Growth, Cell Death and Neoplasia Flashcards

1
Q

define apoptosis

A

programmed cell death

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

define necrosis

A

traumatic cell death - unintended cell death in response to cellular injury

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

why do we need apoptosis?

A

for continuous renewal of cells, working alongside mitosis

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

name some inhibitors of apoptosis

A

growth factors
extracellular cell matrix
sex steroids
some viral proteins

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

name some inducers of apoptosis

A
growth factor withdrawal
loss of attachment of ECM
glucocorticoids
some viruses
free radicals
ionising radiation
DNA damage
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

give some examples of when apoptosis is crucial for normal growth - and what occurs if apoptosis fails

A

separating of fingers - interdigital cell death - syndactyl (webbed fingers)
removal of redundant epithelium on fusion of palatine processes in roof of mouth - cleft palate

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

what is coagulative necrosis?

A

most common.

dead tissue is swollen and firm, then later becomes soft - digestion of macrophages

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

what is colliquative (liquefactive) necrosis?

A

occurs in brain due to lack of supporting stroma - necrotic tissue liquefies

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

what is caseous necrosis?

A

dead tissue is completely structureless. histology shows amorphous eosinophilic area with haematoxyphilic nuclear debris.
seen in TB.

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

what is gangrene?

A

necrosis with putrefaction of tissues. tissues appear black.

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

what is fibrinoid necrosis?

A

occurs in malignant hypertension - increased arterial pressure results in necrosis of SM wall.
eosinophilic and fibrinous deposits seen.

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

what is fat necrosis?

A

focal adipose tissue destruction due to direct trauma, or enzymatic lipolysis (seen in acute pancreatitis)

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

define hypertrophy

A

increase in size of a tissue caused by increase in SIZE of the constituent cells

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

define hyperplasia

A

increase in size of a tissue caused by an increase in NUMBER of constituent cells - decrease in apoptosis

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

define atrophy

A

decrease in size of a tissue caused by a decrease in NUMBER or SIZE of constituent cells - apoptosis

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

define metaplasia

A

change in differentiation of a cell from one fully-differentiated type to another

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

define dysplasia

A

morphological changes seen in cells in the progression to becoming cancer

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

give some examples of physiological hypertrophy/hyperplasia

A

muscle hypertrophy in athletes - in limbs and LV of heart.
hyperplasia of breast tissue at puberty and in pregnancy/lactation.
hypertrophy and hyperplasia of uterine smooth muscle at puberty/pregnancy.

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

give some examples of conditions involving pathological hypertrophy/hyperplasia

A

RVH/LVH.
gynaecomastia.
prostate hyperplasia.
follicular epithelial hyperplasia e.g. in Graves’ disease (thyroid)

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

define congenital

A

present at birth - not necessarily inherited, foetus is affected by environmental factors in womb (eg FAS)

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

give some examples of changes to the body that are typical of ageing

A
loss of dermal elastosis
osteoporosis
cataracts
senile dementia
sarcopaenia (loss of skeletal muscle mass/strength)
deafness
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

define carcinogenesis

A

transformation of normal cells to neoplastic cells through permanent genetic alterations or mututations
- malignant tumours

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

define oncogenesis

A

formation of benign and malignant tumours

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

define carcinogen

A

agents known/suspected to cause cancer - malignant tumours.

act on DNA - mutagenic

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

define oncogenic

A

agents known/suspected to cause tumours - benign or malignant

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

what problems are presented by attempts to identify carcinogens?

A

latent interval may be decades.
environment is complex - difficult to isolate one factor.
ethical constraints - can’t deliberately give people a suspected carcinogen as part of a study

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

list the classes of carcinogen

A
chemical
viral
ionising/non-ionising radiation
hormones, parasites and mycotoxins
misc.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

define neoplasm

A

lesion resulting from autonomous (or relatively autonomous) abnormal growth of cells which persists after the initiating stimulus has been removed
A NEW GROWTH

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

what are the features of neoplasia?

A

autonomous
abnormal
persistent
new growth

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

what makes a neoplasm malignant?

A

if they invade surrounding tissues, and are able to metastasise/spread to other tissues

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

describe the structure of solid tumours

A

neoplastic cells + stroma

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

what induces stroma formation in a tumour?

A

connective tissue fibroblast proliferation is induced by growth factors produced by tumour cells

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

why is angiogenesis needed in tumours?

A

to perfuse the tumour, allowing it to grow - without angiogenesis, tumour may only grow to 1-2mm diameter

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

what induces angiogenesis in tumours?

A

vascular endothelial growth factor (VEGF) - secreted by tumour cells

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

describe the features of a neoplastic cell

A

derived from nucleated cells, usually monoclonal.
growth pattern relates to parent cell.
synthetic activity relates to parent cell - production of collagen/mucin/keratin/hormones etc

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

describe the features of a benign neoplasm

A

localised, non invasive.
slow growth rate, low mitotic activity.
closely resembles normal tissue.
necrosis/ulceration rare.

