🧪Synthetic Biology Unit 12 – Protein Synthesis for Drug Discovery
Protein synthesis is a crucial process in molecular biology, involving transcription of DNA to RNA and translation of RNA to proteins. This complex mechanism is essential for drug discovery, as it allows scientists to understand and manipulate the production of proteins involved in various diseases.
The drug discovery process leverages protein synthesis knowledge to identify targets, design drugs, and develop new therapies. From high-throughput screening to structure-based drug design, researchers use advanced techniques to create innovative treatments that can potentially save lives.
Central dogma of molecular biology describes the flow of genetic information from DNA to RNA to proteins
Transcription process where DNA is used as a template to synthesize complementary RNA strands catalyzed by RNA polymerase enzymes
Translation process where the genetic code in mRNA is decoded by ribosomes to synthesize polypeptide chains that fold into functional proteins
Occurs in the cytoplasm of cells and involves tRNA molecules that carry specific amino acids to the ribosome
Ribosomes read the codons in mRNA and link the corresponding amino acids together to form polypeptide chains
Post-translational modifications can alter the structure and function of proteins after synthesis (phosphorylation, glycosylation)
Protein folding determines the three-dimensional structure of proteins critical for their biological functions
Misfolded proteins can lead to various diseases (Alzheimer's, Parkinson's)
Protein degradation pathways break down and recycle proteins no longer needed by the cell (ubiquitin-proteasome system)
Molecular Biology Fundamentals
DNA double helix structure consists of two complementary strands held together by hydrogen bonds between base pairs (A-T, G-C)
DNA replication occurs during cell division to ensure genetic information is passed on to daughter cells
Involves DNA polymerase enzymes that synthesize new strands using the original DNA as a template
RNA molecules are single-stranded and contain uracil (U) instead of thymine (T) found in DNA
Three main types of RNA: mRNA (messenger), tRNA (transfer), and rRNA (ribosomal)
Genetic code determines how the sequence of nucleotides in mRNA is translated into amino acids
Consists of 64 codons, each specifying a particular amino acid or stop signal
Mutations can alter the genetic code and lead to changes in the amino acid sequence of proteins
Point mutations involve single nucleotide changes (substitutions, insertions, deletions)
Gene expression regulation controls which genes are transcribed and translated in different cell types and conditions
Transcription factors bind to specific DNA sequences to activate or repress gene expression
Protein Structure and Function
Primary structure refers to the linear sequence of amino acids in a polypeptide chain determined by the genetic code
Secondary structure describes the local folding patterns of polypeptide chains stabilized by hydrogen bonds (α-helices, β-sheets)
Tertiary structure represents the three-dimensional shape of a single polypeptide chain resulting from interactions between amino acid side chains
Stabilized by various forces (hydrogen bonds, disulfide bridges, hydrophobic interactions)
Quaternary structure involves the assembly of multiple polypeptide chains into a functional protein complex (hemoglobin)
Protein domains are distinct structural and functional units within a protein that can evolve and function independently
Enzymes are proteins that catalyze biochemical reactions by lowering the activation energy
Active site is the region where substrates bind and the reaction occurs
Specificity depends on the complementary shape and chemical properties of the active site
Protein-protein interactions mediate many cellular processes (signal transduction, cell adhesion)
Interaction interfaces involve complementary surfaces and specific amino acid contacts
Techniques for Protein Engineering
Recombinant DNA technology allows the manipulation and expression of genes in different host organisms
Restriction enzymes cut DNA at specific sequences enabling the insertion of genes into plasmid vectors
Plasmids are circular DNA molecules that can replicate independently and carry foreign genes
Polymerase chain reaction (PCR) amplifies specific DNA sequences using primers and DNA polymerase
Enables the rapid generation of large quantities of DNA for cloning and mutagenesis
Site-directed mutagenesis introduces specific changes to the DNA sequence of a gene to alter the amino acid sequence of the encoded protein
Oligonucleotide primers containing the desired mutation are used in PCR to amplify the modified gene
Protein expression systems produce recombinant proteins in various host organisms (bacteria, yeast, mammalian cells)
Choice of expression system depends on factors such as post-translational modifications and yield
Protein purification techniques isolate the protein of interest from the host cell lysate
Affinity chromatography uses tags (His-tag) that bind specifically to a matrix
Size-exclusion chromatography separates proteins based on their molecular weight
Directed evolution mimics natural selection to evolve proteins with desired properties
Involves rounds of mutagenesis, screening, and amplification to identify improved variants
Drug Discovery Process Overview
Target identification involves selecting a protein or pathway implicated in a disease that could be modulated by a drug
Genomics, proteomics, and computational methods aid in identifying potential targets
Target validation confirms the role of the target in the disease and its suitability for drug development
Uses techniques such as gene knockouts, RNA interference, and small molecule probes
