Module 2.5: Other Flashcards

1
Q

Features of h pylori

A

• It is a spiral shaped/curved organism that increases acid production

• It is a model of disease as it produces a range of diseases:
o Acute gastritis – not very symptomatic
o GU and chronic gastritis: majority of patients (90% of cases of chronic antral gastritis)
o DU: almost all patients (DU: duodenal ulcer)
o Also associated with gastric cancer (adenocarcinoma) and lymphoma

Hence if H.Pylori is treated, a lot of diseases can be treated also.

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

Epidemiology of H Pylori

A
•	H. Pylori infection is associated with:
o	Crowding 
o	Poor sanitation
o	Limited education 
o	Poverty 
o	Immigrant

• Worldwide: 10-90% colonisation rates
o Given that <1% of H. Pylori infected individuals will develop gastric cancer and lymphoma it may not seem like a big problem. However, H. Pylori infection is very prevalent and so this can affect millions of people hence helicobacter is a big cause of gastric cancers (+ lymphoma)

  • In high prevalence countries it is acquired in childhood
  • Various routes of transmission e.g. endoscopes (interestingly ,H. Pylori is highest among gastroenterologists)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Pathogenesis of h pylori

A

• Mainly an antral gastritis associated with increased acid production despite decreased gastrin
o Remember this is an alternate source of acid production that is not affected by gastrin but will contribute to gastrin reduction
• May progress to a multifocal atrophic gastritis with decreased acid
• Adapted to live in the gastric mucin – rarely invades tissue

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

Factors associated with h pylori Virulence

A

• Flagella
• Urease – produces ammonia from urea raising the pH
o This is its protective mechanism  so that it can live in the stomach lining
o This is the basis of screening test

• Adhesins – mediate adherence to surface foveolar cells

• Toxins – cytotoxin-associated gene A (CagA) which is associated with ulcer development and cancer
o Virulent damaging toxin  interacts with p53
o Toxin injected through Type IV secretion system

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

Pathology caused by h pylori

A

• Organism can usually be seen on biopsy in the mucus of the surface and neck regions
• Best diagnosed on an antral biopsy
o On biopsy, H. Pylori gastritis can be acute (neutrophil) or chronic (lymphocyte)

• Inflammation
o Neutrophils in the lamina propria, which may extend into the pits
o Plasma cells in the superficial lamina propria, which produces rugal fold thickening
o Lymphoid follicles often present = induced MALT
 This is a marker of chronic inflammation as it is not present in normal mucosa

• Mucosa:
o Intestinal metaplasia with absorptive cells and goblet cells and Paneth cells
o Atrophy
o Dysplasia  can lead to flat dysplasia pathway

Acute Inflammation – crypts packed with neutrophil polymorphs

Chronic inflammation – crypts contain increased numbers of plasma cells. Lymphoid follicles seen with germinal centres (blue circle).

Intestinal metaplasia – hallmark is goblet cells as can be seen on slide.

Dysplasia
• Nuclei are big (high nuclear:cytoplasmic ratio), hyperchromatin

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

H. Pylori Infection confers an increased risk of:

A
•	Adenocarcinoma
•	Genotype of hlo associated cancers 
•	Lymphoma
•	Reversibility of lymphoma 
o	i.e. in a patient with low grade lymphoma, if you treat the H. Pylori infection, this can be reversed  not reversible in high grade
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Extra-intestinal Manifestations of Helicobacter Pylori

A
•	Cardiovascular diseases 
o	Epidemiological evidence is most convincing for this 
•	Ischaemic heart disease 
•	Noncardioembolic ischaemic stroke 
•	Pre-eclampsia 
•	Raynaud phenomenon 
•	Migraine 
•	Diabetes mellitus 
•	The metabolic syndrome 
•	Neurodegenerative diseases 
•	Multiple sclerosis 
•	Neuromyelitis optica
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

CVS effects of H. Pylori

A
  • Especially caused by CagA+ve strains
  • These induce anti-CagA antibodies from the host, which promote atherosclerotic plaque formation
•	Biopsy
o	Histology
o	Rapid urease test
o	Culture
o	PCR
•	Serology
•	Faecal bacterial detection
•	Urea breath test
o	Detect ammonia production

Treatment
• Antibiotics and proton pump inhibitors (PPIs)  triple therapy
• May relapse

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

Define cancer

A

Definition: a disease caused by an uncontrolled division of abnormal cells in a part of the body.

