Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

Syllabus-BPCIL

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

General Aptitude Syllabus 30 Marks

Section Topics No. of


Questions
1. Quantitative Number Systems, Percentage, Profit and Loss, Simple and Compound Interest, 6
Aptitude: Ratio and Proportion, Time, Speed, and Distance, Time and Work, Average,
Algebra, Geometry, Mensuration, Data Interpretation.
2. Logical Reasoning: Analogy , Classification, Series Completion, Coding-Decoding, Blood Relations, 6
Direction Sense Test, Logical Venn Diagrams, Alphabet Test, Number Ranking
and Time Sequence Test, Mathematical Operations, Logical Sequence of Words,
Arithmetical Reasoning, Inserting the Missing Character, Data Sufficiency.
3. Verbal Ability: Reading Comprehension, Vocabulary, Synonyms and Antonyms, Fill in the 6
Blanks, Error Correction, Sentence Correction, Para-jumbles, Sentence,
Completion, Idioms and Phrases, One-word Substitution.
4. General Current Affairs (National and International), History, Geography, Indian Polity, 6
Awareness: Indian Economy, Science and Technology, Environmental Studies, Important
Dates, Events and Persons, Books and Authors, Awards and Honors, Sports,
Abbreviations, Indian Constitution.
5. Data Tabular Data Interpretation, Bar Graphs, Line Graphs, Pie Charts, Caselet Data 6
Interpretation: Interpretation, Data Sufficiency.

