You are on page 1of 20

CHAPTER 5: GENETIC DISORDERS Mutations within Noncoding Sequences

 Transcription of DNA is initiated and regulated by promoter and enhancer sequences


GENES AND HUMAN DISEASE  Point mutations involving these regulatory sequences interfere w/ binding of
transcription factors  lead to marked reduction or total lack of transcription
o Eg., thalassemia
 Human genetic disorders can be broadly classified into three categories:  Point mutations w/in introns may lead to defective splicing of intervening sequences
 interferes w/ normal processing of initial mRNA and results in a failure to form
Mutations in  Cause the disease or predispose to the disease (exeption: mature mRNA
single genes w/ hemoglobinopathies)  Translation cannot take place  gene product is not synthesized
large effects  Highly penetrant  presence of the mutation is asso. w/ the
disease in a large portion of individuals
 Follow classic Mendelian pattern of inheritance Deletions and Insertions
 Referred to as Mendelial disorders  Small deletions or insertions involving coding sequence have two effects.
Chromosomal  Arise from structural or numerical alteration in the
disorders autosomes and sex chromosomes  If number of base pairs involved is three or multiple of three, the reading frame
 Uncommon but asso. w/ high penetrance will remain intact, and an abnormal protein lacking or gaining one or more amino
Complex  More common acids will be synthesized
multigenic  Caused by interactions between multiple variant forms of  If the number of affected coding bases is not a multiple of three, this will result in
disorders genes and environmental factors  also called an alteration of the reading frame of the DNA strand, producing a frameshift
polymorphism mutation
 Each variant gene confers small increase in disease risk; no
single gene is sufficient to produce a disease
 Several polymorphisms needed to cause a disease 
Trinucleotide-repeat Mutations
multigenic or polygenic
 Belong to a special category of genetic anomaly
 Low penetrance
 Cxd by amplification of a sequence of three nucleotides
 Multifactorial disorders
 Almost all affected sequence share the nucleotides guanine (G) and cytosine (C)
 Atherosclerosis, DM, hypertension, and autoimmune disease
 E.g., fragile X syndrome  familial mental retardation 1 (FMR1)  mental retardation
 Distinguishing feature of trinucleotide-repeat mutations is that they are dynamic
Mutations (degree of amplification increases during gamtogenesis)
o Influence the pattern of inheritance and the phenotypic manifestations of
 Mutation – permanent change in DNA diseases
o Germ cells affected are transmitted to the progeny and can give rise to
inherited diseases
 Transcription may be suppressed by gene deletions and point mutations involving
o Mutations that arise in somatic cells do not cause hereditary diseases;
promoter sequences
important in cancer genesis and congenital malformations
 Abnormal mRNA processing result from mutations affecting introns or splice
 General principles relating to the effects of gene mutation:
junctions or both
Point Mutations within Coding Sequences  Translation is affected if a nonsense mutation creates a stop codon (chain
 Point mutation – change where a single base is substituted with a different base termination mutation) within an exon
 May alter the code in a triplet of bases  lead to replacement of one amino acid   Three major categories of genetic disorders:
alter the meaning of sequence of the encoded protein  called missense mutation o Disorders related to mutant genes of large effect
 Conservative missense mutation – if substituted amino acid is biochemically o Diseases with multifactorial inheritance
similar to the original and causes little change in the function of the protein o Chromosomal disorders
 Nonconservative missense mutation – replaces the normal amino acid w/ a
 Single-gene disorders with nonclassic patterns of inheritance – caused by
biochemically different one
o E.g., sickle mutation affecting the β-globin chain of Hb  CTC (glutamic mutations in single-genes, but they do not follow the Mendelian pattern of inheritance
acid) is replaced by CAC (encodes valine)  alters physicochemical o Triple-repeat mutations, mutations in mtDNA, genomic imprinting or
properties of Hb  sickle cell anemia gonadal mosaicism
 Nonsense mutation – point mutation where amino acid codon is change to a chain  Hereditary disorders – derived from one’s parentes and transmitted in the germ
terminator or stop codon line through the generations and therefore are familial
o E.g., CAG creates a stop codon (UAG) is U is substituted for C  β0-  Congenital – “born with”; some are not genetic (e.g., congenital syphilis)
thalassemia
o Not all genetic diseases are congenital
abmrmtmd
MENDELIAN DISORDERS 2. Key structural proteins, such as collagen and cytoskeletal elements of the red cell
membrane (e.g., spectrin)
 Mendelian disorders - result of mutations in single genes that have large effects o E.g., collagen is a trimer where the three collagen chains are arranged in
o dominant or recessive helical form  must be normal for assembly and stability  normal
collagen trimers cannot be formed if there is a single mutant collagen chain
o Codominance – both alleles of gene pair are expressed or contribute to the
o Dominant negative – mutant allele where it impairs the function of a
phenotype; e.g., histocompatibility or blood group Ag
normal allele
 Sickle cell anemia – caused by substitution of normal Hb (HbA) by HbS
o Homozygote – all Hb is abnormal; disorder is fully expressed  Gain-of-function mutation – transmission is almost always autosomal dominant;
o Heterozygote – proportion of Hb is HbS (the rest is HbA); red cell sickling can take two forms:
occurs only under unusual circumstances (e.g., lowered O2 tension)  o Increase in a protein’s normal function (e.g., excessive enzymatic activity)
sickle cell trait o Impart a whole new activity completely unrelated to the affected protein’s
normal function
 Pleiotropism – single mutant gene may lead to many end effects (sickle cell anemia)
 Huntington disease – gain of function mutation;
 Genetic heterogenecity – mutations at several genetic loci may produce the same o Huntingtin (abnormal protein) – toxic to neurons; even heterozygotes
trait (profound childhood deafness) develop a neurologic deficit

Transmission Patterns of Single-Gene Disorders Autosomal Dominant Disorders


System Disorder
 Mutations involving single genes typically follow one of three patterns of inheritance: Nervous Huntington disease; Neurofibromatosis; Myotonic dystrophy;
o Autosomal dominant Tuberous sclerosis
o Autosomal recessive Urinary Polycystic kidney disease
o X-linked Gastrointestinal Familial polyposis coll
Hematopoietic Hereditary spherocytosis; von Willebrand disease
Skeletal Marfan syndrome; Ehlers-Danlos syndrome; Osteogenesis
Autosomal Dominant Disdorders
imperfect; Achondroplasia
 Manifested in the heterozygous state, so at least one parent of an index case is
Metabolic Familial hypercholesterolemia
usually affected
Acute intermittent porphyria
 Both males and females are affected; both can transmit the condition
 Characterized by the following:

 With every autosomal dominant disorder, some proportion of patients do


not have affected parents Autosomal Recessive Disorders
o New mutations involving egg or sperm from which they were derived   Make up the largest category of Mendelian disorders
related to the effect of the disease on reproductive capability  They occur when both alleles at a given gene locus are mutated; characterized by:
o Siblings are neither affected nor at increased risk
o Many new mutations occur in germ cells of older fathers 1. The trait does not usually affect the parents of the affected individuals; but siblings
 Clinical features can be modified by variations in penetrance and may show the disease
expressivity 2. Siblings have one chance in four of having the trait (25%)
o Incomplete penetrance – inherit the mutant gene but are phenotypically 3. If the mutant gene occurs with a low frequency in the population, there is a strong
normal likelihood that the affected individual (proband) is the product of a consanguineous
o Variable expressivity – trait is seen in all individuals carrying the mutant marriage
gene but is expressed differently among individuals ( neurofibromatosis 1)
 Ff features distinguish AR from AD diseases:
 The biochemical mechanisms of autosomal dominant disorders depend upon the
nature of the mutation and the type of protein affected  Expression of the defect tends to be more uniform than in autosomal dominant
 Most mutations may lead to the reduced production of a gene product or give rise to disorders
dysfunctional or inactive protein  Complete penetrance is common
 Many autosomal dominant diseases arising from deleterious mutations fall into one of  Onset is frequently early in life
a new familiar patterns:  Rarely detected clinically. (heterozygote + heterozygote = affected offspring)
 Many of the mutated genes encode enzymes. In heterozygotes, equal amounts of
1. Those involved in regulation of complex metabolic pathways that are subject to normal and defective enzyme are synthesized
feedback inhibition
o E.g., LDL receptors  in familial hypercholesterolemia, 50% loss of LDL  Autosomal recessive disorders include almost all inborn errors of metabolism
receptors results in secondary elevation of cholesterol
abmrmtmd
Autosomal Recessive Disorders Biochemical and Molecular Basis of Single-Gene (Mendelian) Disorders
System Disorder
Metabolic Cystic fibrosis; Phenylketonuria; Galactosemia; Homocytinuria;  Mendelian disorders result from alterations involving single genes (table 5-4)
Lysosomal storage disease; a1-antitrypsin deficiency; Wilson  Genetic defect may lead to the formation of an abnormal protein or a reduction in the
disease; Hemochromatosis; Glycogen storage disease output of the gene product
Hematopoietic Sickle cell anemia; Thalassemia  Any type of protein may be affected
Endocrine Congenital adrenal hyperplasia
 Mechanisms involved in single-gene disorders are classified into four categories:
Skeletal Ehler-Danlos syndrome; Alkpatonuria
1. Enzyme defects and their consequences
Nervous Neurogenic muscular atrophies; Friedreich ataxia; Spinal muscular
atrophy 2. Defects in membrane receptors and transport systems
3. Alterations in the structure, function, or quantity of nonenzyme proteins
4. Mutations resulting in unsual reactions to drugs

X-Linked Disorders Enzyme Defects and their Consequences


 All sex-linked disorders are X-linked, and almost all are recessive  Mutations may result in the synthesis of an enzyme with reduced activity or a reduced
 Males with mutations affecting the Y-linked genes are usually infertile, and hence amount of a normal enzyme
there is no Y-linked inheritance  Consequence is metabolic block
 X-linked recessive inheritance accounts for small number of well-defined clinical  Biochemical consequences of an enzyme defect in such a reaction may lead to three
conditions major consequences:
 Y chromosome is not homologous to X  male is said to be hemizygous for X-linked
mutant genes  disorders are expressed on males 1. Accumulation of substrate
 Features that characterize these disorders: o May be accompanied by accumulation of intermediates
 Affected male does not transmit the disorder to his sons, but all daughters are o Tissue injury may result if the precursor (intermediates or products) are
carriers. Sons of heterozygous women have one chance in two of receiving the toxic in high concentrations
mutant gene o Ex: galactosemia  deficiency of G1PUT leads to accumulation of
 Heterozygous female does not express the full phenotypic change because of the galactose and tissue damage
paired normal allele; express the disorder partially. o Lysosomal storage disease – excessive accumulation of complex
 X-linked disorders – transmitted by an affected heterozygous female to half her substrates w/in the lysosomes as a result of deficiency of degradative
sons and half her daughters; and by an affected male to all his daughters but none of enzymes
his sons (if the female parent is unaffected) 2. An enzyme defect can lead to a metabolic block and a decreased amount of
o E.g., Vitamin D-resistant rickets end product
o Ex: melanin deficiency may result from lack of tyrosinase  albinism
X-Linked Recessive Disorders o If end product is a feedback inhibitor of enzymes involved in early reactions
System Disease  deficiency of end product may permit overproduction of intermediates
Musculoskeletal Duchenne muscular dystrophy and their catabolic products  toxic at high concentrations
Blood Hemophilia A and B; Chronic granulomatous disease; G6PD deficiency o Ex: Lesch-Nyhan syndrome
Immune Agammaglobulinemia; Wiskott-Aldrich syndrome 3. Failure to inactivate a tissue-damaging substrate
o Ex: a1-antitrypsin deficiency  unable to inactivate neutrophil elastase
Metabolic Diabetes insipidus; Lesch-Nyhan syndrome
in their lungs  destruction of elastin in the wall of lung alveoli 
Nervous Fragile X suyndrome
pulmonary emphysema

