Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                
1 Protein Pioneers: A Comprehensive Study on Amino Acids and Proteins An Analysis of Structure, Function, and Modern Techniques Tonique Swaby June 10, 2024 Rockefeller University Summer Science Research Program 2 Table of Contents Introduction...................................................................................................................................................... 3 Fundamental Biology Concepts.......................................................................................................................4 Amino Acids................................................................................................................................................ 4 Protein.......................................................................................................................................................... 8 Protein Structures.............................................................................................................................................9 Primary Structure...................................................................................................................................9 Secondary Structure.............................................................................................................................11 Tertiary Structure.................................................................................................................................12 Quaternary Structure........................................................................................................................... 13 Enzymatic Roles:............................................................................................................. 14 Protein Purification Techniques.................................................................................................................... 15 Chromatography.........................................................................................................................................15 Affinity Chromatography.................................................................................................................... 16 Ion Exchange Chromatography...........................................................................................................17 Size Exclusion Chromatography (Gel Filtration)................................................................................20 Reverse Phase Chromatography..........................................................................................................23 SDS-PAGE (Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis).....................................26 Western Blotting.................................................................................................................................. 26 Ultracentrifugation.............................................................................................................................. 27 Tools Used to Predict Protein Structure....................................................................................................... 28 AlphaFold...................................................................................................................................................28 Rosetta........................................................................................................................................................29 Swiss Model...............................................................................................................................................31 Phyre2........................................................................................................................................................ 32 Techniques to Validate Predicted Structures............................................................................................... 33 X-ray Crystallography................................................................................................................................33 NMR Spectroscopy (Nuclear Magnetic Resonance)................................................................................. 34 Cryo-Electron Microscopy (Cryo-EM)......................................................................................................36 Conclusion....................................................................................................................................................... 38 3 Introduction Proteins are essential to life, forming the building blocks of cells, tissues, and organs. They perform a wide range of functions, including catalyzing metabolic reactions, replicating DNA, transporting molecules, and providing structural support. Central to understanding proteins are amino acids, the organic compounds that link to form these macromolecules. This paper explores the structures, properties, and functions of amino acids and proteins. We begin with a review of the biological concepts underlying amino acids, noting how their unique side chains contribute to protein functionality. We then discuss the hierarchical levels of protein structure—primary, secondary, tertiary, and quaternary—and how each level shapes protein function. Advances in biotechnology have transformed protein study and manipulation. Techniques such as chromatography are critical for isolating specific proteins for analysis, allowing researchers to understand protein interactions and functions in various contexts. Additionally, computational tools like AlphaFold, Rosetta, and SWISS-MODEL have significantly advanced our ability to predict protein structures with high accuracy. This paper provides a comprehensive overview of amino acids and proteins, detailing their structural complexities, purification methods, and modern predictive technologies. By combining foundational knowledge with recent research, we aim to highlight the crucial role of proteins in biochemistry and molecular biology. The following sections will cover fundamental concepts of amino acids and proteins, levels of protein structure, advanced protein purification techniques, and state-of-the-art tools for predicting protein structures, emphasizing the central role proteins play in life processes. 4 Fundamental Biology Concepts Amino Acids Amino acids are the fundamental building blocks of proteins. Each amino acid consists of an amino group (NH2), a carboxyl group (COOH), a hydrogen atom, and a variable R group that determines its properties. Proteins are long chains of amino acids, each with a unique sequence that dictates the protein's shape and function in the body.(Cleveland Clinic Medical) Amino acids can be compared to letters of the alphabet: when combined in various sequences, they form different proteins, much like letters form different words. Proteins are essential for numerous bodily functions, including breaking down food, growing, repairing tissues, and performing other critical roles. They can also serve as an energy source.(Amino Acids: MedlinePlus Medical Encyclopedia) When proteins are digested, they break down into amino acids. The human body uses these amino acids to synthesize new proteins needed for various bodily functions.(Cleveland Clinic Medical) Amino acids are categorized into three groups: 1. Essential amino acids: Must be obtained through diet, as the body cannot produce them. 2. Nonessential amino acids: Can be synthesized by the body. 3. Conditionally essential amino acids: Usually nonessential but become essential under certain conditions, such as illness or stress. (Cleveland Clinic Medical) Structure Amino acids are the building blocks of proteins, each sharing a basic structure: an α-carbon bonded to hydrogen, an α-carboxyl group, an α-amine group, and a variable R group (side chain) that determines the amino acid's properties. Except glycine, which has a hydrogen atom as its R group, all amino acids possess four different groups attached to the α-carbon, allowing them to exist in L and D forms. In biological systems, amino acids typically exist in the L configuration. (Libretexts) The general formula of an amino acid is R-CH(NH2)-COOH. These molecules combine to form proteins, crucial for many biological and chemical functions in the body, including growth, tissue repair, and metabolic processes. Amino acids generally have high melting and boiling points, exist as white crystalline compounds, and are water-soluble but insoluble in organic solvents. (Libretexts) 5 Common Amino Acids and Their Structures 1. Glycine (NH2-CH2-COOH): The simplest amino acid with a hydrogen atom as its side chain. It plays a critical role in forming alpha-helices in protein secondary structure and is an inhibitory neurotransmitter. (BYJUS) 2. Serine: Contains a hydroxymethyl group as its side chain, classifying it as a polar amino acid. It is nonessential, meaning it can be synthesized by the human body. (BYJUS) 3. Leucine: An essential amino acid with an isobutyl side chain. It must be obtained through the diet from sources like dairy products, meats, and legumes. 4. Cysteine: Has a thiol side chain and participates in enzymatic reactions and the formation of disulfide bonds, important for protein structure. It is semi-essential, often synthesized from methionine.(BYJUS) 5. Valine: An essential amino acid with an isopropyl side chain, crucial for protein biosynthesis and must be obtained through the diet. Basic Amino Acids Three amino acids have basic side chains at neutral pH: arginine (Arg), lysine (Lys), and histidine (His). Their side chains contain nitrogen, resembling ammonia, which gives them their basic properties. Lysine has two amine groups, enhancing its basic nature.(BYJUS) 6 Understanding these fundamental structures and properties of amino acids is crucial for studying proteins, as their specific sequences and combinations lead to the diverse and essential functions that proteins perform in living organisms.(BYJUS) Properties Amino acids exhibit diverse properties based on their side chains (R groups). These properties include polarity, charge, and hydrophobicity, which are critical for the structure and function of proteins.(ChatGPT) Polarity Polarity refers to the distribution of electric charge around the molecule. Amino acids can be polar or nonpolar based on the nature of their side chains. (ChatGPT) ● ● Polar Amino Acids: Have side chains that can form hydrogen bonds with water, making them hydrophilic (water-loving). ○ Examples: Serine, Threonine, Asparagine, Glutamine, Tyrosine. Nonpolar Amino Acids: Have side chains that do not interact well with water, making them hydrophobic (water-fearing). ○ Examples: Glycine, Alanine, Valine, Leucine, Isoleucine, Methionine, Phenylalanine, Tryptophan, Proline. Charge Charge at physiological pH (around 7.4) determines whether amino acids are acidic, basic, or neutral.(ChatGPT) ● ● ● Acidic Amino Acids: Have side chains that contain a carboxyl group, which can donate a proton, giving the amino acid a negative charge. ○ Examples: Aspartic acid (Asp), Glutamic acid (Glu). Basic Amino Acids: Have side chains that contain an amine group, which can accept a proton, giving the amino acid a positive charge. ○ Examples: Lysine (Lys), Arginine (Arg), Histidine (His). Neutral Amino Acids: Do not carry a net charge at physiological pH. They can be polar or nonpolar. ○ Examples of neutral polar: Serine (Ser), Threonine (Thr), Asparagine (Asn), Glutamine (Gln). ○ Examples of neutral nonpolar: Glycine (Gly), Alanine (Ala), Valine (Val). Hydrophobicity Hydrophobicity refers to the tendency of amino acids to avoid water.(ChatGPT) ● ● Hydrophobic Amino Acids: Typically have nonpolar side chains and are found in the interior of proteins, away from the aqueous environment. ○ Examples: Valine (Val), Leucine (Leu), Isoleucine (Ile), Methionine (Met), Phenylalanine (Phe), Tryptophan (Trp). Hydrophilic Amino Acids: Typically have polar or charged side chains and are found on the exterior of proteins, interacting with the aqueous environment. 7 ○ Examples: Serine (Ser), Threonine (Thr), Aspartic acid (Asp), Glutamic acid (Glu), Lysine (Lys), Arginine (Arg). 1. Polarity: ○ Polar: Serine, Threonine, Asparagine, Glutamine, Tyrosine. ○ Nonpolar: Glycine, Alanine, Valine, Leucine, Isoleucine, Methionine, Phenylalanine, Tryptophan, Proline. 2. Charge: ○ Acidic: Aspartic acid, Glutamic acid. ○ Basic: Lysine, Arginine, Histidine. ○ Neutral: (varies with polarity) ■ Polar: Serine, Threonine, Asparagine, Glutamine. ■ Nonpolar: Glycine, Alanine, Valine. 