Path Book: Chapter 5 Neoplasia pg. 178-190 Flashcards

1
Q

Carcinogenesis is a multistep process resulting from the accumulation of multiple genetic alterations that collectively give rise to the transformed phenotype.

A

Many cancers arise from non-neoplastic precursor lesions, which molecular analyses have shown already possess some of the mutations needed to establish a full-blown cancer. Presumably these mutations provide the cells of the precursor lesion with a selective advantage. Once initiated, cancers continue to undergo darwinian selection.

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

What are some phenotypic manifestations of malignant neoplasms?

A

excessive growth, local invasiveness, and the ability to form distant metastases.

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

What is tumor progression?

A

Over a period of time, many tumors become more aggressive and acquire greater malignant potential.

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

T or F. Increasing malignancy often is acquired in an incremental fashion.

A

T. At the molecular level, tumor progression and associated heterogeneity are most likely to result from multiple mutations that accumulate independently in different cells, generating subclones with different characteristics such as ability to invade, rate of growth, metastatic ability, etc

Some of the mutations may be lethal; others may spur cell growth by affecting proto-oncogenes or cancer suppressor genes.

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

Thus even though most malignant tumors are monoclonal in origin, by the time they become clinically evident their con- stituent cells may be extremely heterogeneous.

A

Thus even though most malignant tumors are monoclonal in origin, by the time they become clinically evident their con- stituent cells may be extremely heterogeneous.

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

During progression, how are tumor cells subjected to immune and nominate section pressures?

A

For example, cells that are highly antigenic are destroyed by host defenses, whereas those with reduced growth factor requirements are positively selected.

A growing tumor, therefore, tends to be enriched for subclones that “beat the odds” and are adept at survival, growth, invasion, and metastasis.

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

What is a major significance of the selective pressure of tumors?

A

The recurrent tumor is almost always resistant to the drug regimen if it is given again.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are some of the hallmarks of cancer cell growth/behavior?

A
• Self-sufficiency in growth signals
• Insensitivity to growth inhibitory signals
• Evasion of cell death
• Limitless replicative potential
• Development of sustained angiogenesis
• Ability to invade and 
metastasize

Also, reprogramming of energy metabolism and evasion of the immune system

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

How does normal cell growth/proliferation occur?

A
  1. The binding of a growth factor to its specific receptor on the cell membrane
  2. Transient and limited activation of the growth factor receptor, which in turn activates several signal- transducing proteins on the inner leaflet of the plasma membrane
  3. Transmission of the transduced signal across the cytosol to the nucleus by second messengers or a cascade of signal transduction molecules
  4. Induction and activation of nuclear regulatory factors that initiate and regulate DNA transcription
  5. Entry and progression of the cell into the cell cycle, resulting ultimately in cell division
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

All cells need growth factors to grow. Are most growth factors autocrine, paracrine, or endocrine?

A

paracrine- most cells that make growth factors don’t express their receptor. This prevents positive feedback loops in the cell

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

How do cancer cells evade the growth factor requirement?

A
  • Many acquire the ability to synthesize the same growth factors to which they are responsive. Similar autocrine loops are fairly common in many types of cancer.
  • Cancer cells acquire growth self-sufficiency is by interaction with stroma. In some cases, tumor cells send signals to activate normal cells in the supporting stroma, which in turn produce growth factors that promote tumor growth.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

How can mutated growth receptors stimulate cancer formation?

A

Mutant receptor proteins deliver continuous mitogenic signals to cells, even in the absence of the growth factor in the environment.

However, mutated receptors are not very common causes of cancer formation

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

Growth receptors still play a significant role in cancer formation even though mutation is not common. How?

A

More common than mutations is overexpression of growth factor receptors, which can render cancer cells hyperresponsive to levels of the growth factor that would not normally trigger proliferation.

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

What the result of a mutated HER2/NEU receptor? Treatment? Prognosis?

A

The gene encoding a related receptor, HER2/NEU (ERBB2), is amplified in 25% to 30% of breast cancers and adenocarcinomas of the lung, ovary, and salivary glands.

These tumors are exquisitely sensitive to the mitogenic effects of small amounts of growth factors, and a high level of HER2/NEU protein in breast cancer cells is a harbinger of poor prognosis.

Treatment of breast cancer with anti- HER2/NEU antibody is an elegant example of “bench to bedside” medicine.

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

What are RAS and ABL?