•	Common Carcinomas
o	Lung 
o	Breast (women)
o	Colon 
o	Bladder 
o	Prostate (men)

• Leukaemias
o Bloodstream

• Lymphomas
o Lymph nodes

• Common Sarcomas
o Fat
o Muscle
o Bone

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

two types of programmed cell death

A

o Apoptosis: an energy-dependent process that suppresses inflammation and allows for maintenance of plasma membrane integrity and ordered DNA fragmentation. Process: cells first shrink, nuclei condense and form ‘apoptotic bodies’

o Necrosis: an energy-independent process that induces inflammation whereby plasma membrane integrity is lost and there is random DNA fragmentation. Process: cell swells, ruptures and during its demise there is release of cellular contents

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

Define Transcription Factors

A

proteins involved in the regulation of gene expression that bind to the regulatory regions upstream of genes and either facilitate or inhibit transcription

• Transcription factors are composed of two essential functional regions:

o DNA-binding domain  consists of amino acids that recognise specific DNA bases near the start of transcription
o Transactivator domain  interacts with components of the transcriptional apparatus (RNA Polymerase) and with other regulatory proteins, thereby affecting the efficiency of DNA binding

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is NFkB

A

key nuclear transcription factor

  • Consists of a protein family of inducible dimeric TFs which recognise the kB site
  • This factor is thought to be involved in 80% of cancer. This is not mutated but is the main transcription factor to be dysregulated.

o In normal cells, NFkB is bound and deactivated by inhibitor-kappa B proteins (IkB)
o During stress and exposure to microbial productions/cytokines/mitogens, IkB protein is phosphorylated by inhibitor kappa-B kinase (IKK)
o This inactivation releases NFkB  can enter the nucleus and affect transcription

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Describe the Classical NFkB Pathway

A

• Expression of NFkB’s gene products mediate:
o Immunity
o Inflammation
o Development
o Cell survival  problematic in malignancy as NFB effectively inhibits apoptosis

• Summary:
o IKK  kinases activated by microbial product/mitogens/cytokines/stress
o IKK phosphorylates IkB, thereby releasing NFkB
o NFkB is now activated and so, migrates to the nucleus and binds to specific sites on DNA that are upstream of certain genes  genes involved mediate above

N.B: An alternative NFkB pathway:  activated in response to specific cytokines. This is involved in stimulating lymphoid organogenesis, as well as activating other chemokines and cytokines.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Protein Kinase (Definition)

A

A kinase is an enzyme that modifies other proteins by adding phosphate groups (phosphorylation).

Phosphorylation usually results in a functional change of the target protein (substrate).

The activity of a kinase involves removing a phosphate group from ATP and covalently attaching it to an amino acid that has a free hydroxyl group.

Most kinases act on both serine and threonine (others act on tyrosine).

By contrast, phosphatase is an enzyme that removes phosphate group from its substrate.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Pro-survival activity of NFkB in Disease

A
  • Cellular responses to triggering of TNF-Rs, TRAIL-Rs and Fas
  • B-Lymphopoeisis
  • Bone morphogenesis
  • B- and T-cell Costimulation (CD40, CD28, etc)
  • Liver development
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

NFkB in Disease

A

• Its amplification allows development of cancer
o Not considered an oncogene but is regulated by oncoproteins e.g. Ras which is thought to phosphorylate in cancer
o This causes inhibition of programmed cell death and differentiation, enhancement of proliferation, invasion and metastasis
o Hence NFkB is associated with survival of late stage tumours e.g. breast, Hodgkin’s, colorectal cancer (and resistance to anti-cancer therapy)
• Implicated in cancer chemo- and radio-resistance
• Chronic inflammatory disease (IBD, RA)
• Metabolic and vascular disease (atherosclerosis)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Molecular Mechanism by which NFkB controls PCD

A

• TNF-alpha binding to its receptor – TNF-R1 – stimulates the death through the TRADD/TRAF pathway, causing the production of MAP3K, MAP2K and JNKs (mitogen activated protein kinases)
• JNKs interact with caspases (cysteine-aspartic proteases), leading to activation of cytochrome C and activation of all caspases  DNA fragmentation and cell death
• However, a pro-survival pathway can also activated by NFkB (if dysregulated)
o MEKK3 leads to IKK activation  inhibition of IkB  activation of NFkB
o NFkB transcribes pro-survival genes that inhibit MAPKs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Distinct Mechanisms of PCD Inhibition

A

genes targeted by NFkB at different stages of the TRADD/TRAF pathway
• JNK is the end-product of this pathway and is itself, phosphorylated by MKK7
• Gadd45beta/Myd118 is a specific inhibitor of JNK cascade via inhibition of MKK7 (Papa S, et al, Nat. Cell Biol, 2004)
o Gadd45-beta is a 21kDa peptide predominantly found in the nucleus. It is a member of the Gadd45 family of inducible factors (others: Gadd45-alpha and –gamma)
o It inhibits MKK7 via its ATP-binding site (lysine 149)  no ATP binding site mans no phosphorylation of JNK  no caspases  no cell death
o Peptides can therefore be devised e.g. peptide 1 which are synthetic competitive inhibitors of Gadd45-beta. This would restore MKK7 function and allow programmed cell death to return. This is an example of a peptide currently undergoing clinical trials for multiple myeloma.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

NFkB blockers as a future therapy

A

o Inhibition of NFkB by either glucocorticoid or proteasome inhibitors is beneficial in certain malignancies such as Hodgkin’s lymphoma or multiple myeloma.
o However, current compounds can only achieve partial inhibition of NFkB and have considerable side effects, thereby limiting their use in humans.