3. Hydrophobicity: ○ Hydrophobic: Valine, Leucine, Isoleucine, Methionine, Phenylalanine, Tryptophan. ○ Hydrophilic: Serine, Threonine, Aspartic acid, Glutamic acid, Lysine, Arginine (ChatGPT). Functions Amino acids play a critical role in your body by forming proteins and supporting various physiological functions. The different types of amino acids and their specific sequences determine the function of each protein they compose. Here are the key roles amino acids perform in your body: Breaking Down Food: Amino acids aid in the digestion process, helping to break down food into nutrients that the body can absorb and utilize. Growing and Repairing Body Tissue: They are essential for the growth, maintenance, and repair of tissues, including muscles, skin, and organs. Making Hormones and Neurotransmitters: Amino acids are precursors to hormones and neurotransmitters, which are critical for communication within the body and the brain. For example, tryptophan is necessary for the production of serotonin, a neurotransmitter that regulates mood, appetite, and sleep. Providing an Energy Source: Amino acids can be used as an energy source when carbohydrates and fats are not available, ensuring the body has enough energy to function. Maintaining Healthy Skin, Hair, and Nails: Proteins formed from amino acids are fundamental to the structural integrity of skin, hair, and nails, keeping them strong and healthy. Building Muscle: Amino acids, especially the branched-chain amino acids (BCAAs) like leucine, isoleucine, and valine, are vital for muscle protein synthesis and muscle growth. Boosting the Immune System: They play a role in the production of antibodies and immune cells, which help protect the body against infections and illnesses. 8 Sustaining a Normal Digestive System: Amino acids contribute to the maintenance and repair of the digestive tract, ensuring efficient digestion and absorption of nutrients. Protein Proteins are highly complex substances present in all living organisms, crucial for nutritional value and chemical processes essential for life. Recognized for their importance in the early 19th century, the term "protein" was coined by Swedish chemist Jöns Jacob Berzelius in 1838, derived from the Greek word "prōteios," meaning "holding first place."(Haurowitz and Koshland). Proteins are species-specific and organ-specific, meaning the proteins vary between different species and different organs within a single organism. (Haurowitz and Koshland) A protein molecule is significantly larger than molecules of sugar or salt, consisting of many amino acids joined in long chains, similar to beads on a string. (Haurowitz and Koshland) There are about 20 naturally occurring amino acids in proteins. Proteins with similar functions have similar amino acid compositions and sequences. While the functions of a protein cannot be fully explained by its amino acid sequence alone, established correlations between structure and function are linked to the properties of the constituent amino acids. (Haurowitz and Koshland) Plants have the ability to synthesize all amino acids, whereas animals cannot, even though all amino acids are essential for life. (Haurowitz and Koshland) Plants can thrive in a medium containing inorganic nutrients like nitrogen and potassium and utilize carbon dioxide during photosynthesis to form organic compounds such as carbohydrates. (Haurowitz and Koshland).In contrast, animals must obtain organic nutrients from external sources.(Haurowitz and Koshland) Due to the low protein content in most plants, animals that consume only plant material, such as ruminants (e.g., cows), need large amounts of plant material to meet their amino acid requirements. Nonruminant animals, including humans, primarily obtain proteins from animal sources and products like meat, milk, and eggs. (Haurowitz and Koshland).Additionally, legume seeds are increasingly used to prepare inexpensive, protein-rich foods, contributing to human nutrition. Structure, Roles & Function Proteins are polypeptide structures composed of one or more long chains of amino acid residues, and they play a crucial role in various biological functions(The Functions of Amino Acids | the Power of Amino Acids | Amino Acids | Ajinomoto Group Global Website - Eat Well, Live Well., n.d.): 1. DNA replication: Proteins are involved in the replication process, ensuring accurate copying of genetic material during cell division. 2. Transporting molecules: Proteins act as carriers, transporting molecules such as oxygen (e.g., hemoglobin), nutrients, and ions across cell membranes. 3. Catalyzing metabolic reactions: Protein enzymes catalyze biochemical reactions in cells, speeding up chemical processes necessary for metabolism. 4. Providing structural support: Proteins provide structural support to cells and tissues. For example, collagen provides strength to connective tissues, and cytoskeletal proteins maintain cell shape and structure. 9 5. Mediating signaling: Proteins play crucial roles in cell signaling pathways, transmitting signals from the cell membrane to the nucleus, regulating various cellular processes. 6. Gene expression control: Proteins regulate gene expression by binding to DNA and influencing the transcription process, thereby controlling which genes are turned on or off. Proteins are incredibly versatile molecules, and their functions are determined by their specific structures. The diversity in protein structure allows them to carry out a wide range of functions crucial for life. Enzymes, in particular, are remarkable proteins that catalyze biochemical reactions with great specificity and efficiency, making life processes possible.(The Functions of Amino Acids | the Power of Amino Acids | Amino Acids | Ajinomoto Group Global Website - Eat Well, Live Well., n.d.) Protein Structures Protein structures are hierarchical and complex, beginning with the primary structure, which is the linear sequence of amino acids linked by peptide bonds. This sequence determines the protein's secondary structure, characterised by local folded formations such as alpha helices and beta sheets, stabilized by hydrogen bonds. The tertiary structure involves the overall three-dimensional folding of the protein chain, influenced by various interactions among amino acid side chains. Some proteins further assemble into a quaternary structure, where multiple folded polypeptide subunits join to form a functional complex. Each level of structure is crucial, dictating the protein's specific function and its role in biological processes.(BYJU’S) Primary Structure The primary structure of proteins is foundational to their function. Accurate sequencing of amino acids is essential for the correct folding and function of the protein. Mutations that alter this sequence can lead to significant health issues, illustrating the importance of the primary structure in biological systems. (BYJUS) The primary structure of a protein refers to the specific sequence of amino acids in its polypeptide chain(s). This sequence is crucial because it determines the final three-dimensional shape and function of the protein. Any alteration in the amino acid sequence can significantly impact the protein's properties and its biological activity. (BYJUS) 10 Proteins are composed of one or more polypeptide chains, each formed by linking amino acids in a specific order. This order is dictated by the genetic code in DNA. The primary structure is stabilized by covalent peptide bonds between adjacent amino acids.(Libretexts) The gene, or sequence of DNA, encodes the unique sequence of amino acids in each polypeptide chain. Mutations in the DNA can alter the nucleotide sequence, leading to changes in the amino acid sequence, which can affect the protein's structure and function.(Libretexts) Changes in the amino acid sequence can lead to significant structural and functional alterations in the protein. For example, the oxygen-transport protein hemoglobin is composed of four polypeptide chains: two identical α chains and two identical β chains. In sickle cell anemia, a single amino acid substitution (glutamic acid replaced by valine in the β chain) causes the hemoglobin to fold differently, creating dysfunctional proteins that aggregate under low-oxygen conditions. This aggregation distorts red blood cells into a sickle shape, leading to various health issues, such as breathlessness, dizziness, headaches, and abdominal pain. (Libretexts) Insulin, a hormone that regulates blood sugar levels, consists of two polypeptide chains: the A chain with 21 amino acids and the B chain with 30 amino acids. Each sequence is unique to the insulin protein and is crucial for its proper function. The primary structure of insulin is as follows: Example: Hemoglobin and Sickle Cell Anemia The primary structure of hemoglobin includes specific sequences in its α and β chains. A mutation causing sickle cell anemia involves the substitution of valine for glutamic acid at position 6 in the β chain. This single change causes the hemoglobin to form fibers under low-oxygen conditions, distorting red blood cells and impairing their function. 11 Secondary Structure The secondary structure of a protein refers to local folded structures that form within a polypeptide chain due to interactions between atoms of the backbone. These structures, such as α-helices and β-pleated sheets, play a crucial role in determining the overall three-dimensional shape and stability of the protein. Types of Secondary Structures 1. α-Helix: The α-helix is a right-handed coil formed by hydrogen bonding between the oxygen atom of the carbonyl group in one amino acid and the hydrogen atom of the amino group in another amino acid, located four residues down the chain. Each helical turn consists of approximately 3.6 amino acid residues. The side chains (R groups) of amino acids protrude outward from the helix and do not participate in hydrogen bonding. 2. β-Pleated Sheet: β-Pleated sheets consist of extended polypeptide chains lying alongside each other, forming a sheet-like structure. The structure resembles pleated folds, with hydrogen bonds forming between adjacent chains. β-Pleated sheets can be either parallel or antiparallel, depending on the directionality of the peptide bonds. The side chains of amino acids point outward from the sheet and do not participate in hydrogen bonding within the sheet. Formation and Stability Secondary structures arise from hydrogen bonding between neighboring amino acids in the polypeptide chain. Although a single secondary structure rarely extends throughout the entire chain, they are crucial for the overall protein fold. Certain amino acids have a propensity to form α-helices, while others favor β-pleated sheets, based on their side chain interactions.(“Chapter 3: Investigating Proteins Chemistry”) 12 Impact on Protein Function Secondary structures play a structural role in most globular and fibrous proteins. They contribute to the stability and functional properties of proteins, influencing interactions with other molecules and their biological activities.(“Chapter 3: Investigating Proteins - Chemistry”) Genetic and Structural Influences The sequence of amino acids in the polypeptide chain, determined by the genetic code, influences the formation of secondary structures. Mutations can alter the secondary structure, affecting the protein's function and leading to various disorders.(“Chapter 3: Investigating Proteins - Chemistry”) Tertiary Structure The tertiary structure of proteins represents the overall folding of the polypeptide chains, beyond the secondary structure. It determines the precise three-dimensional shape of the protein, crucial for its function and activity. Molecular Shapes 1. Fibrous and Globular: Tertiary structures give rise to two major molecular shapes: Fibrous: Long, filamentous proteins with repeated secondary structures, providing structural support. Examples include keratin and collagen. Globular: Compact, spherical proteins with complex folding patterns, often with enzymatic or regulatory functions. Examples include enzymes and antibodies.(“Chapter 3: Investigating Proteins - Chemistry”) Stabilizing Forces The tertiary structure is stabilized by various forces: ● Hydrogen Bonds: Form between polar groups within the protein. Hydrogen bonds form between polar groups within the protein, such as between hydrogen and oxygen or nitrogen atoms.They contribute to the folding of the protein by stabilizing secondary structures like alpha helices and beta sheets, as well as maintaining specific interactions between amino acid residues. 