A

A relatively common mechanism by which cancer cells acquire growth autonomy is mutations in genes that encode various components of the signaling pathways down- stream of growth factor receptors.

These signaling proteins couple growth factor receptors to their nuclear targets. They receive signals from activated growth factor receptors and transmit them to the nucleus, either through second messengers or through a cascade of phosphorylation and activation of signal transduction molecules.

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

RAS is the most commonly mutated proto- oncogene in human tumors.

A

RAS is the most commonly mutated proto- oncogene in human tumors.

Indeed, approximately 30% of all human tumors contain mutated versions of the RAS gene, and the frequency is even higher in some specific cancers (e.g., colon and pancreatic adenocarcinomas).

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

What is RAS specifically?

A

a G-receptor coupled protein that is inactivated when bound to GDP and active when bound to GTP

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

How are GRCPs like RAS inactivated?

A

Activated state is short-lived because the intrinsic GTPase activity of RAS hydrolyzes GTP to GDP, releasing a phosphate group and returning the protein to its quiescent GDP-bound state.

In its mutated (aka cancer causing) state, RAS can no longer be inactivated due to point mutations

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

What is BRAF? What kind of cancer is its mutated form present in?

A

a GRCP that lies in the RAS/RAF/ERK/MAP kinase pathway, is mutated in more than 60% of melanomas.

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

What is ABL?

A

a non–receptor-associated tyrosine kinase that functions as a signal transduction molecule.

Mutation is common with cancer formation

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

How is ABL mutated in chronic myelogenous leukemia and certain acute leukemias?

A

A part of the ABL gene is translocated from its normal abode on chromosome 9 to chromosome 22, where it fuses with part of the breakpoint cluster region (BCR) gene.

The BCR-ABL hybrid protein maintains the tyrosine kinase domain; the BCR domain self- associates, a property that unleashes a constitutive tyrosine kinase activity. Of interest, there is cross-talk between BCR-ABL and RAS pathways, since BCR-ABL protein activates all of the signals that are downstream of RAS.

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

How would/could you treat chronic myelogenous leukemia?

A

BCR-ABL kinase inhibitors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is Gleevec?

A

imatinib mesylate (Gleevec) is a BCR-ABL kinase inhibitor used in cancer therapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What is oncogene addiction?

A

case in which a tumor is profoundly dependent on a single signaling molecule (such as BCR-ABL). Common in leukemia

remember: BCR-ABL fusion gene formation is an early, perhaps initiating, event that drives leukemo- genesis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

T or F. The ultimate consequence of signaling through mutated oncoproteins such as RAS or ABL is inappropriate and continuous stimulation of nuclear tran- scription factors that drive the expression of growth- promoting genes.

A

T. Thus, in addition to growth factor pathway mutations, growth autonomy may thus be a consequence of mutations affecting genes that regulate transcription of DNA

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

What are some important transcription factors/genes/proteins that play a role in cancer formation when mutated?

A

A host of oncoproteins, including products of the MYC, MYB, JUN, FOS, and REL oncogenes, function as transcription factors that regulate the expression of growth-promoting genes, such as cyclins.

Of these, the MYC gene is involved most commonly in human tumors (the MYC gene makes MYC protein).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What does the MYC protein do?

A

The MYC protein can either activate or repress the transcription of other genes.

MYC also is a key regulator of intermediate metabolism, upregulating genes that promote aerobic glycolysis (Warburg effect) and the increased utilization of glutamine, two metabolic changes that are hallmarks of cancer cells.

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

Which growth-factor promoting genes does/can MYC activate?

A

Cyclin- dependent kinases (CDKs).

In contrast, they repress transcription of CDK inhibitors (CDKIs)

Thus, dysregulation of MYC promotes tumorigenesis by increasing expression of genes that promote progression through the cell cycle and repressing genes that slow or prevent progression through the cell cycle.

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

How is MYC affected in Burkitt lymphoma?

A

Dysregulation of the MYC gene resulting from a t(8;14) translocation occurs in Burkitt lymphoma, a B cell tumor.

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

The replication of cells is stimulated by growth factors or by signaling from ECM components through integrins.

A

The replication of cells is stimulated by growth factors or by signaling from ECM components through integrins.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

What are the main checkpoints of the cell cycle?

A

The cell cycle has multiple checkpoints, particularly during emergence from G0 into G1 and the transition from G1 to S phase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

The orderly progression of cells through the various phases of the cell cycle is orchestrated by ______.

