• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/204

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

204 Cards in this Set

  • Front
  • Back
Clonality
the entire population of cells within a neoplasm arises from a single cell
- assessed by examining cells in women who are heterozygous for polymorphic X-linked markers (G6PDH)
Choristoma (Heterotopia)
an ectopic rest of normal tissue
ex. neural tissue in the scalp
Hamartoma
a mass of disorganized but mature specialized cells indigenous to a particular site
- developmental abnormalities but not neoplasms
Benign Neoplasms
() cohesive, expansile masses
() remain localized to site of origin
() well differentiated
() slow growth
Malignant Neoplasms
() invasive
() poorly differentiated or undifferentiated
() may grow rapidly
() 'cancer'
Desmoplasia
the formation of an abundant collagenous stroma in response to tumor growth and invasion
Differentiation
the extent to which neoplastic cells resemble comparable normal cells
Anaplasia
lack of differentiation
Morphologic Features of Malignant Neoplasms
() pleomorphism - variation in cell size and shape
() abnormal nuclear morphology - hyperchromasia and inc. nuclear size
() mitoses - b/c rapidly dividing
() loss of cell polarity
() tumor giant cells
() necrosis - b/c they outgrow their blood supply
Dysplasia
() disordered growth
() falls short of malignancy - no invasion
() loss of cell uniformity and architecture
() full-thickness dysplasia = carcinoma in-situ
() may progress to cancer
Metastasis
tumor implant that is discontinuous w/ the primary tumor
Direct seeding
penetration of tumor into a natural open space (peritoneal, pleural, pericardial cavities..)
Lymphatic spread
epithelial malignancies
Hematogenous spread
mesenchymal malignancies
Adenoma
benign epithelial neoplasm
Carcinoma
malignant epithelial neoplasm
Mesenchymal neoplasms
() Benign - end in -oma
() Malignant - end in -sarcoma
Teratoma
germ cell tumors
-blastomas
embryonal tumors
tumors of children, derived from immature cells
Wilms tumor (kidney)
Ewing sarcoma (bone)
Wilms tumor (kidney)
Ewing sarcoma (bone)
Grade
expression of the level of differentiation of a malignant neoplasm (G I-G IV)
the better differentiated the tumor (G I) the better the prognosis
grade correlates with the aggressiveness of the neoplasm
Stage
extent of size and spread of cancer within the patient.
the lower the stage, the better the prognosis
stage dictates therapy
TNM system
For staging:
T = primary tumor size and extent of invasion
N = regional lymph node involvement
M = metastases

