Molecular Basis of Disease Flashcards

1
Q

Monogenic vs Complex Diseases

A

Monogenic

  • stronger genetic component
  • causative gene directly leading to disorder
  • recognisable inheritance patterns, hereditary
  • gene mutations
  • rare

Complex

  • more involvement of environment, multifactorial/polygenic
  • susceptibility gene increases risk but doesn’t directly cause disorder
  • no clear inheritance pattern, familial
  • gene variants
  • common in population e.g. cancer, CVS disease
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Recap basic genetics: chromosomes in our body, DNA bases, alleles, protein synthesis, genotype vs phenotype, epigenetics

A

22 autosomes and 1 sex chromosome
4 DNA bases: adenine, thymine, cytosine, guanine

Alleles = different DNA sequences at a gene or locus (homozygous vs heterozygous)

Protein synthesis: replication of DNA –> transcription into mRNA –> translation into proteins
- every amino acid coded by 3 consecutive bases

Genotype = sequence
Phenotype = anatomical or physiological manifestation

Epigenetics = modifications in gene expression without changing DNA sequence e.g. DNA methylation at CpG islands

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

Sequence variation: Mutation vs Polymorphism

A

Mutation = disease-causing change

Polymorphism = changes found in >1% population which are not necessarily disease-causing

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

Mutations - point mutations (definitions) and trinucleotide repeat expansion

A

Point mutations:

  1. Substitution
    - silent = no change in amino acid sequence of the protein
    - missense = change in codon for ONE amino acid, SIZE of mRNA unchanged but COMPOSITION and possibly FUNCTION of protein does change
    - non-sense = change to a STOP CODON e.g. UAA, UAG, UGA with premature termination and truncated protein
  2. Insertion and deletion: frameshift mutations

Trinucleotide repeat expansion:

  • error in DNA replication
  • dynamic mutations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Structural (chromosomal) variations (3)

A

Copy number variation
- change in copy number of one particular chromosome region (>50bp)

Chromosomal translocation, deletion and inversions

  • translocation = exchange of genetic material between two non-homologous chromosomes
  • -> gene may be DISRUPTED OR NEW FUSION GENE

Aneuploidies
- abnormal copy numbers of one or more chromosome

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

Mendelian inheritance - characteristics of specific inheritance patterns

A

Strong correlation between disease genotype and disease

Autosomal:
= BOTH GENDERS EQUAL
- dominant = each generation affected; 50% risk of passing mutated allele from parent to child
- recessive = not each generation affected; no known FHx of the disease; consanguinity (25% affected if both parents are carriers)

X-linked:
= NO FATHER-SON transmission (affected male passes allele to all daughters)
- mostly recessive = incidence much higher in male (hemizygous for X-linked genes)
- dominant = both male and females affected

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

Non-Mendelian inheritance: Mitochondrial inheritance - mechanism, pattern, heteroplasmy, bottleneck, how to confirm mutation, is prenatal diagnosis useful?

A

Mitochondrial genome (or nuclear genome) mutation

  • either maternally inherited or de novo
  • pattern of inheritance similar to XLR but only females pass and females are also affected

Heteroplasmy

  • co-existence of normal and mutant mtDNA (some functional proteins still present)
  • mutation load above a certain threshold = disease
  • tissue specific mutation load = variable manifestation in different tissues (liver, brain, heart, kidney generally most affected)
  • blood precursor cells has selection against some mutations (die if they carry mutation above load) –> less mutations circulating in blood

Bottleneck effect

  • only a small subset of mtDNA molecules is passed from mother to germ cells
  • completely random (can be high/low proportion of mutant mtDNA)
  • difficult to predict manifestations in child

How to confirm presence of mutation:
- skeletal muscles, urine samples, tissue with high energy demand

Prenatal diagnosis not helpful:

  • load of mutation in foetal tissues not correlated with load in other tissues of newborn
  • mutant load difficult to predict due to heterplasmy and bottleneck
  • mutant load may change with time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Non-Mendelian inheritance: Genomic imprinting

A

Expression of gene only from one allele –> activity depending on whether the variant was inherited from mother or father (parent-of-origin specific)

  • due to transcriptional silencing/epigenetic changes during gametogenesis
  • imprinting pattern different in sperm/oocyte

E.g. Prader-Willi syndrome: deletion in paternal homologue
Angelman syndrome: deletion in maternal homologue

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

Non-Mendelian inheritance: Dynamic mutations - definition/characteristics, examples

A

PROGRESSIVE EXPANSION of “triplet repeats”

ANTCIPATION = symptoms in later generations often more severe and appear at progressively younger ages

Examples: Fragile X, Huntington, Myotonic dystrophy, Friedrich ataxia, Kennedy disease

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

Penetrance and Expressivity

A

Penetrance = % individuals with given genotype who exhibit the associated phenotype

Expressivity = number of symptoms in the presentation of a disease in individuals who have associated phenotypes