A

CDKs, which are activated by binding to the cyclin

The CDK–cyclin complexes phosphorylate crucial target proteins that drive the cell through the cell cycle.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

What happens to cyclin levels when they accomplish their task?

A

cyclin levels decline rapidly.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

What happens if cell surveillance mechanisms such as CDKIs sense that DNA is too damaged to complete a cell cycle?

A

If DNA damage is too severe to be repaired, the cells are eliminated by apoptosis, or enter a nonreplicative state called senescence, primarily through p53-dependent mechanisms.

Mutations in genes regulating these checkpoints allow cells with damaged DNA to divide, producing daughter cells carrying mutations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Which CDK-cyclin complexes regulate entry into the S phase from G1? How?

A

cyclin D–CDK4, cyclin D–CDK6, and cyclin E–CDK2 regulate the G1-to-S transition by phosphorylating the Rb protein (pRb).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Which CDK-cyclin complexes are active in S phase?

A

Cyclin A–CDK2 and cyclin A–CDK1 are active in the S phase.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Which CDK-cyclin complex is essential for the G2-to-M transition?

A

Cyclin B–CDK1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What do INK4 CDKIs do? Members?

A

The so-called INK4 inhibitors, composed of p15, p16, p18, and p19, act on cyclin D–CDK4 and cyclin D–CDK6.

The other family of three inhibitors, p21, p27, and p57, can inhibit all CDKs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

All cancers appear to have genetic lesions that disable the G1-S checkpoint, causing cells to continually reenter the S phase. For unclear reasons, particular lesions vary widely in frequency across tumor types.

A

All cancers appear to have genetic lesions that disable the G1-S checkpoint, causing cells to continually reenter the S phase. For unclear reasons, particular lesions vary widely in frequency across tumor types.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Mutation of which CDK is most common in neoplastic transformation?

A

CDK4. Mutations affecting cyclins B and E and other CDKs also occur, but they are much less frequent than those affecting cyclin CDK4.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Specifically, which kinds of cancers is CDK4 mutation common in?

A

melanomas, sarcomas, and glioblastomas.

42
Q

Do oncogene proteins tend to promote cell growth and proliferation or the opposite?

A

growth and proliferation. TSGs do the opposite.

43
Q

What is RB?

A

The retinoblastoma gene (RB), the first tumor suppressor gene to be discovered (aka the cell cycle governor)

44
Q

Describe the prevalence of sporadic vs. hereditary retinoblastoma

A

Approximately 60% of retinoblastomas are sporadic, and the remaining ones are familial, the predisposition to develop the tumor being transmitted as an autosomal dominant trait.

45
Q

What is the two-hit hypothesis?

A

•Two mutations (hits) are required to produce retinoblas-toma.

These involve the RB gene, which has been mapped to chromosomal locus 13q14. Both of the normal alleles of the RB locus must be inactivated (hence the two hits) for the development of retinoblastoma.

• In familial cases, children inherit one defective copy of the RB gene in the germ line; the other copy is normal.

46
Q

At the patient level, is hereditary retinoblastoma dominant or recessive?

A

Dominant. Thus, you only need one mutated/absent allele to perpetuate transmission

47
Q

At the cellular level, is hereditary retinoblastoma dominant or recessive? Explain

A

Recessive. A child will be born with one messed up allele and then a somatic mutation is needed for disease

In sporadic cases, both normal RB alleles are lost by somatic mutation in one of the retinoblasts. The end result is the same: a retinal cell that has lost both of the normal copies of the RB gene becomes cancerous.

Thus, a cell heterozygous at the RB locus is not neoplastic. Tumors develop when the cell loses its normal RB gene copy and thus becomes homozygous for the mutant allele.

48
Q

Patients with retinoblastoma are at increased of developing what other cancers?

A

osteosarcomas and some soft tissue sarcomas.

49
Q

Where is RB protein expressed? What is its job?

A

The RB gene product is a DNA-binding protein that is expressed in every cell type examined, where it exists in an active hypophosphorylated state and an inactive hyperphosphorylated state.

The importance of Rb lies in its regulation of the G1/S checkpoint, the portal through which cells must pass before DNA replication commences.

50
Q

What is the most important checkpoint of cell division? Why?

A

The transition from G1 to S because once cells cross the G1 checkpoint they can pause the cell cycle for a time, but they are obligated to complete mitosis.

51
Q

If a cell is damaged in G1, what can it do to escape the cell cycle?