• Gadd45beta and FHC blockers may present new targets
o Blocking molecules directly involved in the pro-survival pathway downstream of NFkB should display beter results with fewer side-effects as other functions of NFkB would not be affected

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

define mutation

A

• Definition: a mutation is a change in the normal base pair sequence of DNA

• Mutations can occur in either coding or noncoding regions
o These may be silent  have no effect on resulting protein. This is especially true if they occur in noncoding regions of the DNA. Even small base-pair changes in the coding region may be silent because of the redundancy of the code.
o Point mutations  a single base change that results in the change of an amino acid in the structure
o Deletions, insertions, translocations  involve larger sections of DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Summarise Cancer Development

A

multistage process that involves inherited and somatic mutations of cellular genes

o Somatic mutations
 Occur frequently (1 mutation/cell division)
 Normally inconsequential (not passed on to next generation)
 Dangerous if occurs on specific genes
 E.g. BRCA1

o Germline mutations
 Occurs rarely
 Present in all somatic cells of affected individual
 Cancer develops only in specific tissue/organ
 Increased risk of cancer passed on to future generation

• Cancers are clonal
o Many mutations in several different distinct genes are required for normal cells to become cancerous
o This progressive accumulation of these multiple hits explains the age-incidence of cancer
o All the cells in a tumour originate from a single ancestral cell
o But, not all cells in a tumour have the same genotype because cancer cells are genetically unstable
o Variation gives rise to selection
o Clonal selection of variant progeny with the most robust growth properties play major contributing roles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Describe Cancer-associated Mutations

A
  • Cancer-associated mutations, whether somatic or germline, point mutations or large deletions, alter key proteins and their functions in the human bio-system. A wide variety of mutations arw involved including those in non-coding regions
  • Mutation can result in under- or over-expression of proteins needed for normalcy
•	Mutations can affect:
o	Growth factors, cytokines
o	Receptors genes
o	Cell signalling genes
o	Transcription factor genes
o	Cell cycle control genes
o	Cell death and survival genes
o	DNA repair genes
o	Cellular differentiation genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Describe Normal Cell Growth

A

• Most cells remain in interphase for at least 90% of cell cycle
o First phase of interphase: G1  rapid growth and metabolic activity (including synthesis of RNA and proteins)
o Followed by S phase (DNA synthesis)
o Followed by G2  cell grows and prepares for division
• Cell division happens in M phase (mitosis)
• Cells that do not divide for long periods do not replicate their DNA – G0 phase

  • In normal cells TS genes act as braking signals during G1 to stop or slow the cell cycle before S phase
  • DNA repair genes are active throughout cycle, particularly during G2 after DNA replication
  • Oncogenes allow progression from G1 to S phase
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Definition of a Protooncogone:

A

a normal gene that can become an oncogene, either after mutation or increased expression. Proto-oncogenes code for proteins that help regulate cell growth and differentiation. Proto-oncogenes are often involved in signal transduction and execution of mitogenic signals. Upon activation, a proto-oncogene becomes a tumour-inducing agent, which is named oncogene.

o Growth factors
o Growth factor receptors
o Signal relaying molecules
o Nuclear transcription factors (proteins that bind to genes to start transcription)

  • When proto-oncogene is mutated or overregulated, it is called an oncogene
  • Only one mutation in a single allele enough to trigger oncogenic role
  • The chance will increase as a person ages
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Definition of a TS Gene:

A

gene that protects a cell from uncontrolled cell growth. When this gene is mutated to cause a loss or reduction in its function, the cell can progress to cancer, usually in combination with other genetic changes.

both copies of tumour suppressor genes must be lost or mutated for cancer

o Person who carries a germline mutation in a tumour suppressor gene has only one functional copy of the gene in all cells
o For this person, it is more likely for the second hit to occur for cancer progression

•	Act as negative regulators of cancer growth:
o	inhibit proliferation
o	induce apoptosis,
o	inhibit angiogenesis
o	induce cell adhesion
•	Act to maintain chromosome integrity
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Examples of Oncogenes

A

RAS (e.g. colon cancer)
• Protein subfamily of small GTPases that are involved in cellular signal transduction
• Activation causes cell growth, differentiation and survival