13 ● Disulfide Linkages: Covalent bonds between cysteine residues.Disulfide linkages are covalent bonds formed between cysteine residues.They provide structural stability by covalently linking distant parts of the protein chain, preventing unfolding or denaturation. Disulfide bonds are crucial for maintaining the protein's tertiary structure, particularly in extracellular proteins. ● Van der Waals Forces: Weak attractions between nonpolar groups.They contribute to the compact folding of the protein by stabilizing interactions between hydrophobic residues. Van der Waals forces help bring nonpolar groups close together, promoting the formation of the protein's core structure. Electrostatic Forces: Attraction between oppositely charged groups.They stabilize the protein structure by forming salt bridges between positively and negatively charged amino acid side chains. These interactions can stabilize specific conformations and play a role in protein-protein interactions.(“Chapter 3: Investigating Proteins - Chemistry”) ● Protein Folding Process Interactions between polar, nonpolar, acidic, and basic R-groups determine the complex tertiary structure. Hydrophobic R-groups tend to be buried in the interior of the protein, while hydrophilic R-groups are exposed on the protein's surface. Disulfide linkages between cysteine residues stabilize the protein's structure under oxidizing conditions. These interactions collectively determine the final three-dimensional shape of the protein.(“Chapter 3: Investigating Proteins - Chemistry”) Functional Implications Proper tertiary structure is essential for protein function. Loss of the three-dimensional shape leads to loss of function. Protein folding takes place in the aqueous environment of the body, ensuring correct folding and stability.(“Chapter 3: Investigating Proteins - Chemistry”) Quaternary Structure The quaternary structure of proteins refers to the spatial arrangement of subunits within a protein molecule, formed by the assembly of multiple polypeptide chains. . Quaternary Structure Definition: - Purpose: It describes how subunits are oriented and arranged with respect to each other. Function:Provides stability and functional diversity to multi-subunit proteins.(“Chapter 3: Investigating 14 2. Domains: - Purpose: Domains are parts of the protein chain with their own unique three-dimensional fold and specific function. -Function: Considered as evolutionary and functional building blocks of proteins, domains contribute to the protein's overall structure and function.(“Chapter 3: Investigating Proteins - Chemistry”) Protein Evolution and Functional Insights The exact amino acid sequence of each protein determines its tertiary structure, which is crucial for its biological activity. Protein Evolution: - Purpose: Provides structural insight and connects proteins from different metabolic pathways. - Function:Helps in understanding the evolutionary relationships and functional similarities among proteins. Stabilization of Quaternary Structure In multi-subunit proteins, weak interactions between subunits stabilize the overall structure, often facilitated by enzymes: Enzymatic Roles: - Purpose: Enzymes facilitate bonding between subunits to form the final functional protein. - Function: Enzymes play a crucial role in stabilizing the quaternary structure of proteins, ensuring proper function. 15 Insulin Structure: Insulin is a globular protein composed of two polypeptide chains held together by hydrogen bonds and disulfide bonds. Function:The arrangement of subunits contributes to its stability and function as a hormone. -Silk Fibroin: Structure:Silk is a fibrous protein resulting from hydrogen bonding between different β-pleated chains. Function:The quaternary structure of silk provides strength and resilience to silk fibers. Protein Purification Techniques Protein purification is a series of processes aimed at isolating one or a few proteins from a complex mixture, typically derived from cells, tissues, or whole organisms. This process is essential for characterizing the function, structure, and interactions of the protein of interest. It involves separating the protein from non-protein components and from other proteins. The most laborious aspect is often the separation of one protein from all others, which typically exploits differences in size, physico-chemical properties, binding affinity, and biological activity. The end result is termed a protein isolate.(“Chapter 3: Investigating Proteins - Chemistry”) Protein purification can be either preparative or analytical. Preparative purifications aim to produce large quantities of purified protein for subsequent use, such as in commercial products (e.g., enzymes like lactase, nutritional proteins like soy protein isolate, and biopharmaceuticals like insulin). These steps often involve removing by-products, like host cell proteins, that could be harmful to patients. Analytical purification, on the other hand, produces small amounts of protein for research purposes, such as identification, quantification, and studying the protein's structure, post-translational modifications, and function. Pepsin and urease were among the first proteins purified to the point of crystallization.(“Chapter 3: Investigating Proteins - Chemistry”) Chromatography Protein purification employs various chromatography techniques to isolate specific proteins based on their unique properties.(“Affinity Chromatography | Principles”) Among these, affinity chromatography is particularly effective for purifying tagged proteins, which are engineered with tags like His-tag or GST-tag that bind specifically to ligands on the resin. This method ensures high specificity and efficiency, resulting in highly pure proteins. Other chromatography techniques used include ion exchange chromatography, which separates proteins by charge; size exclusion chromatography, which separates based on molecular size; and hydrophobic interaction chromatography, which exploits differences in hydrophobicity. Each technique leverages distinct protein characteristics, facilitating their separation and purification.(“Affinity Chromatography | Principles”) 16 Affinity Chromatography Purification via affinity chromatography is a powerful technique to isolate your protein of interest from a sample. In this method, proteins are marked with "tags" that confer specific binding properties (Fig. 3, Table 4). These tags allow the protein to bind to an immobilized ligand on the chromatography column (Fig. 1). As a result, the tagged protein is selectively retained on the column, effectively separating it from other components in the sample. This high specificity makes affinity chromatography an efficient and precise method for protein purification.(“Chapter 3: Investigating Proteins - Chemistry”) Principle of Affinity Chromatography Affinity chromatography is a powerful technique used to purify proteins by exploiting specific interactions between the target molecule and a ligand. (“Chapter 3: Investigating Proteins - Chemistry”)The process involves a stationary phase (solid phase) and a mobile phase. The mobile phase typically contains the mixture of biomolecules, such as cell lysate. A ligand that specifically binds the target molecule is covalently attached to the solid phase. (“Chapter 3: Investigating Proteins Chemistry”)The target molecule binds to the ligand as the mobile phase passes through the solid phase, while most other molecules flow through. To elute the target biomolecule, conditions such as pH or salt concentrations are changed, or a free ligand competes with the bound target molecule. The most crucial property of the solid phase is its ability to immobilize the ligand. Materials such as acrylates or silica gels are commonly used for this purpose. An inhibitor, known as a spacer, is attached to the solid phase to avoid steric interference. This spacer, typically a hydrocarbon chain, ensures proper interaction between the target molecule and the ligand. Chemicals like cyanogen bromide or epoxide are used to functionalize the solid phase with varying carbon chain lengths, enhancing the efficiency of the purification process.Affinity chromatography separates biomolecules based on specific binding interactions between the target molecule and a ligand attached to a solid phase.(“Chapter 3: Investigating Proteins - Chemistry”) 17 Applications of Affinity Chromatography Often used to purify proteins with a known affinity tag (e.g., His-tag, GST-tag) by using columns with corresponding ligands (e.g., nickel for His-tagged proteins). Boronate affinity chromatography can be used to analyze hemoglobin A1c (HbA1c), a component of glycated hemoglobin, and to capture glycoproteins like lactoferrin using a capillary boronate affinity monolith structure.(News-Medical) Similarly, cellufine phenyl borate, an affinity ligand, can purify glycoproteins, glycated-proteins, and diol compounds. The beads are packed in a spherical cellulose structure and operate effectively within a pH range of 3 to 12.(News-Medical) Ion Exchange Chromatography Ion-exchange chromatography is a widely used method for protein purification that exploits charge-charge interactions between proteins and charged resins.(Adhikari et al.) There are two types of resins: cation-exchange resins for positively charged proteins and anion-exchange resins for negatively charged proteins. (Adhikari et al.)For cation-exchange chromatography, the pH of the binding buffer must be below the protein's isoelectric point (pI). Cation-exchange resins typically contain sulfate derivatives (S-resins), while anion-exchange resins (CM resins) have carboxylate-derived ions.(Adhikari et al.) Despite its effectiveness, ion-exchange chromatography often does not achieve single-step purification due to its lack of specificity.(Adhikari et al.) At its core, ion exchange chromatography operates on the principles of electrostatic interactions between charged molecules and a solid stationary phase containing fixed charged groups.(Qiu) The stationary phase can be made up of a resin with either positively charged groups (cation exchange) or negatively charged groups (anion exchange). (Qiu). The sample containing the mixture of charged molecules is loaded onto the column, and as the mobile phase flows through, molecules interact with the charged groups on the stationary phase.(Qiu) Depending on their charge, molecules will either be attracted to or repelled by the stationary phase, leading to separation.(Qiu) The separation process involves several steps: 1. Sample loading: The sample mixture is applied to the column, and the charged molecules interact with the oppositely charged groups on the stationary phase. (Qiu)This interaction determines the retention of the molecules. 2. Washing:Unbound or weakly bound molecules are washed away by the mobile phase, typically a buffer solution. (Qiu)This step helps remove impurities and further concentrate the target molecules on the column.(Qiu) 18 3. Elution: Elution is the critical step where a gradient of increasing salt concentration or a change in pH disrupts the electrostatic interactions between the charged molecules and the stationary phase. As the conditions change, molecules are released from the column at different times based on their affinity for the stationary phase.(Qiu) 4. Analysis and collection:The eluted fractions are collected and analyzed using various techniques such as UV spectrophotometry, fluorescence, or mass spectrometry, depending on the nature of the molecules being separated.(Qiu) Cation Exchange Cation exchange chromatography is a type of ion exchange chromatography (IEX) used to separate molecules based on their net surface charge.(“Cation Exchange Chromatography”BioRad) Specifically, it employs a negatively charged ion exchange resin that has an affinity for positively charged molecules. This versatile technique is used for both preparative and analytical purposes and can separate a wide range of molecules, including amino acids, nucleotides, and large proteins.(“Cation Exchange Chromatography”BioRad) A protein's net surface charge varies with pH, determined by its isoelectric point (pI). At the pI, the protein has no net charge; below the pI, it's positively charged, and above, it's negatively charged. A negatively charged cation exchange resin is selected by choosing a buffer pH below the protein's pI, ensuring the protein binds. Proteins with varying pI values bind to the resin with different strengths, enabling their separation based on charge. For instance, at a pH of 7.5, proteins with pI > 7.5 will carry a net positive charge and bind to the resin. A salt gradient then elutes proteins based on their net surface charge, with those closer to 7.5 eluting first and those with higher pI values requiring higher salt concentrations for elution.