A

In G1 cells can remove themselves entirely from the cell cycle, either temporarily (quiescence, or G0) or permanently (senescence).

52
Q

The initiation of DNA replication (S phase) requires what?

A

the activity of cyclin E/CDK2 complexes

53
Q

How is cyclin E activated (so that it can activate CDK2)?

A

It is dependent on the E2F family of transcription factors

54
Q

What prevents the E2F family of transcription factors from activating cyclin E in early G1?

A

Early in G1, Rb is in its hypophosphorylated active form, and it binds to and inhibits the E2F family of transcription factors, preventing transcription of cyclin E.

55
Q

How does hypophosphorylated Rb inhibit E2F factors? 2 ways

A

1) First, it sequesters E2F, preventing it from interacting with other transcriptional activators.
2) Second, Rb recruits chromatin remodeling proteins, such as histone deacetylases and histone methyltransferases, which bind to the promoters of E2F-responsive genes such as cyclin E. These enzymes modify chromatin at the promoters to make DNA insensitive to transcription factors like E2F.

56
Q

How is Rb phosphorylated?

A

Growth factor signaling leads to cyclin D expression and activation of cyclin D–CDK4/6 complexes. These complexes phosphorylate Rb, inactivating the protein and releasing E2F to induce target genes such as cyclin E.

Expression of cyclin E then stimulates DNA replication and progression through the cell cycle.

57
Q

When are Rbs dephosphorylated again?

A

When the cells enter S phase, they are committed to divide without additional growth factor stimulation. During the ensuing M phase, the phosphate groups are removed from Rb by cellular phosphatases, regenerating the hypophosphorylated form of Rb.

58
Q

Would cyclin D be overexposed or underexposed in a tumor?

A

overexpressed

59
Q

Which four cell-cycle mediators are most mutated in human cancer?

A

CDKN2A (p16), cyclin D, CDK4, or Rb

60
Q

How does/can HPV cause cancer?

A

E7 protein binds to the hypophosphorylated form of Rb, preventing it from inhibiting the E2F transcription factors. Thus, Rb is functionally deleted, leading to uncontrolled growth.

61
Q

What is p53?

A

protein product of the p53-encoding tumor suppressor gene, TP53

62
Q

How does p53 prevent neoplastic event?

A

1) activation of temporary cell cycle arrest (termed quiescence),
2) induction of permanent cell cycle arrest (termed senescence), or
3) triggering of apoptosis.

If Rb “senses” external signals, p53 can be viewed as a central monitor of internal stress, directing the stressed cells toward one of these three pathways.

63
Q

In healthy cells, is p53 around long? Why or why not?

A

In nonstressed, healthy cells, p53 has a short half-life (20 minutes) because of its association with MDM2, a protein that targets p53 for destruction.

64
Q

What happens in relation to MDM2/p53 when cells become ‘stressed’ via hypoxia, DNA damage, etc.

A

“sensors” that include protein kinases such as ATM (ataxia telangiectasia mutated) are activated. These activated complexes catalyze post-translational modifications in p53 that release it from MDM2 and increase its half-life and enhance its ability to drive the transcription of target genes.

65
Q

Hundreds of genes whose transcription is triggered by p53 have been found. These genes suppress neoplastic transformation by three mechanisms:

A

1) p53-mediated cell cycle arrest may be considered the primor- dial response to DNA damage
2) p53-induced senescence (permanent cell cycle arrest)
3) p53-induced apoptosis of cells with irreversible DNA damage is the ultimate protective mechanism against neoplastic

66
Q

How does p53 promote quiescence?

A

It occurs late in the G1 phase and is caused mainly by p53-dependent transcription of the CDKI gene CDKN1A (p21).

The p21 protein inhibits cyclin–CDK complexes and prevents phosphorylation of Rb, thereby
arresting cells in the G1 phase for repair.

67
Q

What happens if DNA repair following quiescence is successful?

A

If DNA damage is repaired successfully, p53 upregulates transcription of MDM2, leading to destruction of p53 and relief of the cell cycle block.

68
Q

What happens if DNA repair following quiescence is not successful?

A

If the damage cannot be repaired, the cell may enter p53-induced senescence or undergo p53-directed apoptosis

69
Q

What occurs in senescence?

A

The mechanisms of senescence are unclear but seem to involve global chromatin changes, which drastically and permanently alter gene expression.

