Comprehensive Study Notes: DNA, Chromatin, Replication, Transcription/Translation, Cell Cycle, ANS, Embryology, Cytogenetics, Metabolic Disorders, Pharmacology

DNA Structure and Nuclear Organization

  • Nucleolus: inside the nucleoplasm, the site of rRNA synthesis; rRNA combines with proteins to form ribosomes.

    • Ribosomal proteins are first synthesized in the cytoplasm by existing ribosomes.

    • Proteins imported into the nucleolus, where they join with newly made rRNA to form ribosomal subunits: 40S (small) and 60S (large).

    • Subunits are exported to the cytoplasm and remain free until they bind to mRNA to form a functional ribosome.

  • Fates of ribosomes after assembly in cytoplasm:

    • Free ribosomes: stay in cytoplasm and synthesize proteins used inside the cell.

    • Bound ribosomes: attach to rough ER (including outer nuclear membrane) and synthesize proteins destined for secretion, insertion into membranes, or to lysosomes.

  • Chromatin: DNA + histone proteins; two forms:

    • Heterochromatin: tightly packed; transcriptionally inactive.

    • Euchromatin: loosely packed; histone-DNA interaction weak enough to allow RNA polymerase transcription.

    • During cell replication, chromatin condenses into chromosomes to ensure accurate DNA transmission to daughter cells.

  • DNA (deoxyribonucleic acid): structure and building blocks

    • Repeating units: nucleotides (phosphate, sugar, nitrogenous base).

    • Nitrogenous bases: Purines (Adenine A, Guanine G) with two rings; Pyrimidines (Cytosine C, Thymine T) with one ring.

    • When base attaches only to sugar, it’s a nucleoside; when phosphate is added, it becomes a nucleotide.

    • DNA double helix: two antiparallel strands; complementary base pairing (A with T via 2 hydrogen bonds; C with G via 3 hydrogen bonds).

    • Nucleotide connectivity: phosphodiester bonds link the 5' carbon of one nucleotide to the 3' carbon of the next.

  • Phosphodiester bonds in DNA

    • Definition: Bonds that connect nucleotides together.

    • Bond formation: links the 5' carbon of one nucleotide to the 3' carbon of the next.

    • Result: continuous chain of nucleotides (a DNA strand).

    • Directionality: each strand has a 5' end and a 3' end; orientation is essential for polynucleotide synthesis.

  • Base pairing details

    • Phosphodiester bonds create the sugar-phosphate backbone (negatively charged).

    • A-T pairs via 2 hydrogen bonds; C-G pairs via 3 hydrogen bonds.

  • Double-stranded DNA structure and topology

    • Two strands paired together; antiparallel orientation: one 5'→3', the other 3'→5'.

    • Helical structure with bases packed inside and phosphate backbone outside.

    • Major and minor grooves serve as binding sites for proteins involved in transcription and replication.

  • Packaging of DNA: histones and nucleosomes

    • Histones are rich in positively charged amino acids (lysine, arginine) and bind the negatively charged DNA backbone.

    • DNA wraps twice around a histone core (octamer) composed of 2× each of H2A, H2B, H3, H4.

    • H1 histone secures DNA around the core, forming a nucleosome bead (~146 bp DNA).

    • Euchromatin vs heterochromatin in the nucleosome context; epigenetic regulation controls condensation and gene activity.

  • Epigenetic regulation: acetylation and methylation

    • Histone acetylation reduces positive charge on histones, loosening DNA and promoting transcription (euchromatin).

    • DNA methylation adds methyl groups to bases, often repressing gene expression; histone methylation has variable effects.

    • Epigenetics governs euchromatin vs heterochromatin states and gene expression without changing DNA sequence.

  • Telomeres and telomerase

    • Telomeres: protective repeats at chromosome ends; human telomeres consist of ~2500 TTAGGG repeats.

    • Function: telomeres protect coding DNA from loss during replication; they are sacrificed gradually as a buffer.

    • Telomerase: extends telomeres; active in stem cells and some gametes and cancer cells; limited activity in most somatic cells.

    • Shortening of telomeres links to aging; overactivity of telomerase is associated with cancer.

  • Progeria (clinical correlation)

    • Rare disease caused by LMNA gene mutation; defective lamin A disrupts nuclear envelope structure and chromatin organization, affecting euchromatin/heterochromatin regulation and epigenetic control.