C-MYC (e.g. overexpressed in colon cancer, amplified in lung, rearranged in lymphoma)
• Encodes transcription factor that regulate expression of 15% of all genes through binding on Enhancer Box sequences (E-boxes) + recruiting histone acetyltransferases (HATs)
• So in addition to its role as a classical transcription factor, c-myc also functions to regulate global chromatin structure by regulating histone acetylation both in gene-rich regions and other sites far from known genes

Others
•	CDK4 (familial melanoma)
•	MET (hereditary papillary renal cancer)
•	BCR/ABL (chronic myelogenic leukaemia)
•	BCL2 (follicular lymphoma)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Examples to TSG

A

o Rb
o P53
o APC

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Describe retinablastoma

A

• Paediatric intraocular tumour accounting for 5% of childhood blindness
• Occurs as an inherited disease with autosomal dominant transmission + 90% penetrance
• Sporadic cases also known, but differ from typical hereditary disease
• May cause thickening of optic nerve due to extension of tumour + displace normal retina
• Several curative treatments
o Surgery and radiotherapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Describe the two hit model

A

• The first insult is inherited in the DNA + any second insult
• In non-inherited retinoblastoma, two hits had to take place, explaining the age difference
• The development of malignancy depended both on the activation of proto-oncogene + deactivation of tumour suppressors
o First hit in an oncogene would not necessarily lead to cancer as normally functioning tumour suppressors would counterbalance
o Conversely, a damaged tumour suppressor (Rb1) would not lead to cancer unless there is a growth impetus from activated oncogene

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Function of Rb Protein

A

• 105 kDa peptide known as pRb
• Member of the pocket protein family, consisting of three proteins
o pRB – Retinoblastoma protein
o p107 – Retinoblastoma like protein 1
o p130 – Retinoblastoma like protein 2
• Play crucial roles in cell cycle through interaction with members of E2F transcription factors family
• The three all contain pockets which interact with oncogenic DNA viral proteins such as HPV16 E7 and other proteins including E2F transcription factor
• P107 and p130 are not classical tumour suppressors – their functions may be redundant
• Action
o pRb is normally bound to the E2F-1 transcription factor – prevents it from interacting with the cell’s transcription machinery
o In the absence of pRB, E2F-1 (along with its binding partner DP-1) mediates trans-activation of E2F-1 target genes that facilitate the G1/S transition
o G1-Cdk activity (cyclin D-Cdk4) initiates Rb phosphorylation, which inactivates Rb and causes it to detach from E2F-1
o The inactivation frees E2F to activate the transcription of S-phase genes, including cyclin E and cyclin A

  • So in summary, Rb activity is regulated by phosphorylation
  • When Rb is phosphorylated, transcription factors like E2F are free, which would cause transcription of target genes and entry in to S phase
31
Q

Describe the role of P53 in Cancer

A

• The p53 gene, like the Rb gene, is a tumor suppressor gene.
• If a person inherits only one functional copy of the p53 gene from their parents, they are predisposed to cancer and usually develop several independent tumors in a variety of tissues in early adulthood.
o This condition is rare, and is known as Li-Fraumeni syndrome.
• Mutations in p53 are found in approximately 50% of cancers, and so contribute to the complex network of molecular events leading to tumour formation

• p53 act not only as a transcription factor, but also as regulatory protein.

• Human p53 consists of 393 amino acids long and, like other transcription factors, has three domains:
o An N-terminal transcription-activation domain (TAD), which activates transcription factors
o A central DNA-binding core domain (DBD). Contains zinc molecules and arginine amino acid residues.
o A C-terminal homo-oligomerisation domain (OD). Tetramerization greatly increases the activity of p53 in vivo.
• Mutations that deactivate p53 in cancer usually occur in the DBD.
o Most of these mutations destroy the ability of the protein to bind to its target DNA sequences, and prevent transcriptional activation of these genes.
o Mutations in the DBD are recessive loss-of-function mutations.
• Molecules of p53 with mutations in the OD dimerise with wild-type p53, and prevent them from activating transcription. Therefore OD mutations have a dominant negative effect on the function of p53.

32
Q

Function of P53

A

• Regulates the expression of stress response genes + mediates variety of anti-proliferative processes
• Mediates activities through activation of genes regulating
o Cell cycle checkpoints
o DNA damage and repair
o Apoptosis

33
Q

Role of p53 in apoptosis

A
  • P53 enhances expression of Bcl-2 family members (Bax, BID, PUMA, Noxa)
  • Also regulates APAF-1, co-activator of apoptosis initiator caspase-9
  • Some studies suggest p53 might affect apoptosis via novel transcription independent pathways
  • Apoptosis can still occur in the presence of inhibitors of protein synthesis, or when p53 mutants are incapable of acting as transcription factors are ectopically expressed
34
Q

P53 Activation

A

• 3 activating steps:
o Stress-induced stabilisation mediated by phosphorylation
o DNA binding
o Recruitment of general transcription machinery
• During normal homeostasis, p53 is degraded after Mdm2-mediated ubiquitination
• Stress signal-induced p53 phosphorylation by ATM, ATR and other kinases stabilise p53 and promotes DNA binding
o DNA-bound p53 recruits transcriptional machinery to activate p53 target genes
• Image: Kruse JP, Gu W. (2009) Cell.

