UNIT-II of Principles of Biotechnology | M.Sc. Biotech. & M.Tech. Biotech. Notes

📖 CHAPTER 1: Structure of DNA, RNA & Proteins

Physical and Chemical Properties

Structure of DNA (Deoxyribonucleic Acid)

Historical Background

  • 1869: Friedrich Miescher discovered "nuclein" from white blood cell nuclei
  • 1919: Phoebus Levene identified components - sugar, phosphate, nitrogenous bases
  • 1950: Erwin Chargaff's rules - [A] = [T] and [G] = [C]
  • 1952: Rosalind Franklin's X-ray crystallography (Photo 51)
  • 1953: James Watson & Francis Crick proposed double helix model (Nobel Prize 1962)

Chemical Components of DNA

1. Nitrogenous Bases

  • Purines (Double ring structure):
    • Adenine (A) - C₅H₅N₅
    • Guanine (G) - C₅H₅N₅O
  • Pyrimidines (Single ring structure):
    • Cytosine (C) - C₄H₅N₃O
    • Thymine (T) - C₅H₆N₂O₂ (unique to DNA)

Base Pairing Rules (Chargaff's Rules)

A ≡ T (2 hydrogen bonds)
G ≡ C (3 hydrogen bonds)

Purines always pair with Pyrimidines

2. Pentose Sugar

  • 2'-Deoxyribose (C₅H₁₀O₄)
  • 5-carbon sugar lacking hydroxyl (-OH) group at 2' position
  • Numbered as 1', 2', 3', 4', 5' to distinguish from base numbering

3. Phosphate Group

  • PO₄³⁻ (Phosphoric acid residue)
  • Links nucleotides via phosphodiester bonds
  • Connects 5' carbon of one sugar to 3' carbon of next sugar
  • Gives DNA its negative charge

Nucleotide Structure

  • Nucleoside = Base + Sugar (e.g., Adenosine, Guanosine)
  • Nucleotide = Base + Sugar + Phosphate (e.g., AMP, GMP, dATP)
  • Building block of DNA/RNA
  • Linked by 3'-5' phosphodiester bonds

🧱 DNA Structure Hierarchy

Nucleotide
(Base + Deoxyribose + Phosphate)
Polynucleotide Chain
(Multiple nucleotides linked by phosphodiester bonds)
Double Helix
(Two antiparallel polynucleotide strands)
Chromatin
(DNA + Histone proteins)
Chromosome
(Condensed chromatin during cell division)

Watson-Crick Double Helix Model

  • Two polynucleotide strands coiled around common axis (right-handed helix)
  • Antiparallel orientation: One strand 5'→3', other 3'→5'
  • Complementary base pairing: A with T, G with C (Chargaff's rules)
  • Sugar-phosphate backbone on outside (hydrophilic)
  • Nitrogenous bases on inside (hydrophobic), perpendicular to axis
  • Uniform diameter: 2 nm (20 Å)
  • Pitch: 3.4 nm (34 Å) - one complete turn
  • Base pairs per turn: 10 base pairs
  • Distance between base pairs: 0.34 nm (3.4 Å)
  • Major groove: Wide groove (22 Å)
  • Minor groove: Narrow groove (12 Å)

Forms of DNA

Feature B-DNA A-DNA Z-DNA
Helix direction Right-handed Right-handed Left-handed (zigzag)
Diameter 20 Å 23 Å 18 Å
Base pairs/turn 10 11 12
Helix pitch 34 Å 28 Å 45 Å
Major groove Wide, deep Narrow, deep Flat
Minor groove Narrow, shallow Wide, shallow Narrow, deep
Occurrence Most common (physiological conditions) Dehydrated DNA, RNA-DNA hybrids High GC content, specific sequences
Base position Nearly perpendicular Tilted ~20° Tilted away from axis

Physical Properties of DNA

  • Molecular Weight: Very high (millions to billions of Daltons)
  • Density: 1.7 g/cm³ (in CsCl density gradient)
  • Viscosity: High due to long polymer chains
  • Optical Properties:
    • UV Absorption: Maximum at 260 nm (due to aromatic rings in bases)
    • Hyperchromic effect: Increased UV absorption upon denaturation
    • Hypochromic effect: Decreased absorption in double helix
  • Solubility: Insoluble in alcohol, soluble in water
  • Stability:
    • Stable at neutral pH (7.0-7.5)
    • Depurination at acidic pH
    • Denaturation at alkaline pH or high temperature

Chemical Properties of DNA

  • Denaturation (Melting):
    • Separation of two DNA strands by breaking hydrogen bonds
    • Caused by: Heat, extreme pH, chemicals (urea, formamide)
    • Tm (Melting temperature): Temperature at which 50% DNA is denatured
    • Higher GC content → Higher Tm (3 H-bonds vs 2 in AT)
    • Monitored by hyperchromic shift at 260 nm
  • Renaturation (Annealing):
    • Reformation of double helix on slow cooling
    • Complementary strands re-associate
    • Basis of PCR and hybridization techniques
  • Hydrolysis:
    • Acid hydrolysis: Breaks glycosidic bonds (removes purines)
    • Alkaline hydrolysis: Breaks phosphodiester bonds (more effective on RNA)
    • Enzymatic hydrolysis: Nucleases (DNases) degrade DNA
  • Depurination: Loss of purine bases (A, G) at acidic pH
  • Oxidation: Damage by reactive oxygen species (ROS)

🎯 Key Points for Exams

  • CSIR NET Chargaff's rules: [A] = [T] and [G] = [C], but [A+T] ≠ [G+C]
  • ICAR SRF A-T has 2 hydrogen bonds, G-C has 3 hydrogen bonds
  • CSIR NET B-DNA: 10 bp/turn, pitch = 34 Å, diameter = 20 Å
  • ICAR SRF DNA absorbs UV light maximally at 260 nm
  • CSIR NET Z-DNA is left-handed helix, found in high GC regions
  • ICAR SRF Higher GC content = Higher melting temperature (Tm)
  • CSIR NET Phosphodiester bonds link 3' OH of one nucleotide to 5' phosphate of next

Structure of RNA (Ribonucleic Acid)

Chemical Differences from DNA

DNA

  • Sugar: 2'-Deoxyribose
  • Bases: A, G, C, T
  • Double-stranded
  • More stable
  • Located: Nucleus, mitochondria, chloroplasts
  • Longer molecules

RNA

  • Sugar: Ribose (has 2'-OH group)
  • Bases: A, G, C, U (Uracil)
  • Usually single-stranded
  • Less stable (2'-OH makes it labile)
  • Located: Nucleus, cytoplasm, ribosomes
  • Shorter molecules

Types of RNA

Type Full Name % of Total RNA Function Size
mRNA Messenger RNA 3-5% Carries genetic information from DNA to ribosomes for protein synthesis Variable (500-10,000 nucleotides)
rRNA Ribosomal RNA 80-85% Structural and catalytic component of ribosomes 18S, 28S, 5.8S, 5S (eukaryotes)
tRNA Transfer RNA 10-15% Transfers amino acids to ribosomes during translation 75-95 nucleotides
snRNA Small nuclear RNA <1 td=""> Splicing of pre-mRNA (part of spliceosome) 100-300 nucleotides
miRNA MicroRNA <1 td=""> Gene regulation by RNA interference 21-25 nucleotides
siRNA Small interfering RNA <1 td=""> Gene silencing, defense against viruses 20-25 nucleotides
lncRNA Long non-coding RNA Variable Gene regulation, chromatin remodeling >200 nucleotides

mRNA Structure

  • Monocistronic (eukaryotes): Codes for one protein
  • Polycistronic (prokaryotes): Codes for multiple proteins
  • 5' Cap: 7-methylguanosine (m⁷G) - protection, ribosome recognition
  • 5' UTR (Untranslated Region): Ribosome binding site, regulation
  • Coding sequence: Open Reading Frame (ORF) - translated into protein
  • Start codon: AUG (codes for Methionine)
  • Stop codons: UAA, UAG, UGA
  • 3' UTR: Stability, localization signals
  • Poly-A tail: 100-250 adenine nucleotides - stability, export from nucleus

tRNA Structure (Cloverleaf Model)

  • Shape: Cloverleaf in 2D, L-shaped in 3D
  • Size: 75-95 nucleotides
  • Four arms:
    • Acceptor arm: 3' end with CCA sequence - amino acid attachment site
    • D arm: Contains dihydrouridine (D) - ribosome recognition
    • Anticodon arm: Contains anticodon (3 nucleotides) - complementary to mRNA codon
    • TψC arm: Contains thymine, pseudouridine (ψ), cytosine - ribosome binding
  • Modified bases: Pseudouridine (ψ), dihydrouridine (D), inosine (I)
  • Wobble hypothesis: Third position of codon can pair loosely with anticodon
  • 61 sense codons but only ~45 different tRNAs (due to wobble pairing)

rRNA Structure

  • Prokaryotic ribosomes (70S):
    • Small subunit (30S): 16S rRNA + 21 proteins
    • Large subunit (50S): 23S rRNA + 5S rRNA + 31 proteins
  • Eukaryotic ribosomes (80S):
    • Small subunit (40S): 18S rRNA + 33 proteins
    • Large subunit (60S): 28S rRNA + 5.8S rRNA + 5S rRNA + 49 proteins
  • Functions:
    • Structural framework of ribosome
    • Catalyzes peptide bond formation (peptidyl transferase activity)
    • rRNA is a ribozyme (catalytic RNA)

Physical & Chemical Properties of RNA

  • Less stable than DNA: 2'-OH group makes it susceptible to alkaline hydrolysis
  • UV absorption: Maximum at 260 nm (like DNA)
  • Single-stranded: But can form secondary structures (hairpins, stem-loops)
  • Base pairing: Intramolecular - A-U, G-C, G-U (wobble)
  • Catalytic activity: Some RNAs are ribozymes (self-splicing, peptide bond formation)
  • Hydrolysis: Easily degraded by RNases (omnipresent enzymes)
  • Density: Slightly higher than DNA (~1.9 g/cm³)

🎯 Key Points for Exams

  • CSIR NET RNA has Uracil instead of Thymine
  • ICAR SRF 2'-OH group in ribose makes RNA less stable
  • CSIR NET Prokaryotic ribosome: 70S (30S + 50S), Eukaryotic: 80S (40S + 60S)
  • ICAR SRF tRNA has cloverleaf 2D structure, L-shaped 3D structure
  • CSIR NET Wobble pairing: 3rd codon position can pair non-canonically
  • ICAR SRF Start codon: AUG, Stop codons: UAA, UAG, UGA
  • CSIR NET Ribozymes: Catalytic RNA molecules (discovered by Cech & Altman)

Structure of Proteins

Introduction

  • Proteins: Polymers of amino acids linked by peptide bonds
  • Derived from Greek word "proteios" meaning "of prime importance"
  • Most abundant biological macromolecules (50-60% of cell dry weight)
  • Essential for structure, function, regulation of cells

Amino Acids - Building Blocks

  • General structure: H₂N-CHR-COOH
    • Central carbon (α-carbon)
    • Amino group (-NH₂)
    • Carboxyl group (-COOH)
    • Hydrogen atom (-H)
    • Side chain (R group) - determines properties
  • Zwitterion: Amino acids exist as dipolar ions at physiological pH
  • 20 standard amino acids in proteins

Classification of Amino Acids

Classification Amino Acids Properties
Based on Polarity
Non-polar (Hydrophobic) Gly, Ala, Val, Leu, Ile, Pro, Phe, Trp, Met Found in protein core, away from water
Polar (Hydrophilic) Ser, Thr, Cys, Asn, Gln, Tyr Can form hydrogen bonds, surface of proteins
Acidic (Negatively charged) Asp, Glu Carboxyl group in side chain, pH > pKa
Basic (Positively charged) Lys, Arg, His Amino group in side chain, pH < pKa
Based on Nutritional Requirement
Essential Val, Leu, Ile, Phe, Trp, Thr, Met, Lys, His (Arg for children) Cannot be synthesized, must be obtained from diet
Non-essential Ala, Asn, Asp, Glu, Ser, Gly, Pro, Cys, Tyr Can be synthesized by the body
Special Amino Acids
Aromatic Phe, Tyr, Trp Contain aromatic ring, absorb UV at 280 nm
Sulfur-containing Cys, Met Cys can form disulfide bonds
Imino acid Pro Creates kinks in protein structure

Peptide Bond Formation

  • Peptide bond: Covalent bond between carboxyl group of one amino acid and amino group of another
  • Condensation reaction: Release of H₂O molecule
  • Properties:
    • Planar structure (partial double bond character)
    • Trans configuration (R groups on opposite sides)
    • Rigid - no rotation around C-N bond
    • Resonance stabilized
    • Length: 1.32 Å
  • Nomenclature:
    • Dipeptide: 2 amino acids
    • Tripeptide: 3 amino acids
    • Oligopeptide: 2-20 amino acids
    • Polypeptide: >20 amino acids
    • Protein: Functional polypeptide (usually >50 amino acids)
  • Direction: N-terminus (free amino group) to C-terminus (free carboxyl group)

🏗️ Levels of Protein Structure

Primary Structure (1°)
Linear sequence of amino acids
Peptide bonds
Secondary Structure (2°)
Local folding patterns
α-helix, β-sheet, turns
Hydrogen bonds
Tertiary Structure (3°)
3D structure of single polypeptide
Multiple interactions
Quaternary Structure (4°)
Assembly of multiple polypeptide chains
Subunit interactions

Primary Structure (1°)

  • Definition: Linear sequence of amino acids in polypeptide chain
  • Bonds: Peptide bonds (covalent)
  • Determination: Edman degradation, Mass spectrometry, DNA sequencing
  • Significance:
    • Determines all higher levels of structure
    • Genetic information encoded in DNA
    • Single amino acid change can affect function (e.g., Sickle cell anemia: Glu→Val at position 6 in β-globin)
  • Example: Insulin - first protein sequenced by Frederick Sanger (1951, Nobel Prize 1958)

Secondary Structure (2°)

  • Definition: Regular, repeating local folding patterns of polypeptide backbone
  • Stabilized by: Hydrogen bonds between backbone C=O and N-H groups
  • Types:
Feature α-Helix β-Sheet
Shape Right-handed spiral/coil Extended, pleated sheet
H-bonds Between C=O of residue n and N-H of residue n+4 Between adjacent strands
Residues per turn 3.6 amino acids N/A
Pitch 5.4 Å N/A
R-groups Project outward from helix Alternately above and below sheet
Examples Keratin, myoglobin, hemoglobin Silk fibroin, immunoglobulins
Variants 310 helix, π helix Parallel β-sheet, Antiparallel β-sheet
  • β-Turns (Reverse turns):
    • 180° change in direction of polypeptide chain
    • Usually 4 amino acids
    • Connects antiparallel β-strands
    • Often contains Gly and Pro
  • Random coil/Loop regions:
    • Irregular, non-repetitive structure
    • Connecting regions between α-helices and β-sheets

