Shared Flashcard Set

Details

Pathology
Path Exam Second Half
72
Medical
Graduate
05/20/2012

Additional Medical Flashcards

 


 

Cards

Term
How are acute and chronic inflammation involved in the foreign body response?
Definition
Similar cells and chemical mediators are employed in both inflammation and FBR.
These include cytokines, PMN, macrophages, lymphocytes, and fibroblasts
Term
What are the molecular modifiers of the foreign response?
Definition
Growth factors such as PDGF, FGF, TGFβ > proliferation of fibroblasts, endothelial
cells, and specialized fibrogenic cells

Cytokines such as TNF, IL-1, IL-4, IL-13 > increased collagen synthesis > fibrosis
Term
Give an overview of the sequence of the inflammatory response
Definition
1. vasoconstriction
2. vasodilation (histamine, NO)
3. increased permeability
4. edema
5. decreased blood flow (hypotension)
6. neutrophils (roling, adhesion, transmigration, chemotaxis)
7. phagocytosis
3 outcomes
1. chronic inflammation (angiogenesis, mononuclear cell infiltrate, scar)
2. resolution (resolvin, etc)
3. Scarring
Term
What are the roles of serotonin, prostaglandins, and Leukotrienes in Inflammation?
Definition
serotonin comes from platelets, and does vasodilation and increased permeability.

prostaglandins, from mast and leukocytes do vasodilation, pain and fever

leukotrienes come from mast and leukocytes and do chemotaxis, increased permeability, leukocyte adhesion and activation
Term
What are biomaterials?
Definition
natural or synthetic materials introduced into body as part of medical device
Term
What is the foreign body response?
Definition
body's reaction to introduction or presence of external object/ device

1. non-specific protein deposition in surface
2. recruitment/ interaction of inflammatory cells w/ coated surface
3. repair-like processes
4. foreign body giant cells (multinucleated macrophages fuse)
5. matrix production and encapsulation
Term
What are similarities and differences between wound healing and the foreign body response?
Definition
Similarity-- beginning steps similar - recruit platelets, neutrophils, fibroblasts, macrophages

Differences:
1. depostion of DENSE collagen in FBR (only some collagen syn in wound healing)
2. healing restores normal tissue arch. and vasculature- FBR collagen in parallel, fibrosis, avascular
Term
What are the pathways of the receptors w/ intrinsic tyrosine kinase activity?
Definition
PI3 kinase pathway, MAP-kinase pathway, IP3 pathway > Ca2+ release
Term
What are the pathways of the G-protein coupled receptors?
Definition
G proteins > cAMP
Term
What are the pathways of the receptors w/o intrinsic tyrosine kinase activity?
Definition
JAK/STAT > STATs direct binding to DNA
Term
What are the differences between autocrine, paracrine and endocrine signaling?
Definition
autocrine- cell responds to signals they secrete themselves

paracrine- cells in close proximity signal to neighboring cells

endocrine- synthesized by cells of endocrine organs- target cells distant (blood steam)
Term
What are the 2 main signal effects of growth factors (within the cell)?
Definition
1. stimulate transcription of many genes that were silent in resting cells

2. regulate entry into cell cycle
Term
What are stem cells? (name the liver, skin and skeletal stem cells)
Definition
cells that self renew and differentiate
(asymmetric or symmetric)

Liver= Oval
Skin= Bulge
Skeletal= satallite, satallite glial cells in PNS
Term
What is the difference between labile, quiescent and non-dividing?
Definition
labile = continuously divide eg. surface epithelia

quiescent= low replication rate, rapid w/ stimulus (eg. liver, kidney, pancreas)

Nondividing= senescence (eg. neuron, skeletal and cardiac muscle)
Term
Define regeneration and healing
Definition
regeneration= growth of cells to replace lost structures

healing= regeneration and laying down of fibrous tissue (scar formation)
-after MI can lead to constrictive pericarditis :(