35
Q

Activation of Cell-Cycle Checkpoints via induction of P53

A

• DNA damage activates p53 by indirect mechanism
• In undamaged cells, p53 is highly unstable and present at low concentrations
o Due to Mdm2 that acts as a ubiquitin ligase that targets p53 for destruction by proteasomes
• DNA damage activates protein kinases that phosphorylate p53 and reduce its binding to Mdm2, decreasing degradation
• The increased p53 concentration in cell causes stimulation of gene transcription
o CKI protein is encoded, called p21
• P21 binds to G1/S-Cdk and S-Cdk and inhibits their activities, blocking entry into S phase

36
Q

Apoptotic pathway regulation by p53

A
•	Death receptor pathways
o	KILLER/DR5
o	FAS
o	PIDD
•	Inhibition of survival signals
o	IGF-BP3
o	PTEN
•	Mitochondrial pathways
o	APAF1
o	BAX
o	FDXR
o	NOXA
o	P42AIP1
o	PUMA
37
Q

Regulation of choice of p53

A

• Choice of response to p53 activation is determined by differential regulation of p53 activity in normal and tumour cells
• Normal cells lead to selective expression of cell-cycle-arrest target genes
o CDKN1A, which encodes WAF1, resulting in reversible or permanent inhibition of cell proliferation
• Tumour cells
• Phosphorylation of p53 at Ser46 through:
o Activation of kinases
o Expression of co-activators such as p53DINP1
o Repression of phosphatases such as WIP1
o Functional interaction with apoptotic cofactors (ASPP, JMY, p63/p73)
• Phosphorylation allows activation of apoptotic target genes
• These cofactors can bind p53 as shown for ASPP and JMY or, as with p63/73, assist p53 DNA binding by directly interacting with p53 responsive promoters
• Although not proven, possible that phosphorylation alters conformation of p53 to enhance interaction with apoptotic cofactors or allow binding to apoptotic target promoters

38
Q

Refined Model for p53 Activation

A

• P53 stabilisation
o Occurs through many different mechanisms
o Many act by affecting ability of Mdm2 to ubiquitinate p53
• Anti-repression
o Consists of release of p53 from the repression mediated by Mdm2 and MdmX
o Requires acetylation of p53 at key lysine residues and facilitates activation of specific subsets of p53 target genes
• Promoter-specific activation
o For full activation, p53 recruits and interacts with numerous cofactors
o These modify p53, the surrounding histones, or other transcription factors

39
Q

describe role of APC in cancer

A
•	Inactivated in FAP
•	Mutated in most colorectal cancers
•	Encodes multifunctional protein involved in
o	Cell adhesion and migration
o	Signal transduction
o	Microtubule assembly
o	Chromosome segregation
o	However, the main tumour-suppressing function of APC resides in its capacity to regulate intracellular b-catenin levels
40
Q

APC in the Wnt Signalling Pathway

A

• Absence of Wnt signal
o APC binds to glycogen-synthase kinase 3-beta (GSK3-b) and Axin
o GSK3b phosphorylates b-catenin, allowing it to be recognised by an SCF complex containing the F-box protein beta-TrCP
o Other proteins in the SCF complex catalyse addition of a polyubiquitin chain to b-catenin
o Allows b-catenin to be recognised and degraded by proteasome
o This means that b-catenin cannot reach the nucleus to activate TCF-responsive genes

• Groucho
o Co-repressor also prevents activation of TCF-responsive genes in absence of b-catenin

• Presence of Wnt signal
o The Wnt receptor, Frizzled, complexes with LRP6 to be activated
o This activates Dishevelled, which activates GBP, an inhibitor of GSK3b
o The inhibition of GSK3b leads to b-catenin being freed into the nucleus to be a co-activator for TCF-responsive genes

• Loss of APC function promotes aberrant cell migration
• Prevents migration of the affected enterocytes out of the colonic crypt
• The mutant cells are therefore retained, rather than being lost through emigration and apoptosis
In Summary
• Aberrant cell migration
• Mutant cells fail to migrate upwards towards the gut lumen and are retained in the crypt
• Increased cell proliferation through stabilisation of b-catenin
• Decreased cell differentiation through effects on Wnt signaling
• Chromosomal instability through chromosome mis-segregation during mitosis leading to aneuploidy

