Fact-checked by Grok 2 weeks ago

Protein

Proteins are large, complex macromolecules composed of one or more long chains of amino acids linked by peptide bonds, serving as the fundamental building blocks and functional workhorses of living cells and organisms. These molecules, encoded by genes in DNA, adopt unique three-dimensional structures that determine their diverse roles, including enzymatic catalysis, structural support, transport, signaling, and immune defense. With 20 standard amino acids forming their primary sequence, proteins fold into secondary structures like alpha helices and beta sheets, tertiary folds stabilized by noncovalent interactions, and sometimes quaternary assemblies of multiple subunits, all of which are essential for biological function. The synthesis of proteins, known as , occurs on ribosomes in the , where (mRNA) templates direct the assembly of carried by (tRNA), consuming energy from GTP and often involving post-translational modifications in the and Golgi apparatus. This process ensures that each protein has a specific sequence dictated by the , which not only defines its shape but also its precise function, as even minor sequence variations can lead to diseases like sickle cell anemia. Chaperone proteins assist in proper folding to prevent aggregation, highlighting the intricate balance required for protein stability and activity. In terms of functions, proteins are indispensable for virtually every physiological process: enzymes accelerate chemical reactions by lowering , structural proteins like and provide mechanical support and enable movement, transport proteins such as carry oxygen, hormones like insulin regulate metabolism, and antibodies defend against pathogens. Approximately 80% of the body's biochemical reactions rely on enzymes, underscoring proteins' catalytic dominance, while their denaturation—unfolding due to heat, pH changes, or chemicals—can disrupt these roles, as seen in conditions like from misfolded proteins. Overall, proteins constitute about 15-20% of the human body's mass and are continuously turned over, with dietary replenishing types that cannot be synthesized endogenously.

Introduction and History

Definition and etymology

Proteins are large biomolecules and macromolecules composed of one or more long chains of residues, playing essential roles in the structure, function, and regulation of living organisms. These chains, known as polypeptides, are formed through the linkage of via bonds, resulting in complex structures that enable proteins to carry out a wide array of biological tasks. The building blocks of proteins are the 20 standard , which are covalently joined in specific sequences determined by genetic information. Proteins perform diverse functions, including enzymatic catalysis to accelerate chemical reactions, signaling to transmit messages between cells, and providing to maintain cellular and integrity. For instance, enzymes like facilitate digestion, while structural proteins such as reinforce connective tissues. The term "protein" originates from the Greek word prōteios, meaning "primary" or "of first importance," reflecting the recognition of these molecules' central role in and . It was coined in 1838 by the in a letter to the Dutch chemist Gerrit Jan Mulder, who had observed that various organic substances shared a similar empirical composition; Berzelius proposed the name to emphasize their fundamental importance. This underscores the early understanding of proteins as the "first rank" components of living matter.

Historical development

The study of proteins originated in the early , as chemists began isolating and characterizing organic substances from plant and animal sources. In 1838, Dutch chemist Gerardus Johannes Mulder conducted systematic analyses of substances like from and from , proposing that they shared a common composition and dubbing them "protein bodies" based on their elemental makeup, primarily carbon, , , and oxygen. This work laid the groundwork for recognizing proteins as a distinct class of biological molecules essential to life. In 1838, Swedish chemist coined the term "protein" from the Greek word "proteios," meaning "primary" or "of first importance," to emphasize their fundamental role in living organisms, building on Mulder's findings. By the mid-19th century, investigations expanded into proteins' nutritional significance. In 1840, German chemist demonstrated through animal feeding experiments that proteins were indispensable for growth and maintenance, distinguishing them from carbohydrates and fats as the sole source of nitrogen for tissue building; this established the concept of proteins as the cornerstone of animal chemistry and nutrition. Advancing into the , structural insights emerged. In 1901, German chemist proposed the peptide bond hypothesis, suggesting that proteins are linear polymers of linked by amide bonds, supported by his synthesis of simple di- and tripeptides; this idea was further corroborated by Franz Hofmeister's concurrent studies on protein yielding . A pivotal experimental milestone came in 1926 when American biochemist James B. Sumner crystallized the from jack bean meal, providing the first evidence that enzymes are proteins and earning him the 1946 . Mid-20th-century advances focused on protein structure and function, integrating physical and biochemical methods. In 1951, Linus Pauling and colleagues proposed the alpha-helix and beta-sheet as common secondary structures in proteins, based on model-building and X-ray diffraction data, revolutionizing understanding of polypeptide chain configurations. The year 1958 marked the determination of the first three-dimensional protein structure when John Kendrew's team used X-ray crystallography to resolve the atomic model of sperm whale myoglobin at 6 Å resolution, revealing a compact globular fold with a heme prosthetic group. Techniques for protein analysis also evolved; in the late 1960s, Ulrich K. Laemmli developed sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) in 1970, enabling separation of proteins by molecular weight for purification and characterization. Further progress linked proteins to genetics. The 1953 Watson-Crick model of DNA structure provided the framework for understanding how genetic information encodes proteins. In 1961, Marshall Nirenberg and J. Heinrich Matthaei deciphered the first codon of the genetic code using cell-free protein synthesis, showing that polyuridylic acid directed incorporation of phenylalanine, initiating the full elucidation of how DNA sequences specify amino acid order in proteins. By 1972, Christian Anfinsen's experiments on ribonuclease demonstrated that the native structure of a protein is determined solely by its amino acid sequence under physiological conditions, establishing the thermodynamic hypothesis of protein folding for which he shared the 1972 Nobel Prize in Chemistry.

Chemical Structure

Primary structure

The primary structure of a protein refers to the linear of covalently linked by bonds to form a polypeptide chain, serving as the foundational blueprint that dictates the protein's identity, function, and potential for higher-order folding. This is unique to each protein and arises from the specific order of , which can range from tens to thousands in length, ensuring structural specificity essential for biological roles such as enzymatic or . Proteins are composed of 20 standard amino acids, each characterized by a central alpha carbon atom bonded to a , a carboxyl group, an , and a (R group) that imparts distinct chemical properties. For instance, (Gly) has the simplest (a atom), conferring flexibility, while alanine (Ala) features a , contributing to hydrophobic interactions. properties include hydrophobicity (e.g., in ), charge (acidic in , which carries a negatively charged carboxyl group at physiological pH; basic in , with a positively charged amino group), and polarity, which influence the protein's solubility, stability, and interactions. Peptide bonds form through a between the carboxyl group of one and the amino group of another, releasing a and creating a covalent linkage with partial double-bond character that restricts rotation. The resulting backbone has the repeating general formula -[NH-CHR-CO]-, where R represents the unique of each , forming a rigid, planar around the bond. This linkage connects in an N-terminal to C-terminal direction, with the bearing a free amino group and the a free carboxyl group. Determining the primary structure is crucial for understanding protein function, as it reveals the exact order necessary for proper activity and can identify affecting . A seminal method for this is , developed in the late and refined in the , which sequentially removes and identifies the N-terminal as a phenylthiohydantoin using phenylisothiocyanate, allowing step-by-step sequencing of peptides up to 50-60 residues long. Although primarily linear, the primary structure can include covalent variations such as bridges, formed by the oxidation of sulfhydryl groups (-SH) from two residues, creating a -S-S- linkage that stabilizes the chain. These bridges are considered part of the primary structure due to their covalent nature and are common in extracellular proteins for enhanced stability. The primary sequence ultimately encodes the information guiding into functional three-dimensional forms.