Defects in Receptors and Transport System


KEY CONCEPTS: Transmission Patterns of Single-Gene Disorders
 Biologically active substances have to be actively transported across the cell
 Autosomal dominant disorders – cxd by expression in heterozygous state; affect
 Transport is achieved by receptor-mediated enocytosis
males and females equally, and both sexes can transmit the disorder
 Genetic defect in a receptor-mediated transport system
 Enzyme proteins are not affected in autosomal dominant disorders; instead receptors
o Familial hypercholesterolemia  ↓ fxn or synthesis of LDL receptors 
and structural proteins are involved
defective transport of LDL into cells and secondarily to excessive cholesterol
 Autosomal recessive diseases – occur when both copies of a gene are mutated;
synthesis by complex intermediary mechanisms
enzyme proteins are involved; males and females are affected equally
o Cystic fibrosis  transport system for chloride ions in exocrine glands,
 X-linked disorders – transmitted by heterozygous females to their sons, who
sweat ducts, lungs, and pancreas is defective
manifest the disease. Female carriers usually are protected because of random
o Impaired chloride transport leads to serious injury to the lungs and
inactivation of X chromosome
pancreas

abmrmtmd
Alterations in Structure, Function, or Quantity of Nonenzyme Proteins Clinical Features
 Genetic defects resulting in alterations of nonenzyme proteins often have widespread
secondary effects (e.g., sickle cell disease, hemoglobinopathies)  Skeletal abnormalities are the most striking feature of Marfan Syndrome
 Hemoglobinopathies – quality defect; defects in structure of globin molecule  Px are unusually tall w/ long extremeties and long, tapering fingers and toes
 Thalassemia – quantity defect; mutations in globin genes that affect the amount of o Joint ligaments are lax  px is double-jointed
globin chains synthesized o Thumb can hyperextend back to the wrist
o Asso. w/ reduced amount of structurally normal a-globin or b-globin chains o Head is dolichocephalic (long-headed) w/ prominent frontal eminence and
 Osteogenesis imperfecta – defective structural collagen
supraorbital ridges
 Hereditary spherocytosis – defective spectrin
 Muscular dystrophies – defective dystrophin  Spinal deformities: kyphosis, scoliosis, or rotation or slipping of the dorsal or
lumbar vertebrae
 Chest is classically deformed  pectus excavatum (deeply depressed sternum) or a
Genetically Defective Adverse Reactions to Drugs pigeon-breast deformity
 Certain genetically determined enzyme deficiencies are unmasked only after exposure  Ocular changes  bilateral subluxation or dislocation of lens (ectopia lentis)
of the affected individual to certain drugs  Cardiovascular lesions  most life-threatening features
 Pharmacogenetics – special area of genetics o Two most common lesions: mitral valve prolapse and dilation of the
 G6PD – classic example of drug-induced injury; asso. w/ deficiency of the enzyme ascending aorta due to cystic medionecrosis
G6PD
 Causes of progressive dilation of the aortic valve ring and the root of aorta that give
o Primaquine – cause severe haemolytic anemia
 Genetic factors have major impact on drug sensitivity and adverse reactions rise to aortic incompetence
o Loss of medial support & Excessive TGF-b signalling in the adventitia
 Weakening of adventitia lead to intimal tear  initiate intramural hematoma that
Disorders Associated with Defects in Structural Proteins cleaves layers of media  produce aortic dissection
o Hemorrhage ruptures through the aortic wall
 Marfan Syndrome and Ehlers-Danlos syndromes (EDS) – affect connective o Cause of death in 30-45% of cases
tissue and involve multiple organs  Mitral valve lesions are more frequent but less important than aortic lesions
 Loss of connective tissue support in mitral valve leaflets makes them soft and billowy
Marfan Syndrome  creates floppy valve
 Disorder of connective tissues manifested principally by changes in the  Valvular lesions w/ lengthening of chordate tendinae  lead to mitral valve
skeleton,eyes and cardiovascular system regurgitation
 Prevalence 1 in 5000; autosomal dominant inheritance
 Great majority of deaths are caused by ruptures of aortic dissections followed by
 Pathogenesis: results from an inherited defect in an extracellular glycoprotein called
cardiac failure
fibrillin-1; two mechanisms where loss of fibrillin leads to clinical manifestations:
o Loss of structural support in microfibril rich connective tissue  Variability in clinical expression is seen w/in a family; interfamilial variability is more
o Excessive activation of TGF-b signalling common and extensive  clinical diagnosis of Marfan syndrome is based on “revised
Ghent criteria”
 Fibrillin – major component of microfibrils found in ECM; provide scaffolding where  Mainstay or medical treatment is administration of beta-blockers which likely act by
tropoelastin is deposited to form elastic fibers; fibrillin-1 and fibrillin-2 reducing heart rate and aortic wall stress
o Microfibrils are widely distributed in aorta, ligaments, and ciliary zonules
that support the lens  affected by Marfan syndrome
Classification of Ehlers-Danlos Syndromes
o Fibrillin 1 – encoded by FBN1 on chromosomes 15q21.1; most common EDS Type Clinical Findings Inheritance Gene Defects
defect; inhibit polymerization of fibrillin fibers (dominant negative effect) Classic (I/II) Skin & joint hypermobility, atrophic Autosomal Dominant COL5A1, COL5A2
o Fibrillin 2 – encoded by FBN2 on chromosomes 5q23.31; mutations are scars, easy bruising
less common and give rise to congenital contractural arachnodactylyl Hypermobility Joint hypermobility, pain, dislocations Autosomal Dominant Unknown
(autosomal dominant disorder) (III)
o Reduction of fibrillin content below a certain threshold weakens the Vascular (IV) Thin skin, arterial or uterine rupture, Autosomal Dominant COL3A1
connective tissue  haploinsufficiency bruising, small joint hyperextensibility
 Bone overgrowth and myxoid changes in mitral valves cannot be attributed to Kyphoscoliosis Hypotonia, joint laxity, congenital Autosomal recessive Lysyl hydroxylase
(VI) scoliosis, ocular fragility
changes in tissue elasticity
Arthrochalasia Severe joint hypermobility, mild skin Autosomal Dominant COL1A1, COL1A2
o Loss of microfibrils give rise to abnormal and excessive activation of TGF-b (VIIa,b) changes, scoliosis, bruising
(normal microfibrils sequester TGF-b and thus control bioavailability) Dermatosparaxis Severe skin fragility, cutis laxa, Autosomal recessive Procollagen N-
o Excess TGF-b has deleterious effects on vascular smooth muscle (VIIc) bruising peptidase
development and increase activity of metalloproteases , causing loss of ECM