41
Q

Colon Tumour Progression

A

see p90

42
Q

Background and Epidemiology of CRC

A

• CRC is the second commonest cancer/30,000 new cases
• Major economic health burden
• 50% of those affected die from disease
• Prognosis depends on stage of disease
• Incidence increases with age
• Lifetime risk: 1/20 in general population
o 5%
• 11% of cancer-related deaths
• Adenomatous polyp develops into carcinoma
• Tumor progression may take 10-35 years
• Incidence: rises exponentially after the age of 50, hence rare before then unless familial predisposition

43
Q

Risk Factors of crc

A
•	Genetic
•	Environmental
o	Age
o	Alcohol
o	Smoking
o	Red meat
	Left sided tumours
o	Cholecystectomy
	Right sided tumours
o	Pelvic radiation
44
Q

Protective Factors in crc

A
•	Diet
o	High fruit/vegetables
o	Low red meat
•	NSAIDs
o	Aspirin
•	Physical activity
•	Folic acid
•	HRT
•	Statins
•	Fibre?
o	Probably does not but difficult to remove from data, due to its presence in foods such as fruit but it is likely that it is the antioxidants in the fruit and veg that protect rather than fibre itself
45
Q

Genetic Pathways to CRC

A
  • Chromosomal instability
  • CIMP
  • Microsatellite instability
46
Q

Chromosomal instability mechanism in CRC

A

• Most common mechanism for genetic defects in CRC (60-70% of tumours go down this pathway)
• Changes to the chromosome structure
o Insertions
o Deletions
• May be inherited or acquired
• May be in a particular gene or particular chromosome
• Types of instabilities
o Loss of heterozygosity (LOH) – loss of the entire gene and the surrounding chromosomal region.
o Two hit model for alleles – need an abnormality on both chromosomes in order for disease to manifest (variety of genes involved in CRC require the two hits as they are recessive in nature)

47
Q

Methylation in CRC

A

• Definition of methylation: a process by which methyl groups are added to the DNA molecule
o No structural change but gene becomes inactivated
• 30-40% of proximal sporadic tumours are CIMP (CpG Island methylator phenotype)
• 3-12% of distal and rectal cancers CIMP
• CIMP common in proximal tumours independent of MSI
• CIMP associated with BRAF mutations
• High CIMP in sessile serrated polyps
• Risk factors that increase methylation
o Smoking
 CIMP in bronchial epithelium of smokers
 Related to increased incidence of hyperplastic polyps
o Low folate/ High alcohol
 Interferes with DNA methylation resulting in CIMP

48
Q

MSI in CRC

A

• Repair base pair errors in DNA replication
• Defect in HNPCC
• Two hit model also applies
• hMSH2, hPMS1, hPMS2, hMSH6, hMLH3, MLH1
• Tumours exhibit MSI
o Microsatellites are repeated sequences of DNA. These sequences can be made of repeating units of one to six base pairs in length. Although the length of these microsatellites is highly variable from person to person and contributes to the individual DNA “fingerprint”, each individual has microsatellites of a set length. If a mistake occurs in these which is not repaired by mismatch repair genes, this leads to mutations  hence MSI (right)
• Inherited or acquired

49
Q

Genetic Changes in CRC

A

• Human Genome project has identified approx 140 candidate genes
• The average CRC contains 15 mutated genes
• In addition approx. 60 passenger mutated genes present
• High degree of heterogeneity within CRC
o Significant number of potential candidate genes but all may not be important. This differs in each tumour and it is not known which particular gene may be driving this process. Hence difficulty in finding appropriate targeted therapies.

50
Q

APC in CRC development

A

• Inherited defect in FAP, but common in sporadic CRC
o Present in 80% sporadic cancers
• Chr5q21
• Mutation results in truncated APC protein
• Results in accumulation of beta-catenin
• Resists apoptosis by binding and activating transcription T-cell factor
• Acts as a recessive gene  but in FAP it behaves as dominant as these individuals already have one hit

51
Q

RAS-RAF-MEK-ERK Pathway

A
K-ras
•	GTPase – involved in cell signalling
•	Chr12p
•	Most important oncogene in CRC development
•	90% mutations in codons 12 and 13
•	30-40% sporadic MSS CRC
•	40% HNPCC
BRAF
•	Serine-threonine specific kinase
•	40% sporadic MSI-H tumours
•	Not present in HNPCC 
•	Common mutation (90%) is V600E

Constant activation of raf or ras leads to constant proliferation – oncogenes.