(usually in response 3 injury- 1.wound, 2. inflammation, 3. necrosis)
Term
What is the difference between regeneration and healing?
Definition
regneration restores normal tissue structure while in healing there is a loss of ECM > involves fibrosis and scar formation
Term
What are growth factor functions?
Definition
proliferation, cell survival, locomotion, contractility, differentiation, angiogenesis
Term
Describe the unique angiogenic and fibrogenic response elicited by foreign bodies
Definition
Unique angiogenic response: vessels are pushed away from the encapsulated foreign
body (instead of penetrating the site as during normal inflammation)
b. Unique fibrogenic response: thick and tightly packed collagen network formed around the foreign body (see above for more details)
Term
What is the overlap between granulomas and the foreign body response?
Definition
In both granuloma formation and FBR, inflammatory cells accumulate at the site of foreign object, and fibrosis is initiated to wall off the object. The term granuloma is
commonly used to denote the encapsulation of a pathogen (bacteria or parasites),
whereas the term FBR is used to denote the reaction against artificially introduced devices/materials.
Term
What is a neoplasm?
Definition
New growth (cells growing autonomously, escaping normal feedback mechanisms, and the growth exceeds and is uncoordinated with that of the normal tissues). The growth continues after cessation of the stimuli
Term
What are the two basic components of both benign and malignant tumors?
Definition
1. clonal neoplastic cells (parenchyma)
2. reactive stroma of connective tissue, blood vessels, macrophages and lymphocytes
Term
How are hyperplasia, metaplasia and dysplasia different from neoplasia?
Definition
Hyperplasia may still be regulated by growth factors
Metaplasia is replacement of one type of cell with another type (usually there is still orderly growth and it can be reversible or self-limiting

dyplasia is simply disordered growth (loss of uniformity of individual cells and loss of architectural orientation)

neoplasm is an abnormal mass of tissue, the growth of which exceeds and is uncoordinated with that of the normal tissue and persists in the absence of stimulus.
Term
How do we name benign mesenchymal tumors (suffix?)
Definition
- oma eg. fibroma
Term
How do we name benign epithelial tumors (examples)?
Definition
adenoma - glandular tissue
papilloma- finger like projections
cytoadenomas- form cysts
polyp- projection above mucosal surface into lumen (eg. GI)
Term
How do we name malignant mesenchymal tumors?
Definition
Sacroma (eg. fibrosarcoma)
Term
How do we name malignant epithelial tumors?
Definition
carcinoma (eg. adenocarcinoma)
Term
Compare benign and malignant tumors on differentiation, rate of growth, local invasion and metastasis
Definition
Begnin- well differentiated, progressive/slow, do not invade or metastasize

Malignant- range of differentation (some lack = anaplasia), erratic rates (slow to rapid), locally invasive, frequently metastasize
Term
What is anaplasia?
Definition
Lack of differentiation
Term
How do we characterize tumors?
Definition
classification, grading, and staging
Term
What is classification of tumors?
Definition
Classifying type of cell that the tumor resembles (presumed origin of tumor)
Term
What is grading of tumors?
Definition
The degree of differentiation (1-5) with 1 being very differentated
Term
What is staging of tumors?
Definition
Based on the size of tumor (T), extent of spread to regional lymphnodes (N), presence or absence of metastasizes (M)
Term
What are some techniques for diagnosing cancer?
Definition
excision/ biopsy
fine needle aspiration
pap smear
immunohistochemistry
flow cytommetry
molecular diagnosis
Term
What determines the rate of growth of tumors?
Definition
doubling time of tumor cells, fraction of tumor cells in the replication pool, rate at which cells shed or die
Term
What are some of the clinical manifestations of neoplasia?
Definition
paraneoplastic syndromes (unexplained system complexes
tumor endocrine production
anemia
jaundice
ischemia
fever
masculinization
leucocytosis (increased number of WBC)
leucopenia (decreased number of WBC)
episodic flushing (caused by serotonin and bradykinin)
infection
hemorrhage
obstruction
pain
fracture
Death
Term
What are some serologic markers of tumors?
Definition
tumor associated enzymes
hormones (eg. Hcg, calcitonin)
specific proteins (immunoglobulins)
antigens
molecular markers (p53, RAS)
Term
Name two important oncogenes and explain their mechanism
Definition
RAS- G protein that when mutated is continuously in the active state, binding GTP. This leads to continued cell proliferation (via stimulation of regulators or proliferation like MAP kinase pathway)