DNA Structure Deep Dive: Key Concepts and Terminology

  • DNA nucleotide components: phosphate, deoxyribose sugar, nitrogenous base (A, G, C, T).

  • Purines vs pyrimidines: A/G are purines; C/T are pyrimidines.

  • Nucleoside vs nucleotide: nucleoside lacks phosphate; nucleotide includes phosphate.

  • Base pairing rules: A pairs with T (2 H-bonds); G pairs with C (3 H-bonds).

  • Directionality and antiparallel strands: 5'→3' vs 3'→5' orientation.

  • Phosphodiester bond: linkage between 5' phosphate and 3' sugar of adjacent nucleotides; backbone is negatively charged.

  • Telomere biology: TTAGGG repeats; role in genomic stability; telomerase in stem/cancer cells.

  • Epigenetic marks: DNA methylation (usually represses expression); histone acetylation (generally activates expression); histone methylation can activate or repress depending on context.

Fates and Function of Ribosomes

  • Ribosome assembly occurs in the nucleolus with rRNA and ribosomal proteins.

  • Ribosomal proteins synthesized in cytoplasm, imported into nucleolus, assemble with rRNA into 40S and 60S subunits.

  • Ribosome export to cytoplasm; final assembly occurs upon binding to mRNA to form complete ribosome.

  • Free ribosomes: synthesize intracellular proteins.

  • Bound ribosomes: synthesize proteins for secretion, insertion into membranes, or lysosomes.

Epigenetics and Chromatin Regulation

  • Euchromatin vs heterochromatin: transcriptionally active vs silent.

  • Epigenetic modifications:

    • Histone acetylation: promotes decondensation and gene expression.

    • DNA methylation: generally represses gene expression; histone methylation has context-dependent effects.

  • Clinical corollary: epigenetic regulation can influence disease, development, and aging processes (e.g., progeria links to chromatin structure).

Key Equations and Formulas (LaTeX)

  • Loading dose (accounting for bioavailability):
    ext{Loading dose} = rac{C{ ext{desired}} imes Vd}{F}

  • Maintenance dose (accounting for bioavailability and interval):
    ext{Maintenance dose} = rac{C_{ ext{desired}} imes CL imes au}{F}

  • Telomere repeat unit: TTAGGG

  • Base-pairing summary: A-T with 2 H-bonds; C-G with 3 H-bonds.

  • DNA backbone charge: phosphate groups are negatively charged.

DNA Replication: Core Concepts

  • Semiconservative replication: each daughter DNA contains one old and one new strand.

  • Origins of replication: multiple origins in eukaryotes to speed up copying of long genomes; A-T rich regions facilitate opening.

  • Replication fork: Y-shaped structure where DNA unwinds; helicase unzips; SSBPs keep strands apart; nucleases monitor exposed DNA.

  • Topoisomerases address supercoiling ahead of the fork by cutting and rejoining DNA strands; Top1 (no ATP, mainly eukaryotes), Top2 (ATP-dependent, both eukaryotes and prokaryotes), Top4 (prokaryotes only, separates linked circular DNA).

  • DNA replication enzymes (prokaryotes):

    • Primer synthesis: RNA primase; main DNA synthesis: DNA Polymerase III; primer removal and replacement: DNA polymerase I; ligation: DNA ligase; SSBPs stabilize single strands; helicase unwinds; nucleases remove damaged ssDNA.

  • DNA replication enzymes (eukaryotes):

    • Primer synthesis: DNA polymerase α with primase; main synthesis: DNA polymerase δ (lagging) and ε (leading); primer removal/replacement: DNA polymerase I (in combination with RNase H in places); ligation: DNA ligase; SSBPs stabilize; Topoisomerases relieve supercoiling.

  • Leading vs lagging strand synthesis:

    • Leading strand: continuous synthesis toward the replication fork; primer laid by primase; DNA Pol III (or δ); proofreading occurs 5'→3' polynomial activity.

    • Lagging strand: discontinuous Synthesis in short Okazaki fragments; RNA primers laid repeatedly; DNA Pol I removes primers and replaces with DNA; ligase seals gaps.

  • Primers and proofreading:

    • RNA primers provide 3' OH for DNA polymerase extension.

    • DNA polymerase III proofreading via exonuclease activity; can backtrack and correct mistakes.