Tertiary Structure (3°)

  • Definition: Overall 3D arrangement of all atoms in a single polypeptide chain
  • Stabilizing forces:
    • Disulfide bonds (S-S): Covalent bonds between Cys residues (strongest)
    • Hydrogen bonds: Between R groups
    • Ionic bonds (Salt bridges): Between oppositely charged R groups
    • Hydrophobic interactions: Non-polar R groups cluster in core
    • Van der Waals forces: Weak attractions between atoms in close proximity
  • Domains: Compact, semi-independent folding units within protein
  • Motifs: Combinations of secondary structures (e.g., helix-turn-helix, zinc finger)

Quaternary Structure (4°)

  • Definition: Arrangement of multiple polypeptide chains (subunits) in a multi-subunit protein
  • Subunits: Individual polypeptide chains
  • Stabilized by: Same forces as tertiary structure (except usually no disulfide bonds)
  • Examples:
    • Hemoglobin: 2α + 2β subunits (α₂β₂)
    • Immunoglobulin (IgG): 2 heavy + 2 light chains
    • DNA polymerase III: 10 different subunits
    • Collagen: Triple helix of 3 polypeptide chains
  • Cooperative binding: Binding of ligand to one subunit affects others (e.g., O₂ binding to hemoglobin)

Protein Folding

  • Anfinsen's dogma: Primary structure determines 3D structure
  • Spontaneous folding: Proteins fold to lowest energy state
  • Chaperones: Proteins that assist in proper folding
    • Hsp60, Hsp70 (Heat shock proteins)
    • Chaperonins (GroEL-GroES in bacteria)
    • Prevent aggregation, promote correct folding
  • Misfolding diseases:
    • Alzheimer's disease (amyloid plaques)
    • Parkinson's disease (α-synuclein aggregation)
    • Prion diseases (BSE, CJD) - infectious misfolded proteins

Physical Properties of Proteins

  • Molecular weight: 6,000 Da (insulin) to >1,000,000 Da (titin)
  • Isoelectric point (pI): pH at which protein has no net charge
  • UV absorption: Maximum at 280 nm (due to Trp, Tyr, Phe)
  • Solubility: Depends on surface charges, pH, salt concentration
  • Colligative properties: Osmotic pressure, viscosity
  • Electrophoresis: Separation based on charge and size
    • SDS-PAGE: Based on molecular weight
    • Native PAGE: Based on charge and shape
    • IEF: Based on isoelectric point

Chemical Properties of Proteins

  • Denaturation:
    • Loss of native 3D structure without breaking peptide bonds
    • Causes: Heat, pH extremes, detergents, organic solvents, urea
    • Usually irreversible (except in specific conditions)
    • Loss of biological activity
  • Hydrolysis:
    • Breaking of peptide bonds
    • Acid/alkaline hydrolysis
    • Enzymatic hydrolysis (proteases, peptidases)
  • Color reactions:
    • Biuret test: Detects peptide bonds (violet color with Cu²⁺)
    • Ninhydrin test: Detects free amino groups (purple color)
    • Xanthoproteic test: Detects aromatic amino acids (yellow with HNO₃)
    • Bradford assay: Protein quantification (Coomassie blue dye)
    • Lowry method: Protein quantification (Cu²⁺ and Folin reagent)
  • Precipitation:
    • Salting out: High salt concentration (ammonium sulfate)
    • pH adjustment: At pI
    • Organic solvents: Acetone, ethanol
    • Heavy metals: Pb²⁺, Hg²⁺

🎯 Key Points for Exams

  • CSIR NET 20 standard amino acids, 9-10 essential amino acids
  • ICAR SRF Peptide bond: Planar, trans configuration, partial double bond character
  • CSIR NET α-helix: 3.6 residues/turn, pitch = 5.4 Å
  • ICAR SRF Disulfide bonds between Cysteine residues (strongest covalent bond in proteins)
  • CSIR NET Hemoglobin: Quaternary structure (α₂β₂ tetramer)
  • ICAR SRF Proteins absorb UV at 280 nm (Trp, Tyr, Phe)
  • CSIR NET Anfinsen: Primary structure determines tertiary structure
  • ICAR SRF Chaperones (Hsp70, GroEL) assist protein folding
  • CSIR NET SDS-PAGE: Separates proteins by molecular weight

💡 Memory Tricks & Mnemonics - Chapter 1

  • DNA Bases: "All Good Children Try" (A, G, C, T) - Purines (AG), Pyrimidines (CT)
  • Essential Amino Acids: "PVT TIM HALL"
    • P - Phenylalanine
    • V - Valine
    • T - Threonine, Tryptophan
    • I - Isoleucine
    • M - Methionine
    • H - Histidine
    • A - Arginine (for children)
    • L - Leucine, Lysine
  • Chargaff's Rules: "AT = True, GC = True" (Amount of A = T, G = C)
  • DNA Forms: "BAZ" - B (most common), A (dehydrated), Z (zigzag/left-handed)
  • Ribosome Sizes: "Pro-7, Eu-8" (Prokaryotes 70S, Eukaryotes 80S)
  • Stop Codons: "U Are Away, U Are Gone, U Go Away" (UAA, UAG, UGA)
  • Protein Structure Levels: "Please Send The Quail" (Primary, Secondary, Tertiary, Quaternary)

📝 Common MCQ Patterns - Chapter 1

  • Watson-Crick Model: Year (1953), features (antiparallel, complementary, double helix)
  • Base Pairing: Number of H-bonds (A-T: 2, G-C: 3)
  • DNA vs RNA: Sugar (deoxyribose vs ribose), bases (T vs U), strands (double vs single)
  • Ribosome composition: rRNA types and protein numbers in subunits
  • tRNA structure: Anticodon position, acceptor arm function
  • Amino acid properties: Hydrophobic vs hydrophilic, essential vs non-essential
  • Secondary structures: α-helix parameters (3.6 residues/turn, 5.4 Å pitch)
  • Bonds in proteins: Types and relative strengths (disulfide > ionic > H-bond > Van der Waals)
  • Protein denaturation: Causes and effects

📖 CHAPTER 2: 🧬 DNA FUNCTION

Expression, Exchange of Genetic Material & Mutation

CSIR NET ICAR SRF

1. DNA Expression (Gene Expression)

Gene expression is the process by which information from a gene is used to synthesize functional gene products, primarily proteins. This is the fundamental mechanism by which genotype gives rise to phenotype.

1.1 Central Dogma of Molecular Biology

Proposed by Francis Crick in 1958, the Central Dogma describes the flow of genetic information:

Central Dogma Flow

DNA (Deoxyribonucleic Acid)
Storage of genetic information
TRANSCRIPTION
DNA → RNA (in nucleus)
RNA (Ribonucleic Acid)
Messenger molecule
TRANSLATION
RNA → Protein (in ribosome)
PROTEIN
Functional molecules
1.2 Transcription

Transcription is the process of synthesizing RNA from a DNA template. It occurs in three stages:

Stage Key Events Key Enzymes/Factors
Initiation RNA polymerase binds to promoter region; DNA unwinds RNA Polymerase II (eukaryotes), Transcription factors, TFIID
Elongation RNA polymerase synthesizes RNA in 5' → 3' direction RNA Polymerase, Elongation factors
Termination RNA polymerase reaches terminator sequence; transcript released Termination factors, Rho protein (prokaryotes)

⚠️ Important Differences: Prokaryotic vs Eukaryotic Transcription

Prokaryotes

  • Single RNA polymerase
  • No RNA processing
  • Transcription and translation coupled
  • Polycistronic mRNA
  • No introns (generally)

Eukaryotes

  • Three RNA polymerases (I, II, III)
  • Extensive RNA processing
  • Transcription (nucleus) and translation (cytoplasm) separated
  • Monocistronic mRNA
  • Introns present; require splicing
1.3 RNA Processing (Eukaryotes)

Primary transcript (pre-mRNA) undergoes several modifications before becoming mature mRNA:

  • 5' Capping: Addition of 7-methylguanosine cap at 5' end (protection and ribosome binding)
  • 3' Polyadenylation: Addition of poly(A) tail (~200 adenine nucleotides) at 3' end (stability and export)
  • Splicing: Removal of introns and joining of exons by spliceosome
    • Alternative splicing can produce multiple proteins from one gene
    • Consensus sequences: 5' splice site (GU), 3' splice site (AG), branch point (A)
1.4 Translation

Translation is the process of protein synthesis from mRNA template. It occurs on ribosomes and involves tRNA molecules.

Translation Process

INITIATION
Small ribosomal subunit + mRNA + initiator tRNA (Met)
Recognition of START codon (AUG)
ELONGATION
Codon recognition → Peptide bond formation → Translocation
Aminoacyl-tRNA, Peptidyl transferase, EF-Tu, EF-G
TERMINATION
STOP codon (UAA, UAG, UGA) recognized
Release factors bind → Polypeptide released

🔑 The Genetic Code

  • Triplet code: 3 nucleotides (codon) = 1 amino acid
  • Degenerate: Multiple codons can code for same amino acid
  • Non-overlapping: Each nucleotide part of only one codon
  • Universal: Same in almost all organisms (with minor exceptions)
  • Unambiguous: Each codon specifies only one amino acid
  • 61 codons for amino acids + 3 stop codons (UAA, UAG, UGA)
  • START codon: AUG (codes for Methionine)
1.5 Gene Regulation

Gene expression is tightly regulated at multiple levels:

Level Mechanisms Examples
Transcriptional Promoters, enhancers, silencers, transcription factors Lac operon, Trp operon, TATA box
Post-transcriptional Alternative splicing, RNA stability, miRNA Tropomyosin variants, let-7 miRNA
Translational Ribosome binding, translation factors, uORFs Ferritin regulation by iron
Post-translational Protein modifications, degradation, localization Phosphorylation, ubiquitination
Epigenetic DNA methylation, histone modifications X-inactivation, genomic imprinting
2. Exchange of Genetic Material

Exchange of genetic material is crucial for genetic diversity and evolution. It occurs through various mechanisms in both prokaryotes and eukaryotes.

2.1 Genetic Recombination in Prokaryotes

Prokaryotes exchange genetic material through three primary mechanisms:

TRANSFORMATION

Uptake of naked DNA from environment

Discovered by: Griffith (1928)

TRANSDUCTION

Transfer of DNA via bacteriophages

Discovered by: Zinder & Lederberg (1952)

CONJUGATION

Transfer through physical contact (pilus)

Discovered by: Lederberg & Tatum (1946)

Detailed Mechanisms

A. Transformation:

  • Competent cells can take up exogenous DNA
  • DNA binds to receptor sites on cell surface
  • One strand degraded, other strand integrates into chromosome
  • Natural competence: Bacillus, Streptococcus, Haemophilus
  • Artificial competence: CaCl₂ treatment, electroporation

B. Transduction:

  • Generalized transduction: Any bacterial gene can be transferred (lytic cycle error)
  • Specialized transduction: Only specific genes near prophage integration site (lysogenic cycle)
  • Phage packages bacterial DNA instead of/along with viral DNA
  • Recipient cell receives donor DNA during infection

C. Conjugation:

  • Requires F (fertility) plasmid with tra genes
  • F+ cells (donor) have F plasmid; F- cells (recipient) lack it
  • Sex pilus forms bridge between cells
  • F plasmid replicates via rolling circle mechanism during transfer
  • Hfr cells: F plasmid integrated into chromosome, can transfer chromosomal genes
  • Used for chromosome mapping (interrupted mating experiments)
2.2 Genetic Recombination in Eukaryotes

Eukaryotic genetic recombination primarily occurs during meiosis through two mechanisms:

Independent Assortment

  • Random distribution of homologous chromosomes during Meiosis I
  • Produces 2ⁿ combinations (n = haploid number)
  • Humans: 2²³ = 8,388,608 possible gametes
  • Discovered by Mendel (1866)

Crossing Over (Recombination)

  • Exchange of DNA segments between homologous chromosomes
  • Occurs during Prophase I (Pachytene stage)
  • Mediated by synaptonemal complex
  • Visible as chiasmata
  • Increases genetic diversity exponentially
2.3 Molecular Mechanism of Recombination

Homologous Recombination (Holliday Model)

Step 1: DNA Break
Single-strand nick in one DNA molecule
Step 2: Strand Invasion
Broken strand invades homologous DNA duplex
Step 3: Holliday Junction Formation
Cross-shaped structure forms between four DNA strands
Step 4: Branch Migration
Junction moves along DNA, facilitated by RecA protein
Step 5: Resolution
Resolvase enzymes cut junction → Recombinant DNA molecules

Key Enzymes in Recombination:

  • RecBCD complex: Processes DNA ends, loads RecA
  • RecA protein: Catalyzes strand invasion and homology search
  • Resolvases: Cut Holliday junction (RuvC in prokaryotes)
  • Rad51: Eukaryotic homolog of RecA
  • Spo11: Creates double-strand breaks in meiosis
2.4 Horizontal Gene Transfer (HGT)

Transfer of genetic material between organisms outside of reproduction, especially important in microbial evolution.

Type Mechanism Significance
Transformation Natural uptake of DNA Antibiotic resistance spread
Transduction Phage-mediated transfer Virulence factor transfer
Conjugation Plasmid transfer Rapid adaptation to antibiotics
Transposons Mobile genetic elements Genome plasticity, evolution

📝 CSIR NET & ICAR SRF Exam Tips: Genetic Exchange

  • Know the discoverers and years of each mechanism (frequently asked in MCQs)
  • Understand Hfr mapping: genes transfer in linear order, time-based mapping
  • F+ × F- → both F+ ; Hfr × F- → usually F- (rarely Hfr)
  • Cotransformation frequency used to map closely linked genes
  • Recombination frequency = Map units = centiMorgans (cM)
  • 1% recombination = 1 map unit = 1 cM
  • Three-point test cross determines gene order and map distance
  • Coefficient of coincidence (COC) = Observed DCO / Expected DCO
  • Interference = 1 - COC (measures crossover interference)
3. Mutation

Mutations are permanent changes in DNA sequence. They are the ultimate source of genetic variation and can be beneficial, neutral, or deleterious.