MYC- controls transcription, mutation leads to increased histone acetylation and reduced cell adhesion, increased protein synthesis, and other changes that induce high rate of cell division.
Term
Name specific tumors in which MYC oncogene is involved
Definition
Burkitt's lymphoma (B cell tumor)
Breast, colon, lung cancer
Term
Name specific tumors in which RAS oncogene is involved
Definition
pancreatic and lung adenocarcinomas
colon and thyroid cancer
myeloid leukemias
Term
Name two important tumor suppresor genes and their mechanism
Definition
RB gene- (retinal blastoma) enforces the G1 phase which once crossed leads to obligatory mitosis

p53 gene- prevents propagation of genetically damaged cells, guards the G1-S phase transition, involved in activating DNA repair pathways
Term
Name specific tumors in which RB tumor suppressor gene is involved
Definition
retinoblastoma
glioblastomas
osteosarcomas
breast, bladder cancer
Term
Name specific tumors in which p53 tumor suppressor gene is involved
Definition
Virtually everything!
Term
Name 2 apoptosis regulating genes
Definition
BCL2- antiapoptotic
p53
Term
Name 2 DNA repair genes
Definition
BRCA1
BRCA2
--these are the DNA damage response network that repair intra and interstrand DNA crosslinks

--these are important in breast and ovary carcinoma
Term
Name the 7 physiologic activities required for malignancy
Definition
self-sufficiency in growth signals
insensitivity to growth inhibition
evasion of apoptosis
limitless proliferative potential
sustained angiogenesis
defects in DNA repair
ability to invade and metastasize
Term
How do tumors gain self-sufficiency in growth signals?
Definition
Through activation of oncogenes, which produce oncoproteins that are devoid of important regulatory elements

Additionally they do not depend on external signals of GFs
Term
How are tumor cells able to evade apoptosis?
Definition
Inactivate p53, activate antiapoptotic genes, reduce levels of FAS/CD95 receptors or have high levels of FLIP, which prevents activation of caspase 8.

The best established is to use BCL2 to prevent apoptosis
Term
How does a metastatic tumor invade the ECM?
Definition
1. Alterations in intercellular adhesion molecules.
2. Local degradation of the basement membrane and interstitial connective
tissue. (proteolytic enzymes secreted by themselves or induce stromal cells to elaborate proteases.
3. Changes in attachment of tumor cells to ECM proteins.
4. Locomotion
Term
Why is genomic instability considered the enabler of malignancy?
Definition
Basically lack of DNA repair leads to genomic instability, and defects in DNA repair contribute to most cancer!

(both copies of the DNA repair gene are lost usually but there is evidence that there is a subset of genes that can promote cancer in a haploinsufficient manner)
Term
What are the categories of genetic change that can activate oncogene or inactivate tumor suppressor genes?
Definition
Mutations, translocations, amplification, deletions, epigenetic regulation.
Term
Describe the rationale for activity of at least 2 molecularly targeted cancer drugs
Definition
Herceptin- monoclonal antibody that binds HER2 to block the signaling of the growth factor receptor