looks at size, spread, and metastases
Fine-needle aspiration (FNA)
*can't tell if lesion is invasive or not
used most commonly for readily palpable lesions (thyroid, lymph nodes)
Is the prognosis for a receptor-positive tumor better or worse?
better prognosis
How many mutations are usually needed for a cell to be transformed into a cancer cell?
a minimum of 5 or 6
7 Critical features common to different cancers
() they stimulate their own growth
() they ignore growth-inhibiting signals
() they avoid death by apoptosis
() they develop a blood supply: angiogenesis
() they metastasize
() they replicate continuously
() they evade or outrun the immune response
Tumor rejection Ag's
specific to individual tumors and prevent infections transfer of tumors from one individual to another
Tumor specific Ag's
peptides from mutated cellular 'self' proteins, will then be recognized as being foreign rather than self
Tumor associated Ag's
arise by reactivation of embryonic genes or by overexpression of normal self proteins
3 Mechanisms by which tumors escape immune recognition:
1) low immunogenecity - no MHCs, adhesion molecules, or co-stimulatory molecules
2) antigenic modulation - immune selection of Ag-loss variants
3) tumor-induced immune suppression - factors secreted by the tumor (TGFb) inhibit T cells directly* (most effective)
3 Mechanisms by which tumors escape immune recognition:
1) LOW IMMUNOGENICITY - no MHCs, adhesion molecules, or co-stimulatory molecules
2) ANTIGENIC MODULATION - immune selection of Ag-loss variants
3) TUMOR-INDUCED IMMUNE SUPPRESSION - factors secreted by tumor cells (TGFb) inhibit T cells directly* (most effective)
What percent of lung cancers are associated w/ smoking?
90%
*smoking is associated w/ depression
Common Serum Tumor Markers:
() Prostate Specific Ag (PSA) - for prostate cancer
() Carcinoembryonic Ag (CEA) - for colorectal and pancreatic cancers
() Ca-125 - for ovarian cancer
Neoplasm
an abnormal tissue mass whose growth exceeds and is uncoordinated with that of adjacent normal tissue and persists after cessation of the stimuli that provoked it
General progression of breast cancer:
Normal breast --> Atypical hyperplasia (lobar or ductal) --> carcinoma in situ --> invasive breast cancer --> metastatic breast cancer
Carcinoma in situ
full-thickness dysplasia extending from the BM to the surface epithelium.
applicable only to epithelial neoplasms.
if entire lesion is CIS then risk of metastasis = 0 (b/c no blood vessels above the BM)
Gross Features of Malignancy:
() Irregular borders
() Necrosis
() Metastasis
Disruption of what molecular structure serves as a hallmark for transformation?
disruption of actin filaments
allows for tumor migration and metastasis
What characteristic will be seen with hematogenous spread?
venous invasion of a sarcoma following the venous drainage from the site of the tumor
Where do each of the following cancers tend to spread? Breast, Prostate, Colon
Breast --> lungs
Prostate --> bone
Colon --> liver and pancreas
Where do most genetic changes happen in the development of cancer?
@ the transition from a normal cell --> atypical hyperplasia
*very early
Promotion of progression to invasive breast carcinoma:
Survival when invasive cells separate from the tumor mass
Increased resistance to apoptosis or anoikis
Increased motility
Ability to form motility structures such as lamellipodia, filopodia, membrane ruffles
Ability to relax the constraints of the actin-based cytoskeleton
Ability to activate signals that regulate focal adhesion/contacts
Increased invasive potential
Ability to form podosomes or invadopodia
Upregulate activity of cell surface proteases
Activation of kinase signaling cascades
Receptor tyrosine kinases
ErbB2, EGFr
cMet
VEGFr family
Nonreceptor kinases
Fak
Src
PKC
Angiogenesis pathways
PI3K  Akt  Hif1  VEGF
Tumor Microenvironment Changes
Loss of cell-cell tumor suppressor signals
e.g., loss of TGF from the stroma
Signaling Pathways promoting progression to invasive breast cancer:
() MAP Kinase Cascade - increasing cell proliferation
() VEGF - increasing angiogenesis, proliferation, motility, and invasion
() Rac/Rho - increasing motility
() MMPs and cell surface proteases - increasing invasiveness
Haploinsufficiency
with tumor suppressor genes, one fxnal allele is not enough. two alleles are required for normal fxn, otherwise --> transformation
RAS family of GTP binding proteins
*most common known oncogene in human tumors
considered a signal-transducing oncoprotein
contains: KRAS, NRAS, HRAS
GAP
GTPase-activating proteins - inactivate RAS via GTP hydrolysis
Neurofibromatosis 1
() caused by a mutation in neurofibromin 1, a GAP
Why are breast cancer death rates on the decline?
earlier diagnosis
What is key to making cancers hard to treat with pharmacotherapeutics?
As cancers progress they become more heterogenous
What type of growth fraction makes tumors generally more sensitive to anti-cancer drugs?
High growth fractions
When are tumors most sensitive to treatment?
when they are small and more actively growing
*as a tumor gets larger, the percent of cell mass that is dividing decreases
What is released in tumor lysis syndrome?
IC CaPO4, K, and Urea --> renal failure and coagulopathies
Myelosuppression is normally subject to..
dose limiting toxicity
How do some cancers regulate the MDR transporter?
the MDR transporter is a PGp that normally pumps foreign particles or toxins back into the intestinal lumen for excretion. Some drugs cause the PGp to see them as toxins, upregulating the transporter and in turn making the host more resistant to these drugs.
How do alkylating agents work?
they cause a DNA strand break
Chronic Myeloid Leukemia (CML):
9:22 translocation --> oncogenic tyrosine kinase and uncontrolled cell proliferation
Top 3 Cancer by incidence in men and women:
Men = prostate, lung, colon

Women = breast, lung, coon
Top 3 Cancers by death in men and women:
Men = lung, prostate, colon