  • DNA replication inhibitors (cancer context): drugs can trap topoisomerases in cleaved state, leading to double-stranded breaks and cell death.

Transcription and RNA Processing

  • Central dogma: DNA → RNA (transcription) → Protein (translation).

  • Transcription overview (eukaryotes):

    • Initiation: RNA polymerase II binds promoter elements (TATA box, CAAT box); activators/enhancers can upregulate transcription.

    • Elongation: RNA polymerase moves 5'→3' adding nucleotides to the 3' end of RNA.

    • Termination: polyadenylation signals (AAUAAA) direct cleavage and release of mature RNA; in prokaryotes, Rho-dependent/independent termination mechanisms exist.

  • Prokaryotic vs eukaryotic termination:

    • Prokaryotes: Rho-dependent and Rho-independent termination mechanisms.

    • Eukaryotes: RNA polymerase II continues beyond the gene; mRNA processing signals mark cleavage and polyadenylation; no Rho factor.

  • Post-transcriptional processing (eukaryotes):

    • 5' cap (methylguanosine cap) added to 5' end; protects RNA and aids ribosome recognition.

    • 3' poly-A tail added to 3' end after polyadenylation signal; stabilizes mRNA.

    • Splicing: introns removed; exons joined to form mature mRNA.

  • mRNA fate in cytosol:

    • Translation into protein by ribosomes.

    • Degradation: some mRNAs marked for destruction; processing bodies (P-bodies) involved.

  • Translation: key steps

    • Initiation:

    • Prokaryotes: small subunit binds to Shine-Dalgarno sequence on mRNA; start codon AUG; formyl-methionine tRNA; large subunit joins.

    • Eukaryotes: small subunit binds 5' cap; scans to start codon AUG; methionine tRNA; large subunit joins.

    • Elongation: codon-by-codon tRNA delivery; peptide bonds form; chain elongated.

    • Termination: stop codon reached; release factor binds; protein released.

  • Transcription inhibition: RNA Polymerase II is highly sensitive to alpha-amanitin (mushroom toxin) which inhibits mRNA synthesis.

Protein Synthesis and Cellular Machinery (Overview)

  • Role of ribosomes in translation; basic steps and fidelity.

  • Post-translational considerations (not deeply covered in the slides but often implied in integrated notes).

The Cell Cycle and its Regulation

  • Phases: Interphase (G1, S, G2) and M phase (mitosis + cytokinesis). Some cells enter G0 (quiescent).

  • Checkpoints and regulators:

    • G1/S checkpoint: ensures readiness for DNA replication; key players include Cyclin D, CDK4/6; p53 and Rb help monitor DNA integrity.

    • S-phase: DNA replication occurs; S-CDK ensures replication happens once and only once.

    • G2/M checkpoint: ensures DNA replicated correctly before mitosis; genes and kinases regulate entry into mitosis.

    • Metaphase checkpoint: ensures all chromosomes are properly attached to spindle before anaphase.

  • Cyclins and CDKs:

    • Cyclins: D (G1), E (G1-S), A (S), B (M).

    • CDKs: always present; activity controlled by cyclins and other regulators.

    • Activation and inactivation controls: CAK (CDK-activating kinase) fully activates; Wee1 adds inhibitory phosphates; Cdc25 removes the inhibitory phosphate; CKIs can block activation.

  • The “Must-Knows”:

    • Cyclin D + CDK4/6 phosphorylates Rb, releasing E2F to initiate S phase.

    • p53 halts the cycle when DNA is damaged; can promote apoptosis if damage is irreparable.

    • S-CDK initiates DNA replication and prevents re-replication.

    • M-CDK drives mitosis; Wee1 inhibition and Cdc25 activation are necessary to proceed.

    • APC/C (anaphase-promoting complex) drives destruction of cyclin B and securin to finish mitosis.

  • Mitosis stages and cytokinesis (high level): Prophase, Metaphase, Anaphase, Telophase, Cytokinesis.

Nervous System and Autonomic Nervous System (ANS)

  • ANS overview:

    • Two-neuron chain in both sympathetic and parasympathetic divisions (preganglionic and postganglionic neurons).

    • Preganglionic neurons release ACh onto nicotinic receptors; postganglionic neurons usually release norepinephrine onto adrenergic receptors (exception: sweat glands use ACh).