(“Cation Exchange Chromatography”BioRad) The choice of buffer pH in cation exchange chromatography is crucial. By selecting a pH below the protein's pI, where the protein carries a net positive charge, a negatively charged resin is preferred. (“Cation Exchange Chromatography”BioRad).This ensures the protein of interest binds to the resin. Proteins with different pI values will have varying degrees of charge at a given pH, leading to differential binding to the resin. A salt gradient is then applied to elute bound proteins based on their net surface charge, facilitating the separation of proteins based on their charge characteristics.(“Cation Exchange Chromatography” BioRad) The choice between cation and anion exchange resins depends on the net charge of the molecules being captured. Cation exchange resins are used for positively charged molecules, while anion exchange resins are chosen for negatively charged molecules.(“Cation Exchange Chromatography” BioRad) For tasks like removing negatively charged DNA or endotoxins, a strong anion exchange column like Bio-Rad’s ENrich Q chromatography column is a great option. A negatively charged cation exchange resin wouldn't effectively capture DNA or endotoxins from the sample.(“Cation Exchange Chromatography” BioRad) Proteins, unlike DNA, can carry either a net positive or net negative charge depending on the buffer pH. While theoretically, proteins could be purified with either resin type, their stability at different pH levels 19 must be considered. The choice between cation and anion exchange resins for protein purification is influenced by protein stability and buffer conditions.(“Cation Exchange Chromatography” BioRad) Anion Exchange Anion exchange chromatography is a type of ion exchange chromatography (IEX) utilized to separate molecules based on their net surface charge. Specifically, it employs a positively charged ion exchange resin with an affinity for molecules carrying net negative surface charges. This technique is employed for both preparative and analytical purposes and can effectively separate a wide range of molecules, from small amino acids and nucleotides to large proteins.(“Anion Exchange Chromatography” BioRad) A protein's net surface charge varies with pH, dictated by its isoelectric point (pI). At the pI, the protein has no net charge, below it's positively charged, and above, it's negatively charged.(“Anion Exchange Chromatography” BioRad) The pI, determined by the amino acid sequence, allows selecting a buffer to achieve a known net charge. An anion exchange resin is chosen when the protein carries a net negative charge. (“Anion Exchange Chromatography” BioRad). Different proteins with varying pI values bind to the resin with different strengths, facilitating separation. At a specific buffer pH, appropriately charged proteins bind the resin. For instance, at pH 7.5, proteins with pI < 7.5 carry a net negative charge and bind the positively charged resin. A salt gradient separates proteins based on their net surface charge, with those closer to 7.5 eluting first. This process enables the separation of proteins based on their charge characteristics.(“Anion Exchange Chromatography” BioRad) The choice between anion and cation exchange resins depends on the net charge of the molecules being captured. Cation exchange resins are used for positively charged molecules, while anion exchange resins are chosen for negatively charged molecules.(“Anion Exchange Chromatography” BioRad) For applications like removing negatively charged DNA or endotoxins, a strong anion exchange column like Bio-Rad’s ENrich Q chromatography column is ideal. A negatively charged cation exchange resin wouldn't capture DNA or endotoxins effectively.(“Anion Exchange Chromatography” BioRad) Proteins can carry either a net positive or negative charge depending on the buffer pH. While theoretically, proteins could be purified with either resin type, their stability at different pH levels must be considered. The choice between anion and cation exchange resins for protein purification depends on protein stability and buffer conditions.(“Anion Exchange Chromatography” BioRad) 20 Applications of Ion Exchange Chromatography Protein purification: Ion exchange chromatography is commonly used to purify proteins from complex mixtures, providing high purity and yield by selecting the appropriate stationary phase and elution conditions. Pharmaceutical industry: Ion exchange chromatography is crucial in drug development for separating and purifying active pharmaceutical ingredients, ensuring medication quality and safety. Environmental analysis: This technique is vital in water and soil analysis, separating and quantifying ions, heavy metals, and pollutants to aid environmental monitoring and regulatory compliance. Nucleic acid separation: Ion exchange chromatography is used to separate DNA and RNA fragments based on their charge characteristics, essential in molecular biology research and genetic analysis. Biotechnology: In biotechnology, ion exchange chromatography is employed in downstream processing to isolate and purify biomolecules like enzymes, antibodies, and hormones. Size Exclusion Chromatography (Gel Filtration) Size exclusion chromatography (SEC), also known as gel filtration chromatography, is a versatile protein separation method often overlooked but holds significant value beyond just a polishing step.(Proteins) At Peak Proteins, we recognize it as a powerful technique providing crucial insights into protein behavior and facilitating buffer exchange.(Proteins) In essence, SEC acts as a molecular sieve, separating proteins, salts, and small molecules based on their molecular weight. (Proteins)The stationary phase consists of a porous matrix where molecules too large to enter the pores remain in the mobile phase and flow through the column with the buffer, while smaller molecules enter the pores and are retained, thus separating them by size.(Proteins) 21 Mechanism, Steps & Phases Size exclusion chromatography (SEC) separates molecules based on their size by filtration through a gel matrix. (“Introduction to Size Exclusion Chromatography” BioRad)The gel consists of spherical beads with pores of specific size distribution. Separation occurs when molecules of different sizes are either included or excluded from the pores within the matrix. (“Introduction to Size Exclusion Chromatography” BioRad)Small molecules diffuse into the pores and their flow through the column is slowed according to their size, while large molecules do not enter the pores and are eluted in the column's void volume. Consequently, molecules are separated based on their size as they pass through the column and are eluted in order of decreasing molecular weight (MW).(“Introduction to Size Exclusion Chromatography” BioRad) The choice of operating conditions and gel selection depends on the application and desired resolution. Two common types of separations performed by SEC are fractionation and desalting (or buffer exchange).(“Introduction to Size Exclusion Chromatography” BioRad) 22 The principle of size exclusion chromatography (SEC), also known as Gel-Filtration chromatography, relies on separating molecules based on their size or molecular weight. (Proteogenix)This technique utilizes a resin composed of porous particles or beads, with varying pore sizes.(Proteogenix) These pores enable the entry or exclusion of molecules depending on their size while remaining chemically inert.(Proteogenix) In gel-filtration chromatography: 1. Mobile Phase: The liquid outside the particles constitutes the mobile phase.(Proteogenix) 2. Stationary Phase: Inside the porous beads forms the stationary phase. The process involves: 1. Sample Injection: A mixture of molecules of different sizes is injected into the chromatography column.(Proteogenix) 2. Size Separation: As the mixture flows through the column, molecules move at different rates based on their size. Small and medium-sized molecules are trapped within the pores, while larger elements like aggregates and complexes remain in the mobile phase.(Proteogenix) 3. Elution: Large molecules elute first, followed by medium-sized molecules, and finally smaller molecules, as they are excluded from the pores and pass through the column.(Proteogenix) 23 Applications & When To Use Size Exclusion Chromatography (SEC) offers numerous benefits, particularly when seeking contaminant-free or sensitive proteins.(Proteogenix) It ensures improved purity by removing small-size contaminants and aggregates gently, without harsh conditions. (Proteogenix)Unlike other techniques, SEC doesn't require buffer changes, making it ideal for proteins sensitive to pH fluctuations. Additionally, SEC is adaptable to a wide temperature range and allows flexible denaturation concentrations.(Proteogenix) SEC finds diverse applications: 1. Enhancing Purity: SEC chromatography serves as a purification step to elevate final product purity, especially in preparative SEC.(Proteogenix) 2. Purity Analysis: SEC-HPLC efficiently profiles protein samples and assesses product purity.(Proteogenix) 3. Research Studies: Gel-filtration aids in purification, separates molecules by molecular weights, maintains protein stability, and enables thorough protein characterization.(Proteogenix) 4. Endotoxin Removal: SEC chromatography eliminates endotoxins crucial for pre-clinical and clinical studies.(Proteogenix) 5. Buffer Exchange: It's utilized to switch purified molecules to more appropriate buffers before, between, or after purification.(Proteogenix) 6. Desalting: Separates small molecules and contaminants from large molecules and small contaminants from crude lysates. 7. Aggregate Monitoring: SEC monitors aggregates and separates them from small-size molecules. To optimize SEC performance: - Sample Size and Molecular Weight: Implement SEC in the final purification stages for maximum resolution. - Choice of Eluent: Utilize a high ionic strength elution buffer to minimize electrostatic interactions. - Flow Rate Impact: Opt for a moderate flow rate, tailored to the chosen matrix, for optimal resolution. Reverse Phase Chromatography Reversed-phase chromatography (RPC) is a specialized technique within bonded-phase chromatography, where the mobile phase is more polar than the stationary phase. Typically, the mobile phase consists of an aqueous solution, making it suitable for a wide range of practical applications.(Poole) 24 Reverse-phase chromatography (RPC) stands out as the preferred method for purifying, separating, and analyzing biological molecules due to its excellent resolution and user-friendly nature. Column packings typically consist of silica particles with hydrophobic long-chain alkylsilyl ligands such as n-butyl (C4), n-octyl (C8), n-octadecyl (C18), and alkylphenyl groups, especially for enzyme separation.(Poole) In RPC, nonhydrophobic molecules in the sample elute early as they do not strongly interact with the hydrophobic stationary phase, whereas hydrophobic molecules interact more and elute later.(Poole) Several factors influence the effectiveness of RPC: Column packing material: Silica is commonly used due to its mechanical stability and efficiency in bonding with hydrophobic ligands.(Poole) Particle size and pore diameter: Smaller support particles offer higher resolution but may lead to increased column backpressure. An optimal particle size of ≈20 μm is preferred for enzymatic HPLC.(Poole) Mobile phase: Usually an aqueous solution, which is more polar than the stationary phase.(Poole) Length of hydrophobic ligands: Longer ligands provide stronger retention for hydrophobic molecules. Support stability:Silica supports are widely used but may not be stable under basic conditions (pH>8). Pore size:A pore size of 300 Å is common, but larger pore sizes (≈1000 or greater) are suggested for large enzyme molecules to avoid diffusional issues.(Poole) Principle In Reversed-phase chromatography models, the solid phase is a polar component, and the working stage is a nonpolar organic component. Polar components dispersed in the mobile phase are captivated by the polar stationary phase. However, nonpolar components are dissolved along with the solvent.(Poole) This serves as the foundation for separation. This is referred to as normal phase chromatography. In reverse phase chromatography, the stationary phase is made. For molecule separation, various biochemical activities varying from electrostatic interaction to biology affinity are being used.