Life Sciences Syllabus 70 Marks


Section Topics No. of
Questions
Section 1:  Biochemical Techniques: Principles and applications of various biochemical 7
Biochemistry techniques such as chromatography (paper chromatography, TLC, column
chromatography, HPLC), electrophoresis, centrifugation, spectroscopy (UV-
Vis, fluorescence, NMR, mass spectrometry), immunochemical techniques
(ELISA, immunoblotting), PCR, DNA sequencing, protein purification
techniques.
 Biomolecules: Structure, properties, and functions of carbohydrates, lipids,
proteins, and nucleic acids.
 Enzymology: Classification, mechanism of action, kinetics (Michaelis-Menten
kinetics, allosteric enzymes), enzyme inhibition, regulation of enzyme activity.
 Metabolism: Overview of metabolism: Anabolism and catabolism.
Carbohydrate metabolism: Glycolysis, gluconeogenesis, pentose phosphate
pathway, glycogen metabolism. Lipid metabolism: Fatty acid oxidation and
synthesis, cholesterol metabolism. Amino acid metabolism: Protein catabolism,
urea cycle, biosynthesis of amino acids. Nucleotide metabolism: Purine and
pyrimidine metabolism. Bioenergetics: ATP synthesis, oxidative
phosphorylation, electron transport chain.
 Biochemical Genetics: Mendelian genetics and its application in biochemistry.
Gene expression: Transcription, translation, regulation of gene expression
(transcriptional and post-transcriptional regulation). Recombinant DNA
technology: Gene cloning, PCR, DNA sequencing, gene expression vectors.
 Hormones and Signal Transduction: Overview of hormones and their
functions. Mechanisms of hormone action: Receptor-mediated signal
transduction pathways (G-protein coupled receptors, receptor tyrosine kinases,
intracellular receptors). Second messengers: cAMP, IP3, DAG, calcium ions.
 Biochemical Basis of Diseases: Molecular basis of genetic disorders, metabolic
disorders, and cancer. Biomarkers and diagnostic tests for various diseases.
Therapeutic interventions targeting biochemical pathways in diseases.
 Immunochemistry: Structure and function of antibodies and antigens. Immune
response: Innate and adaptive immunity, antigen presentation, humoral and
cellular immunity. Immunological techniques: ELISA, immunoblotting,
immunoprecipitation.
Section 2 : Molecular  DNA Structure and Function: DNA replication: Enzymes involved, 7
Biology replication fork, leading and lagging strands, replication origins, and
termination. DNA repair mechanisms: Mismatch repair, base excision repair,
nucleotide excision repair, homologous recombination, non-homologous end
joining. DNA packaging: Chromatin structure, nucleosomes, histones,
chromatin remodeling complexes.
 Transcription: RNA synthesis: RNA polymerase, transcription factors,
promoter elements, enhancers, terminators. Transcriptional regulation: Operons,
transcriptional activators and repressors, transcriptional co-regulators, RNA
interference.
 RNA Processing: mRNA processing: Capping, splicing, polyadenylation.
tRNA and rRNA processing.
 Translation: Ribosome structure and function. tRNA charging, initiation,
elongation, termination. Post-translational modifications: Protein folding,
glycosylation, phosphorylation, proteolysis.
 Gene Regulation: Mechanisms of gene regulation in prokaryotes and
eukaryotes. Regulatory sequences, transcriptional regulators, epigenetic
modifications. Transcriptional and post-transcriptional regulation of gene
expression.
 Genomics and Proteomics: Genome organization: Gene structure, introns and
exons, repetitive DNA elements. Comparative genomics, functional genomics,
and metagenomics. Proteome analysis: Protein separation techniques, mass
spectrometry, protein-protein interactions.
 Recombinant DNA Technology: Gene cloning: Restriction enzymes, DNA
ligation, vectors (plasmids, phages, cosmids), transformation, selection,
screening. Polymerase Chain Reaction (PCR) and its variations. DNA
sequencing methods: Sanger sequencing, next-generation sequencing (NGS),
and sequencing by synthesis.
 Gene Expression Systems: Expression vectors and hosts: Bacterial, yeast,
insect, and mammalian expression systems. Protein expression and purification
techniques.
 Genome Editing and CRISPR-Cas Technology: Principles of CRISPR-Cas
systems. Applications of CRISPR-Cas for genome editing, gene regulation, and
functional genomics.
 RNA Interference (RNAi) and Gene Silencing: Mechanisms and applications
of RNAi in gene silencing, functional genomics, and therapeutic interventions.
 Molecular Biology Techniques: Principles and applications of molecular
biology techniques such as Southern blotting, Northern blotting, Western
blotting, DNA/RNA hybridization, gel electrophoresis, and DNA/RNA
manipulation techniques.
Section 3:  Microbial Taxonomy and Diversity: Classification and nomenclature of 7
Microbiology microorganisms: Bacteria, Archaea, Fungi, Protozoa, Algae, and Viruses.
Methods for microbial identification and characterization. Microbial diversity in
different environments: Soil, water, air, extreme environments.
 Microbial Structure and Function: Bacterial cell structure: Cell wall, plasma
membrane, cytoplasm, ribosomes, nucleoid, appendages (flagella, pili,
fimbriae), endospores. Structure and function of viral particles: Capsid,
envelope, genome types (DNA, RNA), replication cycles (lytic and lysogenic).
Structure and function of eukaryotic microorganisms: Fungi, Protozoa, Algae.
 Microbial Physiology: Metabolism: Microbial nutrition, energy generation,
catabolism, anabolism, fermentation, respiration, photosynthesis. Growth
kinetics: Growth curve, factors affecting microbial growth (temperature, pH,
osmotic pressure, oxygen). Bacterial genetics: Gene transfer mechanisms
(transformation, transduction, conjugation), plasmids, mutations, horizontal
gene transfer.
 Microbial Ecology: Microbial interactions: Mutualism, commensalism,
parasitism, competition, predation. Microbial communities: Biofilms, microbial
mats, microbial consortia. Microbial roles in biogeochemical cycles: Carbon,
nitrogen, sulfur cycles.
 Medical Microbiology: Pathogenesis of infectious diseases: Virulence factors,
host-pathogen interactions, mechanisms of infection. Microbial diseases:
Bacterial, viral, fungal, and protozoal infections, epidemiology, prevention, and
control measures. Antimicrobial agents: Antibiotics, antivirals, antifungals,
mechanisms of action, resistance mechanisms.
 Industrial Microbiology: Microbial biotechnology: Microbial enzymes,
biofuel production, bioremediation, biotransformation, biosurfactants,
biofertilizers.
 Fermentation technology: Fermentation processes, microbial culture techniques,
downstream processing, scale-up, and optimization.
 Microbial Biotechnology: Genetic engineering of microorganisms:
Recombinant DNA technology, gene cloning, expression vectors, gene editing
tools (CRISPR-Cas), synthetic biology. Microbial production of biomolecules:
Recombinant proteins, vaccines, antibiotics, industrial enzymes.
 Environmental Microbiology: Microbial interactions in ecosystems: Role of
microorganisms in nutrient cycling, degradation of pollutants, bioremediation
strategies.
 Microbial indicators of environmental quality: Indicator microorganisms for
water and soil quality assessment.
 Food Microbiology: Microorganisms in food production, preservation, and
spoilage. Foodborne pathogens: Sources, transmission, prevention, and control
measures.
 Microbial Genetics and Genomics: Microbial genome structure and
organization. Comparative genomics, metagenomics, and systems biology
approaches in microbial research.