abmrmtmd
Ehlers-Danlos Syndromes (EDS) KEY CONCEPTS
 EDSs comprise a clinically and genetically heterogenous group of disorders that Marfan Syndrome
result from some defect in the synthesis or structure of fibrillar collagen  Caused by a mutation in the FBN1 gene encoding fibrillin, w/c is required for
 Other disorders resulting from mutations affecting collagen synthesis: osteogenesis structural integrity of connective tissues and regulation of TGF-b signalling
imperfect, Alport syndrome, epidermolysis bullosa  Major tissues affeccted are the skeleton, eyes, and cardiovascular system
 Mode of inheritance encompases all three Mendelian patterns  Clinical features ay include tall stature, long fingers, bilateral subluxation of lens,
 Tissues rich in collagen (skin, ligaments, joints) are frequently involved in most mutral valve prolapsed, aortic aneurysm, and aortic dissection
variants of EDS; lack tensile strength  Clinical trials with drugs that inhibit TGF-b signalling (angiotensin receptor blockers) 
o Skin is hyperextensible; joints are hypermobile shown to improve aortic and cardiac function in mouse models
o Bending thumb backward to touch forearm Ehlers-Danlos Syndromes
o Bending knee forward to create right angle  There are six variants of syndromes, all characterized by defects in collagen synthesis
 Skin is extraordinarily stretchable, extremely fragile, and vulnerable to trauma or assembly. Each variants is caused by a distinct mutation involving one of several
 The basic defect in connective tissue may lead to serious internal complications collagen genes or genes that encode other ECM proteins like tenascin-X
o Rupture of colon and large arteries (vascular EDS)  Clinical features: fragile, hyperextensible skin vulnerable to trauma, hypermobile
o Ocular fragility w/ rupture of cornea and retinal detachment (kyphoscoliosis) joints, and ruptures involving colon, cornea, or large arteries. Wound healing is poor
o Diaphragmatic hernia (classic EDS)
 Kyphoscoliosis EDS – most common autosomal recessive form of EDS;
o Lysyl hydroxylase – enzyme need for hydroxylation of lysine during Disorders Associated with Defects in Receptor Proteins
collagen synthesis
 Vascular EDS results from abnormalities of type III collagen Familial Hpercholesterolemia
o Genetically heterogeneous; COL3A1
o Affect rate of synthesis of pro-a1 (III) chains; affect secretion of type III  Familial hypercholesterolemia is a “receptor disease” that is the consequence of a
collagen; others lead to the synthesis of structurally abnormal type III mutation in the gene encoding the receptor for LDL, which is involved in the transport
collagen
and metabolism of cholesterol
o Some allele behave as dominant negatives  severe phenotypic defects
o Results from mutations in structural protein rather than enzyme protein   There is loss of feedback control and elevated levels of cholesterol that induced
autosomal dominant inheritance is expected premature atherosclerosis  greatly increased risk of myocardial infarction
o Blood vessels and intestines are known to be rich in collagen type III   One of the most frequent occurring Mendelian disorders
consistent with severe structural defects  Heterozygotes: 2 to 3-fold elevation of plasma cholesterol  tendinous xanthomas
 Arthrochalasia type and dermatosparaxis type – fundamental defect is in the and premature atherosclerosis in adult life
conversion of type 1 procollagen to collagen
 Homozygotes: 5 to 6-fold elevations in plasma cholesterol  skin xanthomas and
o This step involves cleavage of noncollagen peptides at the N-terminus and
C-terminus of procollagen molecule  accomplished by N- and C- coronary, cerebral, and peripheral vascular atherosclerosis may develop at early age
terminal-specific peptidases o Myocardial infarction may occur before age 20 years
o Arthrochalasia type – defect in the conversion involve mutations that o Familial hypercholesterolemia is present in 3-6% of survivors of myocardial
affect two type 1 collagens genes, COL1A1 and COL1A2; abnormal pro-a1 infarction
or pro-a2 that resist cleavage of N-terminal peptides are formed
o Heterozygotes manifest the disease Normal Process of Cholesterol Metabolism and Transport
o Dermatosparaxis type – caused by mutations in te procollagen-N-
peptidase genes, essential for the cleavage of collagens; caused by enzyme  ~7% of body’s cholesterol circulates in plasma; predominantly in the form of LDL
deficiency and follows an autosomal recessive inheritance
 First step is secretion of VLDL (rich in TAG, less in cholesteryl esters) by the liver into
 Classic type of EDS – 30-50% mutations in genes for type V collagen (COL5A1,
COL5A2) blood stream
o Genes other than those that encode collagen may also be involved o When VLDL particles reach the capillaries or adipose tissue or muscle, it is
o Some cases genetic defects that affect the biosynthesis of other ECM cleaved by lipoprotein lipase  process that extracts most of the TAG
molecules that influence collagen synthesis indirectly may be involved o IDL (resulting molecule)  reduced in TAG content and enriched in
o Ex: mutation in tenascin-X (EDS-like condition)  affects synthesis and cholesteryl esters but retains 2 of 3 apoproteins (B-100 and E) present in
fibril formation of type VI and type I collagens
VLDL particle
 Common thread in EDS is abnormality in collagen  extremely heterogeneous
 After release from capillary endothelium, IDL particles have one of two fates:
o 50% of newly formed IDL is rapidly taken up by the liver by receptor-
mediated transport (the receptor recognized B-100 and E)  LDL receptor
(also involved in hepatic clearance of LDL)
 In liver cells, IDL is recycled to generate VLDL
abmrmtmd
o IDL particles not taken up by liver are subjected to further metabolic  Statins – suppress intracellular cholesterol synthesis by inhibiting the enzyme HMG
processing  removes most of the remaining TAG and apo-E  yields CoA reductase
cholesterol-rich LDL particles o Allow greater synthesis of LDL receptors
 IDL is the immediate and major source of plasma LDL o Used for secondary prevention of ischemic heart disease
 Two mechanisms for removal of LDL from plasma:
o Mediated by an LDL-receptor KEY CONCEPTS: Familial Hypercholesterolemia
o Mediated by a receptor for oxidized LDL (scavenger receptor)  Autosomal dominant disorder caused by mutations in the gene encoding the LDL
 70% of the plasma LDL is cleared by the liver receptor
 Patients develop hypercholesterolemia as a consequence of impaired transport of LDL
o First step: binding of LDL to cell surface receptors clustered in coated
into the cells
pits (specialized regions of the plasma membrane)  Heterozygotes: elevated serum cholesterol greatly increases the risk of atherosclerosis
o After binding: coated pits w/ receptor-bound LDL are internalized by and resultant coronary artery disease
invaginating to form coated vesicles then they migrate w/in the cell to fuse  Homozygotes: have greater increase in serum cholesterol and a higher frequency of
with lysosomes ischemic heart disease. Cholesterol also deposits along tendon sheaths to produce
o LDL dissociates from the receptor, which is recycled to the surface xanthomas
o In the lysosomes, LDL is enzymatically degraded; apoprotein part is
hydrolyzed to amino acids; cholesteryl esters are broken down to free
Disorders Associated with Defects in Enzymes
cholesterol
o Free cholesterol crosses the lysosomal membrane to enter cytoplasm where
Lysosomal Storage Disease
it is used for membrane synthesis and as a regulator of cholesterol
homeostasis
 Lysosomes – key components of the intracellular digestive tract; contain hydrolytic
o Exit of cholesterol from lysosomes requires the action of two proteins,
enzymes that have two special properties:
NPC1 and NPC2
o They function in the acidic milieu of the lysosomes
 Three separate processes are affected by the released intracellular cholesterol:
o These enzymes constitue a special category of secretory proteins that are
1. Cholesterol suppresses cholesterol synthesis w/in the cell by inhibiting the activity of
destined for intracellular organelles (requires special processing in golgi
the enxyme HMG CoA reductase  rate limiting enzyme in the synthetic pathway
apparatus)
2. Cholesterol activates the enzyme acyl-coenzyme A: cholesterol acyltransferase 
 Lysosomal enzyme or acid hydrolases – synthesized in the ER and transported to
favors esterification and storage of excess cholesterol
golgii apparatus
3. Cholesterol suppresses the synthesis of LDL receptors, thus protecting the cells from
o w/in golgi comlex they undergo posttranslational modifications including
excessive accumulation of cholesterol
attachment of terminal mannose-6-phosphate groups to oligosaccharide
 Monocytes and macrophages have receptors for chemically altered (acetylated or
side chains
oxidized) LDL
o phosphorylated mannose residues serves as “address label” recognized by
 In hypercholesterolemia, there is marked increase in scavenger receptor-mediated
receptors on the inner surface of the Golgi membrane
traffic of LDL cholesterol into the cells of the mononuclear phagocyte system and
o Lysosomal enzymes bind receptors and segregated from other secretory
vascular walls  responsible for the appearance of xanthomas and contributes to
proteins
the pathogenesis of premature atherosclerosis
 Genetically determined errors in this remarkable sorting mechanism may give rise to
 Familial hypercholesterolemia is classified into five groups:
one form of lysosomal storage disease
 Lysosomal enzymes catalyze breakdown of complex macromolecules
Class I Uncommon; lead to complete failure of synthesis of the receptor protein (null allele)
mutations o Autophagy – metabolic turnover of intracellular organelles
Class II Common; encode receptor proteins that accumulate in the ER because their folding o Heterophagy – acquired from outside the cells by phagocytosis
mutations defects make it impossible for them to be transported to the Golgi complex  Inherited deficiency of a functional lysosomal enzyme gives rise to two pathologic
Class III Affect the LDLD-binding domain of receptor; encoded proteins reach the cell surface consequences:
mutations but fail to bind LDL or do so poorly
Class IV Encode proteins that are synthesized and transported to the cell surface efficiently;
mutations bind LDL normally but fail to localize in coated pits; bound LDL is not internalized  Primary accumulation – catabolism of the substrate of the missing enzyme remains
Class V Encode proteins that are expressed on the cell surface, can bind LDL, and can be incomplete, leading to the accumulation of the partially degraded insoluble metabolite
mutations internalized; the pH-dependent dissociation of the receptor and the bound LDL fails w/in lysosomes
to occur; such receptors are trapped in the endosome, where they are degraded o Lysosomes are stuffed w/ incompletely digested macromolecules; become
and fail to recycle to the cell surface
large and numerous enough to interfere w/ normal cell functions

abmrmtmd
 Secondary accumulation – caused by impaired autophagy from autophagic Niemann-Pick Disease Types A and B
substrated such as polyubiquitinated proteins and old effete mitochondria  Types A and B are two related disorders that are cxd by lysosomal accumulation of
o Absence of this control mechanism causes accumulation of dysfunctional sphingomyelin due to an inherited deficiency of sphingomyelinase
mitochondria w/ poor calcium buffering capacity and altered membrane  Type A – severe infantile form w/ extensive neurologic involvement
potentials; trigger generation of free radicals and apoptosis o Marked visceral accumulations of sphingomyelin
o Progressive wasting & Early death w/in first 3 years of life
 Type B – px have organomegaly; NO CNS involvement; usually survive into adulthood
 Three general approaches to the treatment of lysosomal storage diseases
 Also common in Ashkenazi Jews
o Enzyme replacement therapy  Gene for acid sphingomyelinase maps to chromosome 11p15.4  one of the
o Substrate reduction therapy imprinted genes expressed from the maternal chromosome as a result of epigenetic
o Molecular chaperone therapy silencing of the paternal gene
 Distribution of stored material and organs affected is determined by two factors:  Typically inherited as an autosomal recessive
o The tissue where most of the material to be degraded is found  Heterozygotes who inherit the mutant allele from mother can develop Niemann Pick
o The location where most of the degradation normally occurs Disease
 Lysosomal storage disease can be divided based on the biochemical nature of
Morphology
accumulated metabolite; creating subgroups: (table 5-6)  Type A – missense mutation cause almost complete deficiency of sphingomyelinase
o Glycogenoses  Enzyme deficiency blocks degradation of the lipid  accumulates in lysosome (MPS)
o Sphingolipidoses (lipidoses)  Affected cells become enlarged (90 um in dm)) due to distention of lysosomes w/
o Mucopolysaccharidoses (MPSs) sphingomyelin and cholesterol
o Mucolipidoses  Innumerable small vacuoles uniform in size imparts foaminess to cytoplasm
 EM: vacuoles are engorged secondary lysosomes that contains cytoplasmic bodies
resembling concentric lamellated myelin figures ” zebra” bodies
Tay-Sachs Disease (GM2 Gangliosidosis: Hexosaminidase α-Subunit Deficiency)
 Lipid-laden phagocytic foam cells are seen in spleen, liver, lymph nodes, BM, tonsilds,
 GM2 gangliosidoses – group of three lysosomal storage diseases caused by an
GIT and lungs
inability to catabolise GM2 gangliosides
 Involvement of spleen generally produces massive enlargement (10x)
 Degradation of GM2 gangliosides requires three polypeptides encoded by three distinct
 Brain: gyri are shrunken and the sulci are widened
genes
 Neural involvement is diffused  affects all parts of nervous system
 Tay-Sachs disease – the most common form of GM2 gangliosidosis; results from
 Vacuolation and ballooning of neurons constitute the dominant histologic change
mutations in the α-subunit locus on chromosome 15; cause severe deficiency in
 leads to cell death and loss of brain substance
hexosaminidase A
 Retinal cherry-red spot is present in 1/3 of cases
 Prevalent among Jews (from Eastern European (Ashkenazic) origin); 1 in 30
Clinical Features
Morphology
 Clinical manifestations in type A are present at birth and become evident by age 6
 Hexosaminidase A – absent in all tissues so GM2 ganglioside accumulate in many
 Infants typically protuberant abdomen because of hepatosplenomegaly
tissues (heart, liver, spleen, nervous system)
 Progressive failure to thrive, vominting, fever, and generalized lymphadenopathy as
 Involvement of neurons in the CNS and ANS and retina dominates clinical pic
well as progressive deterioration of psychomotor follows after appearance of
 Histological: neurons are ballooned w/ cytoplasmic vacuoles  markedly distended
manifestations
lysosome filled w/ gangliosides; Oil red O and Sudan black B positive
 Death at 1st or 2nd year of life
 EM: cytoplasmic inclusions can be seen; prominent whorled configurations w/in
 Diagnosed by biochemical assays for sphingomyelinase activity in liver or BM biopsy
lysosomes composed of onion-skin layers of membranes
 DNA analysis – detects individuals affected w/ types A and B as well as carriers
 Cherry-red spot thus appears in the macula  represents accentuation of the
normal color of macular choroid

