavir hypersensitivity

phenylketonuria:


clinical features

inborn error of metabolism caused by dec activity of liver enzyme phenylalanine hydroxylase, resulting in inc blood levels of AA phenylalanine



untreatd chlidren with PKU risk intellectual disability, seziures, inc irritability, musty odor, eczema


elevated blood levels of phenylalanine in older children/adults with PKU put them at risk for behavioral and learning problems, regardless of good tx


incidence 1/15,000- 1/20,000

inheritance pattern of PKU


mutations


lab tests

AR with variable expression due to varying levels of enzyme activity, other epigenetic factors



mutations: allelic heterogenetiy, with over 500 mutations in the phenylalamine hydroxylase gene on chromosome 12q23 identified



lab tests: screening labs measure phenylalanine levels from samples in newborn nursery


elevated phenyl. detected on blood samples obtained at least 24 hrs after birth


screen for BH4 deficiency in all neonates dx with PKU

management and counseling issues in PKU

management: newborn screening for PKU, intellectual disability prevented if tx initiated before 4 wks of age with a special individualized diet low in phenylalanine


if you have BH4 deficiency, biopterin replacement therapy and NT replacement therapy, dont require low phenyl diet



maternal risks: children born to women with PKU are at inc risk of birth defects and intellectual disability bc of tetragoenic effect of phenylalanine


levels should remain between 120 and 360 throughout pregnancy to prevent defects

MCAD (medium chain acyl coenzyme A deydrogenase deficiency)


clinical features

inborn error of metabolism caused by significantly dec activity of one of enzymes involved in pathway of mitchondrial FA oxidation, resulting in accumulation of medium chain FA


variable presentation (usually occur before age 5)


presentation characterized by hypoketotic hypoglycemia, vomiting and lethargy triggered by prolonged fasting or intercurrent illness


episodes may be accompanied by hepatomegaly, acute liver disease, seizures


without tx of IV glucose, child can progress to coma and death


children at risk for acquired aphasia, ADD secondary to brain injury during acute metabolic event which progresses to coma

MCAD (medium chain acyl coenzyme A deydrogenase deficiency)


inheritance

AR with variable with variable age of onset due to environmental factors (diet, stress, intercurrent illness)


mutation: due to changes in ACADM gene with K304E being the most common mutation


gene located on 1p31



lab test: newborn screening measures alcycarnitine species by tandem mass spectrometry from blood samples on filter paper


perform on infants at least 24 hours old

nagement issues in MCAD


counsleing

detected by newobrn screen


prevent symptoms by avoidance of being in catabolic state, management of illnesses which may precipitate fasting, promst hospitilization during acute metabolic decompensation



all affected indiv should carry emergency letter at all times and provide this to ER



test siblings

neurofibromatosis type 1


clinical features

dx by clinical criteria requires at least 2:


6+ cafe au lait spots (>15mm postpubertal or > 5 mm prepubertal)


2+ neurofibromas or 1 plexiform neurofibroma


axillary or groin freckling


an optic glioma


2+ lisch nodules (benign, raised iris hamartomas seen by slit lamp exam)


distinctive bony lesion


first degree relative with NF1



progresses with age, some features are age dependent


lisch nodules and cutaenous fibromas develop late childhood to adulthood


inc risk for learning problems, seizures, hypertension, malignant neoplasms

NF1


incidence


inheritance


mutations


lab test


1/3000


inheritance: AD, variable expression, nearly complete penetrance, 50% are new mutations


mutations: allelic heterogeneity, NF1 gene chromsome 17q, protein is neurofibromin



lab test: dx depends on clinical features and family history


genetic testing includes protein truncation testing, sequencing, FISH testing, southern blotting detects up to 95%


gene sequencing alone detects 89% gene mutations


more deleted, earlier onset

management issues of NF1

complications increase with age


progression is cafe au lait spots at birth progressing in number, freckling, lisch nodules before adulthood and neurofibromas later in life


intellectual disability presents early, if present



surgical removal of neurofibromas not recommended



discuss genetic risks-- 50% have mild manifestations, 30% have severe complications


first degree relatives need examination

leigh syndrome and NARP


clinical features

mitochondrial DNA associated leigh and NARP are part of continuum of disorders



leigh: progressive neuro disease with motor and intellectual disability, onset typically between 3-12 months, periods of deterioration followed by plateaus with stabalization


early symptoms: poor suck, vomiting, irritability, failure to thrive, loss of head control, loss of motor skills


seizures, hypotonia, muscle weakness, movement disorder, ataxia, optho findings, respiratory difficulty