Section 4: Genetics  Mendelian Genetics: Laws of inheritance: Mendel's laws, deviations from 7
and Genomics Mendelian genetics (incomplete dominance, codominance, multiple alleles,
pleiotropy, epistasis). Pedigree analysis: Autosomal and sex-linked inheritance
patterns, genetic disorders.
 Molecular Genetics: DNA structure and replication: Double helix model, DNA
replication mechanisms (semi-conservative, conservative, dispersive). DNA
repair mechanisms: Mismatch repair, base excision repair, nucleotide excision
repair. DNA recombination and repair: Homologous recombination, site-
specific recombination, transposition. DNA packaging: Chromatin structure,
nucleosomes, histone modifications, chromatin remodeling.
 Gene Expression and Regulation: Transcription: RNA polymerase,
transcription factors, promoters, enhancers, transcriptional regulation
(activators, repressors). RNA processing: Splicing, capping, polyadenylation,
RNA editing. Translation: Ribosome structure, initiation, elongation,
termination, regulation of translation.
 Post-translational modifications: Protein folding, glycosylation,
phosphorylation, ubiquitination.
 Genomics: Genome organization: Gene structure, introns and exons, gene
families, repetitive sequences. Comparative genomics: Genome evolution,
orthologs, paralogs, synteny, genome duplication events.
 Functional genomics: Transcriptomics, proteomics, metabolomics, high-
throughput sequencing technologies (RNA-seq, ChIP-seq, DNA microarrays).
 Epigenetics and Chromatin Dynamics: Epigenetic modifications: DNA
methylation, histone modifications, non-coding RNAs. Epigenetic inheritance:
Role of epigenetics in development, disease, and environmental responses.
Chromatin remodeling complexes: SWI/SNF, ISWI, histone acetyltransferases,
histone deacetylases.
 Genetic Engineering and Biotechnology: Recombinant DNA technology:
Gene cloning, expression vectors, gene editing tools (CRISPR-Cas), synthetic
biology. Genetic manipulation techniques: Gene knockout, gene knock-in, gene
silencing (RNA interference).
 Applications of genetic engineering: Transgenic organisms, gene therapy,
genetically modified crops, molecular diagnostics.
 Population and Evolutionary Genetics: Population genetics: Gene frequency,
Hardy-Weinberg equilibrium, genetic drift, gene flow, natural selection.
Molecular evolution: Molecular clocks, neutral theory, adaptive evolution,
phylogenetics, molecular markers.
 Genetic Diseases and Genetic Counseling: Human genetic disorders:
Monogenic disorders, chromosomal abnormalities, multifactorial diseases.
Principles of genetic counseling: Pedigree analysis, risk assessment, ethical
issues in genetic testing.
 Genome Editing and Gene Therapy: CRISPR-Cas9 system: Mechanism,
applications in genome editing and gene therapy. Gene therapy approaches:
Gene addition, gene correction, gene silencing, ex vivo and in vivo gene
therapy.
 Systems Biology and Network Analysis: Systems biology approaches:
Modeling biological systems, network analysis, signaling pathways, regulatory
networks. Computational methods in genomics: Sequence alignment, gene
prediction, motif discovery, genome annotation.
Section 5: Cell  Cell Structure and Function: Cell theory: Historical perspective, modern cell 7
Biology theory. Cellular compartments: Structure and function of cell organelles
(nucleus, endoplasmic reticulum, Golgi apparatus, mitochondria, chloroplasts,
lysosomes, peroxisomes). Cytoskeleton: Microtubules, microfilaments,
intermediate filaments, their structure, function, and dynamics. Cell membrane:
Structure, fluid mosaic model, membrane proteins, membrane transport (passive
and active transport), cell-cell interactions, cell junctions.
 Cell Cycle and Division: Cell cycle phases: Interphase (G1, S, G2), mitosis,
cytokinesis. Regulation of the cell cycle: Checkpoints, cyclins, cyclin-
dependent kinases (CDKs), tumor suppressor genes. Cell division: Mitosis,
meiosis, comparison between mitosis and meiosis, gametogenesis.
 Cell Signaling: Types of cell signaling: Endocrine, paracrine, autocrine,
juxtacrine. Signal transduction pathways: Receptor-mediated signaling,
intracellular signaling cascades (e.g., G-protein coupled receptors, receptor
tyrosine kinases). Second messengers: cAMP, cGMP, IP3, DAG, calcium ions.
Signaling molecules: Growth factors, cytokines, hormones.
 Cellular Energetics: Cellular respiration: Glycolysis, citric acid cycle (Krebs
cycle), oxidative phosphorylation, electron transport chain. Photosynthesis:
Light-dependent and light-independent reactions, Calvin cycle, chloroplast
structure and function. Energy metabolism: ATP synthesis, energy storage and
utilization, metabolic pathways (anabolism and catabolism).
 Cellular Membrane Dynamics: Membrane trafficking: Endocytosis
(phagocytosis, pinocytosis, receptor-mediated endocytosis), exocytosis, vesicle
transport. Membrane fusion and fission: Mechanisms and regulation. Membrane
remodeling: Lipid rafts, membrane curvature, membrane-associated proteins.
 Cellular Adhesion and Motility: Cell adhesion molecules (CAMs): Integrins,
selectins, cadherins. Cell migration: Mechanisms of cell motility, cytoskeletal
dynamics in cell migration (lamellipodia, filopodia), extracellular matrix (ECM)
interactions. Cell-cell and cell-matrix adhesion: Role in tissue development,
wound healing, and cancer metastasis.
 Cell Differentiation and Stem Cells: Stem cell biology: Types of stem cells
(embryonic stem cells, adult stem cells, induced pluripotent stem cells), stem
cell niches, stem cell markers. Cell fate determination: Signaling pathways
involved in cell differentiation, transcription factors, epigenetic regulation.
Tissue regeneration and repair: Role of stem cells in tissue homeostasis and
regeneration.
 Cellular Responses to Environmental Stimuli:
 Cellular stress responses: Heat shock response, unfolded protein response,
oxidative stress response. Apoptosis and cell death: Mechanisms of
programmed cell death, caspase activation, apoptotic signaling pathways.
 Cell senescence: Molecular mechanisms, role in aging and age-related diseases.

