1/144
A comprehensive set of vocabulary flashcards covering wound healing, inflammation, cancer biology, DNA repair, epigenetics, infectious cancer, and therapeutic concepts drawn from the lecture notes.
Name | Mastery | Learn | Test | Matching | Spaced |
---|
No study sessions yet.
HIP-R
Mnemonic for the four wound-healing phases: Haemostasis, Inflammation, Proliferation, Remodelling.
Haemostasis
Wound-healing phase 0–20 min: vasoconstriction, platelet plug formation, coagulation to form a fibrin clot.
Inflammation
Wound-healing phase 0–6 days: neutrophils (0–48h) followed by macrophages (48–96h) with cytokines IL-1, TNF-α, IL-6, NO, prostaglandins.
Proliferation
Wound-healing phase 4–14 days: fibroplasia (fibroblasts → ECM Type III collagen), VEGF-driven angiogenesis, re-epithelialization (keratinocyte migration; EMT to MET).
Remodelling
Wound-healing phase 8 days–1 year: Type III collagen replaced by stronger Type I; MMPs remodel ECM; end when Type I:III ~4:1.
Neutrophils
First responders in inflammation; perform phagocytosis; typical window 0–48 hours.
Macrophages M1
Inflammatory macrophages that promote debris clearance and host defense.
Macrophages M2
Healing macrophages that promote tissue repair and regeneration.
Cytokines IL-1, TNF-α, IL-6
Key inflammatory mediators driving vasodilation, leukocyte recruitment, and fever/acute phase responses.
Nitric oxide (NO)
Cytokine/vasodilator involved in inflammation and antimicrobial activity.
Prostaglandins
Inflammatory mediators produced during the inflammatory response; contribute to vasodilation and pain.
Fibroplasia
Proliferation of fibroblasts producing extracellular matrix during wound repair.
ECM Type III collagen
Initial collagen in wound repair; later replaced by Type I collagen during remodelling.
Angiogenesis
New blood vessel formation; VEGF-driven process during wound healing and tumor growth.
VEGF
Vascular Endothelial Growth Factor; central driver of angiogenesis.
Re-epithelialization
Migration of keratinocytes to restore epidermal layer; may involve EMT/MET transitions.
EMT
Epithelial-to-mesenchymal transition; increases motility and invasiveness.
MET
Mesenchymal-to-epithelial transition; re-establishment of epithelial traits.
MMPs
Matrix metalloproteinases; degrade ECM during remodeling; TIMPs inhibit MMPs.
PRISH
Signs of inflammation: Pain, Redness, Immobility, Swelling, Heat.
PAMPs
Pathogen-associated molecular patterns detected by pattern-recognition receptors (TLRs).
DAMPs
Damage-associated molecular patterns detected by receptors (NLRs); trigger inflammation.
TLRs
Toll-like receptors that recognize PAMPs and activate innate immunity.
NLRs
Nod-like receptors that recognize DAMPs and initiate inflammatory responses.
VEC-CD-P
Sequence mnemonic for inflammation: Vasodilation, Edema, Chemotaxis, Cytokine amplification, Destruction, Proliferation prep.
Vasodilation
Early inflammatory response increasing blood flow to the site.
Edema (permeability ↑)
Fluid leakage into tissues due to increased vascular permeability.
Chemotaxis
Recruitment of neutrophils and other immune cells via cytokines.
Cytokine amplification
Amplified signaling (e.g., IL-1, IL-6, TNF) driving the inflammatory response.
Destruction (phagocytosis, NETs)
Removal of pathogens by phagocytes; neutrophil extracellular traps (NETs) may form.
Proliferation prep (M2 macrophages, growth factors)
Preparation of tissue for repair; M2 macrophages secrete pro-regenerative signals.
Mast cells
Cells releasing histamine and prostaglandins to promote inflammation.
Histamine
Vasoactive mediator released by mast cells; promotes vasodilation and permeability.
Dendritic cells
Antigen-presenting cells that activate T cells and bridge innate/adaptive immunity.