  • Sympathetic division:

    • Preganglionic cell bodies in T1–L2 lateral horn; postganglionic cell bodies in paravertebral or prevertebral ganglia.

  • Parasympathetic division:

    • Preganglionic neurons originate in the brainstem (CN III, VII, IX, X) and sacral spinal cord (S2–S4); postganglionic neurons often release ACh on muscarinic receptors.

  • Key clinical entities:

    • Horner’s syndrome: ptosis, miosis, anhidrosis due to sympathetic pathway disruption.

    • Hirschsprung disease: congenital absence of enteric ganglion cells; failure to relax, proximal dilation; associated with Down syndrome; rectal suction biopsy shows absence of ganglion cells.

  • CNS integration of ANS:

    • Hypothalamus as primary regulator; receives input from solitary nucleus (visceral sensory input via Vagus) and higher brain regions (limbic system and cortex).

  • Synaptic transmission and receptors:

    • Postganglionic sympathetic neurons typically release norepinephrine acting on adrenergic receptors; sweat glands use ACh on muscarinic receptors.

    • Parasympathetic preganglionic neurons release ACh on nicotinic receptors; postganglionic release ACh on muscarinic receptors.

  • Practical pharmacology notes:

    • Atropine is a muscarinic antagonist; blocks rest-and-digest effects; increases heart rate by removing vagal tone; used to treat bradycardia and organophosphate poisoning.

    • Dale’s phenomenon: vasomotor reversal—after irreversible α-blockade, epinephrine can cause a drop in BP due to unopposed β2 vasodilation; clinical relevance in pheochromocytoma management.

Embryology and Development: Weeks, Germ Layers, and Morphogenesis

  • Fertilization and early development:

    • Fertilization occurs in the fallopian tube within 24 hours of ovulation; zygote forms, followed by morula, then blastocyst.

    • Trophoblast forms placenta; inner cell mass forms embryo.

  • Week-by-week development overview:

    • Weeks 1–2: fertilization, implantation, bilaminar disc; no teratogenic effects if exposure occurs during this “all-or-none” window (conceptus either dies or is unaffected).

    • Week 3 (gastrulation): formation of trilaminar disc—ectoderm, mesoderm, endoderm; neurulation begins later; somites form.

    • Week 4: folding of trilaminar disc; formation of trunk and organ primordia; neural tube forms.

    • Weeks 5–8: organogenesis; formation of organ systems; severe teratogenic effects if exposure occurs during this sensitive window.

    • Weeks 9–38: fetal period; organ maturation and growth; mostly physiologic/minor anomalies if teratogens present.

  • Germ layers and derivatives (summary):

    • Ectoderm: nervous system, skin, neural crest derivatives (peripheral nervous system, adrenal medulla, etc.).

    • Mesoderm: musculoskeletal system, cardiovascular system, kidneys, gonads, dermis; notochord remnants form nucleus pulposus.

    • Endoderm: GI and respiratory tract linings and glands (liver, pancreas, thyroid parafollicular cells, etc.).

  • Neural crest derivatives (extensive list): peripheral nervous system components, sensory ganglia, adrenal medulla, Schwann cells, melanocytes, odontoblasts, facial bones, etc.

  • Organogenesis gene families: Homeobox (HOX), Hox, Sox, Pax, Wnt, Hedgehog (SHH), TGF-β (including BMPs), FGF.

  • Teratogens and morphogenesis:

    • Teratogen timing matters: Weeks 1–2 all-or-none; Weeks 3–8 major malformations; Weeks 9–38 physiologic/minor anomalies.

    • Infections (TORCH), chemical agents (lithium, valproic acid, retinoic acid, thalidomide), substances of abuse (smoking, alcohol, cocaine).

    • Maternal diseases (diabetes, PKU) affect fetal development.

  • Types of morphologic anomalies

    • Malformation: intrinsic developmental defect (e.g., cleft lip, neural tube defects).

    • Disruption: extrinsic damage to a normally developing structure (trauma, amniotic bands).

    • Deformation: external mechanical forces alter development (uterine constraint, oligohydramnios).

    • Dysplasia: abnormal organization of cells within a tissue (e.g., skeletal dysplasias).

  • Sequences and syndromes:

    • Sequence: cascade of anomalies due to a single initiating event (e.g., Potter sequence from oligohydramnios).