Section 6:  Introduction to Pharmaceutical Biotechnology : Overview of biotechnology 7


Pharmaceutical applications in the pharmaceutical industry. Importance and impact of
Biotechnology biopharmaceuticals. Regulatory aspects and ethical considerations in
pharmaceutical biotechnology.
 Recombinant DNA Technology:
 Techniques for gene cloning, including restriction enzymes, vectors, and gene
libraries. Gene expression systems: prokaryotic and eukaryotic hosts, expression
vectors, and promoters.
 Strategies for recombinant protein production and purification. Monoclonal
Antibodies and Therapeutic Proteins Production of monoclonal antibodies:
hybridoma technology and recombinant antibody techniques. Therapeutic
proteins: insulin, growth factors, cytokines, and hormones. Purification and
characterization of therapeutic proteins.
 Vaccine Technology : Types of vaccines: live attenuated, inactivated, subunit,
DNA, and vector-based vaccines. Vaccine production methods: cell culture,
recombinant technology, and adjuvants. Vaccine formulation, stability, and
delivery systems.
 Drug Delivery Systems : Principles of drug delivery: routes of administration,
pharmacokinetics, and pharmacodynamics. Nanotechnology in drug delivery:
nanoparticles, liposomes, and micelles. Targeted drug delivery systems and site-
specific drug release mechanisms.
 Pharmacogenomics and Personalized Medicine : Role of genomics in drug
discovery and development. Pharmacogenetics: genetic variations and drug
response. Implementation of personalized medicine in clinical practice.
 Regulatory Affairs and Quality Control : Regulatory requirements for
biopharmaceutical products: FDA, EMA, and ICH guidelines. Good
Manufacturing Practices (GMP) and quality control standards.Quality assurance
in biopharmaceutical manufacturing. Emerging Trends in Pharmaceutical
Biotechnology
 Biopharmaceuticals for rare diseases and orphan drugs. Bioprospecting and
natural product discovery. Future directions in precision medicine and
biopharmaceutical innovation.