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

Genetic testing: somatic vs germline, de novo vs inherited

A

Detect mutations
- germline - in gametes –> all cells of offspring –> half of affected individual’s gametes carry mutation
vs
- somatic - occur after fertilisation –> affect certain cell types only –> no gametes carry mutation

  • de novo vs inherited
  • -> all somatic mutations are de novo
  • -> germline mutations can be de novo or inherited
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Example: MEN2 - approach to inherited cancer syndrome, implication of genetic testing

A

Approach to inherited cancer syndromes
- clinical criteria –> molecular analysis –> risk assessment models

MEN2 (2a, 2b and FMTC)

  • AD missense “gain of function” mutation of RET oncogene
  • high risk of developing MTC –> MEN2 accounts for 25% of MTC

Implications of genetic testing for MEN2 in MTC

  • screening of other associated conditions/ tumours
  • family members at risk of inheriting MEN2
  • ?assess need for prophylactic surgery
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Example: MEN2 - inherited cancer syndrome, mutation, implication for genetic testing

A

Approach to inherited cancer syndromes

  • inherited mutation causing increased risk of developing certain tumours at early age
  • most are AD, germline mutations
  • clinical criteria –> molecular analysis –> risk assessment models (surveillance and prevention)

MEN2 (2a, 2b and FMTC)

  • AD missense “gain of function” mutation of RET oncogene
  • high risk of developing MTC –> MEN2 accounts for 25% of MTC

Implications for genetic testing for MEN2 in MTC

  • screening of other associated conditions/ tumours
  • family members at risk of inheriting MEN2
  • ?assess need for prophylactic surgery
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Example: Wilson’s disease - mutation, stepwise testing approach, implications for genetic testing

A

AR disorder of copper metabolism
ATP7B mutations with systemic copper accumulation and multi-organ damage
- impaired copper incorporation into apoceruloplasmin
- permanent damage can be prevented if early chelation therapy given

Low serum ceruloplasmin levels has low PPV –> need molecular diagnosis

STEPWISE TESTING APPROACH

  • tiered screening approach based on previous studies identifying most common locations of mutant alleles
  • start with most common then broaden if negative results

Implications for genetic testing in family members

  • overlapping biochemical results between heterozygotes (CARRIERS) and pre-symptomatic (AFFECTED but no organ damage yet) individuals
  • early identification and treatment prevents permanent damage
  • can use direct mutation analysis since know the mutation points from proband
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Example: Fragile X syndrome - mutation, inheritance characteristic

A

XR disorder
Most common single gene cause of autism spectrum disorders
Transcriptional silencing of FMR1 gene –> CGG REPEATS EXPANSION (trinucleotide repeats) at the 5’ untranslated region

Normal = 5-44 repeats
Pre-mutation (carrier) = 50-200 [normal transmitting male and normal female]
Full mutation = >200 [diseased male, milder phenotype in female]

Trinucleotide repeat in the premutation range is unstable and may EXPAND TO FULL MUTATION AS X CHROMOSOME IS PASSED ONTO NEXT GENERATION

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

Genetic basis of complex diseases - characteristics

A

Cumulative and interactive
More than one gene locus involved, susceptibility gene e.g. DM
Each gene contributes modestly to disease
Disease associated with certain POLYMORPHIC VARIANTS of the genes (>1% of population)
Environmental risk involved

e.g. cancer, dementia, CVS disease

17
Q

DNA polymorphism - definition, types

A

Regions where more than one allele can occupy the position = polymorphic sites

  • reasonably common to see variation within population (GREATER THAN 1%)
  • known sites canbe used as marker of genomic location

Types:
- short tandem repeats (2-13 bp)
- **single nucleotide polymorphism (SNP) –> 80% of polymorphisms in human genome
==> analysed in genome wide associated study (GWAS)
- copy number variation (>50bp)

18
Q

Cancer as a complex genetic trait - main type of mutation, importance of FHx

A

SOMATIC MUTATION as the basis for development and progression of all types of cancer

  • only affects specific types of cells
  • cancer associated genes (oncogenes/TSG) with numerous mutations
  • can have different genetic components of same cancer e.g. EGFR mutation in lung cancer –> potential therapeutic target

Germline mutations can predispose to the development of somatic mutations for cancer – FHx is important!

Germline mutation + somatic mutation –> multiple tumours, early onset, bilateral

Normal gene + 2x somatic mutations –> single tumours, unilateral, later onset

19
Q

Genetic testing purposes - reasons for diagnostic/carrier testing, prenatal, preimplantation genetic diagnosis, predictive/presymptomatic, pharmacogenetic testing, genetic screening

A

Diagnostic and Carrier testing

  • diagnostic = symptomatic individual to confirm/exclude
  • carrier = mutation detection that generally has limited or no consequence to health

Prenatal diagnosis

  • during pregnancy where there is increased risk for a certain condition in the foetus
    e. g. chorionic villus sampling/amniocentesis, cicrulating foetal DNA in maternal plasma

Preimplantation genetic diagnosis
- testing the presence of mutation/chromosomeal change in EMBRYO in a family with previously o=known risk for inherited disorders to select unaffected embryos for implantation in IVF