Clinical Features Niemann-Pick Disease Type C


 Affected infants are normal at birth but begin to manifest signs at 6 months  Distinct at biochemical and genetic levels; more common
 Motor and mental deterioration  motor incoordination, mental obtundation leading  Mutations in NPC1 and NPC2 lead to NPC (NPC1 mutations – 95% of cases)
to muscular flaccidity, blindness and increasing dementia  NPC is due to a primary defect in nonenzymatic lipid transport
 Early course of dx  cxc cherry-red spot appears in macula almost in all px  NPC1 – membrane bound; NPC2 – soluble (both are involved in transport of free
 1-2 years complete vegetative state; death at age 2-3 years cholesterol from the lysosomes to the cytoplasm)
 100 mutations in α-subunit gene  unfolded protein  apoptosis  Clinically heterogenous
 Antenatal diagnosis and carrier detection by enzyme assay and DNA-based analysis  May present as hydrops fetalis and stillbirth, as neonatal sepsis or as a chronic form
 Chaperone therapy cxd by progressive neurologic damage (more common)
 **Sandhoff disease - GM2 gangliosidoses β-subunit deficiency  Progressive neurologic damage – marked by ataxia, vertical supranuclear gaze
palsy, dystonia, dysarthria, psychomotor regression
abmrmtmd
Gaucher Disease Clinical Features
 Gaucher disease refers to a cluster of autosomal recessive disorders resulting  Type I – signs and symptoms first appear in adult life and are related to
from mutations in the gene encoding glucocerebrosidase splenomegaly or bone involvement
 Most common lysosomal storage disorder o (+) pancytopenia or thrombocytopenia secondary to hypersplenism
 Affected gene encodes glucocerebrosidase  enzyme that cleaves the glucose o Extensive expansion of marrow space  pathologic fractures and bone pain
residue from ceramide o Disease is compatible with long life
 Glucocerebroside accumulates prinicipally in phagocytes (also in CNS)  Types II and III – CNS dysfunction, convulsions, and progressive mental
 Glucocerebrosides – continually formed from the catabolism of glycolipids derived deterioration dominate, although organs such as the liver, spleen, and lymph nodes
from cell membrane of senescent WBC and RBC are also affected
 Pathologic changes in GD are caused by:  Diagnosis of homozygotes can be made by measurement of glucocerebrosidase
o Burden of storage material activity in peripheral blood WBC or in extracts of cultures skin fibroblasts
o Activation of macrophages and consequent secretion of cytokines (IL-1, IL-6  Heterozygotes can be identified by detection of mutations
and TNF)  Replacement therapy w/ recombinant enzymes is the mainstay for treatment of
 Three subtypes of Gaucher disease: gaucher disease
o Effective; type 1 cases can expect normal life (extremely expensive)
Type I  Chronic nonneuropathic form; most common (99% of cases)  Allogenic hematopoietic stem cell transplantation can be curative  because
 Storage of glucocerebrosides is limited to the mononuclear fundamental defect resides in mononuclear phagocytic cells from marrow stem cells
phagocytes WITHOUT involving the brain
 Splenic and skeletal involvements dominates the pattern of the dx
 Found principally in Jews of European stock Mucopolysaccharidoses (MPS)
 Px have reduced but detectable levels of glucocerebrosidase activity  MPSs are a group of closely related syndromes that result from genetically determined
 Longevity is shortened but not markedly deficiencies of enzymes involved in the degradation of mucopolysaccharides
Type II  Acute neuropathic Gaucher disease (glycosaminoglycans); MPS I to MPS VII
 Infantile acute cerebral pattern  Mucopolysaccharides – long-chain complex carbs that are linked w/ proteins to
 No predilection for Jews form proteoglycans; abundant in the ground substance of CT
 Px have no detectable glucocerebrosidase activity in tissues  Glycosaminoglycans that accumulate in MPS are:
 Hepatosplenomegaly is present; clinical picture is dominated by o Dermatan sulfate
progressive CNS involvement leading to death at early age o Heparan sulfate
Type III  Intermediate between types I and II o Keratan sulfate
 Patients have systemic involvement cxc of type I but have o Chondroitin sulfate
progressive CNS disease that begins in adolescence or early  Enzymes involved in degradation cleave terminal sugars from polysaccharide chain
adulthood o If absent, these chains accumulate w/in lysosomes
 All MPS except one are inherited as autosomal recessive traits
Morphology o Hunter syndrome – X-linked recessive trait
 Gaucher cells – distended phagocytic cells; found in spleen, liver, BM, lymph nodes,  MPS are progressive disorders, cxd by:
tonsils, thymus, Peyer patches o Coarse facial features
o Rarely appear vacuolated; instead have a fibrillary type of cytoplasm likened o Clouding of the cornea
to crumpled tissue paper o Joint stiffness
o Often enlarged (100um in dm); have one or more dark, eccentrically placed o Mental retardation
nuclei  Urinary excretion of the accumulated mucopolysaccharides is often increased
o PAS intensely positive Morphology
 EM: the fibrillary cytoplasm can be resolved as elongated, distended lysosomes,  Accumulated mucopolysaccharides are generally found in mononuclear phagocytic
containing the stored lipid in stacks of bilayers cells, endothelial cells, intimal smooth muscle cells and fibroblasts
 Type I – enlarged spleen (10kg); lymphadenopathy is mild to moderate (body-wide)  Common sites involve are spleen, liver, BM, lymph nodes, blood vessels, heart
o Accumulation of Gaucher cells in BM (70-100% of type 1 cases)   Microscopically: affected cells are distended and have apparent clearing of the
produce areas of bone erosions  pathologic fractures cytoplasm to create so-called balloon cells
 In px w/ cerebral involvement, Gaucher cells are seen in the Virchow-Robin  EM: clear cytoplasm can be seen as numerous minute vacuoles  swollen lysosomes
spaces, and arterioles are surrounded by swollen adventitial cells containing finely granular PAS + material (identified as mucopolysaccharide)
 There no storage of lipids in the neurons, yet neurons appear shrivelled and are  Some of the lysosomes in neurons are replaced by lamellated zebra bodies (similar to
progressively destroyed those seen in Niemann-Pick dx)
 Lipids that accumulate in the phagocytic cells around blood vessels secrete cytokines  Hepatosplenomegaly, skeletal deformities, valvular lesions, and subendothelial arterial
that damage nearby neurons deposits, particularly in the coronary arteries, and lesions in the brain are common
threads that run through all of the MPSs
 Important causes of death: myocardial infarction and cardiac decompensation
abmrmtmd
 Glycogen storage diseases – result from a hereditary deficiency of one of the
Clinical Features enzyme involved in the synthesis or sequential degradation of glycogen
 Hurler syndrome (MPS I-H) – results from a deficiency of α-1-iduronidase; one of  Glycogen – storage form of glucose
the most severe form of MPS o Glycogen synthesis begins w/ the conversion of glucose to G6P by
o Affected children: normal at birth but develop splenomegaly by age 6-24
hexokinase (glucokinase)
months
o Growth is retarded; develop coarse facial features and skeletal deformities o Phosphoglucomutase then transforms G6P to G1P then converted to
o Death occurs by age 6-10 years; often due to cardiovascular complications uridine diphosphoglucose
 Hunter syndrome (MPS II) – differs from Hurler syndrome in mode of inheritance o Highly branched, large polymer is then built , containing 10,000 glucose
(X-linked), absence of corneal clouding and milder clinical course molecules linked together by α-1,4-glucoside bonds
o Glycogen chain & branches continue to elongate by the addition of glucose
molecules mediated by glycogen synthetase
KEY CONCEPTS: Lysosomal Storage Diseases o During degradation, phosphorylases in the liver and muscle pit G1P from
 Lysosomal storage disease – inherited mutations leading to defective lysosomal glycogen until about four glucose residues remain in each branch, leaving a
enzyme functions gives rise to accumulation and storage of complex substrates in the
branched oligosaccharide called limit dextrin  can be degraded only by
lysosomes and defects in autophagy resulting in cellular injury
the debranching enzyme
Tay-Sachs Disease  Caused by an inability to metabolize GM2 gangliosides o Glycogen is also degraded in the lysosomes by acid maltase
due to lack of the α subunit of lysosomal o If lysosomes are deficient in this enzyme, the glycogen contained w/in them
hexosaminidase. is nor accessible to degradation by cytoplasmic enzymes such as
 GM2 gangliosides accumulate in the CNS and cause phosphorylases
several mental retardation, blindness, motor  On the basis of pathophysiology glycogenoses can be divided into three major
weakness, and death by 2-3 years of age
subgroups: (Table 5-7)
Niemann-Pick Disease  Caused by deficiency of sphingomyelinase
Types A & B  In more severe type A variant, accumulation of
sphingomyelin in the nervous system results in Hepatic forms  Liver – key player in glycogen metabolism
neuronal damage  Contains enzymes that synthesize glycogen and break it down
 Lipid also is stored in phagocytes w/in the liver, to free glucose
spleen, BM, and lymph nodes, causing their  Inherited deficiency of hepatic enzymes involved in glycogen
enlargement degradation leads to storage of glycogen and hypoglycaemia
 Type B – neuronal damage is absent  Deficiency of G6Pase (von Gierke Dx, or type 1 glycogenosis)
Niemann-Pick Disease  Caused by a defect in cholesterol transport and – prime example of hepatic-hypoglycemic form of glycogen
Type C resultant accumulation of cholesterol and storage disease
gangliosides in the nervous system  Glycogen is stored in many organs, but hepatic
 Affected children most commonly exhibit ataxia, enlargement and hypoglycaemia dominate in clinical
dysarthria, and psychomotor regression picture
Gaucher Disease  Results from lack of the lysosomal enzyme Myopathic forms  In skeletal muscles, glycogen is used as a source of energy
glucocerebrosidase and accumulation of  Glycolysis  ATP  lactate
glucocerebroside in mononuclear phagocytic cells  If enzyme that fuel glycolytic pathway are deficient, glycogen
 Type I – most common; affected phagocytes become storage occurs in the muscles and asso. w/ muscular
enlarged (Gaucher cells) and accumulate in liver, weakness due to impaired energy production
spleen, and BM  cause hepatosplenomegaly and  Ex: deficiencies of muscle phosphorylase (McArdle Dx, or
bone erosion type V glycogenoses), muscle phosphofructokinase (type VII
 Types II and III – cxd by variable neuronal GSD)
involvement  Typically, individuals w/ the myopathic forms present w/
Mucopolysaccharidoses  Result in accumulation of mucopolysaccharides in muscle cramps after exercise and lactate levels in the blood
liver, spleen, heart, blood vessels, brain, cornea, fail to rise after exercise due to a block in glycolysis
joints. GSD asso. w/ 1)  Asso. w/ glycogen storage in many organs; death in early life
deficiency of α-  Acid maltase – lysosomal enzyme; deficiency leads to
 Affected px have coarse facial features glucosidase (acid
 Hurler syndrome – corneal cloudness, coronary lysosomal storage of glycogen (type II glycogenosis, or Pompe
maltase) and lack of
arterial and valvular deposits, and death in childhood branching enzyme
disease) in all organs
 Hunter syndrome – asso. w/ milder clinical course  Cardiomegaly is the most prominent feature