Women = lung, breast, colon
What proportion of men will develop cancer at some point in their life?
1/2
What proportion of women will develop cancer at some point in their life?
1/3
What percent of cancer pts (all cancers) will live 5+ years?
70%
The decrease in mortality of breast cancer is due to:
90% of pts w/ breast cancer reach 5+yrs. This is due in part from:
() better understanding
() early detection
() effective tx
Genetic risk factors for cancer:
() FHx of cancer
() Cancer appears earlier in life
() Multiple and bilateral tumors
() May include rare tumor types
Known causes of cancer:
() Occupational exposure - asbestosis, mesothelioma, benzene
() Lifestyle factors - smoking, diet (#2 cause of cancer), EtOH
() Biologic agents - infections (HPV, HIV, EBV, Hepatitis)
() Iatrogenic
What is the worldwide #1 cause of cancer?
Hepatitis
Smoking-related cancers:
Lung
Larynx
Oral cavity
Esophagus
EtOH-related cancers:
Esophagus
Head and neck
Large bowel
Liver
Pancreatic
Breast
Ionizing radiation, an iatrogenic contributor to cancer, usually cause which types of cancer?
Breast and Leukemia
What factors are characteristic of cancer cells?
() loss of contact inhibition
() increase in GF secretion
() increase in oncogene expression
() loss of tumor suppressor genes
() neovascularization
Breast cancer screening recommendations:
CBE
() 20-40 = every 3 years
() 40+ = every year

Mammography - 1st by age 40
() 40-49 = every 1-2 yrs
() 50+ every year
Colon cancer screening recommendations:
() yearly fecal occult blood test (FOBT)* or fecal immunochemical test (FIT)
() flexible sigmoidoscopy every 5 years
() yearly FOBT* or FIT plus flexible sigmoidoscopy every 5 years**
() double-contrast barium enema every 5 years
() colonoscopy every 10 years
Cervical cancer screening recommendations:
() All women should begin cervical cancer screening about 3 years after they start having vaginal intercourse, but no later than when they are 21 years old.
Systemic cancer therapy:
() Chemotherapy - cytotoxic agents
() Hormonal therapy - mainly for prostate and breast cancers
() Targeted Txs - not cytotoxic, herceptin (trastuzumab), Imatinib
Most lymphomas are of what type?
B cell lymphomas
What lymphomas are TdT(+)/TdT(-)?
TdT(+) = precursor lymphomas

TdT(-) = mature lymphomas
Polyclonality
Multiple cells divide, giving rise to a population of cells w/ phenotypic and/or molecular differences
*tend to be reactive
Monoclonality
Single cell gives rise to the cell population w/out phenotypic and/or molecular differences
*tend to be malignant
Autonomy
Growth and proliferation outside of the normal body regulatory pathways - independent or non-regulated growth

*Malignancy = Monoclonality + Autonomy
Mantle Cell Lymphoma (MCL) translocation:
11:14 --> overexpression of Cyclin D1 --> uncontrolled proliferation
Follicular Lymphoma (FL) translocation:
14:18 --> overexpression of BCL2 --> anti-apoptotic
Burkitt Lymphoma (BL) translocation:
8:14 --> overexpression of MYC --> uncontrolled proliferation
Risk factors for B lineage lymphomas:
() Primary Immunodeficiency Syndromes
() Autoimmune Disorders
() Acquired Immunodeficiency Syndromes
Prolymphocytes and paraimmunoblasts:
seen in lymph node proliferation
What CDs are normally present on B cells?
19,20,22
Which has a better prognosis in CLL/SLL pts, those w/ a non-somatic mutation or a somatic mutation?
Those w/ a somatic mutation have a much better survival (20yrs) than those with a non-somatic mutation.
This is b/c those w/ a non-somatic mutation will take their hit after the germinal center stage (bad).
Richter Syndrome
Seen in 2-8% of those with CLL/SLL, is a transformation to Diffuse Large Cell Lymphoma
CD10
acts as a follicular marker in Follicular Lymphoma
Diffuse Large B cell Lymphomas:
Can be
Primary - De novo
Secondary - as a result of a transformation
What are the 3 flavors of Burkitt's Lymphoma?
Sporadic
Endemic
Immunodeficiency Associated
The epinym CRAB in plasma cell myeloma stands for:
hyperCalcemia
Renal insufficiency/failure
Anemia
Bone lesions, multifocal osteolytic
What Sx's are seen at diagnosis of T cell lymphomas?
Cytokine related Sx's:
() hypercalcemia - osteoclast activating factor
() hemophagocytic syndromes - malignant T cells tell monocytes to eat 'self'
Types of T cell Lymphomas:
() Leukemic or Disseminated
() Cutaneous
() Nodal Based
() Extranodal Based
What is the diagnostic triad for Sezary Syndrome and what separates it from advanced mycosis fungoides?
Diagnostic Triad
() Erythroderma = Red Man
() Lymphadenopathy
() Circulating Tumor Cells in Blood