Predictive or presymptomatic testing

  • no features of disorder themselves but have family member with genetic disorder
  • -> determine risk, predict onset/severity, prophylactic Tx, disease surveillance, planning for Mx, clinical trials

Pharmacogenetic testing

  • test for genetic susceptibility for adverse drug reactions or efficacy of drug treatment (not a mutation)
    e. g. HLA-B*1502 for carbamazepine SJS

Genetic screening

  • target is not high-risk but systematicaly offered to general population or specific group
    e. g. newborn screening for IEM
20
Q

Reporting genetic testing results (classification of variants and conclusions)

A
Pathogenic (confirmatory) --> treat
Likely pathogenic (consistent) --> treat
Variant of uncertain significance
Likely benign
Benign

(gene, genomic coordinates, reference transcript, nucleotide change, protein change, zygosity)

21
Q

Family tree and pedigrees

A

KNOW HOW TO INTERPRET/DRAW

Affected = have clinical presentations/symptomatic

Carriers are NOT AFFECTED!
- read question carefully and only use the info given - don’t assume carrier if no info given

22
Q

Problems of genetic testing

A

Sensitivity

  • meaning of “negative” result -
  • > false negatives (low sensitivity) OR
  • > if direct mutation approach, can’t rule out presence of other mutations causing disease
  • variant of uncertain significance (VUS) –> don’t know whether it explains the genetic disease (need further workup to see if other studies reported mutation or use functional studies to assess for alterations in protein function)
  • cost –> direct=cheaper; screening=expensive
23
Q

Example: benefits of molecular testing in Menkes Disease

A

XLR disease of copper transport involving ATP7A gene

  • confirm diagnosis
  • disease course, prognosis and treatment
  • information for prenatal diagnosis or PGD
  • family screening (for other female carriers)
  • clinical trials
24
Q

Genetic counselling - elements of pre and post-test counselling

A

Pre-test counselling

  • conditions being tested, pattern of inheritance, symptoms
  • reproductive options
  • nature of test, indications, limitations, possible results
  • alternative options and their risks, benefits, limitations
  • cost
  • possible emotional impacts, the need to inform relatives about results
  • follow-up procedures

Post-test counselling

  • results
  • cope with emotional impact
  • recurrence risk
  • testing of additional relatives
  • course of action, support groups, followup contacts
25
Q

Genetic testing techniques

A

Karyotyping
- whole chromosomes (number, size, shape)

Chromosomal microarray analysis (CMA)

  • microdeletions and microduplications
  • 5-10kb changes can be detected
  • for patients with unexplained developmental delay/intellectual disability/autism spectrum

Sequencing

  • exact nucleotide components in genome
  • Sanger sequencing (single genes)
  • NGS (massively parallel; several genes to whole exome/genome)

Special groups for trinucleotide repeat disorders, mitochondrial genome

26
Q

Example: Huntington disease - dynamic mutation, reasons for declining genetic testing

A

AD neurodegenerative disorder
Trinucleotide repeat disease
- number of CAG repeats in HTT gene
- anticipation

Declining testing:

  • no effective cure
  • concerns about discrimination
  • cost of testing
  • inability to “undo” knowledge once status known
27
Q

Gene transcription - splicing

A

Splicing of introns to form mRNA consisting only of exons

- splice at splice donor sites (2 nucleotides after 1st exon) and splice acceptor sites (2 nucleotides before 2nd exon)

28
Q

DNA Sequencing - materials, procedure

A

For novel mutations - screening

Materials

  • DNA template (denatured into single strand)
  • Taq polymerase (elongate complementary DNA)
  • Primer
  • deoxynucleotides (dNTP)
  • dideoxy (ddNTP) – terminates reaction
  1. DNA denatured by heat
  2. Primer annealed to template strand to allow addition of dNTP
  3. Add: template strand, DNA polymerase and nucleotides into reaction (only add one type of ddNTP to each mixture)
  4. Many fragments generated in each mixture
  5. Polyacrylamide gel electrophoresis of 4 reagents
    - -> smaller fragments move faster,
    - -> each band reflective of ddNTP termination point so read off from smallest fragment upwards
29
Q

Restrict fragment length polymorphism - procedure, interpretation of results

A

For known mutations - targeted approach

  1. Digestion by restriction enzymes (cut DNA at specific recognition sites)
  2. Analyse size of resulting fragments by gel electrophoresis
    - probe specific for RFLP DNA

Possible results:

  • normal - one cutting point –> 2 fragments
  • homozygous mutation - extra cutting point –> 3 fragments
  • heterozygous mutation –> 4 fragments
30
Q

Example: MELAS

A

Mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes

80% pathogenic variant of mitochondrially encoded tRNA leucine 1

31
Q

Example: SMA

A

AR
SMN1 gene mutation
SMN2 produces 15% functional protein –> needed to compensate for SMN1 for survival (normally insignificant in healthy but in SMA it is useful for severity assessment)

Homozygous deletion of SMN1 = SMA diagnosis
>3 copies of SMN2 = milder phenotype