(Poole) The alkyl or aromatic binding sites are tightly linked to the stationary phase in reversed-phase chromatography to provide a hydrophobic nature. A polar solvent comprising the solutes moves through the mobile phase and passes over the hydrogen bonding stationary phase.(Poole) Hydrophobic solutes in the mobile phase can stick to the stationary phase via hydrophilic groups and thus provide the backbone of detachment. This is recognized as “reversed-phase” chromatography since the basic concept is the exact reverse of how it has been used by classical chromatography. Hence the normal phase and reversed-phase chromatography depict the following stationary & mobile phases. ● normal phase chromatography – Exhibit polar stationary phase and a less polar mobile phase ● reverse phase chromatography – Exhibit less polar stationary phase and polar mobile phase 25 Applications 1. Protein and Peptide Analysis: RP-HPLC is widely used for analyzing proteins and peptides, including characterization, quantification, and identification.(UNACADEMY) 2. Pharmaceutical Analysis:It is essential in pharmaceutical analysis for quality control, impurity profiling, and quantification of drug substances and related compounds.(UNACADEMY) 3. Metabolomics: RP-HPLC plays a crucial role in metabolomics studies by separating and quantifying metabolites in biological samples.(UNACADEMY) 4. Environmental Analysis: It is used for the analysis of pollutants, environmental contaminants, pesticides, and other chemicals in environmental samples.(UNACADEMY) 5. Food and Beverage Analysis: RP-HPLC is employed for quality control and analysis of food and beverage products, including detecting additives, contaminants, and nutritional components.(UNACADEMY) 6. Biochemical Research: It is utilized in biochemical research for the separation and purification of biomolecules, such as enzymes, nucleic acids, and lipids.(UNACADEMY) 7. Clinical Diagnostics:RP-HPLC is used in clinical laboratories for analyzing biomarkers, hormones, drugs, and metabolites in biological fluids.(UNACADEMY) 8. Peptide Synthesis and Purification: RP-HPLC is employed in peptide synthesis and purification processes, enabling the isolation of synthetic peptides with high purity.(UNACADEMY) 9. Natural Product Isolation:It is used to isolate and purify natural products from complex mixtures, including plant extracts, marine organisms, and microbial cultures.(UNACADEMY) 10. Forensic Analysis: RP-HPLC is utilized in forensic laboratories for analyzing drugs of abuse, toxicological substances, and trace evidence in criminal investigations.(UNACADEMY) 26 SDS-PAGE (Sodium Dodecyl Sulfate Polyacrylamide Gel Electrophoresis) Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) is a widely used technique for achieving high-resolution separation of complex protein mixtures.(Nowakowski et al.) The method involves denaturing the proteins by treating them with SDS, which imparts a uniform negative charge to the proteins, effectively linearizing them. (Nowakowski et al.)When subjected to an electric current in a polyacrylamide gel matrix, the negatively charged proteins migrate through the gel based on their size. (Nowakowski et al.)Since the migration rate is primarily determined by the molecular weight of the proteins rather than their charge, SDS-PAGE allows for precise separation and visualization of proteins according to their size.(Nowakowski et al.) Essentially, it denatures proteins and gives them a uniform negative charge, allowing separation based on size when an electric current is applied. The complete system is made of sodium lauryl sulfate sodium dodecyl sulfate and the gel, polyacrylamide. (UNACADEMY). The entire process needs to be done at a particular heat. The gel has prime importance as it takes away the basic characteristics of the protein molecules. The protein molecules get separated based on their polypeptide chain length. (UNACADEMY) Applications There are many applications of SDS-PAGE (UNACADEMY): ● It is used in peptide mapping ● It analyzes the post-translational modifications. ● It is used in separating the HIV proteins in the HIV tests. ● It is used in measuring the size of the protein ● It is used in the eradication of the pureness of proteins. Western Blotting Western blotting, a commonly used technique in research, combines SDS-PAGE separation with protein transfer to a membrane and specific antibody detection. Initially, a mixture of proteins is separated based on molecular weight using gel electrophoresis, creating distinct bands for each protein type. Following separation, the proteins are transferred to a membrane, where each protein forms a band. The membrane is then incubated with antibodies that are specific to the protein of interest, allowing for its identification and quantification. This method enables the detection of specific proteins within complex mixtures and is widely utilized in various biological and biomedical studies.(Yang and Mahmood) 27 Applications Western blotting, a versatile technique, is utilized for the study of proteins, quantification of their concentration in a sample, and aiding in the identification of diseases such as HIV. (Yang and Mahmood). It allows the detection of specific proteins within complex mixtures and provides valuable information about their abundance and size. (Yang and Mahmood) Western blotting is also employed in various research areas including molecular biology, cell biology, immunology, and biochemistry. (Yang and Mahmood)Moreover, it is a crucial tool in pharmaceutical development for assessing the expression of therapeutic proteins.(Yang and Mahmood) However, Western blotting requires high-quality reagents and sophisticated equipment to ensure reliable and accurate results, making it a high-throughput technique suitable for diverse applications in biomedical and life sciences research.(Yang and Mahmood) Ultracentrifugation Ultracentrifugation is a powerful technique used for separation based on the centrifugal force generated by extremely rapid rotation, typically exceeding 50,000 revolutions per minute.(“Ultracentrifugation | Chemistry”, Encyclopaedia Brittanica) In this method, particles or molecules within a sample are subjected to high gravitational forces, causing them to sediment based on their mass or size. Heavier or larger species settle at higher speeds compared to lighter or smaller ones, allowing for their efficient separation. (“Ultracentrifugation | Chemistry”, Encyclopaedia Brittanica) Ultracentrifugation is widely employed in various fields such as biochemistry, molecular biology, and biophysics for isolating subcellular components, studying macromolecular complexes, and analyzing molecular interactions.(“Ultracentrifugation | Chemistry”, Encyclopaedia Brittanica) Applications Protein Separation and Purification:Ultracentrifugation is pivotal in purifying proteins, separating cellular components, and eliminating impurities, ensuring high purity levels essential for protein structure analysis and drug development.(Anonymous) Virus Concentration: In virology, ultracentrifugation concentrates and purifies viruses from cell cultures or biological samples, facilitating the study of virus morphology, protein composition, and nucleic acid content.(Anonymous) Particle Size Analysis:Ultracentrifugation aids in precise particle size analysis in nanotechnology and material science, allowing researchers to characterize nanoparticles for drug delivery and materials development.(Anonymous) Isolation of Cellular Organelles: Cell biology relies on ultracentrifugation to isolate and study cellular organelles like mitochondria, nuclei, and ribosomes, providing insights into cellular functions and structures. Density Gradient Separation: Density gradient centrifugation separates components based on buoyant densities, useful for isolating subcellular fractions and separating similar-sized molecules. Viral Vector Production:Ultracentrifugation is crucial for producing viral vectors in gene therapy and vaccine development, concentrating and purifying viral vectors to ensure their safety and efficacy. Lipoprotein Analysis: Researchers in lipid metabolism and cardiovascular disease use ultracentrifugation to separate and analyze lipoproteins in blood samples, aiding in understanding their role in health and disease. 28 Tools Used to Predict Protein Structure Understanding a protein's structure offers profound insights into its function, enabling researchers to formulate hypotheses on influencing, controlling, or modifying it.(Dinesh-Supreme) For instance, having detailed structural information can facilitate the design of site-directed mutations to alter the protein's function. (Dinesh-Supreme) Additionally, structural knowledge allows for predicting potential binding molecules, which is crucial for drug design and therapeutic interventions. (Dinesh-Supreme)This deeper comprehension can drive advancements in fields such as biotechnology, medicine, and pharmacology, where precise manipulation of protein function is often essential. (Dinesh-Supreme) AlphaFold Developed By Google DeepMind & EMBL - European Bioinformatics Institute (Database|Alphafold) Principle AlphaFold, an artificial intelligence developed by DeepMind, employs deep learning algorithms to predict the 3D structures of proteins based solely on their amino acid sequences. This groundbreaking technology has significantly advanced the field of structural biology by providing high-accuracy predictions that previously required extensive experimental work. In collaboration with EMBL-EBI, AlphaFold has also created a publicly accessible database, offering free access to thousands of predicted protein structures. This resource is invaluable for researchers, facilitating advancements in understanding protein functions, drug design, and various other applications in biomedical research.(Dinesh-Supreme) Impact AlphaFold has achieved unprecedented accuracy in predicting protein structures, often rivaling experimental methods. The availability of a large number of highly accurate models has significantly impacted several areas of life sciences. Data providers leverage programmatic access to AlphaFold models to display structures for proteins that previously had no reliable models. Bioinformatics researchers conduct high-throughput analyses to uncover patterns and characteristics on an unprecedented scale. Structural biologists utilize AlphaFold models to solve structures by combining experimental data with theoretical model coordinates. Researchers and pharmaceutical companies employ structure-based drug discovery techniques using these models. Additionally, AlphaFold models have become powerful tools for teaching and understanding structural biology, leading to the development of new and insightful training materials.(Dinesh-Supreme) 1. Data Providers: Platforms like UniProt and PDB now display AlphaFold-predicted structures for proteins lacking reliable experimental models, enhancing data completeness and utility.(Dinesh-Supreme) 2. Bioinformatics Research: High-throughput analyses enabled by AlphaFold models have led to discoveries of new protein families, functional sites, and evolutionary relationships, which were previously challenging to identify.(Dinesh-Supreme) 3. Structural Biology: AlphaFold predictions assist in experimental structure determination by providing initial models that guide X-ray crystallography and cryo-EM 29 studies, thus accelerating the process and improving accuracy.(Dinesh-Supreme) 4. Drug Discovery: Pharmaceutical companies utilize AlphaFold models for virtual screening, structure-based drug design, and identifying potential drug targets, which has streamlined the drug discovery pipeline.(Dinesh-Supreme) 5. Education and Training: AlphaFold models are integrated into educational programs, offering students and researchers an accessible way to learn about protein structure and function, and creating new opportunities for training in computational biology and structural bioinformatics. (Dinesh-Supreme) Applications AlphaFold, with its unprecedented accuracy in predicting protein structures, has become a powerful tool in various aspects of drug discovery, disease research, and protein engineering: 1. Drug Discovery: AlphaFold's ability to accurately predict protein structures aids in identifying potential drug targets and designing therapeutics. By understanding the structure of target proteins involved in diseases, researchers can develop drugs that specifically interact with these targets, leading to more effective treatments.