Sezary Syndrome develops the triad instantaneously while the triad is seen as acme of progression with Mycosis Fungoides
What are the 4 clinical pictures of T cell lymphomas and their respective survival rates?
() Acute - survival 2wks-1yr
() Lymphomatous - survival 2wks-1yr
() Chronic - >2yrs survival
() Smoldering - >2yr survival
What end up causing the death of those w/ T cell leukemia/lymphoma?
Opportunistic pathogens:
Pneumocystis carinii
Cryptococcal Meningitis
Herpes Zoster, Disseminated
Hypercalcemia
Anaplastic Large Cell Lymphoma:
ALK(+) as well as CD30(+)
Reed Sternberg Cells
A minority of the cells seen in Hodgkin Lymphomas that secrete chemokines and cytokines that attract the inflammatory cell background that makes up the majority of the tumor (but are non-neoplastic)
What are the Hodgkin subtypes and how are they distinguished?
() Nodular Lymphocyte Predominant Hodgkins - looks like Hodgkins under the microscope but show presence of B cell gene expression w/ immunophenotyping - presence of OCT2, BOB1, absence of CD15, CD30
() Classical Hodgkins - looks like Hodgkins under the microscope and lacks B cell gene expression w/ immunophenotyping (like the other B cell lymphomas)
What 2 Lymphomas have a tendency to involve the EBV?
Burkitts and Hodgkins Lymphoma
What are the 4 subtypes of Classical Hodgkins Lymphoma?
() Nodular Sclerosis - dense pink bands of fibrosis surrounding tumor nodule, usually mediastinal with lacunar variant RS cells
() Mixed Cellularity - NS - fibrosis, male predominance, high EBV rate
*background for both NS and MC contain a host of immune cells
() Lymphocyte Rich - male predominance, only lymphocytes in background
() Lymphocyte Depleted - male predominance, very high EBV and HIV rates, RS cell or variant fails to secrete chemokines/cytokines --> rather pure population and poor ability to address
What are the B symptoms?
() fever
() night sweats
() weight loss
Auer rods
characteristic in acute myeloid leukemia (AML)
Oral targeted therapy works best for remission of what type of lymphoma?
Chronic Myeloid Leukemia (CML)
Imatinib
a protein tyrosine kinase inhibitor used effectively when the philadelphia chromosome is present (t9:22) - CML
What are the common Myeloproliferative neoplasms and what mutation is present?
() Polycythemia vera
() Essential thrombocytosis
() myelofibrosis
*each has a point mutation leading to constitutively active JAK2 --> GF INDEPENDENT PROLIFERATION
Myc Oncogene
Normally, Myc acts as a TF that enables immediate, high rate proliferation. Continuous expression of Myc is seen in tumor cells, most notably Burkitt's Lymphoma (t8:14)
Retinoblastoma
() autosomal dominant trait
() Rb usually fxns as a regulator of cell cycle by binding to E2F. When phosphorylated by cyclin D/E/.., or neutralized in the case of HPV and similar viruses, E2F is free to activate transcription of S phase genes (cyclin E)
Loss of heterozygosity
refers to the loss of the second allele, normal allele --> the cell now being homozygous for the mutant allele --> cancer development
quiescence vs senescence
quiescence = TEMPORARY cell cycle arrest
senescence = PERMANENT cell cycle arrest
Fxns of p53:
() activating quiescence
() DNA repair - initiated by ATM, ATR. p53-dep transcription of p21 (inhibits cyclin-CDK complexes from phosph Rb). p53 upregulation of mdm2 --> cell cycle block released
() inducing senescence
() triggering apoptosis - if DNA repair fails, p53 induces pro-apoptotic genes BAX and PUMA
() TF --> Mir34 (binds and inhibits pro-proliferative genes like cyclins, and anti-apoptotic genes like BCL2, preventing translation)
Li-Fraumeni syndrome
autosomal dominant inherited gene defect in p53 --> sarcoma
Familial Adenomatous Polyposis (FAP)
mutation in Adenomatous Polyposis Coli (APC) gene, a tumor suppressor --> adenomatous polyps in the colon
*both copies of APC gene must be lost for tumor formation
APC fxn
() APC normally complexes with B-catenin --> ubiquitinization
() Wnt stimulation breaks up this complex, allowing B-catenin to translocate to the nucleus where it stimulates proliferation
*If APC is mutated or absent, destruction of B-catenin does not occur and cells proliferate as if they were under continuous Wnt signaling
Contact inhibition and the normal response to cell injury
Normally, B-catenin binds to the cell surface protein E-cadherin. Loss of cell-cell contact (injury) disrupts the interaction btw B-catenin and E-cadherin allowing B-catenin to translocate and stimulate proliferation.
*loss of contact inhibition is a key characteristic of carcinomas
p16/INK4 and p14/ARF
tumor suppressors that share a genetic locus (CDKN2A)
mutation at this locus affects both p53 and Rb
() p16/INK4 - blocks cyclinD/CDK2 phosph or Rb
() p14/ARF - binds and inhibits MDM2, preventing p53 destruction
TGF-b and it's dual fxns:
() tumor preventative - inhibits proliferation in normal cells by repressing cMyc or by activating Rb
() tumor promotion - promotes immune system suppression and angiogenesis
*net action depends on location of pathway interruption
PTEN (phosphatase and tensin homologue)
a tumor suppressor that downregulates pro-growth PI3K/Akt signaling
Cowden syndrome
an autosomal dominant mutation in PTEN --> uncontrolled PI3K/Akt pro-growth signaling
- benign tumors (hamartomas)
- malignant tumors
- macrocephaly
- mucocutaneous lesions = facial trichilemmomas, papillomatous papules, and mucosal lesions
Tuberous Sclerosis
Normally, Akt inactivates the tumor suppressors TSC1/TSC2 which are in place to inhibit the mTOR pathway which stimulates uptake of nutrients needed for growth.
Suppression of the tumor suppressors TSC1/TSC2 --> hamartomas and other tumors
von Hippel-Lindau (VHL)