Secondary structure

The secondary structure of proteins refers to the local three-dimensional conformations of segments of the polypeptide backbone, primarily stabilized by bonds between the carbonyl oxygen and atoms within the backbone. These structures form regular patterns that are independent of the side chains, driven instead by the inherent geometry of the and the flexibility of the backbone. The most common secondary structural elements are alpha-helices, beta-sheets, turns, and loops, each characterized by specific angles and bonding patterns. The alpha-helix is a right-handed coiled structure in which the polypeptide backbone folds into a spiral with 3.6 residues per turn and a pitch of 5.4 , corresponding to a rise of 1.5 per residue along the helical axis. In this configuration, bonds form between the carbonyl oxygen of residue i and the amide of residue i+4, stabilizing the helix parallel to its axis. Beta-sheets consist of two or more beta-strands—extended polypeptide segments—aligned laterally to form pleated sheets, with bonds between the carbonyl oxygen of one strand and the amide of an adjacent strand. Beta-sheets can be parallel, where strands run in the same N-to-C-terminal direction, or antiparallel, where adjacent strands run in opposite directions, with antiparallel sheets exhibiting more linear bonds and greater stability. Turns and loops are irregular elements that connect alpha-helices and beta-strands, often involving tight reversals in chain direction, such as beta-turns where the chain folds back on itself over four residues. These secondary structures are constrained by the allowed values of the backbone dihedral angles, denoted as \phi (phi, rotation around the N-Cα bond) and \psi (psi, rotation around the Cα-C bond), which are visualized in the . The delineates regions of allowed (\phi, \psi) combinations based on steric hindrance from atomic overlaps, with core areas corresponding to alpha-helices (\phi \approx -60^\circ, \psi \approx -45^\circ) and beta-sheets (\phi \approx -120^\circ, \psi \approx 120^\circ), while disallowed regions reflect unfavorable clashes. The formation of secondary structures arises primarily from the backbone's conformational preferences, with side chains influencing stability secondarily but not dictating the initial folding. Representative examples illustrate these elements in natural proteins. In alpha-keratins, such as those in human hair and , the polypeptide chains predominantly form coiled alpha-helices that dimerize into protofilaments, providing tensile strength and elasticity. Conversely, silk from the silkworm consists largely of antiparallel beta-sheets, where repeating Gly-Ala sequences stack into crystalline layers, conferring high mechanical rigidity and toughness to the fiber. These local conformations contribute to the overall protein fold but are distinct from long-range interactions.

Tertiary and quaternary structures

The tertiary structure of a protein refers to the overall three-dimensional arrangement of a single polypeptide chain, resulting from interactions between side chains that are distant in the primary . This global fold is primarily stabilized by non-covalent forces, including hydrophobic interactions that drive nonpolar residues to cluster in the protein's interior core, away from the aqueous environment; bonds between polar side chains; ionic bonds or bridges between oppositely charged residues; and van der Waals attractions between closely packed atoms. bonds, which are covalent linkages between residues, can further reinforce the structure in some proteins, particularly those in oxidizing environments like the . Molecular chaperones, such as and families, play a crucial role in assisting this folding process by binding to nascent or misfolded polypeptides, preventing aggregation, and facilitating proper conformation through ATP-dependent cycles, thereby ensuring efficient navigation of the folding landscape. The quaternary structure describes the spatial arrangement and non-covalent association of multiple polypeptide subunits into a functional , often enhancing stability, regulation, or cooperative function. These subunit interfaces are typically stabilized by the same types of non-covalent interactions as in tertiary structure—hydrophobic contacts, hydrogen bonds, ionic interactions, and van der Waals forces—without requiring covalent links between chains. A classic example is , which consists of four subunits (two α and two β chains) that assemble to enable cooperative oxygen binding, with interfaces burying significant surface area to maintain the tetrameric form. In contrast, exemplifies a protein with only tertiary structure, as it functions as a monomeric oxygen-storage unit in muscle cells, featuring eight α-helices packed around a without additional subunits. For example, in insulin, the A and B chains are linked by two interchain disulfide bonds (with an intra-chain disulfide in the A chain) as part of its structure; these monomeric units further oligomerize non-covalently into dimers or hexamers ( structure) for storage in pancreatic cells. The integration of structural levels underscores that the primary sequence ultimately dictates the fold, as demonstrated by the Anfinsen dogma, which posits that a protein's native conformation is thermodynamically determined by its sequence under physiological conditions. This was experimentally validated through denaturation and renaturation experiments on ribonuclease A, where the unfolded protein spontaneously refolded into its active form upon removal of denaturants, indicating that all necessary folding information resides in the . The , which questions how proteins achieve their native fold in biologically relevant timescales despite an astronomically large conformational space, is resolved by the existence of directed folding pathways or funnels, where local secondary structures form early and guide progressive stabilization through energy minima, often assisted by chaperones. Disruption of these higher-order structures can lead to denaturation, the loss of tertiary and quaternary conformations due to heat, pH changes, or chemicals, rendering the protein inactive; however, many proteins can renature upon restoration of native conditions, reaffirming sequence-encoded folding. Pathological misfolding, where proteins adopt aberrant conformations resistant to degradation, underlies diseases like Alzheimer's, where accumulation of β-amyloid aggregates and tau tangles disrupts neuronal function and triggers neurotoxicity.

Domains and Motifs

Protein domains

Protein domains are compact, semi-independent structural units within proteins, typically comprising 50 to 350 residues, that fold independently into stable, globular structures with hydrophobic cores and hydrophilic surfaces. These units serve as the fundamental building blocks for protein architecture, enabling modular organization that supports diverse biological roles. Protein domains often evolve through , allowing for the assembly of new proteins by combining existing modules. In multi-domain proteins, individual domains are frequently connected by flexible linker regions, which permit relative movement while maintaining overall structural integrity. A classic example is the molecule, where immunoglobulin heavy and light chains each contain and domains; the domains (VH and VL) form the antigen-binding site, while domains (CH and CL) mediate effector functions. These domains contribute to the protein's fold by packing together to form the complete three-dimensional structure. Protein domains perform specialized functions, such as binding or enzymatic , often within dedicated active sites. The database catalogs these domain families through curated multiple alignments and Markov models, facilitating the and of over 21,000 families across protein . Evolutionarily, protein domains arise and diversify via mechanisms like domain shuffling and accretion, particularly in eukaryotes, where recombination events generate novel combinations that enhance functional complexity. For instance, the , which is specific to , has undergone extensive shuffling to pair with 19 different partner domains, enabling intricate signaling pathways. Domains are identified computationally through searches, such as those using profile hidden Markov models in , or by structure superposition methods that align three-dimensional models from databases like CATH or . These approaches detect conserved features, revealing evolutionary relationships even among distantly related proteins.