Neutrophils (first responders)
Rapid responders in acute inflammation; primary phagocytes.
Macrophages (M1/M2)
M1: inflammatory; M2: healing/regeneration macrophages.
Complement C3a/C5a
Anaphylatoxins that promote inflammation and recruit immune cells.
Complement C3b
Opsonin that enhances phagocytosis of pathogens.
MAC (C5b-C9)
Membrane attack complex; forms pore in target cell membranes leading to lysis.
Complement system
A system of plasma proteins that enhances inflammation and immunity via opsonization, chemotaxis, and lysis.
Inflammation sequence (VEC-CD-P) – activators
Activators include VEGF, TNFα, PDGF, TGFβ promoting angiogenesis and repair.
Angiogenesis in cancer: angiogenic switch
Tumors shift from dormant to vascularized by releasing VEGF (often via MMP activity).
Tumor vasculature abnormalities
Chaotic, leaky vessels with reduced pericytes and abnormal basement membranes.
Warburg effect
Cancer cells preferentially use glycolysis for energy even in the presence of oxygen.
PET scans with FDG
Imaging using 18F-FDG to detect high glucose uptake by tumors.
Immune surveillance
Theory that immune system detects/eliminates transformed cells; supported by cancers in immunosuppressed.
Tumor antigens: TSA vs TAA
TSA: tumor-specific antigens; TAA: overexpressed/aberrant self-antigens.
CD8+ T cells
Cytotoxic T cells that kill target cells via FASL or perforin/granzyme.
CD4+ T cells
Helper T cells secreting cytokines (IL-2, IFN-γ) and activating other immune cells.
NK cells
Natural killer cells that kill cells with low MHCI or stress ligands.
MHC I and MHC II
MHC I presents to CD8+ T cells; MHC II presents to CD4+ T cells.
Immune evasion (tumors)
Strategies include reduced MHCI, decreased stress receptor expression, anti-apoptosis, soluble FASL, and complement inhibitors.
Invasion & metastasis
Breach of basement membrane; routes include lymphatic, hematogenous, transcoelomic, implantation.
Metastatic cascade steps
Detachment, ECM adhesion, protease secretion, intravasation, survival, extravasation, colonization.
EMT in cancer
Epithelial cells acquire mesenchymal traits, gaining motility and invasiveness.
HIF-1α
Hypoxia-inducible factor; stabilized in hypoxia and upregulates VEGF.
Angiogenesis in cancer – regulators
Activators: VEGF, TNFα, PDGF, TGFβ; Inhibitors: Thrombospondin, Endostatin, TIMPs, IL-4.
Cancer stem cells
Self-renewing, tumor-initiating subpopulation linked to EMT and metastasis.
SASP
Senescence-associated secretory phenotype; SASP factors recruit immune cells and remodel ECM.
Telomere shortening (Hayflick limit)
Limit on cell divisions leading to senescence unless telomerase reactivates.
Immortalisation
Escape from senescence via loss of p53/Rb or activation of telomerase.
Telomerase
Ribonucleoprotein that maintains telomeres; promotes replicative immortality.
Crisis in immortalization
Severe telomere shortening causes genome instability; cells may die or reactivate telomerase.
DNA damage and repair overview
Cells suffer numerous DNA lesions daily; unrepaired damage leads to senescence, apoptosis, mutations, cancer.
BER
Base Excision Repair; fixes small non-bulky lesions; enzymes: glycosylase → AP endonuclease → polymerase → ligase.
NER
Nucleotide Excision Repair; removes bulky UV/chemical lesions; ~30 nt segments.
HR
Homologous recombination; accurate DSB repair using sister chromatid; BRCA1/2, RAD51 involved.
NHEJ
Non-homologous end joining; error-prone DSB repair; can cause mutations; linked to SCID when faulty.
MMR
Mismatch Repair; correcting replication errors; key proteins MSH, MLH.
BRCA1 regulates repair signaling; BRCA2 recruits RAD51 for HR; mutations linked to HBOC.