Glycogen Storage Disease (Glycogenoses)


abmrmtmd
KEY CONCEPTS: Glycogen Storage Disease CHROMOSOMAL DISORDERS
 Inherited deficiency of enzymes involved in glycogen metabolism can result in storage
of normal or abnormal forms of glycogen, predominantly in liver or muscles, but also Normal Karyotype
in other tissues as well
Hepatic form  Von Gierke Disease  22 homologous pairs of autosomes; 2 sex chromosomes (XX, XY)
 Liver cells store glycogen because a lack of hepatic G6Pase  Karyotyping – study of chromosomes; basic tool of cytogeneticist
Myopathic  McArdle disease – muscle phosphorylase lacks gives rise to o Arrest dividing cells in metaphase w/ mitotic spindle inhibitors (Colcemid)
form storage in skeletal muscles and cramps after exercise
and stain the chromosomes
Pompe  There is lack of lysosomal acid maltas; all organs are affected
Disease  Heart involvement is predominant o In metaphase spread, individual chromosomes take the form of two
chromatids connected at the centromere
o Stained using Giemsa stain  G banding
o Resolution obtained by banding can be markedly improved by obtaining the
Disorders Associated with Defects in Proteins That Regulate Cell Growth cells in prophase
o 1500 bands per karyotype can be recognized
 Normal growth and differentiation of cells are regulated by two classes of genes:
o Proto-oncogenes Commonly Used Cytogenetic Terminology
o Tumor suppressor genes
 Mutations in both classes of genes are important in the pathogenesis of tumors  Karyotypes are described using a shorthand system of notations in the ff order:
 Cancer-causing mutations affecting somatic cells are not passed in the germ line o Total number of chromosomes
 Familial cancers are inherited in an autosomal dominant fashion o Sex chromosome complement
o Description o abnormalities in ascending numerical order
o E.g., male w/ trisomy 21 47, XY, +21
COMPLEX MULTIGENIC DISORDERS  p – short arm of a chromosome; p for petit
 q – long arm
 Caused by interactions between variant forms of genes and environmental  Each arm of the chromosome is divided into two or more regions bordered by
factors prominent bands
 Gene that has at least two alleles, each occurs at a frequency of at least 1% in the o Regions are numbered from centromere outward
population is polymorphic o Each region is further subdivided into bands and sub-bands and are ordered
o Each variant allele is referred to as a polymorphism numerically
 Complex genetic disorders occur when many polymorphism (each w/ modest effect o E.g., Xp21.2 – chromosomal segment located on the short-arm of X
and low penetrance) are co-inherited chromosome, in region 2, band 1, and sub-band 2
 Complex disorders result from the collective inheritance of many polymorphism
 Some polymorphisms are common to multiple diseases of the same type, while others Structural Abnormalities of Chromosomes
are disease specific
 Environmental influences significantly modify the phenotypic expression of  The aberrations underlying cytogenetic disorders may take the form of an abnormal
complex traits number of chromosomes or alterations in the structure of one or more chromosomes
o Ex: Type 2 DM (multifactorial disorder)  first manifest after weight gain  46,XX or 46,XY – normal chromosome complement
o Obesity (other environmental influences) unmasks the diabetic genetic trait  Euploid – any exact multiple of the haploid number of chromosomes (23)
 Nutritional influences may cause monozygous twins to achieve different heights  Aneuploidy - if an error occurs in mitosis or meiosis and a cell acquires a
 Difficult to distinguish between Mendelian and multifactorial disease chromosome complement that is not an exact multiple of 23; usual cause:
o Nondisjunction
o Anaphase lag
 Nondisjunction – (gamtogenesis) gametes formed have either an extra (n +1) or
less (n – 1) chromosome;
o Fertilization of such gametes results in two types of zygotes:
 Trisomic (2n + 1)
 Monosomic (2n – 1)

abmrmtmd
 Anaphase lag – one homologous chromosome in meiosis or one chromatid in mitosis Isochromosome  Results when one arm of a chromosome is lost and the
lags behind and is left out of the cell nucleus formation remaining arm is duplicated
o Result is one normal cell and one cell w/ monosomy  Results in a chromosome consisting of two short arms only or
 Monosomy involving an autosome generally causes loss of too much genetic two long arms
 Has morphologically identical genetic information in both arms
information to permit live birth or even embryogenesis
 I(X)(q10) – most common isochrome present in live births;
o But several autosomal trisomies do permit survival involves long arm of X
 Mitotic errors in early development give rise to two or more populations of cells w/ Translocation  Segment of one chromosome is transferred to another
different chromosomal complement  referred to as mosaicism if in the same  Balanced reciprocal translocation – there are single breaks
individual in each of two chromosomes , with exchange of material
 Mosaicism – result from mitotic errors during the cleavage of the fertilized ovum or o 46,XX,t(2;5)(q31;p14) – translocation between long
in somatic cells arm of chromosome 2 and short arm if chromosome 5
o There is no loss of genetic material  phenotypically
 Mosaicism in sex chromosomes is common  lead to one of the daughter cells normal
receiving three sex chromosomes , whereas the other receives only one o Carrier is at increased risk for producing abnormal
o E.g., 45,X/47,XXX mosaic  mosaic variant of Turner syndrome (45,X) gametes
 Autosomal mosaicism is less common  leads to nonviable mosaic due to  Robertsonian translocation or centric fusion –
translocation between two acrocentric chromosomes
autosomal monosomy
o The breaks occur close to the centromeres
o Nonviable cell population is lost during embryogenesis, yielding a viable o Transfer of segments leads to one very large
mosaic (e.g., 46,XY/47,XY, +21) chromosome and one extremely small one
 Second category of chromosomal aberration is asso. w/ structural changes o Usually the small product is lost  still compatible
 FISH (fluorescence in situ hybridization) – gain higher resolution; can detect with normal phenotype
changes as small as kilobases o Significance lies in the production of abnormal
 Structural changes usually result from chromosome breakage followed by loss or progeny
rearrangement of material

Cytogenetic Disorders Involving Autosomes


Deletion  Refers to loss of a portion of a chromosome
 Most are interstitial (rarely terminal)
 Interstitial deletions – occur when there are two breaks w/in Trisomy 21 (Down Syndrome)
a chromosome arm, followed by loss of the chromosomal  Most common of the chromosomal disorders and is a major cause of mental
material between the breaks and fusion of the broken ends retardation; chromosome count is 47
 E.g., 46,XY,del(16)(p11.2p13.1) – breakpoints in the short arm  FISH reveals extra copy of extra chromosome 21
of chromosome 16 at 16p11.2 and 16p13.1 w/ loss of material  Extra chromosomal number is present as a translocation
between breaks  Most common cause is meiotic nondisjuntion
 Terminal deletion – result from a single break in a  Maternal age has a strong influence on the incidence of trisomy 21
chromosome arm; produces fragment w/ no centromere, w/c is  In 4% of cases of Down syndrome, the extra chromosomal material
then lost at the next cell division, and a chromosome bearing a derives from the presence of a robertsonian translocation of the long arm of
deletion; the end of a chromosome is protected by acquiring chromosome 21 to another eccentric chromosome (e.g., 22 or 14)
telomeric sequences o Affects chromosome pairing during meiosis resulting to aneuploid
Ring  Special form of deletion; produced when a break occurs at both  Approximately 1% of Down syndrome patients are mosaics, having a mixture of cells
chromosome ends of a chromosome w/ fusion of the damaged ends w/ 46 or 47 chromosomes
 If genetic material is lost, phenotypic abnormalities result o Results from mitotic nondisjunction of chromosome 21
 E.g., 46,XY,r(14) o Symptoms are variable and milder
 Ring chromosomes do not behave normally in meiosis or mitosis  In cases of translocation or mosaic Down syndrome, maternal age is of no importance
and usually result in serious consequences  The diagnostic clinical features of this condition are evident even at birth:
Inversion  Refers to a rearrangement that involves two breaks w/in a single o Flat facial profile
chromosome w/ reincorporation of the inverted, intervening o Oblique palpebral fissures
segment o Epicanthic folds
 Paracentric – inversion involving only one arm of the  Down syndrome is a leading cause of mental retardation  80% has 25-50 IQ
chromosome o Children have a gentle, shy manner and often content w/ life
 Pericentric – if the breaks are on opposite sides of the
centromere
 Inversion are often compatible w/ normal development
abmrmtmd
 40% of the patients have congenital heart disease  Deletion of genes at chromosomal locus 22q11.2 gives rise to malformations affecting
o Affects endocardial cushion, including ostium primum, atrial septal defects, the face, heart, thymus, and parathyroids. The resulting disorders are recognized as:
atrioventricular valve malforamtions, and ventricular septal defects o DiGeorge syndrome – thymic hypoplasia w/ diminished T-cell immunity
o Cardiac problems are responsible for the majority of deaths in infancy and and parathyroid hypoplasia w/ hypocalcemia
early childhood o Velocardiofacial syndrome – congenital heart disease involving outflow
o Also include atresias of esophagus and small bowel tracts, facial dysmorphism, and developmental delay
 Children w/ trisomy 21 have 10-fold to 20-fold increased risk of developing acute
leukemia
o ALL and AML (most commonly acute megakaryoblastic leukemia) occur Cytogenetic Disorders Involving Sex Chromosomes
 All patients w/ older than age 40 develop neuropathologic changes cxc of Alzheimer
disease  Genetic diseases asso. w/ changes involving the sex chromosomes are far more
 Patients have abnormal immune response that predisposes them to serious infections
common than those related to autosomal aberrations.
(lungs, thyroid autoimmunity)
 Imbalances of sex chromosomes are much better tolerated than are similar
imbalances of autosomes
Other Trisomies  Two factors that are peculiar to the sex chromosomes:
 Other trisomies: involves chromosomes 8, 9, 13, 18, 22 1. Lyonization or inactivation of all but one X chromosome
 Trisomy 18 (Edwards syndrome) 2. The modest amount of genetic material carried by the Y chromosome
 Trisomy 13 (Patau syndrome)  Lyon hypothesis (X-inactivation)
 Most cases result from meiotic nondisjunction and carry a complete extra copy of 1. Only one of the X chromosomes is genetically active
chromosomes
2. The other X of either maternal or paternal origin undergoes heteropyknosis
 Association w/ increased maternal age is also noted
 Malformations are much more severe and wide ranging and is rendered inactive
3. Inactivation of either the maternal or paternal X occurs at random among all
the cells of the blastocyst on or about day 5.5 of embryonic life
Chromosome 22q11.2 Deletion Syndrome 4. Inactivation of the same X chromosome persists in all the cells derived from
 Chromosome 22q11.2 deletion syndrome encompasses a spectrum of disorders that each precursor cell
result from a small deletion of band q11.2 on the long arm of chromosome 22  Normal females  one inactivated maternal X and inactivated paternal X
 Clinical features include: o Barr body or X chromatin
o Congenital heart defects  XIST – unique gene involve in X inactivation; product is a long noncoding RNA
o Abnormalities of the palate
o XIST allele is switched off in the active X
o Facial dysmorphism
 Although it was initially thought that all the genes on the inactive X are “switched off”,
o Developmental delay
o Variable degrees of T-cell immunodeficiency more recent studies have revealed that many genes escape X inactivation
o Hypocalcemia  Patients w/ the monosomy of the X chromosome (Turner syndrome: 45,X) have
 Represent DiGeorge syndrome and velocardiofacial syndrome severe somatic and gonadal deficiency
 DiGeorge syndrome – px have thymic hypoplasia w/ T-cell immunodeficiency,  Regardless of the # of X chromosomes, presence of a single Y determines
parathyroid hypolasia that lead to hypocalcemia, cardiac malforamtions and mild facial male sex
abnormalities
o Gene that dictates testicular development (SRY gene) is located on its distal
 Velocardiofacial syndrome – clinical features include: facial dysmorphism
(prominent nose, retrognathia), cleft palate, cardiovascular anomalies, learning short arm
disabilities, immunodeficiency (less frequent)  Features common to all sex chromosomes disorders:
 Individuals w/ the 22q11.2 deletion syndrome are at high risk for psychotic o Sex chromosome disorders cause subtle, chronic problems relating to sexual
illnesses, such as: schizophrenia (25% of adults) and bipolar disorders development and fertility
 Diagnosis: detection of deletion by FISH o Often difficult to diagnose at birth, and many are first recognized at the
time of puberty
o The greater the number of X chromosomes, in both male and female, the
KEY CONCEPTS: Cytogenetic Disorders Involving Autosomes greater the likelihood of mental retardation
 Down Syndrome – asso. w/ an extra copy of genes on chromosome 21, most
 Two most important disorders arising in aberrations of sex chromosomes:
commonly due to trisomy 21 and less frequently from translocation of extra
chromosomal material from chromosome 21 to other chromosome or from mosaicism o Klinefelter Syndrome
 Patients w/ down syndrome have severe mental retardation, flat facial profile, o Turner Syndrome
epicanthic folds, cardiac malformations, higher risk of leukemia and infections, and
premature development of Alzheimer disease
abmrmtmd
Klinefelter Syndrome  Complete or partial monosomy of the X chromosome; characterized primarily
 Male hypogonadism that occurs when there are two or more X by hypogonadism in phenotypic females
chromosomes and one or more y chromosomes  Most common sex chromosome abnormality in females (1 in 2500 cases)
 One of the most frequent form of genetic disease involving sex chromosome; most  Three karyotypic abnormalities are seen:
common cause of hypogonadism in male; 1 in 660 cases 1. ~57% are missing an entire X chromosome, resulting in a 45,X karyotype
 Rarely diagnosed before puberty because testicular abnormality does not develop o 43%  14% have structural abnormalities in X chromosome; 29% are
before early puberty mosaics
 Distinctive body habitus; increase in length between the soles and the pubic bone  2. Common feature of the structural abnormalities is to produce partial
body appears elongated monosomy of the X chromosome
 Eunuchoid body habitus w/ abnormal long legs, small small atrophic testes asso .w/ o Inorder of frequency, the structural abnormalities of the X chromosome
small penis; lack secondary male characteristics as deep voice, beard and male include:
distribution of pubic hair. 1) Isochrome of the long arm, 46,X,i(X)(q10) resulting in the loss of
 Gynecomastia may be present; IQ is lower than normal; mental retardation is the short arm
uncommon; increased incidence in DM2 (insulin resistance) 2) Deletion of portions of both long and short arms resulting in the
 Mitral valve prolapsed (50% in cases); increased incidence of osteoporosis and formation of a ring chromosome, 46,X,r(X)
fractures due to sex hormonal imbalance 3) Deletion of portions of the short or long arm, 46,del(Xq) or
 Consistent finding  hypogonadism 46X,del(Xp)
 ↑ FSH; ↓ testosterone; ↑ estradiol 3. Mosaic patients have a 45,X cell population along with one or more
 Ratio of estrogen and testosterone  determines degree of feminization karyotypically normal or abnormal cell types
 Klinefelter syndrome is an important genetic cause of reduced spermatogenesis and o Examples of mosaic Turner females
male infertility 1) 45,X/46,XX
o Testicular tubules are atrophied and replaced by pink, hyaline, collegnous 2) 45,X/46,XY
ghost 3) 45,X/47,XXX
 Px have a higher risk for breast cancer, extragonadal germ cell tumors, and 4) 45,X/46,X,i(X)(q10)
autoimmune diseases such as SLE o In patients w/ high 45,X proportion, phenotypic changes are more severe
 Classic pattern is 47,XXXY karyotype (90% of cases)  results from than w/ mosaicism w/c only present w/ primary amenorrhea
nondisjunction during meiotic division in germ cells  5-10% have Y chromosome either as a complete (45,X/46,XY) or as fragments of Y
 15% of cases  46,XY/47,XXY; 47,XXY/48,XXXY chromosomes  higher risk for development of gonadal tumor (gonadoblastoma)
 In cases of normal females, all but one X chromosome is inactivated in px w/  Most severely affected px present during infancy w/ edema of the dorsum of the hand
klinefelter syndrome; px have hypogonadism and same features and foot due to lymph stasis swelling of the nape of the neck
o Reason lies in genes on the X chromosome that escape lyonization and in  Latter is related to markedly distended lymphatic channels, producing hygroma
the pattern of X inactivation  As infants develop, the swellings subside leaving bilateral neck webbing and
persistent looseness of skin on the back of the neck
 Uneven dosage compensation during X-inactivation  Congenital heart disease is common (25-50%)  most important cause of
o 15% of X-linked gene escape inactivation increased mortality in children w/ Turner syndrome
o Extra dose of genes  overexpression leads to hypogonadism  Failure to develop normal secondary characteristics  infantile genitalia, inadequate
 Second mechanism involves the gene encoding the androgen receptor, breast development, little pubic hair; Mental status is normal but there is subtle
through w/c testosterone mediates its effect defects in non-verbal, visual-spatial information processing
o Androgen receptor gene maps to X chromosome and contains highly  Important in establishing the diagnosis is the adult is the shortness of stature
polymorphic CAG (trinucleotide) repeats  dictates functional response to (<150 cm ) and amaenorrhea
particular dose of androgen  Turner syndrome is the single most important cause of primary amenorrhea
o Shorter CAG repeats are more sensitive than w/ long CAG repeats  50% has autoantibodies against thyroid gland  hypothyroidism
o In Klinefelter syndrome, X chromosome w/ shortest CAG repeat is  Fetal ovaries develop normally in early embryogenesis, but absence of the second X
inactivated chromosome leads to accelerated loss of oocytes (completed by age 2 years) 
o XXY males w/ low testosterone levels  expression of long CAG repeats “menopause occurs before menarche” and ovaries are reduced to atrophic
exacerbates the hypogonadism  small penis hehehe strands, devoid of ova and follicles (streak ovaries)
 Short stature homeobox (SHOX) gene at Xp22.33 – gene involve in Turner
phenotype; critical regulator of growth
o Haploinsufficiency gives rise to short stature
o Expressed in fetal life in growth plates (radius, ulna, tibia,, fibula and 1 st and
2nd pharyngeal arches)
o Loss f SHOX  short stature
o Excess copy  tall stature