A tumor suppressor that binds hydroxylated HIF1a (hypoxia inducible TF 1a) in oxygenated tissues --> ubiquitinization
In ischemic tissue, HIF1a is not hydroxylated and thus doesn't bind VHL, instead activating transcription of genes for angiogenesis (VEGF, PDGF)
Wilms' tumor
a pediatric kidney cancer associated with mutation in the tumor suppressor WT1
these children also at a higher risk for aniridia
Gorlin syndrome
basal cell carcinomas alongwith jaw keratocysts and distinct dysmorphic features and skeletal anomalies stemming from a mutation in the tumor suppressor PTCH which normally regulates TGF-b, PDGFRa, PDGFRb pathways
Mechanisms for evasion of apoptosis:
() loss of sensors of genomic integrity - p53
() overexpression of inhibitors - BCL2
() reduced level of component proteins - decreased CD95/Fas, increased FLIP (binds death-inducing signaling complex and prevents activation of caspase 8)
What upregulated enzyme allows cancer cells to have limitless replicative potential?
telomerase - normally absent in somatic cells
- shortened telomeres --> nonreplicative senescence
Angiogenic switch
Angiogenesis is needed for tumor growth and metastasis.
The increased production of angiogenic factors (VEGF, FGF) and/or the loss of angiogenic inhibitors (p53) --> angiogenesis.
This is normally stimulated by hypoxia through HIF1a which activates VEGF and FGF
What goes on in the 2 major phases of invasion and metastasis?
() invasion of ECM
() vascular dissemmination, homing of tumor cells, and colonization
Hereditary Nonpolyposis Colon Cancer Syndrome (HNPCC)
an inherited defect in the DNA mismatch repair genes (MLH1, MSH2)
*each individual inherits one defective copy and acquires the 2nd hit in colonic epithelial cells
() microsatellite instability - hallmark of HNPCC, variability in length of repeat regions caused by insufficient proofreading during replication
Xeroderma Pigmentosum
an inherited defect in the DNA nucleotide excision repair system --> increased risk for skin cancer
- UV light causes cross-linking in pyrimidine residues, preventing normal DNA replication
The Warburg Effect:
a metabolic shift in cancer cells to aerobic glycolysis that allows for visualization via PET scanning
miRNAs
small noncoding, single-stranded RNAs
silences mRNA which can down or upregulate tumorigenic factors
Carcinogenic Initiators vs Promoters
Initiators - cause rapid, irreversible DNA damage, not sufficient for tumor formation, may be direct acting or indirect acting (requiring metabolic conversion or not)
Promoters - changes are reversible and don't directly affect DNA, capable of inducing tumors in initiated cells but not by themselves
Aflatoxin B1
a food contaminant associated w/ hepatocellular carcinoma that produces a characteristic 'signature' mutation in p53
HTLV1 (Human T cell Leukemia Virus Type 1)
a T cell leukemia/lymphoma caused by HTLV1 integration into the T cell (CD4+) genome --> monoclonal neoplastic T cell population
Epstein Barr Virus
may induce B cell lymphomas (esp. Burkitt's lymphoma) by facilitating acquisition of t(8:14)
may also lead to self-limited polyclonal B cell proliferation - infectious mononucleosis
Helicobacter pylori
known to cause gastric B cell lymphomas - MALTomas, via stimulation of chronic inflammation and polyclonal B cell proliferation (at first T cell dependent)
The presence of an Auer rod signify that it is what type of leukemia?
myeloid
What are the 2 major groups of AML?
() Not Otherwise Specified
() Recurrent Cytogenic Abnormalities
What type of cells stain (+) for MPO-SBB?
neutrophils
What type of cells stain (+) for NSE?
monocytic cells
M5a vs M5b
M5a - monoblastic, effects young ppl
M5b - monocytic, effects adults (50yrs+)
You get a call from a dentist who has a pt w/ swelling, bleeding gums. What leukemia do you suspect?
AML class subtype M5a - monoblastic or M7 - megakaryoblastic
M6a vs M6b
M6a - Erythroleukemia, >50% erythroid cells, >20% of non-erythroid are myeloblasts, MPO(+), NSE(-)
M6b - Pure Erythroid, >80% erythroid, no myeloblast increase, MPO(-), NSE(-)
Dry Taps may result in which leukemias?
() Primary myelofibrosis
() Hairy Cell Leukemia
() AML-M7
t(8:21) is associated with what AML subtype?
M2
t(15;17) is associated w/ what AML subtype?
M3
t(9;11) is associated w/ what AML subtype?
M5
inv(16) is associated w/ what AML subtype?
M4
A pt comes in and presents with DIC and a hypergranular cell that has many Auer rods. What is your diagnosis?
The pt has Acute Promyelocytic Leukemia (M3). They will have the t(15;17) PML/RARa and 'Faggot' cells upon further testing.
Myeloid vs Lymphoid Acute Leukemias
Myeloid
most of the cases
usually children
auer rods
CD13, CD33, CD34
MPO, NSE
Lymphoid
usually children under 6yo
TdT(+)
CD19/22 (B cell)
CD3, CD2 (T cell)
What is the main difference in presentation btw Precursor B and T Lymphoblastic Leukemia/Lymphoma?
B = blood and bone marrow presentation
T = tissue base presentation
A few common benign tumors:
() Hemangiomas
() Lymphangiomas
() Fibrous tumors
() Teratomas
Most common teratoma of childhood?
Sacrococcygeal teratoma - usually benign
* more common in girls
* most benign teratomas seen in younger infants
WAGR Syndrome
WT1 defect
Aniridia
Genital anomlies
mental Retardation
Beckwith-Wiedemann Syndrome
() enlargement of body organs, macroglossia, hemihypertrophy, omphalocele, adrenal cytomegaly
() involves WT2 gene
Denys-Drash Syndrome
() gonadal dysgenesis and nephropathy
() germline abnormalities in WT1
Asymptomatic pathogenic bacteria of the nasopharynx
Strep pyogenes
Strep pneumoniae
Staph aureus
Small round blue cell tumors:
Neuroblastoma
Lymphoma
Rhabdomysarcoma
Ewing sarcome/PNET
Clinical presentations of neuroblastoma w/ consideration to age
children < 2yrs of age
Lg abdominal mass
Fever
Possible wt loss