Sequence and structural motifs

Sequence and structural motifs refer to short, recurring patterns in protein primary sequences or three-dimensional structures that often signal specific functional sites, such as binding or catalytic regions. These motifs are typically conserved across diverse proteins due to their critical roles in biological processes, allowing researchers to infer from sequence or structural data. Unlike larger protein domains, which are independent folding units, motifs are smaller signatures embedded within sequences or folds that contribute to localized functionality. Sequence motifs are linear patterns of , often 10-20 residues long, that indicate functional elements like nucleotide-binding sites. A classic example is the Walker A motif, also known as the P-loop, with the GXXXXGK[T/S], where X represents any ; this motif binds the phosphate groups of ATP or GTP in numerous ATPases and . The adjacent Walker B motif, featuring a conserved aspartate or glutamate (e.g., hhhhDE, where h is a hydrophobic residue), coordinates a magnesium essential for . These motifs are detected computationally using regular expressions for exact or hidden Markov models (HMMs) for accommodating variations and insertions/deletions. Structural motifs, in contrast, are defined by their three-dimensional arrangements rather than linear sequences, frequently involving specific secondary structure elements that enable interactions like binding. The (HTH) motif, consisting of two connected by a short turn, is a common DNA-binding structure found in many transcription factors, where the recognition helix inserts into the DNA major groove. Another prominent example is the EF-hand motif, a fold approximately 29 residues long, with a 12-residue loop that coordinates calcium ions via oxygen atoms from side chains like aspartate and glutamate; this motif is prevalent in calcium-sensing proteins such as . Specific examples illustrate the functional diversity of these motifs. The zinc finger motif, particularly the Cys2His2 type, features a beta-beta-alpha fold stabilized by tetrahedral coordination of a zinc ion via two cysteines and two histidines, enabling sequence-specific DNA or RNA binding in transcription factors. The leucine zipper motif, characterized by a coiled-coil dimerization interface with leucines spaced every seven residues along an alpha helix, facilitates protein-protein interactions for oligomerization in regulators like bZIP transcription factors. These motifs often reside within larger protein domains, enhancing their modularity. The evolutionary conservation of and structural motifs underscores their importance, as they are preserved across to maintain core functions like enzymatic activity or signaling. This conservation enables function prediction from genomic s alone, aiding annotation of uncharacterized proteins in large-scale studies. Databases like serve as key resources, cataloging thousands of motifs with associated patterns, profiles, and functional annotations derived from curated literature.

Classification

By structure

Proteins are classified by structure primarily according to their three-dimensional and associated properties, which reflect distinct folding patterns at the whole-protein level rather than specific biological roles. This classification encompasses globular, fibrous, , and , with overlaps possible in multi-domain architectures. Globular proteins exhibit a compact, roughly spherical with a hydrophobic core and hydrophilic surface, rendering them soluble in aqueous environments. Their folded structure often allows dynamic conformational changes that support various activities. A representative example is , an with a tightly packed fold stabilized by hydrogen bonds and disulfide bridges. In contrast, fibrous proteins possess elongated, thread-like structures that are typically insoluble in and provide strength. These proteins often form higher-order assemblies such as filaments or coils. For instance, features a triple-helical where three polypeptide chains wind together, contributing to tensile in connective tissues. , another example, consists of alpha-helical coils that dimerize into coiled-coil dimers, forming robust networks in and nails. Membrane proteins are categorized into integral and peripheral types based on their interaction with lipid bilayers. Integral membrane proteins are embedded within the membrane, often via transmembrane alpha-helices or beta-barrels that span the hydrophobic core. G-protein coupled receptors (GPCRs) exemplify this, with seven transmembrane helices forming a bundle that orients extracellular and intracellular domains. Peripheral membrane proteins, by comparison, associate loosely with the membrane surface through electrostatic or hydrophobic interactions without deep embedding. A distinct category includes (IDPs), which lack a stable three-dimensional fold under physiological conditions and instead adopt flexible, extended conformations. These proteins are often enriched in charged and polar residues, enabling rapid adaptability. serves as a key example, existing in a largely unstructured state that allows interactions with multiple partners in neuronal processes.

By function

Proteins are classified by function based on their biological roles in cellular and organismal processes, encompassing , , molecular , signaling, storage, defense, and contraction. This functional highlights how proteins execute diverse tasks essential for , often overlapping with their structural adaptations but prioritized here by purpose. Enzymes represent a primary class, acting as catalysts to accelerate biochemical reactions by lowering , with approximately 90% of cellular reactions relying on enzymes. Structural proteins provide mechanical support and maintain cellular architecture, such as in and in . Transport proteins facilitate the movement of molecules across membranes or within fluids, exemplified by , which binds and carries oxygen in erythrocytes. Hormones and signaling proteins mediate intercellular communication and physiological regulation, including insulin, which modulates blood glucose levels by promoting cellular uptake. Additional subtypes include storage proteins that sequester essential nutrients for later use, like , which binds and stores iron in cells to prevent toxicity while enabling release as needed. Defense proteins protect against pathogens and foreign invaders, with antibodies (immunoglobulins) secreted by B lymphocytes to recognize and neutralize antigens. Contractile proteins enable movement and force generation, such as the interaction between and in muscle fibers, powering through . These functional categories illustrate the versatility of proteins, with representative examples underscoring their specialized roles without exhaustive . The vast functional diversity of proteins arises evolutionarily from mechanisms like , which allows paralogous copies to diverge and acquire new roles while retaining core functions, contributing to the expansion of protein repertoires across species. In humans, approximately 20,000 protein-coding s generate this variety through and post-translational modifications, yielding numerous isoforms that fine-tune functions for specific contexts. This quantification underscores how a relatively modest gene set supports an expansive capable of multifaceted biological tasks.

By cellular location

Proteins are classified by their cellular location, which reflects their roles in specific subcellular environments or extracellular spaces. In eukaryotic cells, the majority of proteins are targeted to distinct compartments post-translationally or co-translationally, with over 50% requiring translocation across at least one to reach their destination. This localization ensures compartmentalization of biochemical processes, such as in the or energy production in mitochondria. Cytosolic proteins are soluble molecules that reside freely in the without specific targeting signals, comprising a significant portion of the cellular dedicated to functions like intermediary . For example, enzymes involved in , such as glyceraldehyde-3-phosphate dehydrogenase (GAPDH), operate in the to break down glucose into pyruvate, supporting energy production under conditions. These proteins often lack hydrophobic regions that would anchor them to membranes, allowing throughout the cytoplasmic matrix. Organelle-specific proteins are directed to intracellular structures like mitochondria, nuclei, or the endoplasmic reticulum (ER) and Golgi apparatus via dedicated import machineries. In mitochondria, proteins of the respiratory chain, such as cytochrome c oxidase subunits, assemble into complexes embedded in the inner membrane to facilitate electron transport and ATP synthesis. Nuclear proteins, including histones like H3 and H4, package DNA into chromatin within the nucleus, regulating gene expression and maintaining genomic stability. In the ER and Golgi, chaperone proteins such as BiP (also known as GRP78) assist in folding nascent polypeptides and ensuring quality control during protein maturation. Membrane-bound proteins integrate into lipid bilayers of cellular or membranes, often spanning the membrane with transmembrane domains. At the plasma membrane, receptors like G-protein-coupled receptors (GPCRs) and () detect extracellular signals and transduce them into intracellular responses, such as flux or activation. These proteins contribute to cellular communication and , with their localization anchoring them to the membrane via hydrophobic alpha-helices. Secreted proteins are exported from the cell through the secretory pathway, functioning in extracellular environments like the bloodstream or matrices. Examples include antibodies, such as , which provide immune defense by binding pathogens, and digestive enzymes like , which hydrolyze proteins in the gut lumen. components, such as , form structural networks that support integrity and outside the plasma membrane. Localization is primarily governed by targeting signals, short amino acid sequences that direct proteins to their destinations. N-terminal signal peptides, typically 15-30 residues long with a hydrophobic core, mediate entry into the for membrane-bound and secreted proteins, where they are cleaved upon translocation. For nuclear import, localization signals (NLS) consist of clusters of basic residues, such as the monopartite sequence PKKKRKV in , which bind receptors to facilitate transport through nuclear pores. These signals ensure precise distribution, with cytosolic proteins generally lacking them to remain in the .