Section 7 : Bioprocess  Introduction to Bioprocess Engineering: Definition, scope, and importance of 7


Engineering bioprocess engineering. Historical development and milestones in bioprocess
technology. Role of bioprocess engineering in biotechnology, pharmaceuticals,
food processing, and other industries.
 Microbial Growth Kinetics: Growth phases of microorganisms: lag phase,
exponential phase, stationary phase, death phase.
 Mathematical modeling of microbial growth: Monod equation, logistic
equation, and other models. Factors affecting microbial growth: Nutrient
availability, temperature, pH, oxygen, etc.
 Downstream Processing: Recovery and purification of biomolecules: Solid-
liquid separation, filtration, centrifugation, chromatography, etc. Unit operations
in downstream processing: Extraction, precipitation, crystallization, drying, and
formulation. Process optimization and efficiency enhancement in downstream
processing.
 Bioreactor Design and Operation: Design criteria for bioreactors: Reactor
geometry, mixing, aeration, and agitation. Heat and mass transfer in bioreactors:
Oxygen transfer, nutrient transfer, heat removal, etc. Bioreactor instrumentation
and control: Sensors, transducers, actuators, feedback control systems.
 Bioprocess Scale-up and Economics: Principles of scale-up in bioprocess
engineering: Geometric similarity, dynamic similarity, and dimensionless
numbers. Economic considerations in bioprocess design: Cost analysis, process
optimization, return on investment.
 Techno-economic analysis (TEA) of bioprocesses: Cost estimation, sensitivity
analysis, profitability assessment.
 Biocatalysis and Enzyme Technology: Enzyme kinetics and mechanisms:
Michaelis-Menten kinetics, enzyme inhibition, allosteric regulation.
Immobilization of enzymes: Methods, applications, and advantages. Industrial
applications of enzymes: Biocatalysis, biotransformation, enzyme-based
biosensors, etc.
 Bioseparation Techniques: Principles and applications of bioseparation
techniques: Centrifugation, filtration, chromatography, electrophoresis, etc.
Advanced separation methods: Membrane separation, supercritical fluid
extraction, affinity chromatography, etc. Purification of recombinant proteins,
antibodies, and other biomolecules.
 Biological Waste Treatment: Biological treatment of wastewater: Aerobic and
anaerobic digestion, activated sludge process, biofiltration, etc. Bioremediation
of environmental pollutants: Biodegradation, phytoremediation, microbial
consortia, etc. Sustainable bioprocesses and green technologies in waste
treatment.
 Emerging Trends in Bioprocess Engineering: Bioprocess intensification:
Miniaturization, microreactors, continuous processing, and integrated
biomanufacturing. Bioprocess automation and digitalization: Industry 4.0,
artificial intelligence, machine learning, and data analytics. Applications of
bioprocess engineering in biopharmaceuticals, biofuels, biopolymers, and
personalized medicine.
Section 8 :  Introduction to Fermentation Technology: 7
Fermentation  Definition, scope, and importance of fermentation in biotechnology,
Technology pharmaceuticals, food, and other industries. Historical development and
milestones in fermentation technology. Role of microorganisms in fermentation
processes: Bacteria, fungi, yeast, and algae.
 Microbial Growth and Metabolism:
 Growth kinetics of microorganisms: Growth phases, factors affecting growth,
and mathematical modeling. Metabolic pathways in microorganisms:
Glycolysis, TCA cycle, pentose phosphate pathway, and fermentation pathways.
Regulation of microbial metabolism: Enzyme induction, repression, feedback
inhibition, and metabolic control mechanisms.
 Substrate Utilization and Product Formation: Utilization of carbon and
energy sources by microorganisms: Sugars, alcohols, organic acids, and
hydrocarbons. Formation of microbial products: Primary metabolites (ethanol,
lactic acid, citric acid, etc.) and secondary metabolites (antibiotics, enzymes,
pigments, etc.).
 Optimization of substrate utilization and product formation in fermentation
processes.
 Fermentation Process Design and Operation:
 Design considerations for fermentation reactors: Reactor types, vessel
geometry, mixing, aeration, and sterilization. Parameters affecting fermentation
process operation: pH control, temperature control, oxygen transfer rate, and
agitation speed. Monitoring and control of fermentation parameters: Online and
offline monitoring techniques, feedback control systems.
 Fermentation Kinetics and Scale-Up: Kinetics of fermentation reactions:
Monod equation, substrate inhibition, product inhibition, and other kinetic
models. Scale-up of fermentation processes: Principles of geometric similarity,
dynamic similarity, and dimensionless numbers. Challenges and strategies in
scaling up fermentation processes from laboratory to pilot and production scale.