Children > 2yrs of age
May present w/ metastatic disease : bone pain, resp sxs, GI complaints
What prokaryote is unique in lacking a rigid layer of peptidoglycan and also has sterols?
mycoplasma
Pseudomembranous colitis
caused by opportunistic pathogenesis of Clostridium difficile
Lysozyme
a component of tears, cleaves the glycan in gram (+) peptidoglycan cell walls
Capsule
() protects against phagocytosis - gives virulence
() protects against drying out
Mycoplasmas vs Mycobacteria
Mycoplasmas - don't have cell walls

Mycobacteria - thick, waxy cell walls; use acid-fast stain
What enzymes are missing in obligate anaerobes?
SOD* and catalase
auxotrophs
strains of bacteria that need an added growth factor
prototroph
can make everything it needs from glucose and inorganics (ex. E. coli)
Virulence factors possessed by bacteria:
() type III secretion effectors (molecular syringes) - mainly gram (-)
() hemolysin (esp. beta) - in group A strep
() secretory IgA proteases - in respiratory pathogens
2^10 =
~ 10^3
Fluoroquinolone
targets bacterial DNA gyrase, disabling its ability to put in negative supercoils to allow replication/transcription. This leads to double strand breaks --> killing
How do bacteria prevent multiple initiations in their DNA replication?
There is a delay in methylation of the new strand (by DAM) that allows the replication machinery (DNA pol III) to check the newly added base.
What 2 enzymes are needed for a bacteria to grow on lactose?
lactose permease (lacY)- transports lactose into the cell