50% die by 3 yrs due to respiratory or cardiac failure



NARP: initial symptoms-- sensory neuropathy, muscle weakness, ataxia, ocular symptoms start with salt and pepper retinopathy which progresses to retinitis pigmentosa


migraines, seizures, hearing loss common

narp/leigh



inheritance pattern


mutation


lab test

interitance: locus heterogeneity, wiht mendelian (AR, x linked) and mitochondrial inheritance



NARP exhibits mitochondrial inheritance-- complicated by heteroplasmy (presence of more than 1 type of mitochondrial DNA)


threshold effect



mutations: 50% have mutation in MT-ATP6 gene


mutant loads <60% are either asymptomatic or have mild symptoms (most commonly pigmentary retinopathy or migraines)


mutant loads 70-90% have NARP and those with >90% have leigh syndrome



lab testing: if suspected, you need info from FH, blood and/or CSF lactate levels, neuroimaging and molecular genetic testing to confirm the diagnosis



management and counseling for NARP/leigh

management: symptomatic tx, regular neurolgoicla, optho, cardiology evaluations recommended



counseling: review recurrence risk, testing can be offered to mom and maternal relatives if interested


symptoms along family members variable, vaiability related to concept of mitochondrial genetic bottleneck

PCR:

amplify single cell into millions of copies of region of interest


rapid and inexpesnive


limited-- target can only be several hundred bases-- used for sensitivity

multiplex ligation dependent probe amplification


positives?

quantitative method detects larger deletions or insertions affecting multiple exons and whole genes


enables testing of all exons of gene in a single rxn

DNA sequencing


postives

allows examination of nucleotide sequence up to 500 bases


detects single base changes as well as small insertions and deletions

limitations of PCR based mutation detection analysis

large gains and losses will not generally be detected (except MLPA)


pcr relies on efficient annealing of oligonucleotides (primers) to target DNA


sequence variants within primer can interfere with annealing and thus amplification


a deletion that includes region targeted by primer will not be amplified

massivley parallel (nextgen) sequencing

micro/nanotechnologies used to sequence millions of rxns simultaneously


results in multiple reads for targeted sequenced regions that are compared to reference genome-- tons of info, need to trust machine


not good for detection of genomic structural rearrangements and repeat regions


can be used for panels of genes assoc w/ dvlpmnt delay, heraing loss, hereditary cancer, somatic mutations


whole exome sequencing or whole genome sequencing

inheritance of CF


what are 3 main reasons to do molecular testing for CF?

carrier status determination


prenatal testing (method that will detect familial mutations)


diagnostic testing (using a tiered approach-- panel of common mutations, sequence the coding regions, look for large duplications/deletions



oligonucleotide ligation assay measures 32 CF mutations for single multiplex rxn

locus heterogeneity


this refers to the presence of a number of differetn mutant alleles at a single locus (ex: >1000 CFTR mutations have been identified)


if testing targets specific known mutations, others may be missed

locus heterogeneity

locus is site of gene of chromosome


heterogeneity occurs when identical clinical symptoms are caused by defects at two or more genetic loci



ex: polcystic kidney disease can be caused by mutations in either PKD 1 or 2



so when testing a proband, a negative result may not rule out carrier or affected status

what disorders have CAG repeats?



CTG repeats?



CGG repeats?

spinocerebellar ataxia


huntington disease



CTG: myotonic dystrophy type 1



CGG repeats: fragile X syndrome (FRAXA)


myotonic dystrophy 1

AD, you have progressive muscle weakness, cardiac conduction abnormalities, cataracts, frontal balding, elevated risk of diabetes, testicular atrophy


normal is 5-39 CTG repeats


premutation: 39-50


mild: 50-100


100-1000- classic


>2000- congenital



shows genetic anticipation


but size of repeat does not predict severity of disease

congential DM (dystrophic myopathy?)



whom is usually inherited from?

hypotonia, intellectual disability, poor feeding, respiratory distress, bilateral facial weakness, myotonia develops at 5-10 yrs, nearly always inherited from MOTHERS



extra CUG repeat expansions in 3 prime untranslated region of DMPK takes up RNA splicing factors


so you have decreased RNA stability, altered protein conformation, differential RNA distribution --> RNA toxicity model

where is fragile X mutation?


what numbers correlate to mutation?


is expansion from maternal or paternal?


on FMR1 gene


6-45 is normal


45-55 is gray zone


55-200 is premutation


>200 is affected


expansion from mom

what is the significance of C in CG dinucleotide?