 Downstream Processing in Fermentation:


 Recovery and purification of fermentation products: Cell disruption, solid-liquid
separation, filtration, and centrifugation.
 Separation techniques: Chromatography (affinity, ion exchange, size exclusion),
crystallization, evaporation, and drying. Optimization of downstream processing
operations: Yield improvement, purity enhancement, and cost reduction.
 Sterilization and Aseptic Techniques: Sterilization methods for fermentation
media and equipment: Heat sterilization (autoclaving), filtration, chemical
sterilization. Aseptic techniques in fermentation: Cleanroom design, air
filtration, gowning procedures, and environmental monitoring. Prevention and
control of contamination in fermentation processes: Bacterial, fungal, viral, and
phage contamination.
 Industrial Applications of Fermentation Technology: Bioproduction of
biofuels: Ethanol, biodiesel, methane, hydrogen, and other renewable fuels.
Production of industrial enzymes: Amylases, proteases, lipases, cellulases, and
other enzyme products.
 Fermentation-based production of pharmaceuticals, antibiotics, organic acids,
amino acids, vitamins, and specialty chemicals.
 Emerging Trends in Fermentation Technology:
 Biorefinery concept and integrated bioprocessing: Utilization of biomass for the
production of multiple products. Metabolic engineering and synthetic biology
approaches in fermentation: Strain improvement, pathway engineering, and
genome editing.
 Sustainable fermentation processes: Waste minimization, energy efficiency, and
environmental impact assessment.
Section 9:  Introduction to Immunology: Historical perspectives and milestones in 7
Immunology immunology. Basic concepts: Immune system, immunity, antigens, antibodies,
and antigens-antibody interactions.
 Types of immunity: Innate immunity and adaptive immunity.
 Innate Immunity: Components of the innate immune system: Physical barriers,
cellular components (macrophages, neutrophils, dendritic cells), and soluble
factors (complement system, cytokines). Recognition of pathogens by pattern
recognition receptors (PRRs). Phagocytosis and intracellular killing
mechanisms.
 Adaptive Immunity: Cells of adaptive immunity: T cells, B cells, and antigen-
presenting cells (APCs). Antigen recognition and activation of lymphocytes:
Major histocompatibility complex (MHC), T cell receptor (TCR), and B cell
receptor (BCR).
 T cell activation and differentiation: Helper T cells (Th1, Th2, Th17), cytotoxic
T cells (CTLs), and regulatory T cells (Tregs).
 B cell activation, antibody production, and class switching.
 Humoral Immune Response: Structure and function of antibodies:
Immunoglobulin classes (IgG, IgM, IgA, IgD, IgE), antibody diversity, and
antibody-mediated effector functions. Generation of antibody diversity: Somatic
recombination, somatic hypermutation, and affinity maturation. Mechanisms of
antibody-mediated immunity: Neutralization, opsonization, complement
activation, and antibody-dependent cellular cytotoxicity (ADCC).
 Cell-Mediated Immune Response: T cell activation and effector functions:
Cytokine production, T cell proliferation, and differentiation into effector and
memory T cells. T cell-mediated cytotoxicity: Recognition of target cells,
perforin-granzyme pathway, and Fas-Fas ligand pathway. Role of T cells in
immune regulation and tolerance.
 Immune System Development and Tolerance: Development of lymphoid
organs: Thymus, bone marrow, and secondary lymphoid tissues (spleen, lymph
nodes). Central and peripheral tolerance mechanisms: Negative selection in
thymus, peripheral deletion, anergy, and regulatory T cells.
 Antigen Processing and Presentation: Intracellular antigen processing
pathways: Endogenous pathway (MHC class I) and exogenous pathway (MHC
class II). Antigen presentation by professional antigen-presenting cells (APCs):
Dendritic cells, macrophages, and B cells.