b-galactosidase (lacZ)- cleaves lactose to a dissaccharide
What are the components of the lac operon and how do they fxn?
() structural genes (lacZ, lackY) - encode the enzyme to cleave lactos --> monosacchardes and a lactose transporter, respectively
() promoter - promotes transcription of the above structural genes
() operator - is activated by the promoter to do the work
() regulatory gene (lacl) = REPRESSOR - binds next to promoter to inhibit RNA pol from binding
How do cAMP and glucose play in (+) feedback of the lac operon?
*in the presence of glucose, very little cAMP is made, inhibiting the synthesis of b-galactosidase (not needed at the time)
w/ no glucose, cAMP levels increase, binding to CRP which helps the promoter and activates RNA pol so b-galactosidase is made = (+) FEEDBACK
Describe the 2-component regulatory system:
Component #1 - a membrane bound sensor binds to a signaling molecule, causing dimerization and activation of internal kinase --> self-phosphorylation

Component #2 - Response Regulator - is phosphorylated by cognate kinase to form a dimer active for DNA-binding. This activated dimer may now activate or repress the DNA.

*2 component systems are impt for virulence and usually signal arrival in a host or compartment in a host
Attenuation
control of transcription after initiation which responds to the level of Trp in the cell and is quieted by conditional termination
What dictates the growth rate of bacteria?
# of ribosomes
S20 and translational repression:
S20 is one of the proteins involved w/ forming the 30S ribosomal subunit.
When translated, it can either bind to rRNA (if present) of, if not present, bind to and repress it's own mRNA = autoregulation
Silent, Missense, Nonsense, and Frameshift mutations:
Silent = mutation in base doesn't change amino acid, due to redundancy of genetic code

Missense = mutation in base causes an amino acid substitution that is accepted and allows the protein to still fxn as well as wild type

Nonsense = a mutation in the base results in transcription of a STOP CODON, usually results in complete loss of gene fxn
Auxotroph
deals w/ nutrition
mutant that cannot make an essential small molecule and won't grow w/out it

contrasted w/ prototrophs which make all the basic 'building blocks'
Complementation
an extra copy of some bacterial DNA is carried on a plasmid (small, circular dsDNA molecule) which can make up if a mutation is present