(Dinesh-Supreme) 2. Understanding Disease Mechanisms: Predicted protein structures provide insights into the molecular mechanisms underlying diseases. By studying the 3D structures of proteins involved in various diseases, such as cancer or neurodegenerative disorders, researchers can uncover disease mechanisms at the molecular level, leading to new therapeutic approaches.(Dinesh-Supreme) 3. Designing Novel Proteins: AlphaFold enables the design of novel proteins with specific functions or properties. By predicting the structures of designed proteins, researchers can engineer proteins for various applications, including enzyme catalysis, drug delivery, or biomaterials.(Dinesh-Supreme) Rosetta Developed By: Rosetta, initially developed in Dr. David Baker's laboratory at the University of Washington, started as a structure prediction tool but has since evolved to address various computational challenges in macromolecular research.(The Rosetta Software | RosettaCommons) Principle: Rosetta employs computational modeling and energy minimization techniques to predict protein structures and design novel proteins. It utilizes sophisticated algorithms to simulate the folding process and interactions within proteins, enabling several applications: 1. Protein Structure Prediction: Rosetta predicts the 3D structure of proteins from their amino acid sequences. It explores the conformational space of proteins and identifies the most energetically favorable structures.(Structure Prediction Applications) 30 2. Protein-Protein Interaction Prediction: Rosetta models the interactions between proteins, helping understand how proteins bind to each other and form complexes. This is crucial for studying signaling pathways and protein networks.(Structure Prediction Applications) 3. Protein Design and Engineering: Rosetta designs new proteins with desired structures and functions. It can engineer proteins for specific applications, such as enzyme catalysis, therapeutic antibodies, or protein-based materials.(Structure Prediction Applications) 4. Ligand Docking and Drug Design: Rosetta predicts how small molecules bind to protein targets, aiding in drug discovery and design. It identifies binding sites and optimizes ligand-protein interactions to develop new therapeutics.(Structure Prediction Applications) 5. Epitope Mapping and Vaccine Design: Rosetta can map epitopes on protein surfaces, helping in vaccine design and understanding immune responses. It predicts antigen-antibody interactions and designs antigens for vaccine development.(Structure Prediction Applications) 6. Protein Loop Modeling: Rosetta models protein loops, which are crucial for protein structure and function. It predicts loop conformations and designs loops with specific properties, such as stability or binding affinity.(Structure Prediction Applications) 7. De Novo Protein Design: Rosetta designs entirely new proteins with specific structures and functions, often beyond those found in nature. This is useful for creating proteins for synthetic biology, biotechnology, and materials science.(Structure Prediction Applications) 8. Structural Refinement: Rosetta refines and improves the accuracy of experimentally determined protein structures. It optimizes protein structures to better fit experimental data and improve model quality.(Structure Prediction Applications) 9. Molecular Dynamics Simulations: Rosetta can perform molecular dynamics simulations to study protein dynamics, conformational changes, and interactions over time.(Structure Prediction Applications) Applications: Rosetta provides a diverse range of applications for predicting protein structures, modeling loops, predicting RNA structures, and modeling antibodies and T-cell receptors. 1. Ab Initio Modeling: Predicting 3D structures of proteins from their amino acid sequences using protocols like Abinitio, NonlocalAbinitio, and Metalloprotein ab initio. 2. Comparative Modeling: Building structural models of proteins using known structures as templates.(Structure Prediction Applications) 3. Loop Modeling: Generating loop conformations using methods such as CCD loop modeling, Kinematic loop modeling, and Loop closing.(Structure Prediction Applications) 4. RNA Structure Prediction: Predicting 3D structures of RNA from their nucleotide sequences using tools like RNA structure prediction and RNA threading.(Structure Prediction Applications) 5. Antibody Modeling: Creating antibody structures with protocols like RosettaAntibody3 and Grafting CDR loops.(Structure Prediction Applications) 31 6. TCR Modeling: Generating T-cell receptor structures from sequences using the TCRmodel protocol.(Structure Prediction Applications) Swiss Model Developed By SWISS-MODEL is a platform developed by the Computational Structural Biology Group at the SIB Swiss Institute of Bioinformatics in collaboration with the Biozentrum of the University of Basel.(SWISS-MODEL) Principle SWISS-MODEL employs homology modeling, a technique that predicts the three-dimensional structure of a protein based on the known structure of a related protein (the template). 1. Template Selection: SWISS-MODEL first identifies a protein with a known structure that shares a significant sequence similarity (homology) with the target protein. This known structure serves as the template.(Schwede) 2. Alignment: The amino acid sequence of the target protein is aligned with the sequence of the template protein. This alignment helps identify corresponding regions between the two proteins.(Schwede) 3. Model Building: Using the aligned sequences, SWISS-MODEL builds a model of the target protein's structure based on the structure of the template protein. It assumes that proteins with similar sequences will have similar structures.(Schwede) 4. Model Refinement: The initial model may undergo refinement to improve accuracy. This step may involve adjusting side-chain conformations, optimizing hydrogen bonding, and minimizing steric clashes.(Schwede) 5. Validation: The generated model is evaluated to ensure that it conforms to known principles of protein structure and chemistry. Various validation criteria are used to assess the quality of the model.(Schwede) 6. Model Availability: Once validated, the predicted structure is made available for users to download and utilize for various research purposes.(Schwede) Applications SWISS-MODEL offers structural insights into proteins by leveraging the principle of homology modeling, which predicts the three-dimensional structure of a protein based on known homologous protein structures. 1. Homology-based Modeling: SWISS-MODEL compares the amino acid sequence of the target protein to known protein structures (homologs) and predicts its structure based on the conserved regions. 2. Structural Conservation: It identifies regions of structural conservation among homologous proteins, providing valuable information about functionally important regions.(Biasini et al.) 3. Functional Inference: The predicted structure allows researchers to infer the function of the target protein by comparing it with structurally characterized homologs. Similar structures often indicate similar functions.(Biasini et al.) 32 4. Insights into Active Sites: SWISS-MODEL can highlight conserved active sites, ligand-binding pockets, or catalytic residues based on structural conservation, aiding in understanding protein function.(Biasini et al.) 5. Protein-Protein Interactions: Predicted structures can elucidate potential binding interfaces with other proteins or ligands, providing insights into protein-protein interactions and signaling pathways.(Biasini et al.) 6. Functional Annotation: By providing structural insights into proteins with known homologs, SWISS-MODEL assists in annotating their functions, which is crucial for understanding biological processes and disease mechanisms.(Biasini et al.) Phyre2 Developed By The PHYRE2 protein structure prediction tool was developed by a team of researchers at Imperial College London, including Lawrence Kelley, Bob Maccallum, Benjamin Jefferys, Alex Herbert, Riccardo Bennett-Lovsey, and Michael Sternberg.(Kelley) Principle These techniques operate based on two key principles: Firstly, protein structure tends to be more conserved in evolution compared to protein sequence. Secondly, evidence suggests there is a finite and relatively small number of unique protein folds in nature, estimated between 1,000 to 10,000. These principles allow the protein structure prediction problem to be approached as matching a sequence of interest to a library of known structures, rather than relying on the more complex and error-prone simulated folding methods. These techniques combine homology modeling and threading approaches to predict protein structures.(Kelley et al.) Applications Phyre2 is particularly useful for predicting structures of proteins with low sequence identity to known structures. Its web-based platform offers easy access to researchers and provides the following benefits: 1. Structure Prediction for Low-Sequence Identity Proteins: Phyre2 utilizes advanced algorithms to predict 3D structures even for proteins with low sequence identity to known structures. This is essential for understanding the structure-function relationship of less characterized proteins.(Kelley et al.) 2. Web-Based Accessibility: As a web-based tool, Phyre2 provides convenient and universal access to researchers worldwide. Users can submit protein sequences online and receive predicted structural models quickly and efficiently.(Kelley et al.) 3. User-Friendly Interface: Phyre2 offers a user-friendly interface, allowing researchers to easily submit protein sequences, customize prediction parameters, and visualize results interactively. This accessibility encourages broader adoption and usage.(Kelley et al.) 4. Rapid Results: Phyre2 provides rapid results for protein structure prediction, typically within a short turnaround time. This quick response enables researchers to proceed with their studies without significant delays.(Kelley et al.) 5. Reliable Predictions: Despite low sequence identity, Phyre2 generates reliable 33 structural predictions by combining homology modeling and threading techniques. These predictions serve as valuable starting points for further experimental validation.(Kelley et al.) 6. Interactive Visualization: Phyre2 offers interactive visualization tools to explore predicted protein structures, allowing researchers to analyze key structural features, domains, and potential binding sites.(Kelley et al.) 7. Customizable Options: Users can customize prediction parameters based on their specific requirements, such as choosing alternative templates, adjusting confidence thresholds, or selecting modeling options.(Kelley et al.) 8. Quality Assessment: Phyre2 provides quality assessment scores and confidence estimates for predicted models, helping researchers evaluate the reliability of the predictions and make informed decisions.(Kelley et al.) 9. Integration with Other Tools: Phyre2 integrates with other bioinformatics tools and databases, allowing users to further analyze predicted structures, perform functional annotations, or explore protein-protein interactions.(Kelley et al.) 10. Educational Resource: Phyre2 serves as an educational resource for students and researchers interested in protein structure prediction, offering insights into computational methods and techniques used in structural biology.(Kelley et al.) Techniques to Validate Predicted Structures X-ray Crystallography X-ray crystallography is the main technique for the determination of protein structures. About 85% of all protein structures known to date have been elucidated using X-ray crystallography.(Papageorgiou et al.) 34 Principle: Applications X-ray crystallography operates on the fundamental principle that crystalline atoms diffract X-rays in specific directions, producing diffraction patterns. By analyzing the intensity and angle of these diffracted beams, a three-dimensional (3D) electron density image is generated, revealing the mean position of atoms in a crystal, their chemical bonds, and any disorder present. This technique remains the most powerful method for determining the atomic structure of macromolecules such as proteins, DNA, drugs, and vitamins. It accomplishes this by measuring the diffraction of X-rays by protein crystals, providing valuable insights into their molecular structures and functions. X-ray crystallography, with its ability to deduce molecular structures from diffraction patterns, finds numerous applications in proteomics. It is instrumental in determining protein structures, studying protein interactions, and elucidating enzyme catalysis mechanisms. By providing atomic-level insights, it significantly advances our understanding of protein function. High-resolution structure determination of proteins using X-ray crystallography is often considered the gold standard to validate predictions and serves as a cornerstone in protein research and drug discovery.