    • Syndrome: multiple anomalies with a common etiology (e.g., Down syndrome).

  • Common congenital syndromes and chromosomal abnormalities (high-yield overview):

    • Down syndrome (Trisomy 21)

    • Edwards syndrome (Trisomy 18)

    • Patau syndrome (Trisomy 13)

    • Turner syndrome (45,X)

    • Klinefelter syndrome (47,XXY)

    • Other sex chromosome variations (46,XX,Y; 46,XY,del X; etc.)

  • Fertilization basics and ploidy

    • Gametes are haploid (1N); zygote is diploid (2N).

    • Meiosis I and II yield 23 chromosomes per gamete.

    • Mosaicism vs chimerism definitions and examples.

  • Practical clinical correlations

    • Folate (B9) deficiency linked to neural tube defects; importance of folic acid supplementation in pregnancy.

Cytogenetics and Chromosomal Abnormalities

  • Chromosome structure and organization

    • Centromere position classes: metacentric, submetacentric, acrocentric.

    • Banding techniques to detect structural abnormalities: G-banding is standard; Q-banding first; R-banding, C-banding, NOR staining provide additional details.

    • High-resolution banding in prophase/prometaphase for microdeletions.

  • Fluorescent in situ hybridization (FISH)

    • Uses fluorescent DNA probes to detect deletions, duplications, and rearrangements.

  • Numerical vs structural abnormalities

    • Numerical: gains or losses of chromosomes (aneuploidy); euploidy is normal multiples of 23; polyploidy is extra complete sets.

    • Structural: deletions, duplications, translocations (reciprocal or Robertsonian), rings, isochromosomes, etc.

  • Robertsonian translocations

    • Fusion of two acrocentric chromosomes at the centromere; long arms fuse; short arms lost.

  • Common genetic disorders and chromosomal etiologies (high-yield highlights)

    • Down syndrome: Trisomy 21; features include facial characteristics and risk for congenital heart disease.

    • Edwards syndrome: Trisomy 18; severe congenital anomalies; poor prognosis.

    • Patau syndrome: Trisomy 13; CNS and facial defects; poor prognosis.

    • Turner syndrome: 45,X; short stature, webbed neck, streak ovaries, cardiovascular anomalies.

    • Klinefelter syndrome: 47,XXY; hypogonadism, infertility, tall stature.

    • Other mosaicisms and sex chromosome abnormalities.

DNA Damage, Repair, and Genomic Integrity

  • DNA damage and sources

    • Free radicals and ROS, UV light causing pyrimidine dimers (thymine-thymine), spontaneous deamination (e.g., cytosine to uracil).

    • Chemotherapy agents deliberately damage DNA to trigger apoptosis.

  • Telomeres and telomerase (recap)

    • Telomeres protect coding DNA ends; telomerase extends telomeres in stem/germ/cancer cells; limited activity in most somatic cells.

    • Telomere shortening is associated with cellular aging; overactivity of telomerase can support cancer cell immortality.

  • DNA repair pathways (overview)

    • Nucleotide Excision Repair (NER): removes bulky lesions like pyrimidine dimers; active in G1; critical genes include endonucleases; XP (Xeroderma pigmentosum) is a defect.

    • Base Excision Repair (BER): fixes oxidative deamination and spontaneous base modifications; uses DNA glycosylase, AP endonuclease, lyase, DNA polymerase β, ligase.

    • Mismatch Repair (MMR): fixes mispaired bases post-replication; MutS/MutL detect; defects cause Lynch syndrome (HNPCC).

    • Double-strand break repair: Homologous recombination (HR) uses sister chromatid; nonhomologous end-joining (NHEJ) ligates ends, error-prone.

  • Clinical correlations and NBME-style associations

    • XP: defect in NER leading to extreme UV sensitivity and skin cancers.

    • Lynch syndrome: autosomal dominant MMR defects; high risk for colorectal and other cancers.

    • BRCA1/BRCA2 and HR defects elevate breast/ovarian cancer risk.

  • Key question patterns (high-yield themes)

    • Endonuclease cuts in NER during removal of pyrimidine dimers.

    • Telomerase activity and cancer relation: inhibition reduces replicative potential of tumor cells.

    • SSBPs prevent reannealing after DNA separation; their absence leads to fork collapse.