C in CG dinucleotides are susceptible to methylation


high level of methylation in a promoter can lead to transcriptional inactivation of a gene



FMR1 is hypermethylated when tract is >200 CGGs, leads to loss of expression of gene

what does genetic anticipation refer to with fragile X?

it refers to the increased probability that individuals in successive generations will be affected, but not increased severity (like in huntingtons)



females who are premutation carriers have a 20% risk of primary ovarian sufficiency



males have high risk of late onset ataxia/tremor syndrome (FXTAS): intention tremor, ataxia, cognitive decline, eventual brain atrophy

where do repeats tend to be in huntingtons and spinocerebellar ataxia?

CAG repeats usually in coding region of genes, resulting in tracts of poly-glutamine in protein products


lengths of repeat tracts leading to these disorders are generally much shorter than pathogenic repeat in noncoding regions


lengths of these repeat tracts leading to disorders are much shorter htan pathogenic repeats in noncoding regions (adult onset is 40-59 repeats)



aggregates of poly-glutamine tracts are formed in nuclei of neurons and are toxic to the cells

what are 2 ways to get PKU?



how do you screen for PKU?

deficiency of enzyme phenylalanine hydroxylase or deficiency in cofactor, tetrahydrobiopterin



untreated PKU have mental retardation, autistic like behavior, hyperactivity, eczema, seizures



screened by tandem mass spectroscopy

how do you screen for hypothyroidism?

screen by FIA for T4 and TSH (fluroimmune assay)


confirmation requires serum T4 and TSH measurements

screening for sickle cell

isoelectric focusing


other hemoglobinopathies can be detected by screening method



sickle cell is due to abnromal synthesis of beta chains in hemoglobin, affected indiv have hemolytic anemia, vaso occlusive crises, functional asplenia, splenic sequestration


can be lethal in infancy due to sepsis/splenic sequestration

galactosemia

due to deficiency of galactose-1-phosphate uridyltransferase (GALT) enzyme



presents in first two weeks of life with juandice, lethargy, cataracts, hepatomegaly, rapidly proceeds to death if untreated



AR inheritance


fluorometric assay for total galactose


confirmation with GALT activity and galactose-1-phosphate testing

congenital adrenal hyperplasia

family of disorders due to defects in biosynthesis of adrenal corticosteroids, 21 hydroxylase deficiency is most common type, produces ambiguous genitalia in girls


fluoroimmune assay for 17-hydroxyprogesterone


confirmation requires serum 17-hydroxy measurements

which disorders are detected by tandem mass spectometry?

----

tx of PKU



what about for moms?

substrate reduction--on a metabolite level



dietary restriction of phenylalanine


good outcome if treatment (PHE<600 micomoles/L) and compliance are adequate


dietary restriciton recommended for life


successful tx requires dietary intervnetion prior to 3 wks of age


compliance is an issue



for moms, phenylalanine levels need to be 120-360

tx for galactosemia



signs?


what happens even if treated since birth?

presents iwth jaundice, lethargy, cataracts, hepatomegaly, failure to thrive, E coli sepsis-- death if untreated


any infant suspected of it should be put on galactose- free diet (soy formula)


life long restriction of galactose and lactose



short stature, ovarian failure, visual-perceptual, speech, and other learning disabilites are common in patients treated from birth, in spite of dietary compliance

how do you tx urea cycle disorders?



signs of hyperammonemia

specific enzyme deficiency involved in ammonia detoxification


wide range of clinical severity



progressive increase in ammonia results in abnormal brain function and cerebral edema


(damage/death)



signs of hyperammonemia: dec mental awareness, vomiting, combativeness, slurred speech, unstable gait, unconciousness



tx: dec endogenous protein breakdown by giving non-protein calories, limit dietary protein, removal of plsama ammonia by hemodialysis,



increase waste nitrogen excretion (diversion)-- through arginine, citrulline, sodium benzoate, sodium phenylacetate or phenylbutyrate

how do you treat gaucher disease?

lysosomal storage disorder caused by def of enzyme glucocerebrosidase, result in glycolipids primary in macrophages and in CNS



clinical features: enlarged spleen and liver, anemia, thrombocytopenia, skeletal involvment (osteoporosis, bone infarcts), neuro dz in severe form



giving n-butyldcoxynojirmycin inhibits synthesis of glucosylceramide

how do you tx glycogen storage disorder?