Gleevac- used in CML, tyrosine kinase inhibitor for the BCR/ABL tyrosine kinase which is constitutively on in CML.
Term
What is the Philidelphia chromosome?
Definition
Fused BCR/ ABL translocation, chimeric gene encodes a constitutively active oncogenic BCR/ABL tyrosine kinase! Leads to CML.
Term
Describe the concept of premalignancy
Definition
Premalignancy is a state in which the tissue is at risk of tumorgenesis due to genetic mutations, yet there is no cancer specific phenotypes (example: leukoplakia)

In FAP Familial Adenomatous Polyposis, the APC gene is mutated
Term
What are some agents that can cause cancer
Definition
Physical like UVB light, ionizing radiation
Chemical like carcinogens
Infectious agents, like HPV, Hep B
Term
What are some conditions that can lead to cancer
Definition
Chronic inflammatory conditions, genetic predisposition, tumorimmunoediting (changes in tumor surveillance), genetic diseases
Term
Compare and contrast acquired vs. germline cancer causing genetic mutations
Definition
They both can lead to tumorgenesis
Germline mutations put people one step ahead compared to acquired mutations, like shortened time
Term
Describe carcinogenesis in terms of initiation and progression
Definition
Initiation occurs when a chemical irreversibly damages DNA and the damage is non lethal, but these cells DO NOT progress to tumors

Progession is the next step that is needed to stimulate proliferaiton of mutant cells. These promoting agents are not tumorgenic by themselves
Term
What is a cancer stem cell?
Definition
A group of self-renewing cells that possess tumorogenic mutations
Term
Define epigenetics
Definition
Reversible heritable changes in gene expression that occur without mutation, for example histone acetylation and DNA methylation
Term
Describe how we use epigenetics for cancer diagnosis and therapies.
Definition
Cancer cells have abnormal epigenetic signatures

Theraputic targets include having inhibitors of DNA methylation and histone acetylation.
Term
Mechanisms that regulate tumor migration, invasion and metastasis!
Definition
Adhesion (cell-cell, cell-matrix)
Proteolysis (proteases and their inhibitors)
Migration (Roles of adhesion and proteolysis)
Tumor cell-endothelial cell interation
Angiogenesis
Term
What are the 5 categories of congenital anomalies?
Definition
malformation, disruption, deformation, sequences, syndromes
Term
What is malformation?
Definition
Results from intrinsically abnormal developmental process (eg. anencaphaly)
Term
What is disruption?
Definition
Results form extrinsic interference on previously normal development (eg. amnionic band can wrap around limbs and impair growth)
Term
What is deformation?
Definition
Localized or generalized compression of the growing fetus by abnormal biochemical forces leading to structural abnormalities (eg. clubbed feet resulting from decreased amnionic fluid and pressure from the uterine wall)
Term
What are sequences?
Definition
Its a cascade of anomalies triggered by one initiating aberration (any of the other congenital anomalies)
Term
What are syndromes?
Definition
Multiple congenital anomalies believed to be pathologically related and explained by a single localized initiating defect
Term
What is prematurity?
Definition
Gestational age less than 37 weeks
Term
What are the causes of prematurity?
Definition
1. preterm premature ruption of placental membranes (PPROM) usually caused by infection
2. Intrauterine infection
3. Uterine, cerviccal and placental structural abnormalities
4. Multiple gestation (twins!)
Term
What are the consequences of prematurity?
Definition
1. Hyaline membrane disease
2. Necrotizing enterocolitis
3. Sepsis
4. Intraventricular hemorrhage
5. Develpmental delay
Term
What is hyaline membrane disease?
Definition
reduced surfactant at premature birth > increased alveolar surface tension > hypoxemia and CO2 retention > accidosis > pulmonary vasoconstriction > pulmonary hypoperfusion > edothelial damage and pithelial damage > plasma leak into alveoli ? fibrin and necrotic cells (hyaline membrane)
Term
What is intraventricular hemorrhage
Definition
Loss of autoregulation of cerebral blood flow leads to hemorrhage into the ventricles
Supporting users have an ad free experience!