(Arora) NMR Spectroscopy (Nuclear Magnetic Resonance) Nuclear magnetic resonance (NMR) spectroscopy is a widely used technique for determining the structure of proteins and protein complexes at atomic resolution.(Hu et al.) It offers detailed insights into conformational and interactional dynamics over a broad range of time scales, from picoseconds to days.(Hu et al.) Additionally, NMR spectroscopy can analyze various sample states, including dilute solutions and living cells, providing comprehensive information on protein behavior in different environments.(Hu et al.) Principles NMR spectroscopy uses the interaction of nuclear spins with an external magnetic field and radiofrequency pulses to detect energy transitions, which are then translated into detailed structural information about molecules. This principle makes NMR an invaluable tool in the study of protein structures and dynamics in various states: 1. Nuclear Spin and Magnetic Properties: - Certain atomic nuclei possess a property called nuclear spin, which is analogous to the spin of electrons. Nuclei with an odd mass number or an odd atomic number have a net spin. - A spinning charged particle, like a nucleus with spin, generates a small magnetic field, effectively making it a tiny magnet.(Nuclear Magnetic Resonance Spectrometer) 2. Magnetic Field Interaction: - When these nuclei are placed in an external magnetic field, they align with the field in a way that depends on their spin. The external magnetic field causes the nuclear spins to align either with (lower energy state) or against (higher energy state) the field. 3. Energy Transitions: - The energy difference between these two states (aligned with or against the magnetic field) depends on the strength of the external magnetic field and the type of nucleus. - By applying a radiofrequency pulse at a specific frequency (resonance frequency), the nuclei can absorb energy and transition from the lower energy state to the higher energy state. This transition is often referred to as "flipping" the spin.(Nuclear Magnetic 35 Resonance Spectrometer) 4. Detection of NMR Signal: - After the radiofrequency pulse is turned off, the nuclei relax back to their lower energy state, releasing the absorbed energy. - This released energy induces a voltage in a detector coil, producing a time-domain signal known as the free induction decay (FID).(Nuclear Magnetic Resonance Spectrometer) 5. Fourier Transformation: - The FID signal is complex and not immediately useful for structural analysis. However, by applying a mathematical process called Fourier transformation, the time-domain signal is converted into a frequency-domain spectrum. - The resulting spectrum provides peaks at specific frequencies corresponding to the resonance frequencies of the nuclei in the sample.(Nuclear Magnetic Resonance Spectrometer) 6. Chemical Shift and Molecular Structure: - The exact position of these peaks (chemical shift) depends on the chemical environment of the nuclei, which is influenced by the surrounding electronic structure and, consequently, the molecular structure. - Each unique chemical environment of a nucleus within a molecule produces a distinct chemical shift, allowing researchers to deduce information about the molecular structure. 7. Quantitative Information: - The area under each peak in the NMR spectrum is proportional to the number of nuclei contributing to that peak, providing quantitative information about different parts of the molecule.(Nuclear Magnetic Resonance Spectrometer) 8. Multidimensional NMR: - More complex experiments, such as two-dimensional (2D) and three-dimensional (3D) NMR, can provide even more detailed information about the spatial relationships between atoms in a molecule, allowing for the determination of three-dimensional structures of proteins and other macromolecules.(Nuclear Magnetic Resonance Spectrometer) Application Nuclear Magnetic Resonance (NMR) spectroscopy has a wide range of applications, particularly in the fields of chemistry, biochemistry, and medicine. Here are some of the key applications: 1. Protein Structure Determination: - Structural Biology: NMR is extensively used to determine the three-dimensional structures of proteins and nucleic acids in solution. It helps in understanding the conformation and dynamics of biological macromolecules.(Puthenveetil and Vinogradova) -Drug Discovery: NMR aids in the design and optimization of drugs by revealing how small molecules bind to target proteins. It helps in identifying the binding sites and conformational changes upon ligand binding.(Puthenveetil and Vinogradova) 2. Study of Protein Dynamics: - Conformational Flexibility: NMR provides insights into the dynamic aspects of proteins, including conformational changes, folding/unfolding processes, and interactions with other molecules. It can measure motions over a wide range of timescales, from picoseconds to seconds.(Puthenveetil and Vinogradova) - Interaction Studies: It allows researchers to study transient interactions and complex formation between proteins and other biomolecules, which are often challenging to capture using other techniques.(Puthenveetil and Vinogradova) 36 3. Metabolomics: - Metabolite Profiling: NMR is used to analyze metabolites in biological samples. It provides quantitative and qualitative information about the metabolic state of cells, tissues, or organisms.(Puthenveetil and Vinogradova) - Biomarker Discovery: It helps in identifying biomarkers for diseases by comparing the metabolic profiles of healthy and diseased states.(Puthenveetil and Vinogradova) 4. Natural Products Chemistry: - Structure Elucidation: NMR is a crucial tool for determining the structures of natural products, including complex organic molecules isolated from plants, fungi, and marine organisms.(Puthenveetil and Vinogradova) - Stereochemistry: It helps in assigning the stereochemistry of chiral centers in organic molecules, providing detailed information about their spatial arrangement.(Puthenveetil and Vinogradova) 5. Material Science: - Polymer Analysis: NMR is used to study the composition, structure, and dynamics of polymers. It helps in understanding the properties and behavior of synthetic and natural polymers.(Puthenveetil and Vinogradova) - Nanomaterials: It provides insights into the structural and dynamic properties of nanomaterials, including nanoparticles and nanocomposites.(Puthenveetil and Vinogradova) 6. Medical Applications: - MRI (Magnetic Resonance Imaging): While not strictly NMR spectroscopy, MRI is based on the same principles and is widely used in medical diagnostics to create detailed images of tissues and organs.(Puthenveetil and Vinogradova) - Disease Diagnosis: NMR spectroscopy is used in clinical research to study metabolic changes associated with diseases such as cancer, diabetes, and neurodegenerative disorders.(Puthenveetil and Vinogradova) 7. Quality Control in Pharmaceuticals: - Purity and Composition Analysis: NMR is employed in the pharmaceutical industry for the quality control of drug substances and products. It ensures the correct chemical composition and purity of pharmaceuticals. - **Polymorph Identification**: It helps in identifying different crystalline forms (polymorphs) of a drug, which can affect its solubility and bioavailability.(Puthenveetil and Vinogradova) 8. Food Science: - Nutritional Analysis: NMR is used to analyze the nutritional content and composition of food products. It helps in detecting adulterants and contaminants. - Flavor and Aroma Compounds: It provides detailed information about the chemical compounds responsible for the flavor and aroma of food and beverages.(Puthenveetil and Vinogradova) Cryo-Electron Microscopy (Cryo-EM) Invented 40 years ago as an offshoot of electron microscopy, cryo-electron microscopy (cryo-EM) flash-freezes samples into a glassy state and probes them with beams of electrons. This technique involves rapidly cooling biological specimens to cryogenic temperatures, preserving their native structures in a 37 vitreous (glassy) ice. By using electron beams to image these frozen samples, cryo-EM captures high-resolution images that can be computationally processed to reconstruct detailed three-dimensional structures of proteins, viruses, and other macromolecular complexes. This method avoids the potential artifacts introduced by traditional staining and dehydration procedures, providing more accurate representations of the biological molecules in their native environments. Cryo-EM has revolutionized structural biology, offering unprecedented insights into the molecular machinery of life.(“Cryogenic Electron Microscopy (cryo-EM): Amazing Views of Life’s Machinery | SLAC National Accelerator Laboratory”) Principle: Cryo-electron microscopy (cryo-EM) operates on the principle of using electron microscopy at cryogenic temperatures to visualize biological samples at high resolution. By leveraging the wave-particle duality of electrons, which have extremely short wavelengths, cryo-EM produces clear images of small biological molecules. The process involves rapidly freezing the sample to prevent ice crystal formation and preserve its natural state, thereby protecting it from damage by the electron beam. This freezing traps the molecules in a glass-like state, allowing detailed three-dimensional structures to be reconstructed from the diffraction patterns generated by electron interactions.(“Cryogenic Electron Microscopy (cryo-EM): Amazing Views of Life’s Machinery | SLAC National Accelerator Laboratory”) Applications Cryo-electron microscopy (cryo-EM) has several important applications. It is instrumental in high-resolution structure determination of large complexes and membrane proteins, often complementing predictive models. This technique is versatile, applicable to a broad range of biological samples, including proteins, viruses, cells, and tissues. Cryo-EM is particularly valuable for studying large, flexible complexes and assemblies that pose challenges for other structural determination techniques, providing detailed insights into their structure and function.(Carreras) 38 Conclusion In conclusion, this document has provided a comprehensive overview of fundamental biology concepts related to proteins, delving into the structure, function, and techniques involved in studying them. We began by exploring the building blocks of proteins, amino acids, and then delved into the intricacies of protein structures, from primary to quaternary levels. Understanding protein structures is crucial for elucidating their enzymatic roles, which we discussed in detail, along with various protein purification techniques such as chromatography and ultracentrifugation. Moreover, this document highlighted cutting-edge tools like AlphaFold, Rosetta, Swiss Model, and Phyre2, which are revolutionizing protein structure prediction. Techniques like X-ray crystallography, NMR spectroscopy, and Cryo-EM were explored for validating predicted structures, offering insights into the experimental methods employed in structural biology. By bridging fundamental concepts with advanced techniques, this document aims to equip readers with a solid foundation in protein biology, empowering them to explore further in this dynamic field. Whether studying protein structures, their enzymatic functions, or employing computational methods for prediction and validation, this document serves as a valuable resource for students, researchers, and enthusiasts alike, fostering a deeper understanding of one of biology's most fascinating molecules. 39 References Adhikari, S., Manthena, P. V., Sajwan, K., Kota, K. K., & Roy, R. (2010a). A unified method for purification of basic proteins. Analytical Biochemistry, 400(2), 203–206. https://doi.org/10.1016/j.ab.2010.01.011 Adhikari, S., Manthena, P. V., Sajwan, K., Kota, K. K., & Roy, R. (2010b). A unified method for purification of basic proteins. Analytical Biochemistry, 400(2), 203–206. https://doi.org/10.1016/j.ab.2010.01.011 Admin. (2021a, November 25). Amino Acid Structure - Definition, Structure, Basicity of Amino Acid with Examples. BYJUS. https://byjus.com/chemistry/amino-acid-structure/ Admin. (2021b, November 25). Amino Acid Structure - Definition, Structure, Basicity of Amino Acid with Examples. BYJUS. https://byjus.com/chemistry/amino-acid-structure/ Affinity Chromatography | Principles. (n.d.