70
Q

Until recently it was thought that the functions of p53 were mediated exclusively by transcriptional activation of genes with antiproliferative, apoptotic, and senescence- inducing functions. But p53 also represses a subset of pro-proliferative and anti-apoptotic genes as well. How could p53, a transcriptional activator, repress gene function?

A

p53 can transcriptionally activate certain miRNAs. MiRNAs activated by p53 can inhibit the translation of pro- proliferative genes such as cyclins and anti-apoptotic genes such as BCL2.

71
Q

Confirming the importance of TP53 in controlling carci- nogenesis, more than 70% of human cancers have a defect in this gene, and the remaining malignant neoplasms have defects in genes upstream or downstream of TP53.

A

Confirming the importance of TP53 in controlling carci- nogenesis, more than 70% of human cancers have a defect in this gene, and the remaining malignant neoplasms have defects in genes upstream or downstream of TP53.

72
Q

Are most TP53 mutation sporadic or inherited?

A

sporadic

73
Q

What is a disease caused by inherited TP53 defect?

A

Li-Fraumeni syndrome. As with the RB gene, inheritance of one mutant allele predisposes affected persons to develop malignant tumors because only one additional hit is needed to inactivate the second, normal allele. Patients with the Li-Fraumeni syndrome have a 25-fold greater chance of developing a malignant tumor by age 50 compared with the general population and will typically develop multiple tumors

74
Q

What kinds of cancers are common in Li-Fraumeni syndrome?

A

In contrast with tumors developing in patients who inherit a mutant RB allele, the spectrum of tumors that develop in patients with the Li-Fraumeni syndrome is varied; the most common types are:

sarcomas, breast cancer, leukemia, brain tumors, and carcinomas of the adrenal cortex.

75
Q

Which viruses can target p53?

A

Proteins encoded by oncogenic HPVs, hepatitis B virus (HBV), and possibly Epstein-Barr virus (EBV) can bind to normal p53 and nullify its protective function.

76
Q

What is a well known signal that transmits anti-proliferative signals to cells?

A

TGF-B.

At some point, however it will actually help growth of a tumor via immunosuppression and by inducing VEGF (angiogenesis).

77
Q

What does TGF-B do to prevent proliferation?

A

In most normal epithelial, endothelial, and hematopoietic cells, TGF-β is a potent inhibitor of proliferation.

It regulates cellular processes by binding to a complex composed of TGF-β receptors I and II. Dimerization of the receptor upon ligand binding leads to a cascade of events that result in the transcriptional activation of CDKIs with growth-suppressing activity, as well as repression of growth-promoting genes such as MYC, CDK2, CDK4, and those encoding cyclins A and E.

78
Q

What are the major mutation targets of the TGF-B pathway?

A

TGF-B receptor II and SMAD4, 1 of 10 proteins involved in TGF-B signal transduction

79
Q

Mutations affecting the type II receptor are seen in cancers of the ____, ____, and _____.

A

colon, stomach, and endometrium.

80
Q

In what condition is SMAD4 commonly mutated?

A

common in pancreatic cancers.

NOTE: In 100% of pancreatic cancers and 83% of colon cancers, at least one component of the TGF-β pathway is mutated.

81
Q

What is “contact inhibition”?

A

When nontransformed cells are grown in culture, they pro- liferate until confluent monolayers are generated; cell–cell contacts formed in these monolayers suppress further cell proliferation. This is absent in tumor growth, allowing stacking to occur

82
Q

What protein does the gene NF2 make?

A

neurofibromin-2, more commonly called merlin

83
Q

What does merlin do?

A

facilitates E-cadherin mediated contact inhibition. E-cadherin is a major player in cell-cell contact

loss of E-cadherin is common in lobular carcinoma of the breast.

84
Q

Homozygous loss of NF2 can cause in what?

A

A form of neural tumors associated with the condition called neurofibromatosis.

85
Q

What does the APC gene do?

A

exerts antiproliferative actions by regulating the destruction of the cytoplasmic protein β-catenin. With a loss of APC, β-catenin is not destroyed, and it translocates to the nucleus, where it acts as a growth-promoting trans-cription factor via the WNT signaling pathway.

86
Q

Loss of both copies of the APC gene result in what?

A

In familial adenomatous polyposis syndrome, inheritance of a germ line mutation in the APC gene and sporadic loss of the sole normal allele causes the development of hundreds of colonic polyps at a young age. Inevitably, one or more of these polyps evolves into a colonic cancer. Somatic loss of both alleles of the APC gene is seen in approximately 70% of sporadic colon cancers.