Biosynthesis and Synthesis

Biological synthesis

Biological synthesis of proteins occurs through the central dogma of molecular biology, involving transcription of DNA into messenger RNA (mRNA) followed by translation of mRNA into polypeptide chains on ribosomes. This process ensures that genetic information encoded in DNA is accurately converted into functional proteins essential for cellular activities. Transcription begins when RNA polymerase binds to specific promoter sequences in the DNA, initiating the synthesis of a complementary mRNA strand from the DNA template. In eukaryotes, RNA polymerase II primarily handles mRNA production, recognizing core promoter elements such as the TATA box, while enhancers—distal regulatory DNA sequences—bind transcription factors to boost transcription rates by looping to the promoter and facilitating polymerase recruitment. In prokaryotes, transcription is regulated by operons, such as the lac operon in Escherichia coli, where a single promoter controls multiple genes, and repressor or activator proteins modulate access based on environmental signals like lactose availability. In eukaryotes, the primary transcript (pre-mRNA) undergoes processing in the nucleus, including addition of a 5' cap (7-methylguanosine), cleavage and addition of a poly(A) tail at the 3' end, and splicing by the spliceosome to remove non-coding introns and join coding exons, producing mature mRNA for export to the cytoplasm. During , ribosomes assemble on the mRNA in the , reading its sequence in triplets known as codons according to the , which consists of 64 possible codons specifying 20 standard plus three stop signals (UAA, UAG, UGA). Transfer RNAs (tRNAs), each carrying a specific , recognize codons via complementary anticodons in the ribosome's A site, ensuring precise incorporation. The process unfolds in three phases: , where the small ribosomal subunit binds mRNA and scans to the AUG, recruiting the initiator tRNA and large subunit to form the complete ; , involving sequential tRNA binding, formation, and translocation; and termination, triggered by stop codons releasing the completed polypeptide. Peptide bond formation, catalyzed by the ribosome's center—a activity of —links the carboxyl group of the peptidyl-tRNA in the to the amino group of the in the A site, releasing the deacylated tRNA: \text{peptidyl-tRNA (P site)} + \text{aminoacyl-tRNA (A site)} \rightarrow \text{peptidyl-aminoacyl-tRNA (A site)} + \text{tRNA (P site)} This reaction proceeds without external energy input beyond GTP for translocation, accelerating the uncatalyzed rate by approximately 10^7-fold. As the nascent polypeptide emerges from the ribosomal exit tunnel—about 30-40 amino acids long—co-translational folding commences, with chaperone proteins like trigger factor in prokaryotes or Hsp70 in eukaryotes assisting to prevent aggregation and guide domain formation vectorially along the chain. This sequential emergence influences folding pathways, allowing early domains to stabilize before later ones are synthesized. Regulation of biological synthesis occurs at multiple levels to fine-tune protein production. Transcriptional control involves enhancers and operons that respond to cellular signals, such as nutrient availability in the lac operon, to activate or repress gene expression. Translational regulation includes microRNAs (miRNAs), small non-coding RNAs that bind mRNA 3' untranslated regions, inhibiting initiation or promoting decay to suppress protein output from specific transcripts. Following translation, proteins frequently undergo post-translational modifications, such as phosphorylation or glycosylation, to attain mature structure and function.

Chemical and recombinant synthesis

Proteins can be produced in the laboratory through or technology, enabling the creation of proteins not reliant on cellular machinery. primarily employs solid-phase synthesis (SPPS), pioneered by Robert Bruce Merrifield in 1963, which anchors the growing chain to an insoluble resin and involves iterative cycles of coupling, deprotection, and washing. In SPPS, protected are added stepwise via formation, typically using or other coupling agents, followed by selective deprotection of the N-terminal group to allow the next addition. This method revolutionized chemistry by facilitating automation and purification through resin filtration, but it is generally limited to peptides under 50 due to cumulative yield losses from incomplete couplings and side reactions that increase with chain length. Recombinant synthesis, in contrast, leverages genetic engineering to express proteins in heterologous host organisms, offering scalability for larger proteins. The process begins with cloning the target gene into an expression vector containing regulatory elements like promoters, ribosome binding sites, and terminators; for example, the T7 promoter system in Escherichia coli drives high-level transcription via bacteriophage T7 RNA polymerase, inducible by IPTG. Common hosts include prokaryotes like E. coli for rapid, cost-effective production; yeasts such as Pichia pastoris for eukaryotic folding and secretion; and mammalian cells like CHO for complex post-translational modifications including glycosylation. Purification is streamlined by fusing affinity tags to the protein, such as the hexahistidine (His6) tag, which enables one-step immobilized metal affinity chromatography (IMAC) under mild conditions. The landmark achievement was the 1978 production of human insulin chains in E. coli, assembled post-expression to yield the first recombinant therapeutic protein approved in 1982. Advances in these methods address limitations of scale and modification. Cell-free systems, derived from crude extracts supplemented with energy sources, , and , allow in vitro transcription-translation without intact cells, supporting rapid prototyping and incorporation of non-natural ; yields have improved to milligrams per milliliter in optimized wheat germ or E. coli-based reactions. extends chemical approaches to larger proteins (>100 residues) by combining recombinant expression of polypeptide segments with chemical ligation; native chemical ligation (NCL), developed in 1994, chemoselectively joins an N-terminal peptide to a C-terminal fragment via thiol-thioester exchange and native formation, enabling site-specific labeling or incorporation of modifications. These techniques underpin applications in therapeutics, such as recombinant insulin and monoclonal antibodies, where E. coli or hosts achieve gram-scale yields, though challenges persist in ensuring proper folding (e.g., avoiding ) and glycosylation fidelity, often requiring mammalian systems for biologics like . Recent innovations incorporate to optimize recombinant expression, particularly through codon usage adaptation. models, trained on host-specific transcriptomes, predict synonymous codon sequences that maximize efficiency and minimize mRNA secondary structures; for instance, recurrent neural networks have boosted E. coli yields by up to 10-fold for diverse proteins since 2021. Tools like CodonTransformer further generalize this across , enhancing scalability for industrial .