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

how might benign neoplasms cause problems?

A
pressure on adjacent structures.
obstruction of flow.
production of hormones.
transformation to malignant neoplasm.
anxiety.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

describe the features of a malignant neoplasm

A

invasive.
metastases.
rapid growth rate.
variable resemblance to normal tissue - closer resemblance = better prognosis.
poorly defined/irregular border.
hyperchromatic nuclei, increased mitotic activity.
necrosis/ulceration common.

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

how might malignant neoplasms cause problems?

A
destruction of adjacent tissue.
metastases.
blood loss from ulcers.
obstruction of flow.
hormone production.
paraneoplastic effects.
anxiety.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

what is a papilloma?

A

benign tumour of non-glandular, non-secretory epithelium.

prefix with cell type of origin.

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

what is a squamous cell papilloma?

A

benign tumour of squamous cell (non-glandular, non-secretory) epithelium

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

what is an adenoma?

A

benign tumour of glandular or secretory epithelium.

prefix with cell type of origin.

43
Q

what is a colonic adenoma?

A

benign tumour of colonic epithelium

44
Q

how would you refer to a benign tumour of thyroid epithelium?

A

thyroid adenoma - benign, secretory/glandular epithelium

45
Q

what is a carcinoma?

A

malignant tumour of epithelial cells.

prefix by name of epithelial cell type.

46
Q

what is an adenocarcinoma?

A

malignant tumour of glandular epithelium

47
Q

how are benign connective tissue neoplasms named?

A

cell of origin, suffix -oma

48
Q

what is a lipoma?

A

benign neoplasm of adipocytes

49
Q

what is a chondroma?

A

benign neoplasm of cartilage

50
Q

what is an osteoma?

A

benign neoplasm of bone

51
Q

what is an angioma?

A

benign vascular neoplasm

52
Q

what is a rhabdomyoma?

A

benign neoplasm of striated muscle

53
Q

what is a leiomyoma?

A

benign neoplasm of smooth muscle

54
Q

how are malignant connective tissue neoplasms named?

A

‘sarcoma’ prefixed by cell type of origin

55
Q

what is a liposarcoma?

A

malignant tumour of adipose tissue

56
Q

what is a rhabdomyosarcoma?

A

malignant tumour of striated muscle

57
Q

what is a leiomyosarcoma?

A

malignant tumour of smooth muscle

58
Q

what is a chondrosarcoma?

A

malignant tumour of cartilage

59
Q

what is an osteosarcoma?

A

malignant tumour of bone

60
Q

what is an angiosarcoma?

A

malignant tumour of blood vessels

61
Q

what does anaplastic mean?

A

cell-type of origin of a carcinoma/sarcoma is unknown

62
Q

how are carcinomas/sarcomas further classified, apart from by cell type of origin?

A

according to degree of differentiation

63
Q

what is screening?

A

process of identifying apparently healthy people who may be at risk of a disease or condition - then offering info, tests and/or treatment

64
Q

what are the four principles of screening to be considered in developing a screening programme?

A
  1. knowledge of disease - important condition, recognisable early/latent stage, course of disease understood.
  2. knowledge of test - suitable and acceptable test. continuous case finding.
  3. treatment for disease - acceptable, available
  4. cost considerations - cost of screening balanced by savings
65
Q

what is meant by the “sensitivity” of a screening test?

A

how successful was the test at identifying all those with cancer/condition screened for?
true positives divided by true positives + false negatives

66
Q

what is meant by the “specificity” of a screening test?

A

how well did the test identify all those without cancer/condition screened for?
true negatives divided by false positives + true negatives

67
Q

what is meant by the “positive predictive value” of a screening test?

A

how accurate was the selection of cases believed to have the condition screened for?
true positives divided by sum of true and false positives
i.e. what percentage of positive results were accurate

68
Q

what is the incidence rate of a disease?

A

the number of new cases per population at risk in a given time period

69
Q

what is the mortality rate of a disease?

A

measure of the number of deaths due to a specific cause in a population, per unit of time

70
Q

describe some limitations of screening

A
  1. reduces risk of developing condition, but doesn’t offer guaranteed protection.
  2. false positives/negatives
  3. misinterpretation/false results leading to false sense of security
  4. cost to society/individuals - funds directed away from other services
71
Q

what cancers commonly spread to bone?

A

breast, prostate, lung, thyroid, and kidney

72
Q

what sort of cancers are treated well by conventional chemotherapy? list some.

A
fast dividing tumours:
germ cell tumours of testis;
acute leukaemias;
lymphomas;
embryonal paediatric tumours;
choriocarcinoma
73
Q

what are the issues with conventional chemotherapy? what side effects does this commonly cause?

A

not selective for tumour cells.
hits normal cells which are dividing:
myelosuppression; hair loss; diarrhoea

74
Q

what is targeted chemotherapy?