Turner Syndrome
abmrmtmd
Hermaphroditism and Pseudohermaphroditism  Polyglutamine diseases – cxd by progressive neurodegeneration in midlife
 Sexual ambiguity – disagreement among criterias for determining sex
o Proteins are misfolded and tend to aggregate; aggregates may suppress
 Genetic sex – determined by the presence or absence of a Y chromosome
transcription of other genes, cause mitochondrial dysfunction, or trigger the
 Gonadal sex – based on the histologic cxc of gonads
 Ductal sex – depends on the presence of derivatives of mullerian or wolffian ducts unfolded protein stress response and apoptosis
 Phenotypic or genital sex – based on the appearance of the external genitalia o Morphologic hallmark  accumulation of aggregated mutant proteins
 Hermaphrodite – presence of both ovarian and testicular tissue in large intranuclear inclusions
 Pseudohermaphrodite – disagreement between the phenotypic and gonadal sex
(e.g., female pseudohermaphrodite has ovaries but male external genotalia) Fragile X Syndrome and Fragile X Tremor/Ataxia
 Fragile X syndrome is the prototype of diseases in w/c the mutation is cxd by a long
repeating sequence of three nucleotides
KEY CONCEPTS: Cytogenetic Disorders Involving Sex Chromosomes  Most affected sequences share the nucleotides guanine (G) and cytosine (C)
 Females: one X chromosome is inactivated during development (Lyon hypothesis)  Fragile X Syndrome is the second most common genetic cause of mental
 Klinefelter syndrome – two or more X chromosomes w/ one Y chromosome as a result retardation after Down syndrome
of nondisjunction of sex chromosomes; testicular atrophy, sterility, reduced body o Caused by a trinucleotide mutation in the familial mental retardation-1
hair, gynecomastia, and eunochoid body habitus; most common cause of male sterility (FMR-1) gene; 1 in 1550 males; 1 in 8000 females
 Turner syndrome – there is partial or comlete monosomy of genes on the short arm of o Cytogenetic alteration was seen as a discontinuity of staining or as a
the X chromosome, most commonly due to the absence of one X chromosome (45,X) constriction in the long arm of X chromosome in a folate deficient-medium
and less commonly from mosaicism, or from deletions involving the short arm of the X o Chromosome appears broke ”fragile site”
chromosome. Short stature, webbing of the neck, cabitus valgus, cardiovascular o Affected males are mentally retarded; 20-60 IQ; long face w. Large
malformations, amenorrhea, lack of secondary sex characteristics, and fibrotic ovaries mandible, large everted ears, large testicles (macro-orchidism; most
are typical clinical features. distinctive feature; 90%)
o Hyperextensible joints, high arched palate, mitral valve prolapsed
 Some patterns of transmission not typically asso. w/ other X-linked recessive
SINGLE-GENE DISORDERS WITH NONCLASSIC INHERITANCE disorders:
 Carrier males  normal transmitting males (20%)
 Affected females  30-50% are affected
 Transmission of certain single-gene disorders does not follow classic Mendelian
 Risk of phenotypic effects
principles  Anticipation  features worsen w/ each successive generation; becomes
 Can be classified into four categories increasingly deleterious as it is transmitted from a man to his grandsons
o Diseases caused by trinucleotide-repeat mutations and great-grandsons
o Disorders caused by mutations in mitochondrial genes  Xq27.3  where FMR1 gene lies; presence of clinical symptoms and fragile site is
o Disorides asso. w/ genomic imprinting related to the amplification of the CGG repeats
o Disorders asso. w/ gonadal mosaicism o Normal transmitting males and carrier females carry 55-200 repeats
o Expansion of size is called premutaions
o Px have full mutations (extremely large expansion; 200-400 repeats) 
Diseases Caused by Trinucleotide-Repeat Mutations
arise from further amplification of the CGG repeats in permutations
 During the process of oogenesis (but not spermatogenesis) permutations
 Expansion of trinucleotide repeats is an important genetic cause of human can be converted to mutations by triple-repeat amplification
disease, particularly neurodegenerative disorders (table 5-8) o Mental retardation is much higher in grandsons  full mutation
 General principles that apply to these diseases:  Molecular basis of mental retardation and somatic changed is related tp loss of
function of FMRP
 Causative mutations are asso. w/ the expansion of a stretch of trinucleotides that o When FMR1 exceed 230 (normal is 55 CGG repeats), DNA of entire 5’ region
becomes abnormally methylated
usually share the nucleotides G and C
o Methylation results in transcriptional suppression of FMR1
o DNA is unstable and impairs gene function o Absence of FMRP  cause of phenotypic changes
 Expansion depends strongly on the sex of the transmitting parent  FMRP – most abundant in brain and testis (two organs most affected by the
o Fragile X syndrome – expansion occurs during oogenesis disease; functions:
o Huntington disease – expansion occurs during spermatogenesis  FMRP selectively binds mRNAs asso. w/ polysomes and regulates their
 Three key mechanisms w/c unstable repeats cause disease intracellular transport to dendrites (binds 4% of mammalian brain mRNAs)
1) Loss of function of the affected gene  FMRP is a translation regulator
 PCR-based detection of the repeats  method of choice for diagnosis
2) A toxic gain of function by alterations of protein structure (Huntington dx)
3) A toxic gain of function mediated by mRNA (fragile X syndrome)
Fragile X Tremor/Ataxia
abmrmtmd
 Although initially assumed to be innocuous, CGG permutations in the FMR1 gene can  Diseases asso. w/ mitochondrial inheritance are rare and affect the neuromuscular
cause a disease that is phenotypically different from fragile X syndrome through a system
distinct mechanism involving a toxic “gain-of-function” o E.g., Leber hereditary optic neuropathy – manifest a progressive
 Premature ovarian failure; progressive neurodegenerative syndrome bilateral loss of central vision (starts at age 15-35) leading to blindness
 Cxd by intention tremors and cerebellar ataxia and may progress to
parkinsonism
Genomic Imprinting
 FMR1 gene instead of being methylated and silenced continues to be transcribed
 CGG-containing FMR1 mRNAs so formed are “toxic”  accumulate in the nucleus and
form intracellular inclusions  Imprinting – epigenetic process that results in between paternal and maternal allele
 Aggregated mRNA recruits RNA-binding proteins o Inactivates either maternal or paternal allele
o Occurs in the ovum or sperm before fertilization and then transmitted to all
somatic cells through mitosis
KEY CONCEPTS: Fragile X Syndrome o Asso.w/ differential patterns of DNA methylation at CG nucleotides, H4
 Pathologic amplification of trinucleotide repeats causes loss-of-function (fragile X deacytlation and methylation
syndrome) or gain-of-function mutations (Huntington disease). Most such mutations o Regulated by cis-acting elements called imprinting control regions
produce neurodegenerative disorders
o Prader-Willi syndrome and Angelman syndrome
 Fragile X syndrome results from loss of FMR1 gene function and is cxd by mental
retardation, macro-orchidism and abnormal facial features  Maternal imprinting – transcriptional silencing of the maternal allele
 In the normal population, there are about 29-55 CGG repeats in the FMR1 gene. The  Paternal imprinting – paternal allele is inactivated
genomes of carrier males and females contain permutations w/ 55-200 CGG repeats
that can expand to 4000 repeats (full mutations) during oogenesis. When full Prader-Willi Syndrome and Angelman Syndrome
mutations are transmitted to progeny, fragile X syndrome occurs  Prader-Willi syndrome – cxd by mental retardation, short stature, hypotonia, profound
 Fragile X tremor/ataxia due to expression of a FMR1 gene bearing a permutation hyperphagia, obesity, small hands and feet, and hypogonadism
develops in some males and females. The accumulation of corresponding mRNA in the o 65-70% of cases – interstitial deletion of band q12 in the long arm of
nucleus binds and sequesteres certain proteins that are essential for normal neuronal chromosome 15, del(15)(q11.2q13)  causes 5-Mb deletion
functions o It is striking that in all cases the deletion affects the paternally derived
chromosome 15
o In contrast w/ Prader-Willi syndrome, px w/ the phenotypically distinct
Mutations in Mitochondrial Genes – Leber Hereditary Optic Neuropathy Angelman syndrome are born w/ a deletion of the same chromosomal
region derived from their mothers
 A feature unique to mtDNA is maternal inheritance  Angelman syndrome – mentally retarded, but in addition they present w/ ataxic
gait, seizures, and inappropriate laughter  “happy puppets”
 mtDNA complement of the zygote is derived entirely from the ovum
 The comparison of two syndromes demonstrates the “parent-of-origin” effects on
 Mothers transmit mtDNA to all their offspring (male and female) gene function
o However, daughters (not sons) transmit the DNA to their progeny  Mechanisms involved in the genomic printing of these disease:
 Features apply to mitochondrial inheritance:
o Deletion
 Human mtDNA contains 37 genes, 22 are transcribed to tRNA and 2 into rRNA  If maternal 15q12 is imprinted, and paternal is functional 
Prader-Willi syndrome
o Remaining 13 genes encode subunits of the respiratory chain enzymes
 If paternal is inactivated; maternal is functional  Angelman
o Since mtDNA is involved in oxidative phosphorylation, mutations affecting
syndrome
them has deleterious effects on organs most dependent on oxidative o Uniparent disomy – inheritance of both chromosome of a pair from one
phosphorylation (CNS, skeletal muscle, cardiac muscle, liver, kidneys) parent  non-functional set of genes from nonimprinted chromosomes
 Each mitochrondrion contains thousands of copies of mtDNA and deleterious  Prader-Willi syndrome  if nondeletion, they have two maternal
mutations of mtDNA affect some but not all of these copies copies of chromosome 15
o Heteroplasmy – tissues and individuals may harbour both wild and mutant  Angelman syndrome – result from uniparental disomy of paternal
chromosome 15
mtDNA
 Second most common mechanism (20-25% of cases)
o Threshold effect – minimum number of mutant mtDNA must be present o Defective imprinting – 1-4% of cases; no functional alleles
before oxidative dysfunction gives rise to disease  Prader-Willi syndrome – paternal chromosome carries the
 During cell division, mitochondrion and their contained DNA are randomly distributed maternal imprint
to the daughter cells  Angelman Syndrome – maternal chromosome carries paternal
imprint