Metabolic and Genetic Disorders (Amino Acids and Teratogens)

  • Phenylketonuria (PKU)

    • Normal pathway: phenylalanine → tyrosine via phenylalanine hydroxylase, requires BH4.

    • PKU: deficient phenylalanine hydroxylase or BH4; phenylalanine accumulates; tyrosine becomes essential; musty odor; intellectual disability if untreated.

    • Treatment: low phenylalanine diet; supplement tyrosine; BH4 in deficiency.

  • Maple Syrup Urine Disease (MSUD)

    • Defect in branched-chain α-ketoacid dehydrogenase complex; accumulation of leucine, isoleucine, valine; urine smells like maple syrup.

    • Treatment: restrict branched-chain amino acids; thiamine supplementation may help some patients.

  • Albinism

    • Tyrosinase deficiency or melanin synthesis defect; pale skin, light hair/eyes; vision problems; increased skin cancer risk; autosomal recessive common.

  • Homocystinuria

    • CBS deficiency (requires B6/pyridoxine as cofactor) disrupts methionine to cysteine metabolism; accumulation of homocysteine; lens subluxation (downward), marfanoid habitus, thrombosis risk.

  • Alkaptonuria

    • Homogentisate oxidase deficiency; homogentisic acid accumulates; dark urine on standing; ochronosis (bluish-black pigment in cartilage); arthritis later in life; usually treated supportively.

Pharmacology and Therapeutics (Key Concepts from the NBME-style content)

  • Enzyme kinetics: competitive vs non-competitive inhibition

    • Competitive: inhibitor competes with substrate at active site; increases Km; Vmax unchanged; can be overcome by adding more substrate.

    • Non-competitive: inhibitor binds elsewhere; reduces Vmax; Km unchanged; not overcome by more substrate.

  • Dosing concepts

    • Loading dose depends on volume of distribution (Vd) and bioavailability (F):
      ext{Loading dose} = rac{C{ ext{desired}} imes Vd}{F}

    • Maintenance dose depends on clearance (CL) and dosing interval (τ):
      ext{Maintenance dose} = rac{C_{ ext{desired}} imes CL imes au}{F}

  • Bioavailability (F)

    • IV administration has F = 1 (100%); oral F < 1 due to first-pass metabolism.

    • When F decreases, both loading and maintenance doses must be adjusted to achieve the same plasma concentration.

  • Pharmacodynamic concepts: potency vs efficacy

    • Efficacy: maximum effect achievable by a drug; higher efficacy means taller dose-response curve.

    • Potency: amount of drug needed to reach a given effect; leftward shift indicates higher potency.

  • Cytochrome P450 (CYP450) inducers and inhibitors

    • Inducers: Rifampin, Carbamazepine, Barbiturates, St. John’s wort, chronic alcohol; increase metabolism, reduce drug levels.

    • Inhibitors: Cimetidine, Ciprofloxacin, Erythromycin, Ketoconazole, Amiodarone, Grapefruit juice; decrease metabolism, increase drug levels.

  • G-protein coupled receptor signaling (second messengers)

    • Gs: stimulates adenylyl cyclase → ↑ cAMP → PKA activation.

    • Gi: inhibits adenylyl cyclase → ↓ cAMP.

    • Gq: activates phospholipase C → IP3 + DAG → ↑ Ca2+ → PKC activation.

    • Common receptor-class mnemonic (QISS/QIQ): α1 (Gq), α2 (Gi), β1/β2 (Gs), M1 (Q), M2 (I), M3 (Q), V1 (Q), V2 (S).

  • Organophosphate poisoning

    • Irreversible AChE inhibitors leading to accumulation of ACh at muscarinic and nicotinic receptors; treatment with atropine (muscarinic antagonist) and pralidoxime (oxime) as antidote.

  • Adrenergic pharmacology and Dale’s reversal

    • After irreversible α-blockade (e.g., phenoxybenzamine), epinephrine can cause a paradoxical drop in BP due to unopposed β2-mediated vasodilation (Dale’s vasomotor reversal).

  • Other drug classes in the notes

    • PDE inhibitors: PDE-5 inhibitors (sildenafil, vardenafil, tadalafil) increase cGMP to promote vasodilation; PDE-3 inhibitors (milrinone) increase cAMP to enhance cardiac contractility and vasodilation; nonspecific PDE inhibitors (theophylline) for bronchodilation.