52
Q

P53 in CRC

A
  • Most common mutation in human cancer
  • Chr17p
  • 50-70% CRC
  • Late event in CRC development
  • Activated during cell stress
  • Acts as transcriptional factor for growth inhibitory genes
  • Induces apoptosis/cell repair
  • BAX inducer of apoptosis equivalent phenotype
53
Q

DCC in CRC

A
  • Chr18q
  • Has role in cell-cell interaction
  • 73% CRC
  • 43% adenomas
  • May confer poor prognosis in patient with Dukes B – found in studies (not checked in clinical practice)
54
Q

SMAD in CRC

A
Chr18q
•	Mutations result in inactivation of TGFb
•	Late event in adenoma carcinoma sequence
•	SMAD2
o	Associated with pancreatic cancer
•	SMAD4
o	Seen in juvenile polyposis
o	43% of adenomas
55
Q

Importance of Adenomas in CRC

A
  • Coexist in same patient
  • Seen in similar distribution as CRC
  • Precede cancers by 10-15yrs
  • Animal models demonstrate associations
  • Tumours seen arising in adenomatous tissue
  • Removing adenomas decrease incidence of cancer
56
Q

Classical Pathway – Adenoma-Carcinoma Sequence

A

• Normal mucosa  APC Mutation (gatekeeper gene)  Other mutations  classical adenoma
o These microsatellite stable
o No methylation
• Accounts for 60%

57
Q

Describe Serrated Adenomas

A

• Account for 15-20% CRC

• Difficult to detect endoscopically
o Covered in mucus
o More common in right colon

  • Difficult to diagnose histopathologically
  • Confusing classification
  • BRAF mutation lead to CIMP

Types of Serrated Polyps

• Hyperplastic polyp
o 75% of serrated lesion
o Mainly distal
o No dysplasia

• Microvesicular Hyperplastic Polyp (MVHP)
o Smaller version of hyperplastic polyps  mat have dysplasia

• Sessile Serrated lesion
o May/may not be dysplastic
o Dysplasia risk factor for progression

• Traditional Serrated Adenoma
o Adenoma with some serrated features

• Mixed polyp

58
Q

Describe the Sessile Serrated Pathway

A

• Various pathways that normal mucosa can take
o Normal mucosa  BRAF mutation  sessile serrated adenoma  MLH1 methylation (mismatch repair gene – this is prone to methylation)  Sessile serrated polyp
 MSI, high methylation, BRAF mutation
 12%
o Normal mucosa  BRAF mutation  MVHP  sessile serrated adenoma  MGMT methylation (mismatch repair gene – this is prone to methylation)  Sessile serrated polyp
 Microsatellite stable, but high methylation, BRAF mutation
 8%
o Normal mucosa  kRAS mutation  MGMT methylation (mismatch repair gene – this is prone to methylation)  Traditional sessile serrated polyp
 Microsatellite stable, but low methylation, kras mutation
 20%

59
Q

Describe Hereditary CRC

A
  • Familial syndromes (have family members with CRC) make up 10-30% and do not necessarily have one particular gene that predisposes unlike syndromes e.g. FAP, HNPCC
  • These people are at increased risk e.g. if you have 2 first degree relatives with CRC your risk goes up to 17% (instead of 5%). Specific familial syndromes e.g. HNPCC increase risk much more (70%).
60
Q

Describe FAP

A

• 1% CRC
• Estimated penetrance for adenomas >90%
• CHRPE may be present
o Congenital hypertrophy of the retinal pigment epithelium
• Risk of CRC = 100% if untreated polyposis
• Surveillance at puberty
• Colectomy if affected

61
Q

Describe attenuated FAP

A

• 20 to 100 polyps, usually more proximal
• Onset later than FAP, average age of onset = 50
• Lifetime risk of CRC = 80%
• Extracolonic tumors occur at same rate as FAP
o Not associated with CHRPE
• Variant of FAP, mutations in same APC gene
• Surveillance:
o annual colonoscopy starting late teens or early 20’s
o Option of subtotal colectomy

62
Q

Describe APC Gene Mutation in Ashkenazi Jews

A

• Missense mutation (I1307K) associated with increased risk of CRC and adenomas in Ashkenazi Jews (AJ)
o Behaves in a Mendelian way
• Found in 6% of the general AJ population
o 12% of AJs with CRC
o 29% of AJs with CRC and a positive family history
• Lifetime risk of CRC in mutation carrier is 10-20%
• Screening: colonscopy every 2-5 yrs starting at 35 yrs

63
Q

Describe Hereditary Non-Polyposis Coli (HNPCC/Lynch Syndrome)

A

• Inherited mutation in mismatch repair gene
• 3-5% CRC
• 30-90% risk of CRC
• Associated malignancies
o Ovary, Endometrial, small bowel, stomach, ureteric (right – paper)
• Biennial colonoscopy
• Associated screening
• More right sided lesions
• Increased mucinous component
• Lymphocytic infiltration
• Poorly differentiated but better prognosis

64
Q

Amsterdam criteria for HNPCC

A

2+ CRC cases in min 2 generations

One affected individual first degree relative

One case diagnosed before 50

CRC may be replaced with endometrial or small bowel carcinoma

FAP must be excluded

If you fulfil criteria above you definitely have HNPCC, but if you don’t it does not necessarily mean you do not have it.