abmrmtmd
Genetic basis of these two imprinting disorders: MOLECULAR GENETIC DIAGNOSIS
Angelman  Affected gene is ubiquitin ligase involved in catalyzing
Syndrome the transfer of activated ubiquitin to target protein  Karyotyping for recognition of cytogenetic disorders (e.g., Down syndrome)
substrates  DNA-based assays (e.g., Southernblotting for Huntington disease)
 UBE3A – gene; maps w/in 15q12 region; imprinted on the  Regardless of the technique used, human genetic markers can be either
paternal chromosome, and is expressed from maternal
o Constitutional (present in each and every cell of the affected persons;
allele primarily in regions of the brain
 Imprinting is tissue specific  UBE3A is expressed from e.g., CFTR)
both alleles in most tissues o Somatic (restricted to specific tissue types or lesions;e.g., KRAS)
Prader-Willi  No single gene has been implicated
syndrome  Series of genes located in the 15q11.2-q13 interval Diagnostic Methods and Indications for Testing
(imprinted on maternal chromosome and expressed from
paternal chromosome) are involved Laboratory Considerations
 SNORP family of genes – encode small nucleolar RNAs
involved in modification of rRNAs  Pathologist focus on the sensitivity, specificity, accuracy, and reproducibility of
 Loss of SNORP function  contribute to Prader-Willi syn.
different methods; and practical factors: cost, labor, reliability, and TAT
 Molecular diagnosis of these syndrome is based on assessment of methylation of
 Critical to first understand the spectrum of genetic anomalies responsible for the dx
marker genes and FISH

Indications for Analysis of Inherited Genetic Alterations


KEY CONCEPTS: Genomic Imprinting
 May be required at any age; mostly performed during prenatal or postnatal/childhood
 Imprinting involves transcriptional silencing of the paternal or maternal copies of
certain genes during gametogenesis. For such genes, only one functional copy exists periods
in the individual. Loss of the functional (not imprinted) allele by deletion gives rise to  Prenatal testing should be offered for all fetuses at risk for a cytogenetic
diseases. abnormality: indications:
 Prader-Willi Syndrome – deletion of band q12 on long arm of paternal o Advanced maternal age
chromosome 15. Genes in this region of maternal chromosome are imprinted so there o A parent known to carry a balanced chromosomal rearrangement (
is complete loss of their functions. Patients have mental retardation, short stature,
increased risk of abnormal chromosome segregation and aneuploidy)
hypotonia, hyperphagia, small hands and feet, hypogonadism
 Angelman syndrome – there is deletion of the same region from the maternal o Fetal abnormalities during ultrasound
chromosome. Since genes on the corresponding region of paternal chromosome 15 o Routine maternal blood screening, indicating risk of Down syndrome or
are imprinted, these patients have mental retardation, ataxia, seizures, and other trisomy
inappropriate laughter  Prenatal testing may also be considered for children at known risk for genetic
disorders (cystic fibrosis, spinal muscular atrophy) using targeted analysis based on
familial mutations or family history
Gonadal Mosaicism  Usually performed on cells obtained by amniocentesis, chronic villus biopsy,
umbilical cord blood
 In some autosomal dominant disorders, exemplified by oteogenesis imperfect,
 Parents known to be at risk for having a child w/ a genetic disorder can choose to
phenotypically normal parents have more than one affected child  cause by
have genetic testing performed on embryos created in vitro prior to uterine
gonadal mosaicism implantation, eliminating the chance of generational transmission of a familial disease
 Results from a mutation that occurs postzygotically during early (embryonic)
 Ff birth, testing is done as soon as the possibility of genetic disease arises. Most
development commonly performed on peripheral blood DNA. In newborns or children,
 If mutation affects only cells to form gonads, the gametes carry the mutation but
indications may be as follows:
somatic cells are normal o Multiple congenital anomalies
 A phenotypically normal parent who has gonadal mosaicism can transmit the disease-
o Suspicion of a metabolic syndrome
causing mutation to the offspring through their mutated gametes o Unexplained mental retardation and/or developmental delay
o More than one child of such parent would be affected
o Suspected aneuploidy (feat. Of Down syndrome) or other syndromic
 Occurrence depends on the proportion of the germ cells carrying the mutation chromosomal abnormality (deletions, inversions)
o Suspected monogenic disease (previously described or unknown)

abmrmtmd
 In older patients, testing is focused in diseases that manifest at later stages of life; and measured by photo detector
indications include:  Most often used when testing for particular sequence and is
o Inherited cancer syndromes more sensitive than Sanger sequencing
o Atypical mild monogenic disease  Allows detection for as little as 5% mutated alleles in a
background of normal alleles
o Neurodegenerative disorders
 Application: used to analyze DNA obtained from cancer
biopsies, in w/c tumor cells are often “contaminated” w/ large
Indications for Analysis of Acquired Genetic Alterations numbers of stromal cells
Single-base  Useful approach for identifying mutations at a specific
 Identify nucleic acid sequences or aberrations specific for acquired diseases; primer nucleotide position (e.g., mutation in codon 600 of the BRAF
indication: extension gene)
o Diagnosis and management of cancer:  An interrogating sequencing primer is added to the PCR product,
 Detection of tumor specific mutations and cytogenetic alterations w/c binds just one base upstream of the target
 Differently colored terminator fluorescent nucleotides are also
that are hallmark of tumors (BCR-ABL)
added and a single base polymerase extension is performed 
 Determination of clonality as an indicator of a neoplastic condition
fluorescence are then detected
 Identification of specific genetic alterations that can direct  Very sensitive – 1-2% mutated alleles
therapeutic choices (HER2)  Disadvantage: produce only one base pair of sequence data
 Determination of treatment efficacy Restriction  Involves digestion of DNA w/ endonucleases known as
 Detection of drug-resistant secondary mutations in malignancies fragment length restriction enzymes that recognize and cut DNA at specific
o Diagnosis and management of infectious disease analysis sequences
 If specific mutation affects a restriction site, then the amplified
 Detection of microorganism-specific genetic material for definitive
PCR product may be digested, and the normal and mutant PCR
diagnosis
products will yield fragments of different sizes
 Identification of specific genetic alterations in the genomes of  Identified as different bands following electrophoresis
microbes that are asso. w/ drug resistance  Less comprehensive than direct sequencing but remain useful
 Determination of treatment efficacy for molecular diagnosis when actual mutation occurs at an
invariant nucleotide position
PCR and Detection of DNA Sequence Alterations Amplicon length  Mutations that affect the length of DNA (deletion,
analysis expansion) can be detected by PCR
 PCR analysis, w/c involves the synthesis of relatively short DNA fragments from a DNA  E.g., Fragile X syndrome asso. w/ alterations in trinucleotide
repeats  two primers flank the region w/ trinucleotide repeats
template, has been a mainstay of molecular diagnostics for the last few decades
at the 5’ end of FMR1 gene are used to amplify the intervening
 Uses heat-stable DNA polymerases and thermal cycling  target DNA (<1000 sequence  large differences in the number of repeats  size
base pairs) lying between primer sites is exponentially amplified of PCR products from normal and permutation individuals are
different
Sanger  The amplified DNA is mixed w/ a DNA polymerase, a DNA  Easily distinguished by electrophoresis
Sequencing primer, nucleotides, and a four dead-end (di-deoxy  This technique will fail if a trinucleotide repeat expansion is
terminator) nucleotides (A,T,G,C) labelled w/ fluorescent tags so large (use Southern blot instead)
 Reaction produces series of DNA molecules labelled w/ a tag Real-time PCR  Use fluorophore indicators that can detect and quantify the
corresponding to the base at w/c the reaction stopped due to presence of a particular nucleic acid sequences in “real time”
addition of terminator nucleotides  during the exponential phase of NA amplification rather than
 After size separation by capillary electrophoresis, the post-PCR
sequence can be read and compared w/ normal sequence to  Most often used to monitor the frequency of cancer cells
detect mutations bearing cxc genetic lesions in the blood or tissues, or the
 Application: analysis of large genes or multiple genes infectious load of certain viruses
 “gold standard” for sequence determination  Can also be used to detect somatic point mutations in
Pyrosequencing  There is a release of pyrophosphate when a nucleotide is oncogenes such as KRAS and BRAF  an approach that has
incorporated into a growing DNA strand advantage of avoiding the need for post-PCR analysis
 Also performed on PCR products using a single sequencing
primer and involves cycling individual nucleotides (A,C,T,G)
 If one or more nucleotides are incorporated into the growing
strand of DNA, pyrophosphate is released and participate in
secondary reaction involving luciferase that produce light