A

exploits some difference between cancer cells and normal cells to target drugs specifcally at the cancer cells - more effective, fewer side effects

75
Q

describe the ‘two-hit hypothesis’ of cancer

A

first ‘hit’ is inheritance of a defective allele of a tumour suppressor gene, the other allele being normal.
second ‘hit’ is acquired mutational loss of function of normal allele - cell has lost all tumour suppressive function.

76
Q

what are tumour suppressor genes? how are they classified?

A

genes that inhibit carcinogenesis.

divided into ‘gate-keepers’ and ‘caretakers’

77
Q

describe how caretaker tumour suppressor genes act

A

maintain the integrity of genome by repairing DNA damage

78
Q

describe how gatekeeper tumour suppressor genes act

A

inhibit the proliferation of, or promote death of, cells with damaged DNA

79
Q

name some examples of tumour suppressor genes

A

gatekeepers - p53, APC

caretakers - BRCA1, BRCA2

80
Q

what is the p53 gene?

A

gatekeeper tumour suppressor gene.

transcription factor that responds to DNA damage - mutated in c.50% of human cancers

81
Q

where is the p53 gene located?

A

short arm of chromosome 17

82
Q

what are the normal functions of p53 protein?

A

enable repair of damaged DNA before S phase in cell cycle, by arresting cycle in G1 until damage is repaired.
enable apoptotic cell death if there is extensive DNA damage.

83
Q

what is the important consequence of loss of function of p53?

A

cells with damaged DNA (inc. mutated oncogenes or TSGs) undergo mitosis instead of apoptosis.

84
Q

how are oncogenes classified? (5 groups)

A

growth factors;
receptors for growth factors;
signalling mediator with tyrosine kinase activity;
signalling mediator with nucleotide binding activity (disrupting intracellular signalling);
nuclear-binding transcription factor oncoproteins - involved in regulation of cellular proliferation

85
Q

how may oncogenes be activated?

A

by mutation;

by excessive production of a normal oncoprotein

86
Q

list a few oncogene examples and how they function

A

sis - growth factor (PDGF)
erb-B - receptor (for EGF)
src - intracellular signalling (protein-tyrosine kinase)

87
Q

define a somatic mutation

A

mutations that develop after conception in any cell of the body, and are passed down only to daughter cells of that original cell

88
Q

define a germline mutation

A

germline mutations are present in the germ cells at birth and are passed down from generation to generation

89
Q

list factors that influence tumour invasion

A

decreased cellular adhesion.
secretion of proteolytic enzymes.
abnormal/increased cellular motility.

90
Q

what is the role of matrix metalloproteinases in neoplastic invasion?

A

secreted by malignant neoplastic cells, enabling them to digest surrounding connective tissue

91
Q

what counteracts the action of matrix metalloproteinases?

A

tissue inhibitors of metalloproteinases (TIMPs) - net effect determined by balance of TIMPs and metalloproteinases.

92
Q

what is “pagetoid infiltration”?

A

invasion within epithelium

93
Q

what is the significance of invasion?

A

most important criterion for malignancy.

metastases are a consequence of invasion.

94
Q

what is metastasis?

A

the process of malignant tumours spreading from the primary tumour to form other, secondary tumours at distant sites

95
Q

define carcinomatosis

A

extensive metastatic disease

96
Q

what are the 6 steps involved in the metastatic sequence?

A
  1. detachment of tumour cells from neighbours
  2. invasion of surrounding connective tissue to reach blood/lymph vessels
  3. intravasation into lumen of vessels
  4. evasion of host defences (e.g. NK cells in blood)
  5. adherence to endothelium at remote location
  6. extravasation of cells from vessel lumen into surrounding tissue
97
Q

describe the possible routes of metastasis

A
  1. haematogenous - by blood stream
  2. lymphatic - secondary tumours in regional lymph nodes
  3. transcoelomic - in pleural/pericardial/peritoneal cavities - neoplastic effusion
  4. implantation - accidental spillage of tumour cells in surgery
98
Q

what types of metastatic spread do carcinomas and sarcomas prefer, respectively?

A
carcinomas = lymphatic
sarcomas = haematogenous
99
Q

describe what the term “tumour grade” means

A

an assessment of the degree of malignancy or aggressiveness of a tumour -assessed histologically

100
Q

list the most important features contributing to tumour grade

A

mitotic activity.
nuclear size, hyperchromasia and pleomorphism.
degree of resemblance to normal tissue (i.e. differentiation)

101
Q

what does the “tumour stage” describe?

A

the extent of spread

102
Q

what do the letters and numbers stand for in the TNM staging system?

A

T = primary tumour. number indicates tumour size/local extent. number varies with organ harbouring tumour.
N = lymph node status. number indicates number of lymph nodes/groups of nodes containing mets.
M = anatomical extent of distant metastases
TNM is then used to derive a stage score.

103
Q

what is meant by a “stage 1” or “stage 4” cancer?

A

varies with type of cancer. derived from TNM status.
stage 1 = confined to organ of origin.
stage 4 = disseminated widely.