This also is targeted in producing avirulent bacteria, where both virulent genes need to be destroyed
The low rate of mutation in bacterial DNA is a result of:
1) base complementarity - assessed during insertion of the base
2) proofreading or editing fxn of DNA pol III - occurs just after base is inserted
3) mismatch repair - a mismatch distorts the dbl helix and is recognized via methyl-directed mismatch repair and fixed w/ DNA pol III

*overall frequency of spontaneous mutation = 3(10^-3) = 10^-9/generation
Reversion
change in a phenotype from mutant back to wild type as a result of a mutation (auxotrophs undergoing a mutation that allows them to once again be prototrophs)
Why are amino-penicillins considered broad spectrum? (ampicillin, amoxicillin)
they possess amino groups that allow penetration of the outer membranes, making it useful against gram (-) bacteria as well as gram (+)
Do penicillins work on dormant, static bacteria?
No
Antibiotic resistance
diminishing efficacy caused by decreased microbial susceptibility to abx
penicillin-binding proteins (PBPs)
are transpeptidases
PBP1 - binds penicillins
PBP2a
PBP3 - binds cephalosporins

If an organism loses a PBP, then those drugs that would usually bind and interfere w/ the transpeptidase are ineffective

*if only PBP2a = MRSA
penicillinase
aka b-lactamase
possessed by bacteria resistant to the b-lactams (overcome by cephalosporins)
MIC (minimum inhibitory concentration)
the lowest concentration of an abx that causes a significant reduction in the growth of a pt derived strain of pathogen (static)

*MIC_90 is the increased MIC common to 90% of all pt isolates
MBC (minimum bacteriocidal concentration)
the lowest concentration of an abx that will inhibit growth of a clinical isolate, even after subculture (cidal)

*favored for tx of endocarditis, osteomylitis, and infections w/ underlying immunocompromised conditions
Breakpoints S,I,R for an abx
S = susceptible
I = intermediate
R = resistant

reflect
1) pharmacological properties of abx
2) the pts disease
3) the microbe and its properties
AST
antibody susceptibility test
What is the classic example of "cidal-static" antagonism?
penicillin (cidal) + tetracycline (static)
Bacterial Transformation
DNA from a bacterium, either chromosomal or plasmid, can present outside the cell (via lysis) and bind to specific surface proteins of another bacterium which incorporates it.
This occurs w/out cell-cell contact and is susceptible to DNase.
What bacterial species are naturally transformable?
() Neisseria
() Haemophilu
() S. pneumoniae
*all respiratory pathogens
Broad host range plasmid
plasmids (usually R plasmids) containing vectors for abx resistance which have an origin of replication that can fxn in many different bacterial species
Bacterial Conjugation*
the transfer of LINEAR plasmid DNA (F or R plasmid) btw bacteria using cell-cell contact (utilizing a pilus). The plasmid encodes the fxnal genes required for conjugation.

*main mechanism for spread of bacterial resistance
How do R plasmids acquire resistance genes?
Through the action of transposons.
Bacterial Transduction
transfer of DNA (usually for toxins) btw bacteria mediated by a bacterial virus (bacteriophage)
Phages
Phages - can be lytic (virulent) or lysogenic (temperate). this latter allows for the recipient bacteria to have the ability to express changed phenotype.
Specialized vs Generalized transduction
Specialized transduction - toxin gene is permanently assoc w/ the phage DNA

Generalized transduction - host DNA packaged into virus by mistake
Transposable elements and transposons
Transposable elements are DNA segments that can move. The simplest ones are called insertion sequences (IS) which consists of a gene for a transposase enzyme which catalyzes the transfer rxn and the flanking sites that mark the boundaries of the transferred DNA.

A transposon is one type of complex transposable elements which has 2 IS elements acting as one to move themselves plus the intervening DNA.
Salmonella typhimurium - sophisticated pathogen
• adsorbs to and travels through gut epithelium
• uses macrophages like taxis to travel in the body
• eventually grows in spleen and liver

• acquired by "horizontal transfer"
• Salmonella has 2 big ones:
SPI-1 required for growth in epithelial cells
SPI-2 required for growth in macrophages
Knudson's "two-hit" hypothesis
sporadic form - both mutations in Rb gene are acquired by the retinal cells after birth
familial form - all cell inherit 1 mutant Rb gene, the second mutation affects the Rb locus after birth