abmrmtmd
Molecular Analysis of Genomic Alterations
Array-Based Comparative Genomic Hybridization (Array CGH)
 Hybridization-based techniques  The test DNA and a reference (normal) DNA are labelled w/ two different
fluorescent dyes  both are cohybridized to an array spotted w/ DNA probes
Fluorescence in Situ Hybridization (FISH) (cover all 22 autosomes and sex chromosomes)
 FISH uses DNA probes that recognize sequences specific to particular  At each chromosomal probe location, binding of the labelled DNA from two
chromosomal regions samples is compared
 Human DNA inserts in clones in order of 100,000-200,000 base pairs  these DNA  If two samples are equal  diploid; all spots will fluoresce yellow (green and
clones are labelled w/ fluorescent dyes and applied to metaphase chromosome red dyes)
spreads to interphase nuclei that are pretreated to “melt” the genomic DNA  If test sample shows even a focal deletion or duplication, the probe spots
 Ability of FISH to circumvent the need for dividing cells is invaluable when a rapid corresponding to it will show skewing toward red or green allowing highly accurate
diagnosis is warranted determinations of copy number variant across the genome
 FISH can be performed on: prenatal samples, peripheral blood cells, touch
preparations from cancer biopsies, and even fixed archival tissue sections SNP Genotyping Arrays
 Fish is used to detect numeric abnormalities of chromosomes (aneuploidy),  Designed to identify single nucleotide polymorphism (SNP) sites genome-
subtle microdeletions or complex translocations that are not demonstrable by routine wide
karyotyping , and gene amplification  SNP’s serve as both a physical landmark within the genome and as a genetic
 Chromosome painting is an extension of FISH marker whose transmission can be followed from parent to child
 Number chromosomes that can be detected is limited by the availability of fluorescent  Methods involving SNPs can be used to make copy number variations (CNV)
dyes  overcome by introduction of spectral karyotyping or “multicolour FISH”   This technology is the mainstay of genome wide association studies (GWAS)
powerful that it might called “spectacular karyotyping”  In the clinical laboratory, SNP arrays are routinely used to uncover copy number
abnormalities in pediatric patients when the karyotype is normal but a structural
chromosomal abnormality is still suspected
 Common indications include;
Multiplex Ligation-Dependent Probe Amplification (MLPA)
o Congental abnormalities
 MLPA blends DNA hybridization, DNA ligation, and PCR amplification to detect
o Dysmorphic features
deletions and duplications of any size, including anomalies that are too large to
o Developmental delay
be detected by PCR and too small to be identified by FISH
o autism
 Each MLPA reaction uses a pair of probes that can hybridize side-by-side to one strand
of the target DNA  probes are joined via ligase reaction
 Probes also contain additional sequences at their ends that can be used as a primer
sequences in a PCR Polymorphic Markers and Molecular Diagnosis
 Ligated probes create a template that can be amplified by PCR  quantification yields
accurate info regarding the amount of starting material  If exact nature of genetic aberration is unknown or if testing for primary disease is
 MLPA can be performed on very small amounts of genomic DNA challenging or unfeasible  diagnostic lab uses linkage
 The two types of genetic polymorphism most useful for linkage analysis are SNPs and
repeat-length polymorphism knwon as minisatellite and microsatellite
Southern Blotting  Human DNA contains short repetitive sequences of DNA giving rise to “repeat-
 Changes on the structure of specific loci can be detected by Southern blotting  length polymorphism”  subdivide into microsatellite and minisatellite repeats
involves hybridization of radio labeled sequence-specific probes to genomic DNA that
 Microsatellite - <1 kilobase; cxd by a repeat size of 2-6 base pairs
has been first digested w/ a restriction enzyme and separated by gel
electrophoresis o Scaterred through the human genome; high level of polymorphism
 The probe detect one germ line band in normal individuals and a different size band o Ideal for differentiating between two individuals and follow transmission of
depending on the genetic anomaly the marker from parent to child
 Rarely used; useful in the detection of certain large-trinucleotide-expansion o Also use in determining paternity and criminal investigation
diseases, including fragile X syndrome  Minisatelllite – larger (1-3 kilobases); repeat motif is usually 15-70 base pairs
 Assays to detect genetic polymorphisms are also important in many other areas of
medicine, including in the determination of relatedness and identity in transplantation,
Cytogenomic Array Technology cancer genetics, paternity testing, and forensic medicine

 Genomic abnormalities can also be detected w/o prior knowledge by using microarray
technology to perform a global genomic survey

abmrmtmd
Polymorphism and Genome-Wide Analysis Next-Generation Sequencing

 In GWAS (genome wide association studies), large cohort of patients w/ and  Next-generation sequencing (NGS) is a term used to describe several newer DNA
w/o a disease (rather than families) are examined across the entire genome for sequencing technologies that are capable of producing large amounts of sequence
common genetic variants with the disease data in a massively parallel manner
 GWAS also led to the identification of genetic loci that modulate common quantitative  NGS allows to perform impossible analyses at extremely low relative cost
traits in humans (height, body mass, hair and eye color, and bone density)  The fundamental factor that sets NGC apart from traditional Sanger sequencing is its
input sample requirements
Epigenetic Alterations o NGS has no requirements: any DNA from almost any source can be used
o NGS instruments are well suited to heterogenous DNA samples due to the
 Epigenetics – defined as the study of heritable chemical modification of DNA or application of these common basic processes:
chromatin that does not alter the DNA sequence itself
o Ex: methylation of DNA, methylation and acetylation of histones Spatial  At the beginning, individual input DNA molecules are physically
 Gene expression correlates w/ the level of methylation of DNA, usually of cytosines separation isolated from each other in space
specifically in CG dinucleotide-rich promoter regions known as CpG islands  Specifics of this process are platform-dependent
 Increased methylation of these loci is associated with decreased gene expression and Local  After separation, the individual DNA molecules are amplified in
is accompanied by concomitant specific patterns of histone methylation and amplification situ using a limited number of PCR cycles
 Amplification is necessary so that sufficient signal can be
acetylation
generated to ensure detection and accuracy
 Treatment of genomic DNA w/ sodium bisulfite  a chemical that converts Parallel  The amplified DNA molecules are sequenced by addition of
unmethylated cytosine to uracil, w/c acts like thymine in downstream reactions sequencing polymerase  yield a “read” that corresponds to its sequence
o Methylated cytosines are protected from modifications and remain  Sequence reads from NGS instruments are generally short
unchanged (<500bp)
o After treatment, it is then straightforward to discriminate the unmethylated
(modified) DNA from the methylated (unmodified) DNA on the basis of
sequence analysis Bioinformatics

RNA Analysis  NGS instruments can generate a staggering amount of sequence data  enough to
produce a high-quality sequence spanning an entire human genome
 Mature mRNA – contains the coding sequences of all expressed genes  Basic steps necessary to process this type of data in a generic human DNA context:
 RNA can substitute for DNA in a wide range of diagnostic applications o Alignement – process by w/c the sequencing reads from a sample are
o However, DNA-based diagnosis is preferred because DNA is much more mapped to a genome where they can be viewed and interpreted
stable o Variant calling – process involves “walking” across the reference genome
o Detection and quantification of RNA viruses  HIV and Hep C and evaluating all of the sequence data that mapped to each position and
o mRNA expression profiling is emerging as an important tool for molecular compare it w/ reference genome
stratification o Variant annotation and interpretation
 Cancer cells bearing particular chromosomal translocations are detected w/ greater
sensitivity by analyzing mRNA Clinical Applications of NGS DNA Sequencing
o Principal reason for this is that most translocations occur in the scattered
location w/in particular introns, s/c can be very large, beyond the capacity  Basic approaches:
of conventional PCR amplification
o Since introns are removed by splicing during the formation of mRNA, PCR  Targeted sequencing
analysis is possible if RNA is first converted to cDNA by reverse o Just one gene or a panel of genes minimizes sequencing costs as well as
transcriptase the time and expense required for manual interpretation and clinical
 Real-time PCR performed on cDNA is the method of choice for monitoring residual reporting
o Sample preparation  subselecting relevant clones from a whole genome
disease in patients w/ CML and certain other hematologic malignancies
library via custom complementary probes or by alternate preparations from
genomic DNA such as multiplex PCR

abmrmtmd
 Whole exome sequencing (WES)
o Type of targeted sequencing
o Uses hundreds of thousands of custom probes to pull out 1.5% of the
genome consist of protein-encoding exons
o WES enables a broad survey for protein coding mutations (responsible for
80% of Mendelian disease) at reduced cost
 Whole genome sequencing (WGS)
o Most comprehensive type of DNA analysis
o Current cost and informatics challenges still preclude its routine use in
clinical practice
o Indications: mostly limited to cases where exome sequencing has failed to
provide an answer but the clinical suspicion of genetic disease remains high
o Cancer applications: WGS is the only NGS application that can detect
novel structural rearrangements (insertions, deletions, translocations) that
may be clinically relevant
o WGS is generally performed to lower sequencing depth than either targeted
panels or exomes, and may suffer from a lack of statistical power to detect
low percentage mutations in heterogenous tumor samples

 The choice of approach is mainly a function of sequencing cost and interpretive


workload

Future Applications

 Microbiome analysis and blood screening for early markers of diseases, including
cancer

abmrmtmd

You might also like