Cellular Functions

Catalysis and enzymes

Enzymes are proteins that function as biological , accelerating the rate of biochemical reactions by lowering the required for the reaction to proceed, without being altered or consumed in the process. This catalysis occurs primarily at the enzyme's , a specific three-dimensional formed by residues that bind the through non-covalent interactions such as hydrogen bonds, electrostatic forces, and hydrophobic effects. The binding orients the substrate molecules in a way that facilitates the , stabilizing it and thus reducing the energy barrier. Two primary models describe the interaction between the enzyme's and the . The lock-and-key model, proposed by in 1894, posits that the active site has a rigid, complementary shape to the substrate, allowing precise akin to a key fitting a lock, which ensures specificity. In contrast, the induced fit model, introduced by Daniel E. Koshland in 1958, suggests that the active site is flexible and undergoes a conformational change upon substrate , optimizing the alignment for and further enhancing specificity and efficiency. Enzyme kinetics quantifies the rates of these catalyzed reactions, with the Michaelis-Menten equation providing a foundational description for many enzymes following hyperbolic kinetics: v = \frac{V_{\max} [S]}{K_m + [S]} where v is the initial reaction velocity, V_{\max} is the maximum velocity achieved at saturating substrate concentration [S], and K_m is the Michaelis constant, representing the substrate concentration at which v = \frac{1}{2} V_{\max} and serving as a measure of the enzyme's for the (lower K_m indicates higher ). This equation, derived from the work of and in 1913, assumes steady-state conditions where the enzyme-substrate complex formation and breakdown are balanced. Enzymes are classified into seven major classes based on the type of reaction they catalyze, as defined by the Enzyme Commission (EC) numbering system established by the International Union of Biochemistry and (IUBMB). These include EC 1 oxidoreductases, which catalyze oxidation-reduction reactions (e.g., dehydrogenases transferring electrons); EC 2 transferases, which transfer functional groups like methyl or (e.g., kinases); EC 3 hydrolases, which cleave bonds using water; EC 4 lyases, which form or break double bonds; EC 5 isomerases, which rearrange atoms within molecules; EC 6 ligases, which join molecules using ATP; and EC 7 translocases, which transport ions or molecules across membranes. Each enzyme receives a unique four-digit EC number reflecting its class, subclass, sub-subclass, and specific reaction. Many enzymes require non-protein cofactors to achieve full catalytic activity. Prosthetic groups are tightly or covalently bound organic molecules, such as the group in , which contains an iron atom essential for in the . Coenzymes, which are loosely bound and often derived from vitamins, act as transient carriers of chemical groups; for example, (NAD^+) serves as an in reactions, facilitating hydride transfer during and the . Enzyme activity is tightly regulated to maintain cellular homeostasis, with key mechanisms including allostery and inhibition. Allosteric regulation involves binding of effectors at sites distinct from the active site, inducing conformational changes that either activate or inhibit the enzyme; this was formalized in the Monod-Wyman-Changeux model in 1965, which describes cooperative transitions between tense (low-affinity) and relaxed (high-affinity) states. Inhibition can be competitive, where the inhibitor competes with the substrate for the active site, increasing apparent K_m without affecting V_{\max}, or noncompetitive, where the inhibitor binds an allosteric site, decreasing V_{\max} without altering K_m. A representative example is hexokinase, the enzyme catalyzing the first step of glycolysis (phosphorylation of glucose to glucose-6-phosphate), which is allosterically inhibited by its product glucose-6-phosphate in mammalian cells, preventing excessive glycolytic flux when downstream intermediates accumulate.51600-3/fulltext)

Structural and mechanical roles

Proteins play essential roles in providing structural integrity and mechanical support within and organisms, forming scaffolds that maintain shape, enable movement, and withstand physical stresses. In the , filaments, composed of globular monomers polymerized into double-helical structures, provide tensile strength and facilitate and division, while , assembled from α- and β-tubulin dimers, offer compressive resistance and serve as tracks for intracellular . Intermediate filaments, such as those made from or keratins, form rope-like networks that resist mechanical stress and link the to the , ensuring cellular resilience. The () relies on proteins like , which forms triple-helical that impart high tensile strength—exceeding that of on a weight basis—and elasticity to tissues such as tendons and . , a cross-linked of tropoelastin, provides reversible elasticity in organs like lungs and arteries, allowing repeated stretching and without damage.46358-3/fulltext) , a multidomain , mediates by binding on the cell surface to components, thereby anchoring the to the external environment and facilitating organization.00845-6) Mechanical functions are exemplified by , a that interacts with filaments to generate contractile forces in muscle cells, enabling movement through ATP-driven sliding of filaments. , transmembrane heterodimers, transduce mechanical signals by linking the to the , regulating cell shape and migration in response to physical cues. These proteins exhibit dynamic assembly and disassembly, allowing rapid remodeling in response to cellular needs, such as during or embryonic development. Mutations in structural proteins can lead to diseases; for instance, defects in , a rod-like protein that connects the to the in muscle cells, cause by compromising membrane stability and force transmission. Overall, these proteins' mechanical properties, including their ability to bear loads far exceeding biological scales, underscore their in maintaining architecture.

Signaling and transport

Proteins play crucial roles in cellular signaling and molecular , enabling communication between cells and the movement of ions, nutrients, and other molecules across membranes or through the bloodstream. Signaling proteins, such as receptors, detect extracellular cues and initiate intracellular cascades that regulate processes like , , and response to stimuli. Transport proteins facilitate the selective passage of substances, either passively down concentration gradients or actively against them using energy. These functions are essential for maintaining and coordinating physiological responses across organisms. In transport, channel proteins form pores in cell membranes to allow passive diffusion of specific ions or small molecules. For instance, aquaporins are integral membrane proteins that selectively conduct molecules across cell membranes, preventing osmotic imbalances in diverse tissues like kidneys and . Unlike broader porins, aquaporins exhibit high specificity and selectivity, restricting passage to water while excluding protons and other ions through a narrow, hourglass-shaped pore. Carrier proteins, another class of passive transporters, undergo conformational changes to bind and translocate substrates without forming open channels. The (GLUT) family exemplifies this, with facilitating of glucose into cells to support energy needs, particularly in erythrocytes and the - barrier. Active transport, powered by , enables uphill movement against gradients. The sodium-potassium (Na+/K+ ) is a prototypical example, pumping three sodium ions out of the cell and two potassium ions in per cycle, establishing electrochemical gradients vital for nerve impulses and nutrient uptake. Hemoglobin, a soluble transport protein in , binds oxygen in the lungs and releases it in tissues, leveraging among its four subunits to enhance efficiency under varying oxygen levels. In neurons, voltage-gated ion channels, such as sodium and potassium channels, mediate rapid ion fluxes during action potentials, enabling electrical signaling over long distances.02767-X) Signaling begins with receptor proteins that bind ligands like hormones or neurotransmitters, transducing signals across the membrane. G protein-coupled receptors (GPCRs), the largest family of cell surface receptors, activate heterotrimeric G proteins upon binding, leading to dissociation and modulation of effectors. This initiates diverse pathways, including those producing second messengers like cyclic AMP (), which is generated by and amplifies signals by activating . Receptor tyrosine kinases (RTKs), such as the , autophosphorylate upon dimerization, recruiting adaptor proteins to propagate signals through cascades like the (MAPK) pathway. The MAPK cascade involves sequential of kinases (Raf, MEK, ERK), culminating in activation to regulate and . Peptide hormones, including , exemplify extracellular signaling; binds its GPCR on liver cells, elevating levels to promote and glucose release. Regulation of these proteins ensures precise and timely responses, preventing overstimulation. by kinases, such as kinases (GRKs), modifies receptor activity; for GPCRs, GRK-mediated recruits s, uncoupling the receptor from G proteins and promoting internalization. Desensitization mechanisms, including homologous desensitization via binding, rapidly attenuate signaling after prolonged exposure, while heterologous desensitization involves cross-talk from other pathways. These regulatory steps maintain signaling fidelity and allow adaptation to changing environments.