3.1 Classification of Mutations

Based on Cell Type

Somatic Mutations

  • Occur in body cells
  • Not transmitted to offspring
  • Can cause cancer
  • Example: Skin cancer from UV exposure

Germline Mutations

  • Occur in gametes or gamete precursors
  • Heritable, passed to offspring
  • Basis of evolution
  • Example: Hemophilia, color blindness
Based on Molecular Nature
3.2 Types of Point Mutations

Point Mutation Categories

TRANSITION

Purine ↔ Purine

Pyrimidine ↔ Pyrimidine


A ↔ G
C ↔ T

More common (67%)

TRANSVERSION

Purine ↔ Pyrimidine

Pyrimidine ↔ Purine


A/G ↔ C/T

Less common (33%)

Effects of Point Mutations
Type Description Examples
Point Mutations Single nucleotide change Transition, transversion
Insertion Addition of nucleotide(s) Can cause frameshift
Deletion Loss of nucleotide(s) Can cause frameshift
Duplication Repetition of DNA segment Gene family expansion
Inversion Reversal of DNA segment Chromosomal rearrangement
Translocation Transfer between chromosomes Philadelphia chromosome
Mutation Type Effect on Protein Example
Silent (Synonymous) No change in amino acid (same codon family) UUU → UUC (both code Phe)
Missense (Non-synonymous) Different amino acid substitution Sickle cell: GAG→GUG (Glu→Val)
Nonsense Creates premature stop codon UAU→UAA (Tyr→Stop)
Frameshift Insertion/deletion (not multiple of 3) shifts reading frame Tay-Sachs disease (insertion)

🔬 Classical Examples

  • Sickle Cell Anemia: Point mutation in β-globin gene (A→T at 6th codon)
    • GAG (Glutamic acid) → GUG (Valine)
    • Causes hemoglobin polymerization under low oxygen
    • Heterozygote advantage against malaria
  • Tay-Sachs Disease: Frameshift mutation in HEXA gene
    • 4-base insertion in exon 11
    • Deficiency of hexosaminidase A enzyme
    • Accumulation of GM2 ganglioside in neurons
3.3 Causes of Mutations

A. Spontaneous Mutations

  • DNA Replication Errors: Polymerase misincorporation (10⁻⁹ to 10⁻¹⁰ per base per replication)
  • Tautomeric Shifts: Rare forms of bases cause mispairing (A*-C, G*-T)
  • Depurination: Loss of purine bases (~5,000 per cell per day)
  • Deamination: Cytosine → Uracil (100 per cell per day)
  • Oxidative Damage: ROS cause 8-oxoguanine formation
  • Slipped Strand Mispairing: In repetitive sequences (microsatellites)

B. Induced Mutations (Mutagens)

Mutagen Type Examples Mechanism
Physical UV radiation (254 nm), X-rays, γ-rays Thymine dimers, DNA breaks
Base Analogs 5-Bromouracil (5-BU), 2-Aminopurine Incorporated into DNA, cause mispairing
Alkylating Agents EMS, MMS, Mustard gas Add alkyl groups to bases
Deaminating Agents Nitrous acid (HNO₂) C→U, A→hypoxanthine
Intercalating Agents Ethidium bromide, Acridine dyes Insert between bases, cause frameshift
Oxidizing Agents H₂O₂, hydroxyl radicals Oxidative DNA damage
3.4 DNA Repair Mechanisms

Cells have evolved sophisticated mechanisms to detect and repair DNA damage:

Major DNA Repair Pathways

1. DIRECT REPAIR
Damage reversed without removing bases
Example: Photolyase (photoreactivation), O⁶-methylguanine-DNA methyltransferase
2. BASE EXCISION REPAIR (BER)
Removes damaged/modified single bases
Steps: DNA glycosylase → AP endonuclease → DNA polymerase → DNA ligase
3. NUCLEOTIDE EXCISION REPAIR (NER)
Removes bulky lesions (thymine dimers)
Defect causes: Xeroderma pigmentosum (XP)
4. MISMATCH REPAIR (MMR)
Corrects base-base mismatches and small loops
MutS, MutL, MutH proteins; Defect → Lynch syndrome
5. HOMOLOGOUS RECOMBINATION (HR)
Repairs double-strand breaks using sister chromatid
Key proteins: BRCA1, BRCA2, Rad51
6. NON-HOMOLOGOUS END JOINING (NHEJ)
Direct ligation of broken ends (error-prone)
Ku70/80, DNA-PKcs, DNA ligase IV

⚠️ Clinical Significance of Repair Defects

Disease Defective Pathway Symptoms
Xeroderma Pigmentosum NER (XPA-XPG genes) UV sensitivity, skin cancer, neurodegeneration
Lynch Syndrome (HNPCC) MMR (MLH1, MSH2) Hereditary colorectal cancer
Ataxia Telangiectasia DSB repair (ATM gene) Neurodegeneration, immunodeficiency, cancer
Fanconi Anemia ICL repair (FANC genes) Bone marrow failure, developmental abnormalities
3.5 Mutagenesis Testing

Ames Test (Salmonella Mutagenicity Test)

Developed by Bruce Ames (1971) to test chemical carcinogenicity

Principle
Uses histidine-requiring Salmonella typhimurium strains
Reversion to His+ indicates mutagenicity
Procedure
1. Bacteria + Test chemical + Rat liver extract (S9)
2. Plate on histidine-deficient medium
3. Incubate and count revertant colonies
Interpretation
More revertants = Higher mutagenic potential
S9 activates pro-mutagens to active mutagens

Advantages: Rapid, inexpensive, ~90% correlation with carcinogenicity

Limitations: Bacterial system may not reflect mammalian responses

3.6 Transposable Elements

Mobile DNA sequences that can move within genome, causing mutations

Class I: Retrotransposons

  • Mechanism: "Copy and paste" via RNA intermediate
  • Process: DNA → RNA → DNA (reverse transcriptase)
  • Examples: LINEs, SINEs, Alu elements
  • Prevalence: ~45% of human genome
  • Autonomous: LINEs (encode reverse transcriptase)
  • Non-autonomous: SINEs, Alu (use LINE machinery)

Class II: DNA Transposons

  • Mechanism: "Cut and paste"
  • Process: Excision and reintegration
  • Examples: Ac/Ds (maize), P elements (Drosophila)
  • Prevalence: ~3% of human genome (mostly inactive)
  • Enzyme: Transposase
  • Structure: Terminal inverted repeats (TIRs)

🌽 Barbara McClintock's Discovery

Discovered transposable elements in maize (1940s-1950s)

  • Observed color variegation in maize kernels
  • Identified Activator (Ac) and Dissociation (Ds) elements
  • Ac: Autonomous element with transposase
  • Ds: Non-autonomous, requires Ac for movement
  • Nobel Prize in Physiology or Medicine (1983)
  • Called them "controlling elements" or "jumping genes"

Significance of Transposons:

  • Major source of genetic variation and evolution
  • Can cause insertional mutagenesis (disrupt genes)
  • Regulate gene expression (provide regulatory elements)
  • Source of chromosomal rearrangements
  • Used as molecular tools in genetic engineering
  • Associated with diseases (hemophilia A, neurofibromatosis)
3.7 Mutation Rate and Factors
Organism Mutation Rate Notes
Bacteriophages 10⁻⁶ - 10⁻⁸ per base per replication RNA viruses higher than DNA viruses
Bacteria 10⁻⁹ - 10⁻¹⁰ per base per generation Efficient proofreading and repair
Eukaryotes 10⁻⁹ - 10⁻¹⁰ per base per cell division Varies by cell type and gene
Humans (germline) ~1.2 × 10⁻⁸ per base per generation ~70 new mutations per individual

Factors Affecting Mutation Rate:

  • Fidelity of DNA polymerase: Proofreading activity (3' → 5' exonuclease)
  • DNA repair efficiency: More repair systems = lower mutation rate
  • Environmental factors: Mutagens, temperature, radiation
  • Replication timing: Late-replicating regions more prone
  • Chromatin structure: Heterochromatin vs euchromatin
  • CpG islands: Cytosine methylation hotspots
  • Paternal age effect: More mutations from older fathers

📝 CSIR NET & ICAR SRF Exam Tips: Mutations

  • Remember key numbers:
    • Human genome: ~3 × 10⁹ bp, ~70 new mutations per generation
    • Spontaneous depurination: ~5,000 per cell per day
    • Spontaneous deamination: ~100 per cell per day
  • Classical mutagens and their mechanisms:
    • 5-BU: Base analog (T analog), causes A-T → G-C transitions
    • EMS: Alkylating agent, creates O⁶-ethylguanine
    • UV light: Creates thymine dimers (cyclobutane pyrimidine dimers)
    • Nitrous acid: Deaminates C→U and A→hypoxanthine
    • Acridine dyes: Intercalate, cause frameshift mutations
  • Repair pathway specificity:
    • BER: Small base modifications (oxidation, alkylation)
    • NER: Bulky lesions (thymine dimers, large adducts)
    • MMR: Replication errors, microsatellite instability
  • Important concepts:
    • Transitions more common than transversions (2:1 ratio)
    • SOS response in bacteria: Error-prone repair under stress
    • Adaptive mutation: Controversial concept of directed mutation
    • Mutator phenotype: Defective repair genes increase mutation rate
  • Know disease associations:
    • BRCA1/2 mutations → Breast/ovarian cancer (HR defect)
    • TP53 mutations → Li-Fraumeni syndrome (guardian of genome)
    • Microsatellite instability → Lynch syndrome (MMR defect)
  • Transposons:
    • McClintock's Ac/Ds system in maize (Nobel Prize 1983)
    • LINEs (Long Interspersed Elements): ~6 kb, autonomous
    • SINEs (Short Interspersed Elements): ~300 bp, non-autonomous
    • Alu elements: Most abundant SINE in humans (~1 million copies)
    • P elements: Used for Drosophila mutagenesis and transgenesis

📝 CSIR NET & ICAR SRF Exam Tips: Gene Expression

  • Remember the differences between prokaryotic and eukaryotic transcription (frequently asked)
  • Know the consensus sequences: Pribnow box (-10), TATA box (-25), CAAT box, GC box
  • Understand wobble hypothesis (Crick) - explains codon degeneracy
  • Lac operon vs Trp operon (inducible vs repressible systems)
  • Alternative splicing increases protein diversity from limited genes
  • Know key experiments: Beadle and Tatum (one gene-one enzyme), Nirenberg and Khorana (genetic code)
  • RNA polymerases: Pol I (rRNA), Pol II (mRNA), Pol III (tRNA, 5S rRNA)
4. Integration: DNA Function in Context
4.1 Central Dogma Exceptions

Variations from Standard Central Dogma

  • Reverse Transcription: RNA → DNA (retroviruses, telomerase, retrotransposons)
  • RNA Replication: RNA → RNA (RNA viruses like poliovirus, influenza)
  • Direct DNA to DNA: DNA replication
  • RNA catalysis: Ribozymes (self-splicing introns, ribosomal peptidyl transferase)
  • RNA-dependent RNA polymerases: In plants, fungi (RNAi, viral defense)
4.2 Epigenetics and Gene Expression

Heritable changes in gene expression without changes in DNA sequence:

Mechanism Description Effect
DNA Methylation Addition of methyl group to cytosine (5-methylcytosine) Gene silencing, X-inactivation
Histone Modifications Acetylation, methylation, phosphorylation, ubiquitination Chromatin remodeling, transcription regulation
Chromatin Remodeling ATP-dependent complexes alter nucleosome positioning Access to DNA for transcription factors
Non-coding RNAs miRNA, siRNA, lncRNA regulate gene expression Post-transcriptional regulation

Histone Code:

  • H3K4me3: Trimethylation of lysine 4 on histone H3 → Active transcription
  • H3K9me3: Trimethylation of lysine 9 → Heterochromatin, silencing
  • H3K27me3: Polycomb repression, developmental genes
  • H3K27ac: Acetylation → Active enhancers
  • H4K16ac: Acetylation → Chromatin decompaction

📝 CSIR NET & ICAR SRF: High-Yield Integration Topics

  • Splicing mechanisms:
    • Spliceosome: snRNPs (U1, U2, U4, U5, U6)
    • Self-splicing introns: Group I (requires GTP), Group II (no cofactor)
    • Alternative splicing increases protein diversity
  • RNA interference (RNAi):
    • Discovery: Fire and Mello (Nobel Prize 2006)
    • Dicer cleaves dsRNA → siRNA (~21-23 nt)
    • RISC complex mediates mRNA cleavage
    • miRNA: Endogenous regulators from pre-miRNA hairpins
  • Recombination applications:
    • Cre-lox system: Site-specific recombination in mice
    • FLP-FRT system: Yeast recombinase
    • CRISPR-Cas9: Uses HDR for precise editing
  • Mutation screening:
    • SSCP: Single-strand conformation polymorphism
    • DGGE: Denaturing gradient gel electrophoresis
    • HRM: High-resolution melting analysis
    • NGS: Next-generation sequencing for comprehensive analysis
5. Practice Questions & Exam Preparation
5.1 Multiple Choice Questions (MCQ) - CSIR NET Style

Q1. Which of the following statements about the genetic code is INCORRECT?

  • A) It is degenerate with multiple codons for most amino acids
  • B) It is non-overlapping with each nucleotide part of only one codon
  • C) All organisms use exactly the same genetic code without exceptions
  • D) AUG serves as both start codon and codes for methionine

Answer: C (Mitochondria and some microorganisms show variations)

Q2. In the Holliday model of recombination, which enzyme is responsible for catalyzing strand invasion?

  • A) DNA ligase
  • B) RecA protein
  • C) Resolvase
  • D) Topoisomerase

Answer: B (RecA in prokaryotes, Rad51 in eukaryotes)

Q3. A mutation changes codon GAA to UAA. This is an example of:

  • A) Silent mutation
  • B) Missense mutation
  • C) Nonsense mutation
  • D) Frameshift mutation

Answer: C (Creates premature stop codon - nonsense)

Q4. Which DNA repair mechanism is defective in Xeroderma Pigmentosum?

  • A) Base Excision Repair (BER)
  • B) Nucleotide Excision Repair (NER)
  • C) Mismatch Repair (MMR)
  • D) Non-Homologous End Joining (NHEJ)

Answer: B (NER repairs UV-induced thymine dimers)

Q5. In bacterial conjugation, an Hfr strain transfers:

  • A) Only the F plasmid to recipient cells
  • B) Chromosomal DNA in a linear, time-dependent manner
  • C) Random segments of chromosomal DNA
  • D) DNA through bacteriophage intermediates

Answer: B (Hfr = High frequency recombination with integrated F plasmid)

5.2 Statement-Based Questions - ICAR SRF Style

Q6. Consider the following statements about RNA processing in eukaryotes:

I. 5' capping involves addition of 7-methylguanosine

II. Splicing always produces the same mRNA from a given gene

III. Poly(A) tail is added at the 3' end by poly(A) polymerase

IV. Introns are removed by spliceosomes containing snRNPs

Which statements are correct?