High-throughput screening (HTS) rapidly tests large libraries of compounds against the target to identify hits that show desired activity
Automated robotic systems and miniaturized assays enable screening of millions of compounds
Hit-to-lead optimization improves the potency, selectivity, and pharmacokinetic properties of the initial hits
Medicinal chemistry modifies the chemical structure to enhance drug-like properties
Lead optimization further refines the lead compounds to improve their efficacy and safety
In vitro and in vivo studies assess the pharmacology, toxicology, and pharmacokinetics
Preclinical development involves extensive testing of the optimized lead in animal models to establish safety and efficacy
Investigational New Drug (IND) application filed with regulatory agencies to begin clinical trials
Clinical trials test the drug candidate in humans to determine its safety, dosage, and effectiveness
Phase 1 assesses safety in healthy volunteers, Phase 2 evaluates efficacy in a small patient group, Phase 3 confirms safety and efficacy in a larger patient population
FDA approval is required before the drug can be marketed and prescribed to patients
New Drug Application (NDA) submitted with data from preclinical and clinical studies
Protein-Based Drug Design
Structure-based drug design (SBDD) uses the three-dimensional structure of the target protein to guide the design of small molecule inhibitors
X-ray crystallography and NMR spectroscopy determine the atomic structure of proteins
Computational methods (molecular docking) predict the binding of ligands to the target
Ligand-based drug design (LBDD) uses the structure of known ligands that bind to the target to design new compounds with similar properties
Pharmacophore modeling identifies the essential features of ligands responsible for their activity
Quantitative structure-activity relationship (QSAR) models correlate the chemical structure of ligands with their biological activity
Protein-protein interaction inhibitors disrupt the formation of protein complexes involved in disease pathways
Peptidomimetics are small molecules that mimic the structure of peptides and can bind to protein interfaces
Antibody-based therapeutics use monoclonal antibodies that specifically bind to target proteins and modulate their function
Antibody engineering optimizes the affinity, specificity, and effector functions of antibodies
Targeted protein degradation induces the degradation of disease-causing proteins using small molecules
Proteolysis-targeting chimeras (PROTACs) recruit the ubiquitin-proteasome system to degrade the target protein
Synthetic Biology Applications
Metabolic engineering modifies the metabolic pathways of organisms to produce desired compounds (drugs, biofuels)
Uses enzymes and regulatory elements to optimize the flux through the pathway
Examples include the production of artemisinin (antimalarial drug) and isobutanol (biofuel) in engineered microbes
Biosensors are engineered proteins that detect specific molecules and generate a measurable output signal
Transcription factor-based biosensors activate gene expression in response to the target molecule
Fluorescent protein-based biosensors change their fluorescence properties upon binding the target
Genome editing tools (CRISPR-Cas9) enable precise modification of DNA sequences in living cells
Guide RNA directs the Cas9 nuclease to cut the target DNA, allowing for gene knockouts or insertions
Potential applications in gene therapy, disease modeling, and agricultural biotechnology
Synthetic gene circuits are engineered genetic networks that perform complex functions in living cells
Toggle switches and oscillators create bistable and oscillatory gene expression patterns
Logic gates (AND, OR) process multiple input signals to control gene expression
Cell-free systems use purified components (enzymes, ribosomes) to carry out biochemical reactions outside of living cells
Enable rapid prototyping and optimization of metabolic pathways and genetic circuits
Applications in point-of-care diagnostics, vaccine production, and biomanufacturing
Challenges and Future Directions
Protein misfolding and aggregation pose challenges for the production and stability of recombinant proteins
Chaperone engineering and directed evolution strategies can improve protein folding and solubility
Off-target effects of drugs can lead to adverse side effects and limit their therapeutic window
Improving the specificity of drug-target interactions and targeted delivery methods can mitigate off-target effects
Drug resistance can emerge due to mutations in the target protein or activation of alternative pathways
Combination therapies and targeting multiple nodes in a pathway can reduce the risk of resistance
Immunogenicity of protein-based drugs can elicit an immune response and reduce their efficacy
Humanization of antibodies and PEGylation can reduce immunogenicity
Scalability and cost-effectiveness of synthetic biology applications remain challenges for industrial implementation
Advances in bioprocess engineering and metabolic optimization can improve the economics of biomanufacturing
Ethical and societal considerations surrounding the use of synthetic biology and genome editing technologies
Public engagement, regulatory oversight, and responsible innovation frameworks are needed to address these concerns
Integration of artificial intelligence and machine learning methods can accelerate the drug discovery process
Deep learning can aid in target identification, compound screening, and de novo drug design
Personalized medicine approaches tailor drugs to an individual's genetic makeup and disease profile
Pharmacogenomics and companion diagnostics can guide the selection of optimal therapies for each patient