GSD type 1 is characterized by hypoglcyemia 2-3 hrs after eating


secondary to inability to release free glucose from liver


(results in hepatomegaly, poor growth, lactic acidosis, hyperlipidemia, easy bruising, possibly seizures



tx with frequent feedings with glucose or glucose polymers-- raw cornstarch

biotinidase deficiency



what does it cause?


tx?

seizures, ataxia, hypotonia, developmental delay, skin rash, alopecia



treated with oral biotin in free, unbound form

how does chaperone therapy work?


what is it used to treat?

chaperones aid in normal folding of proteins


30% of normal proteins do not become functionally active bc they misfold and/or aggregate and are degraded by proteasome



increase in enzyme activity from chaperone is small, but is sufficient to improve or normalize metabolic activity



used in tx of homocystinuria and lysosomal storage disorders

what is stop-codon-read through therapy?

premature stop codons are common in pts with metabolic disorders


aminoglycoside antibiotics and other chemical compounds allow the ribosome to read through or bypass the premature stop signals in mRNA and continue the translation process to make a full length and functional protein



PTC124 drug now exists-- acts on premature, but not natural stop codons


works in pts with cystic fibrosis and muscular dysrtophy

where does recombinant enzymre replacement therapy work?

lysosomal storage disorders


enzymatic correction possible at hte cellular level in LSD fibroblasts (release correction factors)


efficient mannose 6 phosphate receptor mediated enzyme uptake occurs in fibroblasts



also used in gaucher disease (glucocerebrosidase and cerezyme)


also for mucopolysacrrhardisses (MPS)-- due to breakdown of glycosaminoglycans (GAG)


recombinant enzyme helps the somatic skeletal disease but not the CNS symptoms-- prevention more important than tx



official list: gaucher disease, fabry disease, MPS, pompe, niemann pick

what hepatic based metabolic disorders are sutiable for liver transplantation?

alpha 1 antitrypsin


wilsons


urea cycle disorders


organoacidopathies


CF

what are hematopoietic stem cell transplantation used for?

tx for LSD and MPS



microglia of CNS are of bone marrow origin, source of enzyme in brain after HSCT-- can reduce neuro progression


recommended for MPS 1 under 2 yrs of age


not recommneded with MPS 2 or 3



has CNS benefit for MLD and krabbes

when are most structural birth defects determined?


what is most important stage of life?

during first 2 months after conceptions (weeks 3-8)


gastrulation

what is a malformation?



examples?

non-progressive, congential morphologic anomaly of single organ or body part due to an alteration of primary developmental program



intrinsically abnormal developmental process


inborn error of morphogenesis



ex: cardiovascular: VSD, conotruncal defects


cleft lip/cleft palate


neural tube defects (anencephaly, meningomyelocele, encephalocele)


craniosyntoses (premature closure of one of the cranial sutures)

deformation



when does it most likely occur?

altered position or shape of body part due to an aberrant mechanical force that distorts and otherwise normal structure (caused by mechanical forces)


ex: bowed legs, tibial torsion, clubfoot (talipes deformities), dislocated hip, craniofacial asymmetry, positional plagiocephaly (abn shape head)



frequently a third trimester phenomenon associated with fetal crowding


disruption

non-progessive, congential morphologic anomaly due to breakdown of body structure that had a normal devleopmental potential



extrinsic breakdown or interference with normal developmental process


amniotic bands

dysplasia



types of dysplasia

morphologic anomaly arising prenatally or postnatally from dynamic or ongoing ateraltion of cellular consitution, tissue organizaiton, or function within a specific organ or tissue type


ex:


skeletal dysplasia: abn bone and jiont: osteogenesis imperfecta, achondroplasia-- most are mendelian


some teratogens (warfarin/coumadin cause strippled epiphyses)



may have assoc malformations: cleft palate, polydactylyl, craniosynostosis


may have assoc deformations: talipes, long bone bowign

achondroplasia

short arms and leg bones but average sized trunk

2 collagenopathies

short trunk


high myopia


cleft palate



stickler syndrome

ectodermal dysplasia

affects skin, sweat pores, hair, teeeth, nails, hearing


most are mendelian (single gene) disorders


may have minor anomalies/dysmorphic features (sparse/absent hair, everted lower lip, thin nails)

what are minor anomalies?