  • A) I, II, and III only
  • B) I, III, and IV only
  • C) II, III, and IV only
  • D) All are correct

Answer: B (Statement II is wrong - alternative splicing produces different mRNAs)

Q7. Match the following mutagens with their mechanisms:

Mutagen Mechanism
I. 5-Bromouracil a. Intercalation causing frameshift
II. Acridine orange b. Base analog causing transitions
III. EMS c. Deamination of cytosine
IV. Nitrous acid d. Alkylation of guanine

Correct matching:

  • A) I-a, II-b, III-c, IV-d
  • B) I-b, II-a, III-d, IV-c
  • C) I-c, II-d, III-a, IV-b
  • D) I-d, II-c, III-b, IV-a

Answer: B

5.3 Most Frequently Asked Topics

🎯 Top 20 High-Yield Topics for CSIR NET & ICAR SRF

Gene Expression:

  1. Central Dogma and exceptions
  2. Prokaryotic vs Eukaryotic transcription
  3. Lac operon and Trp operon regulation
  4. RNA processing (capping, splicing, polyadenylation)
  5. Genetic code properties
  6. Translation mechanism and regulation
  7. Alternative splicing mechanisms
  8. Ribozymes and catalytic RNA
  9. microRNA and RNAi pathways
  10. Epigenetic regulation

Recombination & Mutation:

  1. Transformation, transduction, conjugation
  2. Holliday junction and recombination models
  3. Types of mutations and their effects
  4. Mutagens and their mechanisms
  5. DNA repair pathways (BER, NER, MMR)
  6. Diseases due to repair defects
  7. Transposable elements (Class I & II)
  8. Ames test for mutagenicity
  9. Chromosomal aberrations
  10. Molecular basis of cancer
5.4 Important Experiments to Remember
Scientist(s) Year Experiment/Discovery Significance
Beadle & Tatum 1941 One gene-one enzyme hypothesis Established genes code for enzymes (Neurospora)
Griffith 1928 Transformation in pneumococcus Discovered transforming principle
Lederberg & Tatum 1946 Bacterial conjugation Demonstrated genetic recombination in bacteria
Zinder & Lederberg 1952 Transduction in Salmonella Phage-mediated gene transfer
Jacob & Monod 1961 Lac operon model Gene regulation in prokaryotes (Nobel 1965)
Nirenberg & Khorana 1961-66 Genetic code deciphering Cracked the genetic code (Nobel 1968)
Holliday 1964 Recombination model Molecular mechanism of crossing over
Barbara McClintock 1940s-50s Transposable elements Discovered jumping genes in maize (Nobel 1983)
Bruce Ames 1971 Ames test Bacterial mutagenicity assay
Fire & Mello 1998 RNA interference Discovered RNAi in C. elegans (Nobel 2006)
Sharp & Roberts 1977 Split genes (introns) Discovered RNA splicing (Nobel 1993)
Crick 1966 Wobble hypothesis Explained codon degeneracy
5.5 Key Formulas and Calculations

1. Recombination Frequency (RF):

RF = (Number of recombinants / Total progeny) × 100
1% recombination = 1 map unit = 1 centiMorgan (cM)

2. Coefficient of Coincidence (COC):

COC = Observed double crossovers / Expected double crossovers
Expected DCO = (RF₁ × RF₂) × Total progeny

3. Interference:

Interference = 1 - COC
Range: 0 (no interference) to 1 (complete interference)

4. Mutation Rate:

μ = Number of mutations / (Number of cells × Generation time)
Or: μ = m/N where m = mutants, N = total individuals

5. Luria-Delbrück Fluctuation Test:

Variance/Mean ratio >> 1 indicates pre-existing mutations
Variance/Mean ≈ 1 indicates induced mutations

5.6 Common Mistakes to Avoid

⚠️ Exam Pitfalls

  • Don't confuse: Transformation (naked DNA uptake) with Transduction (phage-mediated)
  • Remember: F+ × F- → Both F+ (not Hfr), but Hfr × F- usually remains F-
  • Transitions vs Transversions: Transitions are MORE common (purine↔purine, pyrimidine↔pyrimidine)
  • BER vs NER: BER for small modifications, NER for bulky lesions
  • Wobble position: 3rd position of codon (5' end of anticodon), not 1st position
  • Spliceosome: Contains snRNPs (not snRNAs alone) - protein-RNA complexes
  • Start codon: AUG (not ATG in RNA context)
  • Frameshift mutations: Insertion/deletion NOT in multiples of 3
  • LINEs vs SINEs: LINEs are autonomous (have reverse transcriptase), SINEs are not
  • Gene vs Cistron: In prokaryotes, operon has multiple cistrons; in eukaryotes, one gene = one mRNA
5.7 Quick Revision Tables

RNA Polymerases in Eukaryotes

RNA Pol Products Sensitivity to α-amanitin Location
Pol I 18S, 5.8S, 28S rRNA Insensitive Nucleolus
Pol II mRNA, most snRNA, miRNA, lncRNA Highly sensitive Nucleoplasm
Pol III tRNA, 5S rRNA, U6 snRNA Moderately sensitive (high conc.) Nucleoplasm

Stop Codons - Nomenclature

Codon Name Mnemonic
UAA Ochre "U Are Away"
UAG Amber "U Are Gone"
UGA Opal "U Go Away"

Consensus Sequences

Sequence Location Function Organism
Pribnow box (TATAAT) -10 position RNA Pol binding Prokaryotes
-35 sequence (TTGACA) -35 position RNA Pol recognition Prokaryotes
TATA box (TATAAA) -25 position TBP binding, transcription start Eukaryotes
CAAT box -75 position Enhances transcription rate Eukaryotes
GC box Variable Sp1 transcription factor binding Eukaryotes
Shine-Dalgarno (AGGAGGU) 5' UTR of mRNA Ribosome binding site Prokaryotes
Kozak sequence (GCCRCCAUGG) Around start codon Translation initiation Eukaryotes

📚 Last-Minute Revision Tips

Day Before Exam:

  • Review all flowcharts and diagrams
  • Memorize key numbers (mutation rates, genome sizes)
  • Go through exception cases (wobble, suppressor tRNAs)
  • Revise all scientist names and years
  • Practice drawing Holliday junction
  • Review disease associations

During Exam:

  • Read questions carefully - "NOT" and "EXCEPT"
  • Eliminate obviously wrong answers first
  • For mechanism questions, draw if needed
  • Don't spend too long on one question
  • Mark difficult questions for review
  • Double-check numerical calculations

🌟 Final Success Mantra

Understand the concepts, don't just memorize

Practice previous year questions

Connect topics (expression → mutation → repair)

Visualize processes (draw mechanisms)

Revise regularly with active recall

"Success is the sum of small efforts repeated day in and day out!"

📖 Recommended Reference Books

For CSIR NET:

  • Molecular Biology of the Cell - Alberts et al.
  • Molecular Biology - Weaver
  • Genes - Lewin
  • Principles of Gene Manipulation - Primrose

For ICAR SRF:

  • Principles of Genetics - Snustad & Simmons
  • Genetics - Strickberger
  • Molecular Genetics - Gupta
  • Concepts of Genetics - Klug & Cummings

🎯 Summary: Key Takeaways for Exams

DNA Expression:

  • Transcription: DNA → RNA (RNA polymerase, promoters, terminators)
  • Translation: RNA → Protein (ribosomes, tRNA, genetic code)
  • Regulation at multiple levels (transcriptional, post-transcriptional, translational, post-translational)
  • Prokaryotes: Operons, coupled transcription-translation
  • Eukaryotes: RNA processing (capping, splicing, polyadenylation)

Genetic Exchange:

  • Prokaryotes: Transformation, transduction, conjugation
  • Eukaryotes: Meiotic recombination, crossing over
  • Holliday junction model, RecA/Rad51 proteins
  • Horizontal gene transfer in evolution and antibiotic resistance

Mutation:

  • Types: Point, frameshift, chromosomal
  • Causes: Spontaneous (errors, damage) vs Induced (mutagens)
  • Effects: Silent, missense, nonsense, frameshift
  • Repair: BER, NER, MMR, HR, NHEJ
  • Transposons: Class I (retrotransposons) vs Class II (DNA transposons)
  • Clinical relevance: XP, Lynch syndrome, cancer genetics

📖 CHAPTER 3: Recombinant DNA Technology & Nucleic Acid Hybridization

Methods, Techniques & Applications

Recombinant DNA Technology (rDNA Technology)

Overview

  • Definition: Technology to artificially construct DNA molecules by joining DNA from different sources
  • Also called: Gene cloning, Molecular cloning, Genetic engineering
  • Pioneers: Stanley Cohen & Herbert Boyer (1973) - First recombinant DNA organism
  • Basic Principle: Cut DNA with restriction enzymes → Join with DNA ligase → Insert into vector → Transform into host

🔬 Steps of Gene Cloning

1. Isolation of DNA
Extract genomic DNA or mRNA from source organism
If mRNA → Convert to cDNA using reverse transcriptase
2. Fragmentation of DNA
Cut DNA with restriction enzymes (Type II)
Generate compatible sticky or blunt ends
3. Isolation of Gene of Interest
PCR amplification or gel purification
Select specific DNA fragment
4. Vector Preparation
Cut vector with same restriction enzyme
Treat with alkaline phosphatase (prevent self-ligation)
5. Ligation
Join insert DNA with vector using T4 DNA ligase
Forms recombinant DNA molecule
6. Transformation
Introduce recombinant DNA into host cells
(E. coli, yeast, plant, animal cells)
7. Selection
Select transformed cells using antibiotic selection
Screen for recombinants (blue-white screening)
8. Characterization
Confirm insert by PCR, restriction mapping, sequencing
Expression analysis

Polymerase Chain Reaction (PCR)

Overview

  • Inventor: Kary Mullis (1983) - Nobel Prize 1993
  • Definition: In vitro enzymatic amplification of specific DNA sequences
  • Principle: Repeated cycles of denaturation, annealing, and extension
  • Amplification: Exponential (2n where n = number of cycles)
  • 30 cycles: ~1 billion copies (2³⁰ = 1,073,741,824)

Components of PCR

  • Template DNA: Contains target sequence (ng quantities sufficient)
  • Primers: Two oligonucleotides (18-25 bp)
    • Forward primer (5' end of target)
    • Reverse primer (3' end of target, reverse complement)
    • Tm (melting temperature): 55-65°C
  • DNA Polymerase: Thermostable (Taq, Pfu, Phusion)
    • Taq: Most common, from Thermus aquaticus
    • Optimal activity: 72°C
    • Synthesizes DNA at ~1000 bp/min
  • dNTPs: dATP, dGTP, dCTP, dTTP (building blocks)
  • Buffer: Maintains pH and ionic strength
    • Mg²⁺ (cofactor for polymerase, 1.5-2.5 mM)
    • KCl (50 mM)
    • Tris-HCl pH 8.3-8.8

🔄 PCR Cycle Steps

1. Denaturation (94-95°C, 30 sec)
Double-stranded DNA separates into single strands
Hydrogen bonds broken
2. Annealing (50-65°C, 30-60 sec)
Primers bind to complementary sequences
Temperature depends on primer Tm
3. Extension (72°C, 1 min/kb)
DNA polymerase synthesizes new strand
5' → 3' direction from primer
Repeat 25-40 cycles
Exponential amplification of target DNA
Final Extension (72°C, 5-10 min)
Complete synthesis of all products

Types/Variants of PCR

PCR Type Principle Applications
RT-PCR (Reverse Transcription PCR) RNA → cDNA → Amplification Gene expression analysis, viral RNA detection (COVID-19)
Real-Time PCR (qPCR) Quantification during amplification using fluorescent dyes/probes Quantitative gene expression, viral load measurement
Nested PCR Two rounds with two primer sets (outer + inner) Increased specificity, rare target detection
Multiplex PCR Multiple primer pairs in single reaction Amplify multiple targets simultaneously, genetic testing
Inverse PCR Amplifies unknown flanking regions Chromosome walking, unknown sequence determination
Long-range PCR Amplifies long DNA fragments (up to 40 kb) Amplification of large genes, genomic regions
Touchdown PCR Gradually decreasing annealing temperature Reduces non-specific amplification
Hot-Start PCR Polymerase activated only at high temp Prevents primer-dimer formation, increases specificity
Colony PCR Direct amplification from bacterial colonies Screening recombinant clones
RACE PCR Rapid Amplification of cDNA Ends Amplify 5' or 3' ends of mRNA
Digital PCR Absolute quantification by partitioning Precise quantification, rare mutation detection

Applications of PCR

  • Molecular Cloning: Amplify gene of interest
  • DNA Sequencing: Amplify template for Sanger sequencing
  • Medical Diagnostics: Detect pathogens, genetic diseases
  • Forensics: DNA fingerprinting, paternity testing
  • Gene Expression Analysis: RT-PCR, qPCR
  • Evolutionary Studies: Ancient DNA, phylogenetics
  • Site-Directed Mutagenesis: Introduce mutations
  • Prenatal Diagnosis: Detect genetic disorders

Advantages & Limitations

✅ Advantages

  • Rapid (2-4 hours)
  • Highly sensitive (single DNA molecule)
  • Specific (primers determine specificity)
  • Requires minimal template
  • Automated (thermal cycler)
  • Cost-effective

❌ Limitations

  • Requires sequence information (for primers)
  • Prone to contamination
  • Taq polymerase lacks proofreading (error rate 10⁻⁴)
  • Difficult to amplify GC-rich regions
  • Size limitation (~5-10 kb typically)
  • Primer-dimer and non-specific amplification

Gel Electrophoresis

Principle

  • Definition: Separation of charged molecules based on size and charge in electric field
  • DNA/RNA: Negatively charged (phosphate groups) → Migrate toward positive electrode (anode)
  • Separation: Smaller fragments move faster through gel matrix

Types of Gel Electrophoresis

Type Gel Matrix Used For Resolution Range
Agarose Gel Electrophoresis Agarose (0.5-2%) DNA, RNA fragments 50 bp - 50 kb
Polyacrylamide Gel (PAGE) Polyacrylamide (3.5-20%) Small DNA, RNA, proteins 5-500 bp (DNA), Proteins
SDS-PAGE Polyacrylamide + SDS Proteins (by molecular weight) 5-250 kDa
Native PAGE Polyacrylamide (no SDS) Proteins (by charge and size) Maintains protein structure
2D Gel Electrophoresis IEF + SDS-PAGE Complex protein mixtures Proteomics applications
Pulsed-Field Gel (PFGE) Agarose with alternating fields Very large DNA (chromosomes) 10 kb - 10 Mb

Agarose Gel Electrophoresis (AGE) - Detailed

  • Gel Preparation:
    • Agarose dissolved in buffer (TAE or TBE) by heating
    • Poured into casting tray with comb (forms wells)
    • Concentration: 0.5% (large DNA), 2% (small fragments)
  • Buffer Systems:
    • TAE (Tris-Acetate-EDTA): pH 8.3, better for large DNA, DNA recovery
    • TBE (Tris-Borate-EDTA): pH 8.3, higher resolution, better for small fragments
  • Loading Dye:
    • Glycerol/Ficoll (density - samples sink into wells)
    • Tracking dyes: Bromophenol blue, Xylene cyanol
  • DNA Ladder/Marker: Known size fragments for comparison (100 bp, 1 kb ladder)
  • Running Conditions: 50-150 V, 30-90 minutes
  • Visualization:
    • Ethidium Bromide (EtBr): Intercalates DNA, fluorescent under UV (302/366 nm)
      • Carcinogenic - handle with care
    • SYBR Safe/Green: Safer alternatives
    • Gel Red/Gel Green: Non-toxic dyes