    • Atropine: muscarinic antagonist; used for bradycardia and ocular procedures; antidote in organophosphate poisoning.

    • Alpha blockers: prazosin, doxazosin (reversible), phentolamine (reversible nonselective), phenoxybenzamine (irreversible); used for hypertension, BPH, pheochromocytoma; Dale reversal relevance.

    • Neuromodulation and autonomic control: hypothalamic integration; parasympathetic/sympathetic balance; autonomic reflex pathways.

Imaging Modalities (Clinical Tools)

  • Computed Tomography (CT)

    • Uses X-ray photons; cross-sectional imaging via rotating X-ray tubes and detectors; assesses density differences; quick and widely available.

  • Magnetic Resonance Imaging (MRI)

    • Uses magnetic fields and radiofrequency waves; superior soft-tissue contrast; T1-weighted images highlight fat; T2-weighted images highlight fluids.

  • Ultrasound

    • Uses high-frequency sound waves; real-time imaging; safe; visualizes organs, soft tissues, and blood flow.

Practice Questions and Clinical Correlations (Representative Examples)

  • LMNA mutation in progeria-like syndrome reflects defective lamin proteins affecting nuclear envelope integrity and chromatin organization; this impacts euchromatin/heterochromatin balance and epigenetic regulation (Question 1 in the set).

  • Euchromatin vs. heterochromatin states are best described as: euchromatin = acetylated/histone modifications enabling transcription; heterochromatin = methylated and transcriptionally silent (Question 2).

  • Blocking ribosomal protein import into the nucleus halts ribosome assembly in the nucleolus (Question 3).

  • DNA melting temperature correlates with GC content: higher GC content (more H-bonds) yields higher Tm (Question 4).

  • Telomerase inhibition in dividing tumor cells leads to telomere shortening and reduced replication potential (Question 5).

Embryology and Teratology: Key Windows and Concepts

  • Teratogen windows of sensitivity

    • Weeks 1–2: all-or-none effect (death or no effect).

    • Weeks 3–8: major morphologic anomalies (organogenesis).

    • Weeks 9–38: physiologic or minor anomalies.

  • TORCH infections and congenital defects: Toxoplasmosis, Other agents, Rubella, Cytomegalovirus, Herpes; each has characteristic fetal effects.

  • Critical gene families in development (transcription factors and signaling): Homeobox, Hox, Sox, Pax, Wnt, Hedgehog (SHH), TGF-β (BMPs), FGF.

  • Morphogenesis errors and congenital anomalies

    • Malformation, disruption, deformation, and dysplasia definitions with examples.

    • Sequences and syndromes: Potter sequence; Down syndrome; other common associations.

  • Cytogenetics in clinical practice

    • Karyotyping and chromosomal analysis; use of FISH and various banding methods; Robertsonian translocations and translocation carriers.

Quick References and High-Yield Takeaways

  • Base pairing rules and the chemistry of DNA structure are foundational for replication and transcription.

  • The cell cycle is tightly controlled by cyclins and CDKs; checkpoints ensure genome integrity.

  • The ANS relies on a two-neuron chain; neurotransmitter identity drives the response in target tissues; pharmacology heavily leverages receptor pharmacodynamics.

  • Teratogens exert stage-specific effects; folate supplementation is crucial during weeks 3–8 to mitigate neural tube defects.

  • Genetic diseases often arise from enzymatic defects (PKU, MSUD, albinism, homocystinuria, alkaptonuria) or chromosomal abnormalities (Down, Edwards, Patau, Turner, Klinefelter).

  • NBME-style pharmacology patterns emphasize: loading dose depends on Vd; maintenance dose depends on clearance; changes in bioavailability alter both doses accordingly.

Notes on Notation and LaTeX Usage

  • Equations are presented in LaTeX format for clarity:

    • Loading dose: ext{Loading dose} = rac{C{ ext{desired}} imes Vd}{F}

    • Maintenance dose: ext{Maintenance dose} = rac{C_{ ext{desired}} imes CL imes au}{F}

    • Telomere repeats: ext{TTAGGG}

  • All chemical and genetic notation is kept in standard scientific format for consistency with exam content.

If you’d like, I can tailor these notes further into a shorter study guide focused on specific topics you’ll be tested on, or extract a subset of high-yield questions with concise explanations for quick review.