87
Q

How does the apoptotic pathway work?

A

The apoptotic pathway can be divided into upstream regulators and downstream effectors. The regulators are divided into two major pathways, one interpreting extracellular or extrinsic signals and the other interpreting intra- cellular signals. Stimulation of either pathway results in activation of a normally inactive protease (caspase-8 or caspase-9, respectively), which initiates a proteolytic cascade involving “executioner” caspases that disassemble the cell in orderly fashion. The cellular remains are then efficiently consumed by the cellular neighbors and profes- sional phagocytes, without stimulating inflammation.

88
Q

How is the extrinsic (death receptor) pathway initiated?

A

when a TNF receptor, such as CD95 (Fas), is bound to its ligand, CD95L, leading to trimerization of the receptor and its cytoplasmic death domains, which attract the intracellular adaptor protein FADD.

89
Q

What does FADD do?

A

This protein recruits procaspase-8 to form the death-inducing signaling complex. Procaspase-8 is activated by cleavage into smaller subunits, generating caspase-8.

90
Q

What does Caspase-8 do?

A

then activates downstream caspases such as caspase-3, an executioner caspase that cleaves DNA and other substrates to cause cell death.

91
Q

The intrinsic (mitochondrial) pathway of apoptosis is triggered by a variety of stimuli, including withdrawal of survival factors, stress, and injury. Activation of this pathway leads to what?

A

permeabilization of the mitochondrial outer membrane and release of molecules, such as cytochrome c, that initiate apoptosis.

92
Q

The integrity of the mitochondrial outer membrane is regulated by pro-apoptotic and anti-apoptotic members of the BCL2 family of proteins. How does this work?

A

The pro-apoptotic pro- teins BAX and BAK are required for apoptosis and directly promote mitochondrial permeabilization. Their action is inhibited by the anti-apoptotic members of this family exemplified by BCL2 and BCL-XL.

93
Q

What do BH3-only proteins, which include BAD, BID, and PUMA do?

A

The BH3-only proteins promote apoptosis by neutralizing the actions of anti-apoptotic proteins like BCL2 and BCL-XL. When the sum total of all BH3 proteins expressed “overwhelms” the anti-apoptotic BCL2/ BCLXL protein barrier, BAX and BAK are activated and form pores in the mitochondrial membrane, causing cytochrome c leaks into the cytosol

Because of the pro-apoptotic effect of BH3 only proteins, efforts are underway to develop BH3 mimetic drugs to promote death of tumor cells.

94
Q

What does cytochrome c in the cytosol do?

A

binds to APAF-1 to activate caspase-9.

95
Q

What does caspase-9 do?

A

Like caspase-8 of the extrinsic pathway, caspase-9 can cleave and activate the executioner caspases.

96
Q

How can caspases be inhibited besides not getting their appropriate cell-death signals?

A

by a family of proteins called inhibitor of apoptosis proteins (IAPs).

97
Q

How can tumors evade the apoptotic pathway?

A

via upregulation of BCL2

98
Q

How is Bcl2 unregulated?

A

Approximately 85% of B cell lymphomas of the follicular type carry a characteristic t(14;18) (q32;q21) translocation.

As noted earlier, 14q32, the chromosomal locus for immunoglobulin heavy-chain genes, also is involved in the pathogenesis of Burkitt lymphoma.

Juxtaposition of this transcriptionally active locus with BCL2 (located at 18q21) causes overexpression of the BCL2 protein. This overabundance in turn increases the BCL2/ BCL-XL buffer, protecting lymphocytes from apoptosis and allowing them to survive for long periods; there is therefore a steady accumulation of B lymphocytes, resulting in lymphadenopathy and marrow infiltration.

Because BCL2- overexpressing lymphomas arise in large part through reduced cell death rather than explosive cell proliferation, they tend to be indolent (slow-growing) compared to other lymphomas. In some instances, reduced levels of CD95 may render the tumor cells less susceptible to apoptosis by Fas ligand (FasL).

99
Q

What is FLIP?

A

Some tumors have high levels of FLIP, a protein that can bind death-inducing signaling complex and prevent activation of caspase 8.

100
Q

How is autophagy performed?

A

During autophagy, cellular organelles are sequestered from the rest of the cell by an autophagosome and then fused to a lysosome, where they are degraded and utilized for cellular energy generation.

Later, tumors can use autophxgy by supplying the nutrients needed for survival in nutrient-poor environments