Epigenetics
Heritable gene expression changes without DNA sequence change; includes DNA methylation and histone modification.
DNA methylation
Methylation of CpG islands in promoters; generally represses gene expression.
Histone modifications
Post-translational modifications (acetylation, methylation, phosphorylation) modulating chromatin.
Cross-talk: DNA methylation and histones
Methylated DNA recruits HDACs leading to deacetylation and gene silencing.
Epigenetics in cancer – predisposition to mutations
Global hypomethylation promotes genome instability; hypermethylation silences tumor suppressors.
Epigenetic therapy
Therapies targeting epigenetic marks: DNMT inhibitors, HDAC inhibitors, BRD4 inhibitors, IDH inhibitors.
DNA demethylating agents
Agents like 5-azacytidine, decitabine used to reactivate silenced genes.
HDAC inhibitors
Drugs like Vorinostat; re-expression of silenced genes via histone hyperacetylation.
BRD4 inhibitors
Epigenetic therapy targeting bromodomain proteins (e.g., JQ1) to disrupt gene regulation.
IDH1/2 inhibitors
Target mutant IDH enzymes affecting epigenetic state and tumor metabolism.
Inherited cancer syndromes
Genetic predisposition patterns: Sporadic, Familial, Inherited; examples include HBOC (BRCA1/2), HNPCC (MLH1/MSH2), Li-Fraumeni (TP53), MEN2 (RET).
BRCA1/BRCA2 roles
DNA damage response; BRCA1 links damage sensors to repair; BRCA2 recruits RAD51 for HR.
HBOC
Hereditary breast and ovarian cancer syndrome; BRCA1/2 mutations confer risk.
HNPCC (Lynch)
Hereditary nonpolyposis colorectal cancer; MLH1/MSH2 etc.; DNA Mismatch Repair defects.
Paraneoplastic syndromes
Systems-wide effects caused by cancer (hormonal/immune) not due to local tumor.
Tumor markers
Substances produced by tumors or in response to tumors; used mainly for monitoring (e.g., PSA, AFP, CEA, CA 19-9, CA 125).
Molecular diagnostics in cancer
Tools like FISH, PCR, sequencing (NGS); also liquid biopsy for noninvasive tumor DNA.
Oncogenic viruses – HPV
DNA virus; high-risk types (HPV 16/18) cause cervical/anal/oropharyngeal cancers; E6 inhibits p53, E7 inhibits Rb; vaccines Gardasil, Cervarix.
EBV (Epstein-Barr Virus)
DNA virus linked to Burkitt lymphoma and nasopharyngeal carcinoma; establishes latency in B cells.
HHV-8 (KSHV)
Kaposi’s sarcoma herpesvirus; associated with Kaposi sarcoma and Castleman disease; LANA proteins modulate host pathways.
HBV/HCV and cancer
HBV integration and chronic inflammation promote hepatocellular carcinoma (HCC); HCV linked via chronic inflammation.
HTLV-1
Retrovirus causing adult T-cell leukemia/lymphoma; Tax oncoprotein activates oncogenes.
HIV and cancer
Indirect carcinogenesis via immunosuppression; AIDS-defining cancers include Kaposi sarcoma, CLL/NHL.
Antitumor immunity – CD8+ T cells
Cytotoxic T cells killing tumor cells via perforin/granzyme or FasL pathways.
Checkpoint inhibitors
Immunotherapy blocking CTLA-4 or PD-1/PD-L1 to unleash T cells against cancer.
Monoclonal antibodies (mAbs)
Target tumor antigens (e.g., Rituximab targets CD20); can block receptors or recruit immune effectors.
CAR-T cells
Engineered T cells with tumor-specific receptors; highly effective in some B-cell cancers; early-stage.
Oncolytic viruses
Engineered viruses that selectively infect and kill tumor cells (e.g., PVSRIPO).
Non-specific cancer therapies
Surgery, radiotherapy, and conventional chemotherapy that affect both cancer and normal cells.
Surgery (cancer)
Goal: remove tumor mass; may remove micrometastases in select cancers.