Factors Affecting Migration

  • Size of DNA: Smaller = faster migration
  • Gel concentration: Higher % = better resolution of small fragments
  • Voltage: Higher voltage = faster but less resolution
  • DNA conformation: Supercoiled > Linear > Relaxed circular (same size)
  • Buffer ionic strength: Affects conductivity

Nucleic Acid Hybridization

Principle

  • Definition: Formation of double-stranded nucleic acid by base pairing between complementary single strands
  • Based on: Watson-Crick base pairing (A-T/U, G-C)
  • Process:
    1. Denaturation: Separate DNA strands (heat/alkali)
    2. Annealing: Complementary strands hybridize upon cooling
    3. Detection: Labeled probe detects target sequence
  • Stringency: Conditions that determine specificity
    • High stringency: Only perfect matches hybridize (high temp, low salt)
    • Low stringency: Mismatches tolerated (low temp, high salt)

Probes

  • Definition: Single-stranded DNA/RNA with known sequence, labeled for detection
  • Types of Labels:
    • Radioactive: ³²P, ³⁵S (highly sensitive, but hazardous, short half-life)
    • Non-radioactive:
      • Biotin (detected by streptavidin-enzyme conjugate)
      • Digoxigenin (DIG) - detected by anti-DIG antibody
      • Fluorescent dyes (FITC, Rhodamine, Cy3, Cy5)
  • Probe Preparation:
    • Nick translation
    • Random priming
    • PCR labeling
    • In vitro transcription (for RNA probes)

Blotting Techniques

Overview

  • Principle: Transfer of molecules from gel to membrane, followed by probe hybridization
  • Membranes Used:
  • Nitrocellulose: Binds proteins well, fragile
  • Nylon: More durable, binds nucleic acids strongly
  • PVDF (Polyvinylidene difluoride): For proteins, durable, high binding capacity

1. Southern Blotting

  • Developed by: Edwin Southern (1975)
  • Purpose: Detection of specific DNA sequences
  • Principle: DNA fragments separated by gel electrophoresis → transferred to membrane → hybridized with labeled probe

🧬 Southern Blotting Steps

1. DNA Digestion
Genomic DNA digested with restriction enzymes
2. Gel Electrophoresis
Separate DNA fragments on agarose gel
3. Depurination (Optional)
Brief acid treatment (0.25 M HCl)
Breaks large DNA for efficient transfer
4. Denaturation
Treat gel with alkali (0.5 M NaOH)
Converts dsDNA to ssDNA
5. Neutralization
Buffer treatment (Tris-HCl, pH 7.5)
6. Transfer
Capillary transfer, vacuum transfer, or electroblotting
DNA moves from gel to membrane
7. Fixation
UV crosslinking or baking (80°C, 2 hrs)
DNA permanently bound to membrane
8. Prehybridization
Block membrane with blocking solution
Prevents non-specific probe binding
9. Hybridization
Incubate with labeled probe (42-65°C)
Probe binds to complementary sequences
10. Washing
Remove unbound probe
High stringency washes
11. Detection
Autoradiography (radioactive) or
Chemiluminescence/Fluorescence (non-radioactive)
  • Applications:
    • Gene mapping and RFLP analysis
    • Detecting gene rearrangements, deletions, insertions
    • DNA fingerprinting in forensics
    • Diagnosis of genetic diseases (sickle cell, thalassemia)
    • Confirming transgene integration
    • Copy number determination

2. Northern Blotting

  • Named after: Southern blotting (not a person's name - directional pun)
  • Purpose: Detection and quantification of specific RNA molecules
  • Principle: Similar to Southern but for RNA
  • Key Differences:
    • RNA is already single-stranded (no denaturation needed before transfer)
    • Requires RNase-free conditions
    • Formaldehyde added to gel (prevents RNA secondary structure)
    • More stringent conditions
  • Gel: Formaldehyde-agarose or glyoxal gel
  • Transfer: Capillary or electroblotting to nylon membrane
  • Probe: Labeled DNA or RNA complementary to target mRNA
  • Applications:
    • Gene expression analysis (measure mRNA levels)
    • Study developmental regulation
    • Analyze RNA processing (splicing variants)
    • Detect viral RNA
    • Determine RNA size
  • Note: Largely replaced by RT-PCR and RNA-seq for gene expression studies

3. Western Blotting (Immunoblotting)

  • Developed by: W. Neal Burnette, Harry Towbin, George Stark (1979)
  • Purpose: Detection of specific proteins using antibodies
  • Principle: Protein separation → transfer → antibody detection

🔬 Western Blotting Steps

1. Sample Preparation
Protein extraction and quantification
Mixed with sample buffer (SDS, β-mercaptoethanol, glycerol)
2. Denaturation
Boiling at 95-100°C (5 min)
Proteins unfold, disulfide bonds reduced
3. SDS-PAGE
Proteins separated by molecular weight
SDS coats proteins with negative charge
4. Transfer (Blotting)
Electroblotting to PVDF or nitrocellulose
Wet transfer, semi-dry, or dry transfer
5. Blocking
Block with BSA or non-fat dry milk (1-5%)
1 hr at RT or overnight at 4°C
6. Primary Antibody Incubation
Specific antibody binds to target protein
1-2 hrs at RT or overnight at 4°C
7. Washing
TBST or PBST (3 × 10 min)
Remove unbound antibody
8. Secondary Antibody
Enzyme-conjugated (HRP, AP) or fluorescent
Binds to primary antibody, 1 hr at RT
9. Washing
TBST or PBST (3 × 10 min)
10. Detection
Chemiluminescence (HRP + substrate)
Colorimetric or fluorescence
  • Detection Systems:
    • HRP (Horseradish Peroxidase): Chemiluminescent substrate (ECL)
    • AP (Alkaline Phosphatase): Colorimetric substrates (BCIP/NBT)
    • Fluorescent: Fluorophore-conjugated secondary antibodies
  • Loading Control: β-actin, GAPDH, Tubulin (ensure equal protein loading)
  • Applications:
    • Protein expression analysis
    • Confirm protein size and identity
    • Detect post-translational modifications (phosphorylation)
    • Disease diagnosis (HIV, Lyme disease)
    • Quality control (recombinant protein production)

Comparison of Blotting Techniques

Feature Southern Blot Northern Blot Western Blot
Target Molecule DNA RNA Protein
Separation Method Agarose gel Formaldehyde-agarose SDS-PAGE
Separation Basis Size Size Molecular weight
Denaturation Required (alkali) Not required (already ss) Required (heat + SDS)
Membrane Nylon or nitrocellulose Nylon PVDF or nitrocellulose
Probe/Detection Labeled DNA/RNA probe Labeled DNA/RNA probe Antibodies (1° and 2°)
Hybridization Type DNA-DNA DNA-RNA or RNA-RNA Antigen-Antibody
Developer Edwin Southern (1975) No specific person Burnette/Towbin (1979)

Other Blotting Techniques

  • Southwestern Blotting: Detection of DNA-binding proteins
    • Proteins separated by PAGE → transferred → incubated with labeled DNA
  • Northwestern Blotting: Detection of RNA-binding proteins
    • Similar to Southwestern but uses labeled RNA probe
  • Eastern Blotting: Detection of post-translational modifications
    • Glycoproteins, lipid modifications
  • Far-Western Blotting: Detection of protein-protein interactions
    • Uses labeled protein as probe instead of antibody
  • Dot Blot/Slot Blot: No electrophoresis
    • Sample directly spotted on membrane
    • Quick qualitative/semi-quantitative analysis

Other Hybridization Techniques

1. Colony Hybridization

  • Purpose: Screen bacterial colonies for recombinant clones containing specific DNA insert
  • Procedure:
    1. Grow colonies on agar plate
    2. Transfer colonies to membrane (replica plating)
    3. Lyse bacteria, denature DNA in situ
    4. Fix DNA to membrane
    5. Hybridize with labeled probe
    6. Detect positive colonies
    7. Pick corresponding colony from master plate
  • Advantages:
    • Screen thousands of colonies simultaneously
    • Original colonies remain intact on master plate
    • Simple and rapid
  • Applications: Genomic library screening, identifying recombinant clones

2. Plaque Hybridization

  • Purpose: Screen bacteriophage plaques for specific DNA sequences
  • Similar to colony hybridization but for phage libraries
  • Procedure:
    1. Grow phage plaques on bacterial lawn
    2. Transfer to membrane
    3. Denature DNA, fix to membrane
    4. Hybridize with labeled probe
    5. Detect positive plaques
    6. Pick corresponding plaque and amplify
  • Applications: λ phage library screening, cDNA library screening

3. In Situ Hybridization (ISH)

  • Definition: Hybridization technique performed directly on tissue sections or whole cells
  • Purpose: Localize specific nucleic acid sequences within cells/tissues
  • Principle: Labeled probe hybridizes to target DNA/RNA in fixed cells/tissues
  • Types:
    • Chromogenic ISH (CISH): Colorimetric detection
    • Fluorescence ISH (FISH): Fluorescent probes
    • RNA-FISH: Detects RNA in cells
  • Procedure:
    1. Tissue fixation (formaldehyde, paraformaldehyde)
    2. Permeabilization (protease treatment)
    3. Denaturation (for DNA targets)
    4. Hybridization with labeled probe
    5. Washing to remove unbound probe
    6. Detection and microscopy
  • Applications:
    • Gene mapping on chromosomes
    • Prenatal diagnosis of chromosomal abnormalities
    • Cancer diagnostics (HER2 amplification)
    • Detecting viral infections in tissues
    • Study gene expression patterns in development
    • Evolutionary studies

4. Fluorescence In Situ Hybridization (FISH)

  • Most widely used ISH technique
  • Probes: Labeled with fluorochromes (FITC, Rhodamine, Cy3, Cy5, Texas Red)
  • Detection: Fluorescence microscopy or flow cytometry
  • Types of FISH Probes:
    • Centromeric probes: Detect chromosome number (aneuploidy)
    • Locus-specific probes: Detect specific gene deletions/amplifications
    • Whole chromosome probes: Paint entire chromosome
    • Telomeric probes: Detect chromosome ends
  • Multicolor FISH:
    • M-FISH: Each chromosome labeled with different color
    • SKY (Spectral Karyotyping): All 24 chromosomes (22 + X + Y) in different colors
  • Applications:
    • Prenatal diagnosis (Down syndrome - Trisomy 21, Turner syndrome)
    • Cancer cytogenetics (Philadelphia chromosome, gene amplifications)
    • Microdeletion syndromes detection
    • Sex determination
    • Comparative genomic studies
  • Advantages over conventional ISH:
    • Rapid (hours vs days)
    • Multiple targets detected simultaneously (different colors)
    • Higher resolution
    • Non-radioactive

5. Comparative Genomic Hybridization (CGH)

  • Purpose: Detect DNA copy number changes across entire genome
  • Principle: Compare test and reference DNA by competitive hybridization
  • Procedure:
    • Label test DNA (e.g., tumor) with green fluorophore
    • Label reference DNA (normal) with red fluorophore
    • Mix and hybridize to normal metaphase chromosomes or DNA microarray
    • Analyze fluorescence ratio
  • Interpretation:
    • Green > Red: Gain/amplification in test
    • Red > Green: Loss/deletion in test
    • Yellow (equal): Normal copy number
  • Array CGH (aCGH): Higher resolution using microarrays instead of chromosomes
  • Applications:
    • Cancer genomics (oncogene amplification, tumor suppressor deletion)
    • Prenatal diagnosis
    • Genetic disorder screening

🎯 High-Yield Points for CSIR NET & ICAR SRF

  • CSIR NET rDNA Technology: Stanley Cohen & Herbert Boyer (1973) - First recombinant organism
  • ICAR SRF PCR: Invented by Kary Mullis (1983), Nobel Prize 1993
  • CSIR NET PCR Amplification: Exponential (2n), 30 cycles = ~1 billion copies
  • ICAR SRF Taq polymerase: From Thermus aquaticus, optimal at 72°C
  • CSIR NET PCR Steps: Denaturation (94-95°C), Annealing (50-65°C), Extension (72°C)
  • ICAR SRF RT-PCR: RNA → cDNA → Amplification, used for gene expression
  • CSIR NET Real-Time PCR (qPCR): Quantitative, uses fluorescent dyes/probes
  • ICAR SRF Agarose concentration: 0.5% for large DNA, 2% for small fragments
  • CSIR NET TAE vs TBE: TAE better for large DNA, TBE better for small fragments
  • ICAR SRF Ethidium Bromide: Intercalates DNA, fluoresces under UV (302/366 nm)
  • CSIR NET Southern Blot: Edwin Southern (1975), detects DNA
  • ICAR SRF Northern Blot: Detects RNA, uses formaldehyde-agarose gel
  • CSIR NET Western Blot: Detects proteins using antibodies, SDS-PAGE separation
  • ICAR SRF Blot Membranes: Nitrocellulose (proteins), Nylon (nucleic acids), PVDF (proteins)
  • CSIR NET Colony Hybridization: Screen bacterial colonies for recombinant clones
  • ICAR SRF FISH: Fluorescence in situ hybridization, uses fluorescent probes
  • CSIR NET Stringency: High stringency = perfect match only (high temp, low salt)
  • ICAR SRF Probe Labels: Radioactive (³²P), Non-radioactive (Biotin, DIG, Fluorescent)
  • CSIR NET CGH: Comparative genomic hybridization, detects copy number changes
  • ICAR SRF M-FISH/SKY: Each chromosome in different color, 24 human chromosomes

💡 Memory Tricks & Mnemonics

  • Gene Cloning Steps: "I Feel I'm Very Little (to) See Clearly"
    • Isolation, Fragmentation, Isolation of gene, Vector prep, Ligation, Transformation, Selection, Characterization
  • PCR Steps: "DAE" - Denaturation, Annealing, Extension
  • PCR Temperatures: "95-55-72" (Denaturation-Annealing-Extension)
  • Blotting Techniques: "Directions"
    • Southern (South) = DNA
    • Northern (North) = RNA
    • Western (West) = Protein
    • Eastern (East) = Post-translational modifications
  • Southern Blot Steps: "Don't Drink During Nice Trips From Prague Hoping Weather Doesn't Disappoint"
    • Digestion, Denature, Depurination (optional), Neutralize, Transfer, Fix, Prehybridize, Hybridize, Wash, Detect, Detection
  • Western Blot: "Some People Drink Beer Before Primary School Wash Days"
    • Sample prep, PAGE, Denaturation, Blot/transfer, Blocking, Primary antibody, Secondary antibody, Wash, Detect
  • FISH Applications: "Cancer Patients Get Sad"
    • Cancer diagnosis, Prenatal diagnosis, Gene mapping, Sex determination