Defense and regulation

Proteins play crucial roles in immune defense by recognizing and neutralizing pathogens. Antibodies, also known as immunoglobulins, are Y-shaped glycoproteins produced by B cells that bind specifically to on pathogens or infected cells, marking them for destruction. The (IgG) subclass, the most abundant in human serum, consists of two heavy chains and two light chains linked by bonds, with the regions responsible for antigen binding and the region mediating effector functions like complement and . Complement proteins form a cascade of over 30 plasma proteins that amplify immune responses by opsonizing pathogens, recruiting inflammatory cells, and directly lysing microbes through the membrane attack complex. occurs via classical, , or alternative pathways, all converging on cleavage to initiate downstream effects. Cytokines such as interferons coordinate innate and adaptive immunity; type I interferons (e.g., IFN-α and IFN-β) are rapidly induced by viral infections and inhibit while enhancing and activity. Major histocompatibility complex (MHC) proteins present antigenic peptides to T cells, enabling adaptive immune recognition. molecules display intracellular peptides to cytotoxic + T cells, triggering elimination of infected or malignant cells, while molecules on antigen-presenting cells present extracellular peptides to helper + T cells, promoting activation and production. These proteins ensure immune specificity and tolerance by binding diverse peptides in polymorphic grooves, with (HLA) loci encoding the most variable MHC genes. In cellular regulation, proteins maintain and respond to . Transcription factors like act as tumor suppressors by binding response elements to activate genes involved in , arrest, and following genotoxic . integrates signals from DNA damage sensors, with its activity modulated by posttranslational modifications such as and . , a small 76-amino-acid protein, tags substrates for proteasomal degradation via E1-E2-E3 enzyme cascades, regulating protein levels critical for progression and ; polyubiquitin chains typically linked via 48 serve as degradation signals. Heat shock proteins (HSPs), including and , function as molecular chaperones that assist in , prevent aggregation under , and facilitate refolding or degradation of misfolded proteins to preserve cellular . Apoptosis, or , is executed by , a family of proteases activated in a proteolytic . Initiator (e.g., , -9) cleave and activate effector (e.g., caspase-3, -7), which dismantle cellular structures by cleaving substrates like PARP and , ensuring orderly cell demise without inflammation. Hormones such as insulin-like growth factors (IGFs), which are single-chain polypeptides, regulate cellular proliferation and metabolism by binding receptor tyrosine kinases, activating PI3K/Akt and MAPK pathways to promote growth and survival. Adaptive immunity, characterized by antigen-specific receptors on lymphocytes, evolved uniquely in vertebrates, emerging around 500 million years ago in jawed species with the development of RAG-mediated V(D)J recombination for antibody and T cell receptor diversity. Invertebrates rely solely on innate mechanisms, lacking this somatic diversification. This evolutionary innovation provided vertebrates with memory and specificity against evolving pathogens, distinguishing it from the conserved innate systems shared across metazoans.

Metabolism and Degradation

Protein turnover

Protein turnover refers to the dynamic process by which cells continuously degrade and recycle proteins to maintain , balancing synthesis rates to ensure cellular . This degradation is essential for removing damaged or unnecessary proteins, with rates varying widely across cell types and conditions. The primary intracellular pathway for selective protein degradation is the ubiquitin-proteasome system (UPS), which targets individual proteins for ATP-dependent breakdown. In this process, (E1) forms a thioester bond with using ATP, transferring it to ubiquitin-conjugating enzymes (E2), which then work with ubiquitin ligases () to covalently attach polyubiquitin chains to residues on proteins. The ubiquitinated proteins are recognized by the 26S proteasome, a large multiprotein that unfolds and degrades them into short peptides while recycling . This system handles the majority of short-lived regulatory proteins, such as cyclins involved in control. In parallel, lysosomal pathways mediate the degradation of bulk cytoplasmic components and membrane proteins. Autophagy, a key lysosomal process, engulfs portions of the cytoplasm or organelles into double-membrane vesicles called autophagosomes, which fuse with lysosomes to form autolysosomes where hydrolases degrade the contents into amino acids and other building blocks. For membrane proteins, the endosomal-lysosomal pathway internalizes them via endocytosis, sorting them into multivesicular bodies that deliver cargo to lysosomes for proteolytic digestion. These mechanisms complement the UPS by handling larger aggregates or organelles that cannot be processed by proteasomes. Protein half-lives span a broad range, from minutes for unstable regulators like cyclins to days or longer for structural proteins such as , allowing rapid responses to cellular needs while preserving stable components. is often regulated by the pathway, where the identity of the N-terminal determines degradation susceptibility; for instance, N-terminal or signals rapid ubiquitination and proteasomal breakdown via E3 ligases like UBR1. This rule, first elucidated by Alexander Varshavsky, ensures precise control over protein stability. Protein turnover serves multiple critical functions, including by eliminating misfolded or damaged proteins to prevent , signaling through regulated degradation of key factors like IκB to activate pathways in immune responses, and nutrient recycling by breaking down proteins into reusable during . These processes maintain integrity and adapt cellular to environmental changes. Dysregulation of contributes to diseases, notably in where impaired UPS and lead to accumulation of aggregates, triggering neuronal death through lysosomal dysfunction and collapse. In such cases, oligomers inhibit autophagosome-lysosome fusion, exacerbating protein buildup.

Digestion and absorption

Protein digestion begins in the , where the , secreted as inactive pepsinogen by chief cells, is activated in the acidic environment created by gastric . This low , typically ranging from 1.5 to 3.5, denatures dietary proteins and enables to cleave bonds, primarily those involving aromatic like and , producing large polypeptides. 's activity is optimal at pH 2, ensuring initial breakdown without complete . In the , further occurs primarily in the and , facilitated by pancreatic enzymes secreted into the . and , released from the as inactive zymogens ( and ) and activated by enterokinase on the intestinal , hydrolyze peptide bonds at the carboxyl side of / () and aromatic residues (), respectively, breaking polypeptides into smaller peptides and oligopeptides. Additional brush-border enzymes, such as aminopeptidases and dipeptidases from enterocytes, complete the process by liberating free and di- or tripeptides. The products of —free , dipeptides, and tripeptides—are absorbed across the apical of enterocytes, mainly in the , via specific transporters. The proton-coupled transporter PEPT1 facilitates the uptake of di- and tripeptides using a proton , while individual are transported by sodium-dependent carriers like B^0AT1 for neutral types. Inside enterocytes, peptides are further hydrolyzed by cytosolic peptidases into , which then exit basolaterally via transporters such as LAT4 into the for delivery to the liver, where they undergo first-pass metabolism for protein synthesis, energy production, or other pathways. Regulation of protein digestion involves hormonal signals that coordinate gastric and pancreatic secretions. , released by G cells in the antrum in response to proteinaceous , stimulates secretion to optimize activity. , secreted by I cells in the upon detection of peptides and fats, promotes pancreatic enzyme release, including and , and enhances contraction for delivery to aid overall . Disruptions, such as in celiac disease—an autoimmune disorder triggered by —lead to villous atrophy in the , impairing enzyme activity and resulting in incomplete digestion of gluten peptides, which exacerbates . Dietary proteins must supply essential amino acids, which cannot be synthesized by the and are required for protein synthesis; examples include , vital for formation and immune function, typically obtained from sources like , , and . Inadequate intake of these, such as , can limit overall protein utilization, underscoring the importance of sources in the diet.