which traits are AD?


what do 3+ traits suggest?

minimal health consequence but may have modest impact on appearance


calvaria shape, ear sieze, eye spacing, scalp hair patterning, palmar creases



AD traits: preauricular tags/pits, polydactyly, clinodactyly (curving of fingers), transverse palmar crease



3+ suggests defect in morphogenesis

what is a sequence?



3 specific examples

single primary defect with secondary or tertiary anomalies



one or more secondary morphologic anomalies konwn or presumed to cascade from a single malformation, deformation, disruption, dysplasia



ex: pts with neural tube defect have clubfoot, hydrocephalus that arise secondary to NTD



potter sequence: renal ageneisis, oligohydramnios, lung hypoplsia, fetal compression wiht deformities (flat face ,talipes)



turner syndrome: upper lymphatic channels fail to develop normally-- dsitends jugular lymphatic sac (fetal edema-- swelling of face and hands, overgrowth of skin, webbed neck, ear anomalies, puffy hands and feet, deep set nails

syndrome:

more than one tissue, organ system, region of body-- associated with minor or major anomalies that are not secondary effects


multiple features thought to be pathogenetically related



caused by chromosomal abnromalities, single gene mutations, gestational/prenatal tratogenic effects



common teratogens

prenatal alcohol


ACE inhibitors (lopril drugs)


carbamazepine, cocaine, lithium, tetracyclines, valproate, others)



timing is important-- worse when admin during gastrulation stage-- third week of devleopment (day 17)



at this time, you get FAS facies


can also get alcohol related neurodevelopmental disorders and alcohol related birth defects



(short palpebral fissures, long upper lip with deficient philtrum)

ARBD

alcohol related birth defects


heart/CV


CNS: structural and functional


poor linear growth and weight gain


kidney, ears, eyes, face

what are nonsyndromic malformations

isolated, only one primary malformation (orofacial clefts, NTDS, CV malformations, renal agenesis, etc)



most do NOT follow mendelian patterns


recurence rates are requently in range of 1-5%


usually complex-- combinatio nof single gene and multifactorial causation



complex human diseases:


birth defects, intellectual disability, ADHD, autism, cancer, asthma



adults: HTN, stroke, diabetes, cancer, asthma, alcoholism, etc

what are discontinuous traits?

medical conditions or abormal triats


recognized significant deviations from the norm



signs and symptoms are either PRESENT or ABSENT: short stature, hypertension, microcephaly, intellectual disability


can invoke a threshold

which neuroimaging is better?



RI is better than CT for most patients-- higher resolution with no exposure to x rays


CT better for craniosynostosis and calcifications

williams syndrome

characteristic behavioral and developmental phenotype


very friendly, ADD problems, anxiety, low average


deletion 7 q11/23-- elastin gene


distinctive facial features


supravalvular aortic stenosis, pulm artery stenosis, hypercalcemia



most cases are de novo deletions, but can also involve incidences of inheritance from affected parents with balanced chromosome rearrangement involving WS critical region

which syndromes have cleft lips?

trisomy 13--microcephaly, scalp defects, clefts, micropthalmia, polydactyly, cardiac defects, renal anomalies


mean life expectancy 130 days



wolf-hirschhorn syndrome: 4p minus-- heart defects, dymorphic features, heart defects, intellectual disability, cleft lip/palate



22q deletio nsyndrome-- also have conotruncal heart defects, hypocalcemia, immune dysfunction, learning disabilities, psych disorders


(digeorge)



van der woude syndrome-- lip ptis, cleft lip, cleft palate--AD with variable expression



native american myopathy-- AR-- cleft palate, club feet, risk of malignant hyperthermia with general anesthesia, due to mutation of STAC3 gene



teratogenic: can be seen in FAS



robin sequence: micrognathia (undersized jaw), glossoptosis (retracted tongue), cleft palate-- all due to undersized jaw)



cleft palate can also be due to disruption (amniotic bands)



prenatal testing can detect cleft lip but not cleft palate