📝 Common MCQ Patterns

  • Cohen & Boyer: Year (1973), contribution (first recombinant DNA)
  • Kary Mullis: Invented PCR (1983), Nobel Prize 1993
  • PCR Components: Template, primers, polymerase, dNTPs, buffer
  • PCR Temperature: Which step at which temperature
  • Taq vs Pfu: Taq has no proofreading, Pfu has proofreading (high fidelity)
  • RT-PCR vs Real-Time PCR: RT-PCR uses RNA, Real-Time is quantitative
  • Gel Concentration: Higher % for smaller fragments
  • TAE vs TBE: Uses and advantages
  • Blotting Inventor: Only Southern has a person's name (Edwin Southern)
  • Blot Detection: Southern/Northern use probes, Western uses antibodies
  • Membrane Types: Which membrane for which application
  • Colony vs Plaque Hybridization: Colony for bacteria, Plaque for bacteriophage
  • CGH Interpretation: Green > Red (gain), Red > Green (loss), Yellow (normal)
  • FISH Applications: Prenatal diagnosis, cancer, chromosome mapping

📖 CHAPTER 4: DNA/RNA Libraries & Gene Cloning Applications

Library Construction, Screening & Applications

DNA/RNA Libraries

Definition

  • Gene Library: Collection of cloned DNA fragments representing entire genome or expressed genes of an organism
  • Purpose: Store and maintain genetic information for research and applications
  • Types: Genomic library and cDNA library

Genomic Library

Overview

  • Definition: Collection of clones containing all DNA sequences of entire genome
  • Includes: Coding sequences (exons), non-coding sequences (introns), regulatory regions, repetitive DNA
  • Represents: Complete genetic makeup of organism
  • Species-specific: Each organism has unique genomic library

🧬 Construction of Genomic Library

1. Isolation of Genomic DNA
Extract high molecular weight DNA from organism
Quality check (intact, no degradation)
2. Fragmentation
Partial digestion with restriction enzymes OR
Physical shearing (sonication, nebulization)
Generate overlapping fragments (10-50 kb)
3. Size Selection
Agarose gel electrophoresis
Select fragments of appropriate size
4. Vector Preparation
Choose appropriate vector (λ phage, cosmid, BAC, YAC)
Digest vector with compatible restriction enzyme
Dephosphorylate with alkaline phosphatase
5. Ligation
Mix DNA fragments with vector
T4 DNA ligase (overnight at 16°C)
Creates recombinant molecules
6. Packaging (for phage/cosmid)
In vitro packaging into phage particles
OR transformation into bacterial cells (for plasmids)
7. Transformation/Transfection
Introduce into host cells (E. coli, yeast)
Plate on selective medium
8. Library Storage
Collect and store all clones
Glycerol stocks at -80°C

Library Size Calculation

  • Formula: N = ln(1-P) / ln(1-f)
  • Where:
    • N = Number of clones needed
    • P = Probability of finding desired sequence (usually 0.99 or 99%)
    • f = Fraction of genome in each clone = Insert size / Genome size
  • Example: Human genome (3 × 10⁹ bp), insert size 20 kb
    • f = 20,000 / 3,000,000,000 = 6.67 × 10⁻⁶
    • N = ln(1-0.99) / ln(1-6.67×10⁻⁶) ≈ 690,000 clones needed

Screening Genomic Library

  • Colony/Plaque Hybridization:
    • Transfer colonies to membrane
    • Lyse cells, fix DNA
    • Hybridize with labeled probe
    • Detect positive clones
  • PCR Screening:
    • Pool clones and perform PCR
    • Deconvolute pools to identify positive clone
  • Expression Screening:
    • For genes encoding proteins
    • Use antibodies to detect expressed protein
  • Functional Complementation:
    • Transform mutant strain
    • Select clones that restore function

Advantages & Limitations

✅ Advantages

  • Contains complete genetic information
  • Includes regulatory sequences (promoters, enhancers)
  • Preserves gene structure (exons + introns)
  • Can study gene organization
  • Useful for chromosome walking

❌ Limitations

  • Very large size required
  • Contains non-coding DNA (98% in humans)
  • Difficult to screen
  • Eukaryotic genes not expressed in prokaryotes (introns)
  • Time-consuming and expensive

cDNA Library (Complementary DNA Library)

Overview

  • Definition: Collection of cloned cDNA sequences representing all mRNAs expressed in specific cell/tissue/developmental stage
  • cDNA: DNA synthesized from mRNA template using reverse transcriptase
  • Contains only: Coding sequences (exons only, no introns)
  • Tissue/Stage-specific: Reflects gene expression pattern
  • Smaller than genomic library: Only represents ~2% of genome (protein-coding genes)

🔬 Construction of cDNA Library

1. mRNA Isolation
Extract total RNA from tissue/cells
Purify mRNA using oligo(dT) column
(Binds to poly-A tail of mRNA)
2. First Strand cDNA Synthesis
Oligo(dT) primer anneals to poly-A tail
Reverse transcriptase synthesizes cDNA (3'→5' on mRNA)
Forms mRNA-cDNA hybrid
3. Second Strand Synthesis
Method A: RNase H creates nicks in mRNA
DNA Pol I uses RNA fragments as primers
Method B: Degrade mRNA with alkali
Use random primers or specific primers
4. End Modification
Blunt ends created (Klenow fragment or T4 DNA polymerase)
Add linkers/adapters with restriction sites
OR add poly-C/poly-G tails (homopolymeric tailing)
5. Ligation into Vector
Plasmid or λ phage vector
T4 DNA ligase
6. Transformation
Introduce into E. coli
Plate on selective medium
7. Library Storage
Collect all clones
Store as glycerol stocks at -80°C

Screening cDNA Library

  • Hybridization Screening:
    • Colony/plaque hybridization with labeled probe
    • Heterologous probes from related species
  • Immunological Screening:
    • Use antibodies against target protein
    • Requires expression vector
    • Works only if protein is expressed and antigenic
  • Expression Cloning:
    • Functional assay for specific activity
    • Screen for phenotype
  • Differential Screening:
    • Compare with probes from different tissues/conditions
    • Identify tissue-specific or condition-specific genes

Advantages & Limitations

✅ Advantages

  • Smaller library size
  • Only coding sequences (no introns)
  • Can be expressed in bacteria
  • Direct protein production possible
  • Reflects gene expression pattern
  • Easier to screen
  • Less storage space required

❌ Limitations

  • No regulatory sequences
  • Tissue/stage-specific (incomplete representation)
  • Highly expressed genes over-represented
  • Rare transcripts may be missed
  • Full-length clones difficult to obtain
  • No information about gene structure
  • Cannot study introns or regulatory regions

Genomic Library vs cDNA Library

Feature Genomic Library cDNA Library
Source Genomic DNA mRNA (via reverse transcriptase)
Sequences Present Exons + Introns + Regulatory regions Only exons (coding sequences)
Size Very large (millions of clones) Smaller (thousands to hundred thousands)
Representation Complete genome Only expressed genes
Tissue Specificity Same for all tissues Different for each tissue/stage
Expression in E. coli Difficult (introns present) Possible (no introns)
Protein Production Not directly possible Directly possible
Gene Structure Preserved (exon-intron organization) Lost (only mature mRNA sequence)
Screening Difficulty More difficult Easier
Applications Gene mapping, regulatory studies, chromosome walking Protein production, expression studies, functional analysis
Vectors Used λ phage, Cosmids, BAC, YAC (large inserts) Plasmids, λ phage (smaller inserts)

Applications of Gene Cloning

A. Basic Research Applications

1. Gene Structure & Function Studies

  • Determine DNA sequence of genes
  • Identify regulatory elements (promoters, enhancers, silencers)
  • Study gene organization (exon-intron structure)
  • Analyze alternative splicing patterns
  • Investigate gene expression regulation

2. Protein Structure-Function Analysis

  • Produce large quantities of protein for structural studies
  • Site-directed mutagenesis to study functional domains
  • X-ray crystallography and NMR studies
  • Enzyme kinetics studies
  • Protein-protein interaction studies

3. Evolutionary Studies

  • Phylogenetic analysis by comparing gene sequences
  • Study molecular evolution
  • Analyze conserved vs variable regions
  • Determine evolutionary relationships

4. Model Organism Studies

  • Create transgenic model organisms (Drosophila, C. elegans, mice)
  • Gene knockout/knockin studies
  • Study gene function in development
  • Disease modeling

B. Applied Research & Biotechnology Applications

1. Recombinant Protein Production

  • Therapeutic Proteins:
    • Insulin: First recombinant therapeutic (1982, Humulin)
      • Produced in E. coli and yeast
      • Treats diabetes
    • Human Growth Hormone (hGH):
      • Treats growth hormone deficiency
      • Replaced pituitary-derived hormone
    • Interferons (IFN-α, IFN-β, IFN-γ):
      • Antiviral and anticancer properties
      • Treat hepatitis B, C, multiple sclerosis
    • Erythropoietin (EPO):
      • Stimulates red blood cell production
      • Treats anemia in kidney disease patients
    • Blood Clotting Factors:
      • Factor VIII, Factor IX for hemophilia
      • Safer than blood-derived products
    • Tissue Plasminogen Activator (tPA):
      • Dissolves blood clots
      • Treats heart attack and stroke
  • Industrial Enzymes:
    • Amylases for starch processing
    • Proteases for detergents
    • Lipases for biodiesel production
    • Cellulases for biofuel production

2. Vaccine Development

  • Subunit Vaccines:
    • Hepatitis B Vaccine: Surface antigen (HBsAg) produced in yeast
    • Safer than traditional killed/attenuated vaccines
    • No risk of infection
  • DNA Vaccines:
    • Plasmid DNA encoding antigen
    • Host cells produce antigen
    • Elicits both humoral and cell-mediated immunity
  • Edible Vaccines:
    • Antigen genes expressed in plants (potato, banana)
    • Oral delivery system
    • Cost-effective for developing countries

3. Gene Therapy

  • Definition: Treatment of genetic diseases by introducing functional genes
  • Types:
    • Somatic Gene Therapy: Modifies body cells (not heritable)
    • Germline Gene Therapy: Modifies gametes/embryos (heritable, controversial)
  • Approaches:
    • Gene Replacement: Replace defective gene
    • Gene Augmentation: Add functional copy of gene
    • Gene Inactivation: Silence harmful gene
  • Vectors for Gene Therapy:
    • Viral Vectors:
      • Retroviruses: Integrate into genome, but only infect dividing cells
      • Lentiviruses: Can infect non-dividing cells
      • Adenoviruses: High efficiency, but transient expression
      • Adeno-associated Virus (AAV): Safe, but small insert capacity
    • Non-viral Vectors:
      • Liposomes (lipid vesicles)
      • Naked DNA injection
      • Electroporation
      • Gene gun
  • Clinical Applications:
    • SCID (Severe Combined Immunodeficiency): ADA-SCID treated successfully
    • Hemophilia B: Factor IX gene therapy
    • Inherited blindness: RPE65 gene for Leber's congenital amaurosis
    • Beta-thalassemia: β-globin gene transfer
    • CAR-T cell therapy: Cancer immunotherapy
  • Challenges:
    • Immune response to vector
    • Insertional mutagenesis (cancer risk)
    • Transient expression
    • Targeting specific cells
    • Ethical concerns

4. Molecular Diagnostics

  • DNA Probes:
    • Detect specific DNA sequences in samples
    • Diagnose infectious diseases (HIV, HPV, TB)
    • Identify genetic mutations
  • PCR-based Diagnostics:
    • Rapid detection of pathogens
    • COVID-19 RT-PCR testing
    • Genetic disease screening
  • Genetic Testing:
    • Carrier screening (cystic fibrosis, sickle cell)
    • Prenatal diagnosis
    • Presymptomatic testing (Huntington's disease)
    • Pharmacogenomics (drug response prediction)
  • Cancer Diagnostics:
    • Detect oncogene amplifications (HER2 in breast cancer)
    • Identify tumor suppressor mutations (p53, BRCA1/2)
    • Personalized cancer treatment

5. Forensic Applications

  • DNA Fingerprinting/Profiling:
    • Uses STRs (Short Tandem Repeats) and VNTRs (Variable Number Tandem Repeats)
    • 13-20 STR loci analyzed (CODIS system in USA)
    • Probability of match: 1 in billions
  • Applications:
    • Criminal investigations
    • Paternity testing
    • Disaster victim identification
    • Missing person identification
    • Wildlife forensics (poaching cases)
  • Y-Chromosome Analysis: Paternal lineage tracing
  • Mitochondrial DNA: Maternal lineage, degraded samples

6. Agricultural Applications

  • Transgenic Crops:
    • Insect resistance (Bt crops)
    • Herbicide tolerance (Roundup Ready)
    • Disease resistance (virus-resistant papaya)
    • Improved nutritional quality (Golden Rice)
    • Extended shelf life (Flavr Savr tomato)
    • Drought/salinity tolerance
  • Transgenic Animals:
    • Produce pharmaceuticals in milk (pharming)
    • Disease-resistant livestock
    • Enhanced growth (AquAdvantage salmon)
    • Organ donors for xenotransplantation (pigs)

7. Environmental Applications

  • Bioremediation:
    • Engineered bacteria for oil spill cleanup
    • Heavy metal removal (phytoremediation)
    • Degradation of toxic compounds (PCBs, TNT)
  • Biosensors:
    • Detect environmental pollutants
    • Monitor water quality
    • Early warning systems
  • Biofuel Production:
    • Engineered microbes for enhanced ethanol production
    • Algae for biodiesel
    • Cellulosic biofuel production

8. Industrial Biotechnology

  • Bioplastics:
    • Polyhydroxybutyrate (PHB) production
    • Biodegradable plastics from bacteria
  • Fine Chemicals:
    • Amino acids (lysine, glutamate)
    • Vitamins (B12, riboflavin)
    • Organic acids (citric acid, lactic acid)
  • Biocatalysis:
    • Enzyme production for industrial processes
    • Chiral compound synthesis

C. Expression Systems for Recombinant Proteins

System Advantages Disadvantages Best For
E. coli • Fast growth
• High yield
• Low cost
• Well-characterized
• No post-translational modifications
• Inclusion bodies
• No glycosylation
Simple proteins, industrial enzymes
Yeast (S. cerevisiae, P. pastoris) • Eukaryotic modifications
• Secretion possible
• GRAS status
• High yield
• Hyperglycosylation
• Different glycan patterns
Insulin, hepatitis B vaccine, industrial enzymes
Insect Cells (Baculovirus) • Proper folding
• Post-translational modifications
• High expression
• Expensive
• Time-consuming
• Different glycosylation
Viral proteins, vaccines, research proteins
Mammalian Cells (CHO, HEK293) • Human-like modifications
• Proper folding
• Correct glycosylation
• Very expensive
• Slow growth
• Low yield
• Complex culture
Therapeutic proteins (antibodies, EPO, Factor VIII)
Transgenic Animals • Large-scale production
• Natural purification (milk)
• Human modifications
• Very expensive
• Long time to establish
• Ethical concerns
Complex proteins, antithrombin, α-1-antitrypsin
Transgenic Plants • Very low cost
• Scalable
• Safe
• Oral delivery
• Variable expression
• Different glycosylation
• Low yield often
Vaccines, antibodies, industrial proteins