-a). Cube Biotech. https://cube-biotech.com/knowledge/protein-purification/affinity-chromatography/ Affinity Chromatography | Principles. (n.d.-b). Cube Biotech. https://cube-biotech.com/knowledge/protein-purification/affinity-chromatography/ Amino acids: MedlinePlus Medical Encyclopedia. (n.d.). https://medlineplus.gov/ency/article/002222.htm Anion Exchange Chromatography. (n.d.). Bio-Rad Laboratories. https://www.bio-rad.com/en-jm/applications-technologies/anion-exchange-chromatography ?ID=MWHAZ4C4S Anonymous. (2023, November 30). Inside the Lab: Real-World Applications of Ultracentrifugation | GMI - Trusted Laboratory Solutions. GMI - Trusted Laboratory Solutions. https://www.gmi-inc.com/inside-the-lab-real-world-applications-of-ultracentrifugation/ Arora, A. (2017, October 15). X-ray Crystallography & Its Applications in Proteomics [Slide show]. SlideShare. 40 https://www.slideshare.net/slideshow/xray-crystallography-its-applications-in-proteomics/8 0833430 Biasini, M., Bienert, S., Waterhouse, A., Arnold, K., Studer, G., Schmidt, T., Kiefer, F., Cassarino, T. G., Bertoni, M., Bordoli, L., & Schwede, T. (2014). SWISS-MODEL: modelling protein tertiary and quaternary structure using evolutionary information. Nucleic Acids Research, 42(W1), W252–W258. https://doi.org/10.1093/nar/gku340 BYJUS. (2023a, May 19). Four Types of Protein Structure - Primary, Secondary, Tertiary &amp; Quaternary Structures. BYJUS. https://byjus.com/chemistry/protein-structure-and-levels-of-protein/ BYJUS. (2023b, May 19). Four Types of Protein Structure - Primary, Secondary, Tertiary &amp; Quaternary Structures. BYJUS. https://byjus.com/chemistry/protein-structure-and-levels-of-protein/ Carreras, H. Z., PhD. (2024, May 24). Cryo Electron Microscopy: Principle, Strengths, Limitations and Applications. Analysis & Separations From Technology Networks. https://www.technologynetworks.com/analysis/articles/cryo-electron-microscopy-principlestrengths-limitations-and-applications-377080 Cation Exchange Chromatography. (n.d.). Bio-Rad Laboratories. https://www.bio-rad.com/en-jm/applications-technologies/cation-exchange-chromatography ?ID=MWHB018UU Chapter 3: Investigating Proteins - Chemistry. (2020, September 22). Chemistry. https://wou.edu/chemistry/courses/online-chemistry-textbooks/ch450-and-ch451-biochemist ry-defining-life-at-the-molecular-level/chapter-3-investigating-proteins/ ChatGPT. (n.d.). https://chatgpt.com/c/7c8b0fe3-f6e0-490e-8a5e-1a92045f901a Chromatography for Protein Purification | Sino Biological. (n.d.). https://www.sinobiological.com/resource/protein-review/chromatography-purification 41 Cleveland Clinic Medical. (n.d.-a). Amino Acids. Cleveland Clinic. https://my.clevelandclinic.org/health/articles/22243-amino-acids Cleveland Clinic Medical. (n.d.-b). Amino Acids. Cleveland Clinic. https://my.clevelandclinic.org/health/articles/22243-amino-acids Cleveland Clinic Medical. (n.d.-c). Amino Acids. Cleveland Clinic. https://my.clevelandclinic.org/health/articles/22243-amino-acids Cryogenic electron microscopy (cryo-EM): amazing views of life’s machinery | SLAC National Accelerator Laboratory. (n.d.). SLAC National Accelerator Laboratory. https://www6.slac.stanford.edu/research/slac-science-explained/cryo-em Data Set AlphaFold Predictions. (n.d.). https://giardiadb.org/giardiadb/app/record/dataset/DS_dc1c8cb24c#category:topic-0219-syn onym Database, A. P. S. (n.d.-a). AlphaFold Protein Structure Database. https://alphafold.ebi.ac.uk/ Database, A. P. S. (n.d.-b). AlphaFold Protein Structure Database. https://alphafold.ebi.ac.uk/ Dinesh-Supreme. (2023, April 11). Why Structure Prediction Matters | DNASTAR. DNASTAR. https://www.dnastar.com/blog/protein-analysis-modeling/why-structure-prediction-matters/ Get a detailed understanding of the Reversed-Phase Chromatography. (2022, March 1). Unacademy. https://unacademy.com/content/kerala-psc/study-material/bioinstrumentation/reversed-phas e-chromatography/ Haurowitz, F., & Koshland, D. E. (2024, June 11). Protein | Definition, Structure, & Classification. Encyclopedia Britannica. https://www.britannica.com/science/protein Hu, Y., Cheng, K., He, L., Zhang, X., Jiang, B., Jiang, L., Li, C., Wang, G., Yang, Y., & Liu, M. (2021). NMR-Based Methods for Protein Analysis. Analytical Chemistry, 93(4), 1866–1879. https://doi.org/10.1021/acs.analchem.0c03830 42 Introduction to Size Exclusion Chromatography. (n.d.). Bio-Rad Laboratories. https://www.bio-rad.com/en-jm/applications-technologies/introduction-size-exclusion-chro matography?ID=LUSMV015 Kelley, L. (n.d.). PHYRE2 Protein Fold Recognition Server. http://www.sbg.bio.ic.ac.uk/phyre2/html/page.cgi?id=index Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N., & Sternberg, M. J. E. (2015a). The Phyre2 web portal for protein modeling, prediction and analysis. Nature Protocols, 10(6), 845–858. https://doi.org/10.1038/nprot.2015.053 Kelley, L. A., Mezulis, S., Yates, C. M., Wass, M. N., & Sternberg, M. J. E. (2015b). The Phyre2 web portal for protein modeling, prediction and analysis. Nature Protocols, 10(6), 845–858. https://doi.org/10.1038/nprot.2015.053 Libretexts. (2023, October 31). 3.9: Proteins - Protein Structure. Biology LibreTexts. https://bio.libretexts.org/Bookshelves/Introductory_and_General_Biology/General_Biology _(Boundless)/03%3A_Biological_Macromolecules/3.09%3A_Proteins_-_Protein_Structure Libretexts. (2024a, March 21). 2.2: Structure & Function - Amino Acids. Biology LibreTexts. https://bio.libretexts.org/Bookshelves/Biochemistry/Book%3A_Biochemistry_Free_For_All _(Ahern_Rajagopal_and_Tan)/02%3A_Structure_and_Function/202%3A_Structure__Funct ion_-_Amino_Acids Libretexts. (2024b, March 21). 2.2: Structure & Function - Amino Acids. Biology LibreTexts. https://bio.libretexts.org/Bookshelves/Biochemistry/Book%3A_Biochemistry_Free_For_All _(Ahern_Rajagopal_and_Tan)/02%3A_Structure_and_Function/202%3A_Structure__Funct ion_-_Amino_Acids Libretexts. (2024c, March 21). 2.3: Structure & Function- Proteins I. Biology LibreTexts. https://bio.libretexts.org/Bookshelves/Biochemistry/Book%3A_Biochemistry_Free_For_All _(Ahern_Rajagopal_and_Tan)/02%3A_Structure_and_Function/203%3A_Structure__Funct ion-_Proteins_I 43 Mukherjee, A., & Sasikala, W. D. (2013). Drug–DNA Intercalation. In Advances in protein chemistry and structural biology (pp. 1–62). https://doi.org/10.1016/b978-0-12-411636-8.00001-8 News-Medical. (2019a, February 26). Affinity Chromatography Applications. https://www.news-medical.net/life-sciences/Affinity-Chromatography-Applications.aspx News-Medical. (2019b, February 26). Affinity Chromatography Applications. https://www.news-medical.net/life-sciences/Affinity-Chromatography-Applications.aspx Nowakowski, A. B., Wobig, W. J., & Petering, D. H. (2014). Native SDS-PAGE: high resolution electrophoretic separation of proteins with retention of native properties including bound metal ions. Metallomics, 6(5), 1068–1078. https://doi.org/10.1039/c4mt00033a Nuclear Magnetic Resonance Spectrometer. (n.d.-a). https://neist.res.in/neistsaif/nmr500.php Nuclear Magnetic Resonance Spectrometer. (n.d.-b). https://neist.res.in/neistsaif/nmr500.php Papageorgiou, A. C., Poudel, N., & Mattsson, J. (2020). Protein Structure Analysis and Validation with X-Ray Crystallography. In Methods in molecular biology (pp. 377–404). https://doi.org/10.1007/978-1-0716-0775-6_25 Poole, C. (2000). CHROMATOGRAPHY. In Elsevier eBooks (pp. 40–64). https://doi.org/10.1016/b0-12-226770-2/00021-1 Professional, C. C. M. (n.d.). Amino Acids. Cleveland Clinic. https://my.clevelandclinic.org/health/articles/22243-amino-acids Proteins, P. (2023, June 19). Size Exclusion Chromatography: Size Does Matter. Peak Proteins. https://peakproteins.com/size-exclusion-chromatography-size-does-matter/ Proteogenix. (2023a, August 3). Size exclusion chromatography for protein purification ProteoGenix. ProteoGenix. https://www.proteogenix.science/protein-purification/no-tag-purification/size-exclusion-chr omatography/ 44 Proteogenix. (2023b, August 3). Size exclusion chromatography for protein purification ProteoGenix. ProteoGenix. https://www.proteogenix.science/protein-purification/no-tag-purification/size-exclusion-chr omatography/ Proteogenix. (2023c, August 3). Size exclusion chromatography for protein purification ProteoGenix. ProteoGenix. https://www.proteogenix.science/protein-purification/no-tag-purification/size-exclusion-chr omatography/ Puthenveetil, R., & Vinogradova, O. (2019a). Solution NMR: A powerful tool for structural and functional studies of membrane proteins in reconstituted environments. Journal of Biological Chemistry/˜the œJournal of Biological Chemistry, 294(44), 15914–15931. https://doi.org/10.1074/jbc.rev119.009178 Puthenveetil, R., & Vinogradova, O. (2019b). Solution NMR: A powerful tool for structural and functional studies of membrane proteins in reconstituted environments. Journal of Biological Chemistry/˜the œJournal of Biological Chemistry, 294(44), 15914–15931. https://doi.org/10.1074/jbc.rev119.009178 Puthenveetil, R., & Vinogradova, O. (2019c). Solution NMR: A powerful tool for structural and functional studies of membrane proteins in reconstituted environments. Journal of Biological Chemistry/˜the œJournal of Biological Chemistry, 294(44), 15914–15931. https://doi.org/10.1074/jbc.rev119.009178 Qiu, L. (2023a). Principle and Applications of Ion Exchange Chromatography. Longdom. https://doi.org/10.35248/2471-2698.23.8.205 Qiu, L. (2023b). Principle and Applications of Ion Exchange Chromatography. Longdom. https://doi.org/10.35248/2471-2698.23.8.205 Sanvictores, T., & Farci, F. (2022a, October 31). Biochemistry, Primary Protein Structure. StatPearls - NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK564343/ 45 Sanvictores, T., & Farci, F. (2022b, October 31). Biochemistry, Primary Protein Structure. StatPearls - NCBI Bookshelf. https://www.ncbi.nlm.nih.gov/books/NBK564343/ Schwede, T. (2003). SWISS-MODEL: an automated protein homology-modeling server. Nucleic Acids Research, 31(13), 3381–3385. https://doi.org/10.1093/nar/gkg520 SDS-PAGE. (2022, April 19). Unacademy. https://unacademy.com/content/kerala-psc/study-material/bioinstrumentation/sds-page/ Structure Prediction Applications. (n.d.-a). https://www.rosettacommons.org/docs/latest/application_documentation/structure_predictio n/structure-prediction-applications Structure Prediction Applications. (n.d.-b). https://www.rosettacommons.org/docs/latest/application_documentation/structure_predictio n/structure-prediction-applications Structure Prediction Applications. (n.d.-c). https://www.rosettacommons.org/docs/latest/application_documentation/structure_predictio n/structure-prediction-applications SWISS-MODEL. (n.d.). https://swissmodel.expasy.org/ The Functions of Amino Acids | The Power of Amino Acids | Amino Acids | Ajinomoto Group Global Website - Eat Well, Live Well. (n.d.). Ajinomoto Group Global Website - Eat Well, Live Well. https://www.ajinomoto.com/amino-acids/amino-acids-function The Rosetta Software | RosettaCommons. (n.d.). https://www.rosettacommons.org/software Ultracentrifugation | chemistry. (n.d.). Encyclopedia Britannica. https://www.britannica.com/technology/ultracentrifugation UNACADEMY. (2022a, March 1). Get a Detailed Understanding of the Reversed-Phase Chromatography. Unacademy. https://unacademy.com/content/kerala-psc/study-material/bioinstrumentation/reversed-phas e-chromatography/ 46 UNACADEMY. (2022b, March 1). Get a Detailed Understanding of the Reversed-Phase Chromatography. Unacademy. https://unacademy.com/content/kerala-psc/study-material/bioinstrumentation/reversed-phas e-chromatography/ Varadi, M., & Velankar, S. (2022). The impact of AlphaFold Protein Structure Database on the fields of life sciences. Proteomics, 23(17). https://doi.org/10.1002/pmic.202200128 Western Blot: Overview & Applications. (n.d.-a). https://www.excedr.com/resources/western-blot-overview-applications Western Blot: Overview & Applications. (n.d.-b). https://www.excedr.com/resources/western-blot-overview-applications Yang, P., & Mahmood, T. (2012a). Western blot: Technique, theory, and trouble shooting. North American Journal of Medical Sciences, 4(9), 429. https://doi.org/10.4103/1947-2714.100998 Yang, P., & Mahmood, T. (2012b). Western blot: Technique, theory, and trouble shooting. North American Journal of Medical Sciences, 4(9), 429. https://doi.org/10.4103/1947-2714.100998 Yang, P., & Mahmood, T. (2012c). Western blot: Technique, theory, and trouble shooting. North American Journal of Medical Sciences, 4(9), 429. https://doi.org/10.4103/1947-2714.100998