Methods of Study

Purification and analysis

Protein purification involves isolating target proteins from complex biological mixtures, such as lysates or culture media, to achieve high purity for downstream applications in and . This process typically combines multiple techniques to exploit differences in protein physicochemical properties, including size, charge, solubility, and specific affinities. Effective purification maintains protein and activity while minimizing from host proteins, nucleic acids, or . Centrifugation serves as an initial step in to separate cellular debris and organelles from soluble proteins. applies increasing centrifugal forces to pellet components based on density and size, while density gradient centrifugation, using media like or cesium , further refines separation by forming bands at equilibrium positions corresponding to buoyant densities. For instance, density gradient ultracentrifugation isolates protein complexes by rates, achieving resolutions sufficient for native complex analysis. Chromatography is the cornerstone of , enabling scalable separation based on specific interactions. Ion-exchange chromatography separates proteins by net surface charge using charged , such as anion exchangers (e.g., DEAE) for negatively charged proteins or cation exchangers (e.g., ) for positively charged ones, with via salt or pH gradients. leverages biospecific interactions, such as between a fused tag (e.g., ) and immobilized (e.g., Ni-NTA ), allowing one-step purification with yields often exceeding 90% purity; this method was pioneered in 1968 for . , also known as gel filtration, resolves proteins by hydrodynamic volume through porous matrices, separating monomers from aggregates without altering native structure. Electrophoretic techniques provide high-resolution analysis and preparative separation of purified proteins. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) denatures proteins with SDS to impart uniform negative charge, separating them by molecular weight in a polyacrylamide gel under an ; this method, developed in 1970, remains standard for estimating purity and size. (IEF) separates proteins by (pI) in a pH gradient, where migration ceases at the point of zero net charge. Two-dimensional () gel electrophoresis combines IEF in the first dimension with in the second, resolving up to thousands of proteins for comprehensive profiling, as introduced in 1975. Protein quantification ensures accurate yield assessment post-purification. The Bradford assay measures microgram quantities via G-250 dye binding to basic and aromatic , producing a color shift detectable at 595 nm; it is rapid and compatible with detergents but sensitive to interferents like . The bicinchoninic acid () assay detects protein via reduction of Cu²⁺ to Cu⁺ in alkaline medium, followed by with BCA for absorbance at 562 nm, offering compatibility with reducing agents and sensitivity down to 0.5 μg/mL. Activity assays, such as enzymatic kinetic measurements, quantify functional protein levels by monitoring substrate conversion rates. Purity assessment confirms isolation success through orthogonal methods. Ultraviolet (UV) spectroscopy at 280 nm quantifies protein concentration based on aromatic residue absorbance (tyrosine and tryptophan), with purity inferred from the A280/A260 ratio to detect nucleic acid contamination. Mass spectrometry (MS), including electrospray ionization (ESI) or matrix-assisted laser desorption/ionization (MALDI), verifies protein identity by accurate mass measurement and detects impurities via peptide mapping, often achieving >95% sequence coverage. Challenges in protein purification include maintaining stability during isolation, as proteins may aggregate, degrade, or lose activity due to shear forces, shifts, or ; stabilizers like or inhibitors mitigate these issues. for biotechnological production demands robust processes that transition from lab-scale (milligrams) to industrial-scale (grams to kilograms) without yield loss, often requiring optimization of capacities and systems.

Structure determination

Protein structure determination involves experimental techniques that resolve the three-dimensional atomic coordinates of proteins, providing essential insights into their function, interactions, and mechanisms. These methods have evolved from early pioneering efforts in the mid-20th century to high-resolution approaches capable of near-atomic detail, enabling the study of proteins in various states and complexes. remains the most widely used technique for determining protein structures at atomic resolution, typically achieving resolutions better than 2 . In this method, proteins are crystallized, and an beam is directed at the crystal, producing a pattern from which maps are reconstructed. The problem, which arises because diffraction experiments measure only intensities and not phases of the scattered waves, is commonly solved using multiple isomorphous replacement (MIR), where heavy atoms like mercury are introduced into isomorphous crystals to provide phase information via differences in diffraction patterns. The first complete three-dimensional structure of a protein, , was determined this way by and colleagues in 1960 at 2 resolution, revealing the protein's folded polypeptide chain and group for the first time. Nuclear magnetic resonance (NMR) determines s in solution, capturing dynamic ensembles rather than static crystal forms, and is particularly suited for smaller proteins under 50 kDa. It relies on measuring nuclear spin interactions, such as through-space (NOE) constraints between nearby atoms (typically <5 Å apart), along with restraints from coupling constants and chemical shifts, to computationally refine models that fit the spectral data. The first full by NMR, that of bovine pancreatic (BPTI), was achieved by Kurt Wüthrich's group in 1985 at approximately 2.5 Å effective resolution, demonstrating the technique's ability to resolve backbone and side-chain conformations in aqueous environments. Cryogenic electron microscopy (cryo-EM) has revolutionized structure determination for large macromolecular complexes and membrane proteins that resist , using single-particle analysis to average thousands of two-dimensional projections into a three-dimensional density map without requiring crystals. Samples are flash-frozen in vitreous ice to preserve native states, imaged with beams, and computationally reconstructed; advances in detectors and plates since the have driven the "resolution revolution," routinely achieving ~2 resolutions by the for complexes over 100 kDa. For example, the structure of the , a massive assembly exceeding 2.5 MDa, was first resolved at 3.5 in 2000 and improved to near-atomic detail in subsequent studies, highlighting cryo-EM's power for dynamic assemblies. Hybrid methods integrate data from multiple techniques, such as low-resolution cryo-EM maps with high-resolution NMR or fragments, using computational modeling to resolve structures of complex systems that individual methods cannot fully address alone. These integrative approaches employ restraints from diverse sources—like distance constraints from NMR NOEs and shape envelopes from (SAXS)—to generate ensemble models, as demonstrated in studies of the complex where cryo-EM provided overall architecture and NMR detailed flexible domains. Resolved structures are archived in the Protein Data Bank (PDB), a public repository established in 1971 that now holds over 200,000 entries, facilitating global research and validation. Early depositions included myoglobin (PDB ID: 1MBN, deposited 1989 but based on 1960 data) and hemoglobin, marking the foundation for structural biology. As of November 2025, the PDB holds over 260,000 entries.

Structure prediction and design

Protein structure prediction involves computational methods to determine the three-dimensional (3D) arrangement of atoms in a protein from its amino acid sequence, a challenge that has evolved from physics-based simulations to advanced machine learning approaches. Traditional techniques include homology modeling, which builds target protein structures by aligning sequences to known homologs in databases like the Protein Data Bank (PDB) and refining the model using energy minimization. Ab initio methods, such as those implemented in the Rosetta software, assemble protein structures from scratch using fragment-based assembly and Monte Carlo sampling to minimize an energy function derived from physical principles, achieving success for small proteins without close homologs. The field underwent a revolution with artificial intelligence (AI), particularly DeepMind's AlphaFold2, which in 2020 dominated the Critical Assessment of Structure Prediction (CASP14) competition by predicting structures with atomic accuracy for diverse proteins, even those lacking homologs, using deep learning on multiple sequence alignments and evolutionary data. This breakthrough earned the 2024 Nobel Prize in Chemistry for Demis Hassabis and John Jumper (for AlphaFold's development) and David Baker (for computational protein design). AlphaFold3, released in 2024, extends this capability to predict joint structures of protein complexes with ligands, DNA, and RNA, improving accuracy for biomolecular interactions by up to 50% over prior models in blind tests. Open-source alternatives like ESMFold, based on language models trained on evolutionary-scale sequence data, enable rapid single-sequence structure prediction without alignments, achieving near-AlphaFold accuracy for many targets in seconds on standard hardware. Similarly, RoseTTAFold from the lab uses a three-track (, 1D distance map, 3D coordinates) for high-accuracy predictions and has facilitated experimental structure determination via and cryo-electron microscopy. These tools complement experimental methods by generating hypotheses for validation, accelerating research in . Protein design leverages these prediction advances to create novel structures with desired functions, starting with de novo approaches like Baker's 2003 design of Top7, the first artificial protein fold with no natural counterpart, achieved through Rosetta's computational optimization and confirmed by to match the intended with 1.2 RMSD. Recent AI-driven methods, such as RFdiffusion (2023), fine-tune RoseTTAFold into a for generating diverse backbones conditioned on motifs or symmetries, enabling designs of binders, enzymes, and symmetric assemblies with experimental success rates over 20% for novel folds. Applications span and ; predictions have identified novel drug targets by revealing cryptic pockets in disease-related proteins, such as SARS-CoV-2 enzymes, aiding inhibitor design. In enzyme engineering, tools like RFdiffusion redesign active sites for enhanced catalysis, as in creating luciferases with shifted emission spectra for bioimaging. Despite progress, challenges persist: AI models like struggle with protein dynamics, often outputting static snapshots that overlook conformational ensembles critical for function. Accuracy drops for intrinsically disordered regions (IDRs), which lack stable folds and comprise ~30% of eukaryotic proteomes, due to insufficient evolutionary signals in alignments. proteins, embedded in bilayers, pose additional hurdles from incomplete environmental modeling, though post-2024 refinements in AlphaFold3 and specialized datasets have improved predictions for ~70% of targets. Looking ahead, AI-driven is poised to transform therapeutics by 2025, with generative models enabling custom biologics like antibodies and enzymes for , potentially reducing development timelines from years to months.