🎯 High-Yield Points for CSIR NET & ICAR SRF

  • CSIR NET Genomic Library: Contains entire genome (exons + introns + regulatory sequences)
  • ICAR SRF cDNA Library: Only coding sequences (exons), no introns, made from mRNA
  • CSIR NET Library Size Formula: N = ln(1-P) / ln(1-f), where P = 0.99 (99% probability)
  • ICAR SRF Reverse Transcriptase: Synthesizes cDNA from mRNA template (first strand)
  • CSIR NET Oligo(dT) primer: Anneals to poly-A tail of mRNA for cDNA synthesis
  • ICAR SRF Genomic library vectors: λ phage, Cosmids, BAC, YAC (large insert capacity)
  • CSIR NET cDNA library vectors: Plasmids, λ phage (smaller inserts)
  • ICAR SRF First recombinant therapeutic: Insulin (Humulin, 1982)
  • CSIR NET Hepatitis B vaccine: HBsAg produced in yeast (first recombinant vaccine)
  • ICAR SRF Gene Therapy Types: Somatic (not heritable) vs Germline (heritable)
  • CSIR NET Viral Vectors: Retroviruses, Lentiviruses, Adenoviruses, AAV
  • ICAR SRF SCID-X1: First successful gene therapy (ADA-SCID)
  • CSIR NET DNA Fingerprinting: Uses STRs (Short Tandem Repeats) and VNTRs
  • ICAR SRF CODIS: 13-20 STR loci used in forensic DNA profiling
  • CSIR NET E. coli expression: Fast, high yield, but no glycosylation
  • ICAR SRF CHO cells: Mammalian expression, proper glycosylation, for therapeutics
  • CSIR NET Baculovirus system: Insect cells, proper folding, used for vaccines
  • ICAR SRF Golden Rice: β-carotene (Vitamin A), developed by Ingo Potrykus
  • CSIR NET Pharming: Production of pharmaceuticals in transgenic animals/plants
  • ICAR SRF Molecular pharming examples: α-1-antitrypsin in sheep milk, antithrombin in goat milk

💡 Memory Tricks & Mnemonics

  • Library Types: "GC"
    • Genomic = Gene structure (all DNA)
    • CDNA = Coding sequences only
  • cDNA Synthesis Steps: "MFSE"
    • mRNA isolation, First strand, Second strand, End modification
  • Recombinant Therapeutics: "I Help Everyone Fight Illness"
    • Insulin, Heparin (hGH), Erythropoietin, Factors (VIII, IX), Interferons
  • Gene Therapy Vectors: "RALA" (viral)
    • Retroviruses, Adenoviruses, Lentiviruses, AAV
  • Expression Systems: "EYIM" (increasing complexity)
    • E. coli, Yeast, Insect cells, Mammalian cells
  • DNA Fingerprinting: "STRs are Short, VNTRs are Variable"

📝 Common MCQ Patterns

  • Genomic vs cDNA Library: Differences in source, content, size, applications
  • Library Construction: Steps, enzymes used, vectors required
  • Screening Methods: Which method for which library type
  • Reverse Transcriptase: Function, source (retroviruses), applications
  • First Recombinant Products: Insulin (1982), hGH, Interferons
  • Hepatitis B Vaccine: HBsAg, produced in yeast, subunit vaccine
  • Gene Therapy: Somatic vs germline, vectors, diseases treated
  • Viral Vectors: Properties, advantages, limitations of each
  • SCID Treatment: ADA-SCID, first successful gene therapy
  • Expression Systems: E. coli (no glycosylation), Yeast (hyperglycosylation), Mammalian (correct)
  • DNA Fingerprinting: STRs, VNTRs, applications in forensics
  • Transgenic Applications: Bt crops, Golden Rice, Flavr Savr, pharming
  • Molecular Diagnostics: PCR-based, probes, genetic testing applications

📖 CHAPTER 5: Plant Transformation & GM Crops

Gene Transfer Methods & Applications

Plant Transformation - Overview

Definition

  • Plant Transformation: Introduction of foreign DNA into plant cells resulting in stable integration and expression
  • Transgenic Plant: Plant containing stably integrated foreign gene(s) from another species
  • Goal: Improve crop traits - yield, quality, resistance to biotic/abiotic stress

Methods of Gene Transfer

  • Biological Methods:
    • Agrobacterium-mediated transformation
    • Viral vectors (limited use)
  • Physical/Direct Methods:
    • Biolistics (Gene gun/Particle bombardment)
    • Electroporation
    • PEG-mediated transformation
    • Microinjection
    • Silicon carbide whiskers

Agrobacterium-Mediated Transformation

Overview

  • Most widely used method for dicot transformation
  • Agent: Agrobacterium tumefaciens (causes crown gall disease)
  • Natural Genetic Engineer: Transfers part of its DNA (T-DNA) into plant genome
  • Discovered by: Georges Morel and colleagues (1970s)

🌿 Agrobacterium-Mediated Transformation Process

1. Wounding
Plant tissue wounded to release phenolic compounds
(Acetosyringone, hydroxyacetosyringone)
2. Chemotaxis
Agrobacterium attracted to wound site
Phenolics induce vir genes
3. Vir Gene Activation
VirA (sensor) detects phenolics
Activates VirG (transcription factor)
All vir genes expressed
4. T-DNA Processing
VirD1/D2 create single-stranded T-DNA
VirD2 remains attached to 5' end
VirE2 coats T-DNA strand
5. T-DNA Transfer
VirB complex forms channel
T-complex (T-DNA + proteins) transferred to plant cell
6. Nuclear Import
VirD2 and VirE2 have nuclear localization signals
T-DNA imported into nucleus
7. Integration
T-DNA integrates into plant chromosome
Random integration, usually single copy
8. Expression
Transgenes expressed
Plant promoters drive expression

Ti Plasmid Components

  • T-DNA Region (15-30 kb):
    • Transferred to plant
    • Flanked by 25 bp direct repeats (left and right borders)
    • Contains:
      • aux genes: Auxin biosynthesis
      • cyt genes: Cytokinin biosynthesis
      • ocs/nos genes: Opine synthesis
  • Vir Region (~35 kb):
    • NOT transferred to plant
    • Essential for T-DNA transfer
    • Contains virA, virB, virC, virD, virE, virG operons
  • Opine Catabolism Region:
    • Allows Agrobacterium to use opines as carbon/nitrogen source
  • Origin of Replication: For plasmid maintenance

Binary Vector System

  • Problem: Ti plasmid too large (~200 kb) to manipulate
  • Solution: Separate into two plasmids
    • Binary Vector: Small (~10-15 kb)
      • Contains T-DNA with foreign gene
      • Selectable marker genes
      • Multiple cloning site
      • 25 bp border repeats
      • Can replicate in E. coli and Agrobacterium
    • Helper Plasmid: Contains vir genes
      • Provides vir functions in trans
      • Not transferred to plant
  • Examples: pBI121, pCAMBIA vectors

Selectable Marker Genes

  • Antibiotic Resistance:
    • nptII: Kanamycin/neomycin resistance
    • hpt: Hygromycin resistance
    • bar: Phosphinothricin resistance
  • Herbicide Resistance:
    • bar/pat: Bialaphos/phosphinothricin resistance
    • cp4-epsps: Glyphosate resistance

Reporter Genes

  • GUS (β-glucuronidase):
    • From E. coli uidA gene
    • Converts X-Gluc substrate to blue product
    • Visual detection of transgene expression
  • GFP (Green Fluorescent Protein):
    • From jellyfish Aequorea victoria
    • Fluoresces green under UV/blue light
    • Non-destructive, real-time visualization
  • luciferase: Bioluminescence (firefly, Renilla)

Advantages & Limitations

✅ Advantages

  • Natural mechanism
  • High efficiency (dicots)
  • Single/low copy integration
  • Precise integration (mostly)
  • Large DNA fragments (up to 150 kb)
  • Cost-effective
  • Well-characterized system

❌ Limitations

  • Limited host range (mainly dicots)
  • Monocots less susceptible
  • Random integration
  • Agrobacterium genes may transfer
  • Position effect on expression
  • Time-consuming
  • Tissue culture required

Overcoming Monocot Recalcitrance

  • Use of super-virulent strains (EHA105, AGL1)
  • Prolonged co-cultivation (3-7 days)
  • Addition of acetosyringone (vir gene inducer)
  • Use of meristematic tissues (immature embryos)
  • Vacuum infiltration or sonication
  • Now successfully used in rice, maize, wheat, barley

Biolistics (Gene Gun / Particle Bombardment)

Overview

  • Developed by: John Sanford and colleagues (1987)
  • Principle: High-velocity microprojectiles coated with DNA penetrate plant cells
  • Also called: Microprojectile bombardment, Particle gun method
  • Advantage: Works for all plant species (monocots, dicots, gymnosperms)

Components

  • Microprojectiles:
    • Gold particles (0.6-1.6 μm diameter) - most common
    • Tungsten particles (1.0-1.2 μm) - cheaper but toxic
    • Coated with plasmid DNA
  • Gene Gun Device:
    • Particle Inflow Gun (PIG)
    • PDS-1000/He (Bio-Rad)
    • Uses helium or gunpowder as propellant
  • Target Tissue:
    • Embryogenic callus
    • Immature embryos
    • Meristematic tissues

🔫 Biolistics Procedure

1. DNA-Particle Coating
Mix plasmid DNA with gold/tungsten particles
Precipitate DNA onto particles (CaCl₂, spermidine)
2. Target Preparation
Place plant tissue on medium
Osmotic pretreatment (0.2-0.4 M mannitol/sorbitol)
3. Bombardment
Vacuum chamber (27-28 inches Hg)
Helium pressure (1100-1550 psi)
Distance: 6-12 cm from target
4. Recovery
Incubate on non-selective medium (1-2 days)
Allow cells to recover from bombardment
5. Selection
Transfer to selective medium
Select transformed cells (antibiotic/herbicide)
6. Regeneration
Shoot induction → Root induction
Plantlet development
7. Hardening & Transfer
Acclimatize to greenhouse → field conditions

Parameters Affecting Efficiency

  • Particle size: 0.6-1.6 μm (smaller = deeper penetration)
  • Helium pressure: 1100-1550 psi (higher = deeper penetration)
  • Target distance: 6-12 cm (closer = more damage)
  • Vacuum level: 27-28 inches Hg (reduces air resistance)
  • DNA concentration: 0.5-5 μg
  • Tissue type: Embryogenic cells best
  • Pre-bombardment treatment: Osmotic (plasmolysis protects cells)

Advantages & Limitations

✅ Advantages

  • Universal - all plant species
  • Especially good for monocots
  • Can target organelles (chloroplast, mitochondria)
  • No biological vector needed
  • Multiple genes can be delivered
  • Works on recalcitrant species

❌ Limitations

  • Expensive equipment
  • Low transformation efficiency
  • Multiple copy integration
  • Gene silencing issues
  • Tissue damage
  • Rearrangement of transgenes
  • Labor-intensive

Applications

  • Transformation of monocots (rice, maize, wheat, barley)
  • Organelle transformation (chloroplast, mitochondria)
  • Gymnosperm transformation (conifers)
  • Algae transformation
  • Transient expression studies

Other Gene Transfer Methods

1. Electroporation

  • Principle: Electric pulses create transient pores in cell membrane
  • Protocol:
    • Prepare protoplasts (remove cell wall with cellulase, pectinase)
    • Mix protoplasts with plasmid DNA
    • Apply electric pulse (100-400 V/cm, 10-50 ms)
    • DNA enters through pores
    • Pores reseal after pulse
  • Advantages: Simple, efficient for protoplasts, works for many species
  • Limitations: Requires protoplast preparation, regeneration difficult, low survival
  • Applications: Rice, maize, tobacco, potato transformation

2. PEG-Mediated Transformation

  • Principle: Polyethylene glycol (PEG) facilitates DNA uptake by protoplasts
  • Mechanism: PEG destabilizes membranes, promotes DNA-membrane interaction
  • Protocol:
    • Prepare protoplasts
    • Incubate with DNA in PEG solution (40% PEG-4000 or 6000)
    • Add CaCl₂ (enhances uptake)
    • Wash and culture protoplasts
  • Advantages: Simple, inexpensive, no special equipment
  • Limitations: Low efficiency, protoplast regeneration needed
  • Applications: Rice, maize, tobacco, carrot

3. Microinjection

  • Principle: Direct injection of DNA into plant cell nucleus or cytoplasm
  • Requires: Micromanipulator, micropipette, microscope
  • Target: Large cells (protoplasts, embryos, egg cells)
  • Advantages: Precise, can target specific cells/organelles
  • Limitations: Extremely labor-intensive, low throughput, requires expertise
  • Applications: Mainly research, not commercial scale

4. Silicon Carbide Whiskers (WHISKERS Method)

  • Principle: Silicon carbide fibers create wounds in cell walls allowing DNA entry
  • Protocol:
    • Mix cells/tissues with DNA and silicon carbide whiskers
    • Vortex vigorously (creates micro-wounds)
    • DNA enters through wounds
  • Advantages: Simple, no special equipment, cost-effective
  • Limitations: Variable efficiency, cell damage
  • Applications: Maize, rice, cotton

5. Pollen Tube Pathway Method

  • Principle: DNA injected into ovary through cut style enters ovule via pollen tube
  • Developed in China (Zhou et al., 1983)
  • Protocol:
    • Pollinate flowers
    • Cut style after pollination
    • Apply DNA solution to cut end
    • DNA travels to ovule through pollen tube pathway
  • Advantages: No tissue culture needed, simple
  • Limitations: Very low efficiency, reproducibility issues, controversial

Comparison of Gene Transfer Methods

Method Host Range Efficiency Integration Best For
Agrobacterium Dicots, some monocots High Single/low copy Dicots, stable transformation
Biolistics Universal (all plants) Low-Medium Multiple copies Monocots, organelles
Electroporation Protoplasts (all species) Medium Variable Species with easy regeneration
PEG-mediated Protoplasts (all species) Low-Medium Variable Cereals, simple system
Microinjection All (single cells) Low Precise Research, specific targeting