65
Q

HNPCC Pathway

A

• Normal mucosa  MMR mutation  TSA  other mutations HNPCC tumour
o MSI, but no methylation

66
Q

MAP Syndrome/MYH Gene

A

• Multiple adenomatous polyposis (MAP) syndrome
o Autosomal recessive; mutations in the MYH gene
o Median number of polyps = 55
o Mean age of polyp diagnosis = 30-50 years
• Repairs oxidative damage to guanine
• Often associated with sporadic APC mutation
• Phenotype identical to FAP (but APC would be normal)
• 30% of individuals with 15-100 polyps have homozygous mutations in the MYH gene
• Genetic testing should be offered if >15 polyps (and APC gene testing negative)

67
Q

Peutz-Jeghers Syndrome

A

• <1% of all CRC cases
• Hamartomatous polyps of GI tract as early as 1st decade
• Mucocutaneous hyperpigmentation
o Lips, mouth, buccal mucosa, fingers
o Usually seen in children < 5 yrs
• Cancer risk:
o colon, small intestine, stomach, pancreas, breast, ovaries, uterus, testes, lungs, kidneys
• Mutations in STK11 gene
o Found in 70% of familial cases and 30-70% of sporadic cases

68
Q

Familial Juvenile Polyposis

A

• <1% of all CRC cases
• Autosomal dominant
• 5 or more juvenile polyps in colon or GI tract
o Appear in 1st or 2nd decade
o 50% lifetime risk of CRC; AOO in 30’s
o Increased risk gastric, GI, pancreatic CA
• ~50% of cases have mutations in either the SMAD4 or BMPR1A genes

69
Q

Hyperplastic/Serrated Polyposis (HPSP)

A

• 5 HP proximal to SC of which 2 are > 10mm
• HP polyps proximal to the SC with a FH of HPSP
• More than 20 HP throughout the colon
o Type 1: Large serrated lesion, TSA and HP associated with high risk of CRC
o Type 2: Multiple small HPs <5mm. Low risk of CRC
• Unknown gene

70
Q

Prostaglandin Signalling in CRC

A

• Increased COX2 in 67% tumours
• Decreased 15-prostaglandin dehydrogenase in 80% tumours
• COX-2 inhibitors prevents new adenomas and mediates regression of established adenomas
• PPAR
o Codes for proteins that regulate lipid metabolism and cell growth
o Downstream of the APC gene – may also be involved in COX2 pathway
o May confer increased risk in patients with acromegaly

71
Q

GFs in drug tx

A

• Epidermal Growth Factor (EGF)
o Trophic effect on intestinal cells
o Cetuximab is antibody directed against EGF receptor
• Vascular endothelial growth factor (VEGF)
o Promotes angiogenesis
o Bevacizumab (not as effective) is antibody directed against VEGF
o Adds 4.7 months to overall survival of CRC patients

72
Q

Clinical Importance of Genetic Changes

A
Predictive Factors
•	K-Ras
o	No response to Cetuximab
•	BRAF – ?
o	No response to Cetuximab
•	BRAF
o	No response to BRAF inhibitors
•	Sporadic mismatch repair
o	No response to 5FU but good response to irinotecan
•	PIK3CA
o	Longer survival with aspirin use

Prognosis
• Sporadic microsatelite instability – good prognosis
• 18q LOH – poor prognosis
• Farsenyl Transferase Inhibitors – kRAS inhibitor

73
Q

Genetics of IBD CRC

A

• The mean age 10-20 yrs earlier
• K-Ras mutations less common and occur later
• LOH for p53 occurs earlier than in sporadic cancers
• Dysplasia in UC can be found at distant sites
• Abnormalities of the p53 found in non-dysplastic mucosa
o i.e More of a field effect  not simply one lesion, dysplastic mucosa occurs in whole colon
• Do not get polyps, and can develop cancer very quickly (within 2-3 years) so care must be taken with screening these patients – must be regular, thoroughly searched and especially so if they have particularly active disease

74
Q

Acromegaly – PPAR

A

• Codes for proteins that regulate lipid metabolism and cell growth
• Is downstream of the APC gene and may also be involved in COX 2 pathway
• May confer increased risk in patents with acromegaly
o Especially if they have very high growth factor hormone levels
• Difficult to screen due to large colons