 Hypersensitivity and Autoimmunity: Types of hypersensitivity reactions:
Type I (immediate), Type II (cytotoxic), Type III (immune complex-mediated),
and Type IV (delayed-type). Mechanisms of autoimmune diseases: Loss of self-
tolerance, molecular mimicry, and genetic predisposition. Examples of
autoimmune diseases: Rheumatoid arthritis, systemic lupus erythematosus
(SLE), multiple sclerosis, and type 1 diabetes.
 Vaccines and Immunotherapy: Principles of vaccination: Types of vaccines
(live attenuated, inactivated, subunit, conjugate, and DNA vaccines), adjuvants,
and vaccination strategies. Immunotherapeutic approaches: Monoclonal
antibodies, cytokine therapy, adoptive T cell transfer, and immune checkpoint
inhibitors.
 Immunological Techniques: Immunological assays: ELISA (enzyme-linked
immunosorbent assay), western blotting, immunoprecipitation, and flow
cytometry. Molecular techniques in immunology: Polymerase chain reaction
(PCR), DNA sequencing, and gene expression analysis.
Section 10:  Introduction to Bioinformatics: Definition, scope, and applications of 7
Bioinformatics and bioinformatics. Historical development and milestones in bioinformatics. Role
Computational of computational methods in biological research.
Biology  Biological Databases: Types of biological databases: Nucleotide sequence
databases, protein sequence databases, structure databases, and others. Retrieval
and analysis of biological data from databases. Overview of major biological
databases: NCBI, GenBank, UniProt, PDB, etc. Database search techniques:
BLAST, FASTA, PSI-BLAST, etc.
 Sequence Analysis: Sequence alignment algorithms: Pairwise alignment,
multiple sequence alignment. Scoring matrices: BLOSUM, PAM, substitution
matrices. Sequence similarity and homology. Methods for sequence motif
discovery and analysis.
 Phylogenetic analysis: Construction of phylogenetic trees, evolutionary
relationships.
 Structural Bioinformatics: Protein structure prediction: Homology modeling,
ab initio modeling, threading. Protein structure analysis: Structural motifs,
domains, and folds. Protein-ligand interactions: Docking, virtual screening, drug
design. Molecular dynamics simulations and computational chemistry methods.
 Genomics and Transcriptomics: Genome sequencing technologies: Sanger
sequencing, next-generation sequencing (NGS), third-generation sequencing.
Genome assembly, annotation, and analysis. Transcriptome analysis: RNA
sequencing (RNA-seq), gene expression profiling, alternative splicing.
Identification of non-coding RNAs: microRNAs, long non-coding RNAs
(lncRNAs).
 Proteomics and Metabolomics:
 Protein identification and quantification techniques: Mass spectrometry, 2D gel
electrophoresis. Protein-protein interaction networks and analysis.
Metabolomics: Metabolite identification, metabolic pathway analysis.
 Systems Biology: Integration of omics data: Genomics, transcriptomics,
proteomics, metabolomics. Reconstruction and analysis of biological networks:
Regulatory networks, metabolic networks, signaling networks. Modeling and
simulation of biological systems: Boolean networks, ordinary differential
equations (ODEs), agent-based models.
 Bioinformatics Tools and Software: Overview of commonly used
bioinformatics tools and software packages. Command-line tools, web servers,
and graphical interfaces. Programming languages for bioinformatics: Python, R,
Perl, etc.
 Machine Learning and Data Mining in Bioinformatics: Applications of
machine learning algorithms in bioinformatics.
 Classification, clustering, and regression techniques. Feature selection,
dimensionality reduction, and model evaluation.
 Ethical, Legal, and Social Issues (ELSI) in Bioinformatics: Privacy, data
security, and intellectual property rights in bioinformatics research. Ethical
considerations in handling biological data and conducting computational
analyses. Regulations and policies governing bioinformatics research and
applications.

You might also like