Applications of Genetically Modified (GM) Crops

1. Insect Resistance

Bt Crops

  • Gene Source: Bacillus thuringiensis (soil bacterium)
  • Bt Toxins (Cry Proteins):
    • Cry1Ac: Lepidoptera (caterpillars)
    • Cry1Ab: Lepidoptera
    • Cry2Ab: Lepidoptera
    • Cry3A: Coleoptera (beetles)
    • Cry9C: Lepidoptera
  • Mechanism:
    1. Insect ingests Bt toxin (inactive protoxin)
    2. Alkaline gut pH activates toxin
    3. Toxin binds to gut receptors
    4. Creates pores in gut membrane
    5. Gut cells lyse → insect dies
  • Examples:
    • Bt Cotton (India, 2002): Cry1Ac, Cry2Ab - bollworm resistance
    • Bt Corn/Maize: Cry1Ab - corn borer resistance
    • Bt Brinjal (Bangladesh, 2013): Cry1Ac - fruit and shoot borer
    • Bt Potato: Cry3A - Colorado potato beetle
  • Benefits:
    • Reduced insecticide use (70-80%)
    • Lower production costs
    • Higher yields
    • Reduced pesticide exposure
    • Environmental benefits
  • Resistance Management:
    • Refuge strategy: Plant non-Bt crops nearby (20% area)
    • Gene pyramiding: Multiple Cry genes (Cry1Ac + Cry2Ab)
    • High dose strategy: Express toxin at high levels

2. Herbicide Tolerance

A. Glyphosate Tolerance

  • Herbicide: Glyphosate (Roundup)
  • Target Enzyme: EPSPS (5-enolpyruvylshikimate-3-phosphate synthase)
  • Mechanism: Glyphosate inhibits EPSPS → blocks aromatic amino acid synthesis → plant death
  • Tolerance Gene: cp4-epsps from Agrobacterium strain CP4
    • Glyphosate-insensitive EPSPS enzyme
  • Crops: Roundup Ready Soybean, Cotton, Corn, Canola, Alfalfa
  • Benefits:
    • Broad-spectrum weed control
    • Post-emergence application
    • No-till farming possible
    • Reduced soil erosion

B. Glufosinate Tolerance

  • Herbicide: Glufosinate/Phosphinothricin (Basta, Liberty)
  • Target Enzyme: Glutamine synthetase
  • Tolerance Gene: bar or pat (phosphinothricin acetyltransferase)
    • From Streptomyces bacteria
    • Acetylates glufosinate → inactivates it
  • Crops: LibertyLink Soybean, Cotton, Corn, Canola

C. 2,4-D Tolerance

  • Gene: aad-1 (aryloxyalkanoate dioxygenase)
  • Crops: Enlist corn, soybean

3. Virus Resistance

Coat Protein-Mediated Resistance

  • Strategy: Express viral coat protein (CP) gene in plant
  • Mechanism: CP interferes with virus uncoating/replication
  • Examples:
    • Rainbow Papaya (Hawaii, 1998): PRSV (Papaya Ringspot Virus) coat protein
      • Saved papaya industry
      • First commercialized virus-resistant fruit
    • Squash: CMV, WMV, ZYMV resistance
    • Potato: PVY (Potato Virus Y) resistance

RNA Interference (RNAi)

  • Strategy: Express dsRNA complementary to viral genes
  • Mechanism: siRNA degrades viral RNA
  • More effective than coat protein method
  • Examples: Bean golden mosaic virus resistance in beans

4. Nutritional Enhancement (Biofortification)

A. Golden Rice

  • Developers: Ingo Potrykus & Peter Beyer (2000)
  • Target: Vitamin A deficiency (affects 250 million children)
  • Genes Introduced:
    • psy (phytoene synthase) from daffodil/Zea mays
    • crtI (phytoene desaturase) from Erwinia uredovora
  • Result: β-carotene accumulation in endosperm (provitamin A)
  • Golden Rice 2: 23× more β-carotene (uses maize psy)
  • Status: Approved in Philippines (2021), Bangladesh (2023)

B. Other Biofortified Crops

  • Iron-enriched rice: Ferritin gene
  • High-lysine corn: Enhanced essential amino acid
  • High-oleic soybean: Healthier oil profile
  • Folate-enriched tomato: Vitamin B9
  • Purple tomato: Anthocyanins (antioxidants)

5. Enhanced Shelf Life & Quality

Flavr Savr Tomato

  • First GM food approved: FDA 1994, Calgene Inc.
  • Gene Modified: Polygalacturonase (PG) - antisense technology
  • Result: Delayed softening, extended shelf life
  • Mechanism: Reduced pectin degradation during ripening
  • Status: Withdrawn (1997) - poor commercial performance

Arctic Apple

  • Trait: Non-browning
  • Gene Silenced: PPO (polyphenol oxidase) - RNAi
  • Result: No enzymatic browning when cut
  • Approved: USA (2015), Canada (2015)

Innate Potato

  • Traits: Reduced bruising, lower acrylamide
  • Genes Silenced: PPO (browning), Asparagine synthetase (acrylamide precursor)

6. Abiotic Stress Tolerance

A. Drought Tolerance

  • Strategies:
    • DREB genes (Dehydration-Responsive Element Binding proteins)
    • Late Embryogenesis Abundant (LEA) proteins
    • Proline biosynthesis genes (P5CS)
  • Example: DroughtGard corn (Monsanto) - cspB gene from Bacillus subtilis

B. Salinity Tolerance

  • Genes: Na⁺/H⁺ antiporters, aquaporins, osmoprotectant synthesis
  • Research stage: Rice, tomato, wheat

C. Cold/Heat Tolerance

  • Cold: Antifreeze proteins from fish
  • Heat: Heat shock proteins (HSPs)

D. Heavy Metal Tolerance

  • Phytoremediation: Accumulate heavy metals for soil cleanup
  • Genes: Metallothioneins, phytochelatins

7. Male Sterility (Hybrid Seed Production)

  • Barnase-Barstar System:
    • Barnase: RNase that causes male sterility (expressed in tapetum)
    • Barstar: Barnase inhibitor (restorer)
  • Application: Canola, tobacco hybrid seed production
  • Benefit: No manual emasculation needed, cost-effective

8. Industrial & Pharmaceutical Applications

Molecular Pharming (Plant-Made Pharmaceuticals)

  • Edible Vaccines:
    • Hepatitis B surface antigen in potato/banana
    • Cholera toxin B subunit in potato
    • Norwalk virus capsid protein in tomato/potato
  • Therapeutic Proteins:
    • Human serum albumin in tobacco
    • Hirudin (anticoagulant) in canola
    • Human growth hormone in rice
  • Monoclonal Antibodies (Plantibodies): In tobacco, soybean

Industrial Products

  • Bioplastics: PHB (Polyhydroxybutyrate) in Arabidopsis
  • Modified starch: High-amylose potato (Amflora)
  • Spider silk proteins: In tobacco, potato (strong fibers)

Regulatory Aspects of GM Crops

India - Regulatory Framework

  • Apex Body: GEAC (Genetic Engineering Appraisal Committee)
    • Under Ministry of Environment, Forest and Climate Change
    • Approves environmental release and commercialization
  • Other Bodies:
    • RCGM (Review Committee on Genetic Manipulation): Reviews research proposals
    • IBSC (Institutional Biosafety Committee): At research institutions
    • DBT (Department of Biotechnology): Nodal agency
  • Approved GM Crops in India:
    • Bt Cotton (2002): Only commercial GM crop in India
    • Bt Brinjal: Approved by GEAC (2009) but moratorium imposed (2010)

Global Regulatory Approaches

  • USA: Coordinated framework (FDA, USDA, EPA), product-based regulation
  • EU: Precautionary principle, process-based, strict regulations, mandatory labeling
  • Cartagena Protocol: International agreement on biosafety (2000)

Safety Assessment

  • Food Safety: Toxicity, allergenicity, nutritional composition
  • Environmental Safety: Gene flow, impact on non-target organisms, biodiversity
  • Substantial Equivalence: Comparison with conventional counterpart

Benefits & Concerns of GM Crops

✅ Benefits

  • Increased crop yields
  • Reduced pesticide use
  • Enhanced nutritional quality
  • Improved stress tolerance
  • Lower production costs
  • Environmental benefits
  • Food security
  • Medical applications

⚠️ Concerns

  • Gene flow to wild relatives
  • Development of resistance in pests
  • Potential allergenicity
  • Impact on non-target organisms
  • Corporate control of seeds
  • Ethical concerns
  • Long-term health effects unknown
  • Biodiversity loss

Global Status of GM Crops

  • Area under GM crops (2022): ~200 million hectares
  • Top countries: USA, Brazil, Argentina, Canada, India
  • Major GM crops: Soybean (50%), Maize (31%), Cotton (13%), Canola (5%)
  • Traits: Herbicide tolerance (47%), Insect resistance (12%), Stacked traits (41%)
  • Farmers growing GM crops: ~17 million (90% small-scale)

🎯 High-Yield Points for CSIR NET & ICAR SRF

  • CSIR NET Agrobacterium tumefaciens: Natural genetic engineer, causes crown gall disease
  • ICAR SRF T-DNA borders: 25 bp direct repeats, define region transferred to plant
  • CSIR NET vir genes: Required for T-DNA transfer, NOT transferred to plant
  • ICAR SRF Binary vector system: Separates T-DNA (small plasmid) from vir genes (helper)
  • CSIR NET Acetosyringone: Phenolic compound that induces vir gene expression
  • ICAR SRF Biolistics: John Sanford (1987), gene gun, works for all plants
  • CSIR NET Gold particles: 0.6-1.6 μm diameter, most common microprojectile
  • ICAR SRF Selectable markers: nptII (kanamycin), hpt (hygromycin), bar (phosphinothricin)
  • CSIR NET GUS gene: β-glucuronidase from E. coli, blue color with X-Gluc
  • ICAR SRF Bt toxin mechanism: Alkaline gut activates → binds receptors → pore formation → cell lysis
  • CSIR NET Cry1Ac: Lepidoptera (caterpillars), used in Bt cotton and Bt brinjal
  • ICAR SRF Refuge strategy: Plant 20% non-Bt crops to delay resistance
  • CSIR NET cp4-epsps: Glyphosate-tolerant EPSPS from Agrobacterium strain CP4
  • ICAR SRF bar/pat gene: Phosphinothricin acetyltransferase, glufosinate tolerance
  • CSIR NET Golden Rice: psy (phytoene synthase) + crtI (phytoene desaturase)
  • ICAR SRF Rainbow Papaya: PRSV coat protein gene, first virus-resistant fruit (1998)
  • CSIR NET Flavr Savr tomato: First GM food (FDA 1994), antisense PG gene
  • ICAR SRF Bt Cotton: Only commercial GM crop in India (approved 2002)
  • CSIR NET GEAC: Genetic Engineering Appraisal Committee (India's apex body)
  • ICAR SRF Cartagena Protocol: International biosafety agreement (2000)

💡 Memory Tricks & Mnemonics

  • Agrobacterium Mechanism: "WCVPTIN"
    • Wounding, Chemotaxis, Vir activation, Processing, Transfer, Integration, Nuclear import
  • Gene Transfer Methods: "ABEP-M"
    • Agrobacterium, Biolistics, Electroporation, PEG, Microinjection
  • Bt Cry Proteins: "1-Lepido, 3-Coleo"
    • Cry1 = Lepidoptera (caterpillars), Cry3 = Coleoptera (beetles)
  • Golden Rice Genes: "Pretty Colorful Rice Improves"
    • psy (phytoene synthase), crtI (carotene desaturase)
  • Herbicide Tolerance Genes: "CBPA"
    • Cp4-epsps (glyphosate), Bar (glufosinate), Pat (glufosinate), Aad-1 (2,4-D)
  • Regulatory Bodies in India: "GEAC Rules India's Biotechnology"
    • GEAC (apex body), RCGM, IBSC, DBT
  • First GM Products: "Flavr First, Rainbow Fruit"
    • Flavr Savr (first GM food, 1994), Rainbow Papaya (first virus-resistant fruit, 1998)

📝 Common MCQ Patterns

  • Agrobacterium: Ti plasmid components (T-DNA vs vir region), transformation mechanism
  • T-DNA Borders: 25 bp direct repeats, define transferred region
  • Binary Vector: Separation of T-DNA and vir genes, advantages
  • Phenolic Inducers: Acetosyringone induces vir genes
  • Vir Genes Function: VirA (sensor), VirG (activator), VirD2 (pilot protein), VirE2 (ssDNA binding)
  • Gene Transfer Methods: Which method for which plant type (Agrobacterium for dicots, Biolistics for monocots)
  • Biolistics Parameters: Particle size (0.6-1.6 μm), pressure (1100-1550 psi), distance (6-12 cm)
  • Selectable Markers: Which gene confers which resistance
  • Reporter Genes: GUS (blue color), GFP (green fluorescence)
  • Bt Toxins: Which Cry protein for which insect order
  • Resistance Management: Refuge strategy (20% non-Bt), gene pyramiding, high dose
  • Herbicide Tolerance: Gene-herbicide pairs (cp4-epsps for glyphosate, bar for glufosinate)
  • Golden Rice: Genes (psy, crtI), product (β-carotene), developers (Potrykus & Beyer)
  • Flavr Savr: First GM food, antisense PG gene, delayed ripening
  • Rainbow Papaya: PRSV coat protein, saved papaya industry in Hawaii
  • Bt Cotton in India: Approved 2002, only commercial GM crop
  • GEAC: Apex regulatory body in India for GM crop approval

🎓 Important Years to Remember

  • 1987: Biolistics developed (John Sanford)
  • 1994: Flavr Savr tomato approved (first GM food)
  • 1998: Rainbow Papaya released (first virus-resistant fruit)
  • 2000: Golden Rice developed (Potrykus & Beyer), Cartagena Protocol
  • 2002: Bt Cotton approved in India
  • 2009: Bt Brinjal approved by GEAC (moratorium 2010)
  • 2013: Bt Brinjal released in Bangladesh
  • 2015: Arctic Apple approved (USA, Canada)
  • 2021: Golden Rice approved in Philippines

Future Perspectives

Emerging Technologies

  • CRISPR-Cas9 Gene Editing:
    • Precise genome editing without foreign DNA integration
    • Regulatory advantages (not considered GMO in some countries)
    • Applications: Disease resistance, yield improvement, quality traits
  • RNA-based Strategies:
    • RNAi for pest control (spray-on dsRNA)
    • Host-Induced Gene Silencing (HIGS)
  • Synthetic Biology:
    • Design and construct new biological systems
    • Synthetic promoters, circuits, pathways
  • Genome Editing:
    • TALEN, Zinc Finger Nucleases
    • Base editing, Prime editing

Future GM Crop Traits

  • Climate-Smart Agriculture:
    • Multi-stress tolerance
    • Carbon sequestration crops
    • Nitrogen-fixing cereals
  • Nutritional Enhancement:
    • Multiple micronutrient enrichment
    • Allergen-free foods
    • Enhanced functional compounds
  • Sustainable Agriculture:
    • Perennial grains
    • Enhanced photosynthesis efficiency
    • Reduced methane emission (rice)

About the author

M.S. Chaudhary
I'm an ordinary student of agriculture.

Post a Comment