Proteomics and interactomics

encompasses the large-scale study of the entire set of proteins, or , within a , , or , enabling the , quantification, and characterization of proteins in complex biological samples. Liquid chromatography coupled to (LC-MS/MS) serves as the cornerstone technique for , where proteins are digested into peptides, separated by liquid chromatography, and analyzed by to determine their mass-to-charge ratios and fragmentation patterns for and relative quantification. This approach has revolutionized protein profiling by allowing the detection of thousands of proteins simultaneously with high , outperforming traditional antibody-based methods in throughput and coverage. A key application of proteomics involves the analysis of post-translational modifications (PTMs), such as , which regulate protein function, localization, and interactions. Phosphorylation sites are identified through LC-MS/MS by detecting characteristic mass shifts (e.g., +80 Da for phosphate addition) on peptides, often enriched using techniques like to improve detection of low-abundance modified proteins. Mass spectrometry enables site-specific mapping and quantification of events across entire proteomes, revealing dynamic signaling networks in response to stimuli. For instance, phosphoproteomics has identified thousands of phosphorylation sites in mammalian cells, providing insights into kinase-substrate relationships. Quantitative proteomics methods enhance the ability to measure protein abundance changes across conditions. Stable isotope labeling by amino acids in cell culture (SILAC) incorporates heavy isotopes into proteins during cell growth, allowing direct comparison of samples by mass differences upon mixing and LC-MS/MS analysis, considered a gold standard for its accuracy in metabolic labeling. Isobaric tags for relative and absolute quantification (iTRAQ) enable multiplexing of up to eight samples by attaching tags that release reporter ions during fragmentation, facilitating precise relative quantification without altering peptide masses prior to analysis. These techniques have been pivotal in studying protein dynamics, such as in response to drug treatments or disease states. Interactomics focuses on mapping protein-protein interactions (PPIs) to understand functional networks. The yeast two-hybrid (Y2H) system detects interactions by reconstituting a in cells, where a bait protein fused to a interacts with a prey protein fused to an activation domain, driving expression. Co-immunoprecipitation (co-IP) captures native complexes by using antibodies to pull down a target protein and its interactors from cell lysates, followed by identification via . These methods contribute data to PPI networks, such as the database, which integrates experimental, computational, and literature-derived interactions for over 12,000 organisms, scoring associations based on confidence levels to predict functional partnerships. In applications, identifies disease by comparing proteomes from healthy and pathological samples, such as elevated levels of specific phosphoproteins in cancer signaling pathways, aiding early and prognosis. In , and interactomics data integrate with other to model cellular processes, revealing how PPI networks respond to perturbations like infections. For example, LC-MS/MS-based profiling has uncovered biomarker panels for cardiovascular diseases and cancers, while STRING-facilitated networks illuminate pathway dysregulation in complex disorders. Recent advances in the include single- proteomics, where miniaturized LC-/MS platforms like nanoPOTS enable proteome analysis of individual cells, quantifying over 1,000 proteins per cell to uncover heterogeneity in tumors or immune responses. Spatial proteomics combines with imaging to map protein distributions in tissues, as in Deep Visual Proteomics, which uses to segment and analyze laser-microdissected regions for proteotoxicity studies in diseases like alpha-1-antitrypsin deficiency. Additionally, integration post-2024 has improved prediction; deep learning models like those using prompt-based fine-tuning on sequence data achieve high accuracy in forecasting sites by learning from large datasets, enhancing proteome annotation without exhaustive experimentation.

Mechanical and Physical Properties

Mechanical properties

Proteins exhibit a range of mechanical properties that enable them to withstand and respond to physical forces, including elasticity, strength, and , which are crucial for their structural integrity under . These properties arise from the of chains into secondary and tertiary structures, allowing proteins to deform reversibly or unfold under applied forces. Measurements of these properties often involve techniques that probe single molecules or bulk assemblies, providing insights into how proteins behave as biological materials. Atomic force microscopy (AFM) is a primary for assessing single-molecule mechanical properties, such as force-induced elongation and unfolding, by applying tensile forces at the piconewton scale and measuring deformation with nanometer . For larger assemblies like protein fibers, evaluates bulk mechanical responses, including stress-strain behavior, by stretching samples until failure to determine parameters like and breaking strength. These techniques reveal how proteins balance rigidity and flexibility in response to mechanical loads. A key aspect of protein is , where deformation is time-dependent, combining elastic recovery with viscous dissipation, as seen in the of protein networks under constant strain. Unfolding forces represent another critical type, where applied tension disrupts non-covalent interactions, leading to domain extension; for instance, immunoglobulin domains in the muscle protein unfold at forces of 100-200 pN, contributing to muscle elasticity during contraction. These behaviors highlight proteins' ability to absorb and prevent . Mechanical properties are influenced by intramolecular interactions, including hydrogen bonds that provide reversible linkages for elasticity, disulfide cross-links that enhance tensile strength through covalent stabilization, and beta-sheets that confer rigidity via extended hydrogen-bonded networks. Disulfide bonds, in particular, lock protein conformations, increasing resistance to deformation, while beta-sheet motifs form stiff scaffolds in fibrous proteins. These factors allow proteins to tune their mechanical response based on environmental demands. Representative examples illustrate these properties: proteins, composed of beta-sheet nanocrystals embedded in amorphous regions, achieve a tensile strength of approximately 1 GPa, rivaling synthetic high-performance fibers due to sacrificial bonds that dissipate . In contrast, exhibits high extensibility, with single stretching up to 10-15% of their length before nonlinear stiffening, enabling tissues like tendons to endure repeated loading without fracture. These mechanical attributes underpin applications in biomaterials, where engineered protein hydrogels or fibers mimic natural toughness for scaffolds in , leveraging for dynamic cell interactions. In biomolecular motors, such as , mechanical properties like stall forces around 5-7 pN enable directed transport along cytoskeletal filaments, inspiring nanoscale devices for .

Biophysical characteristics

Proteins' biophysical characteristics encompass their thermodynamic , , and dynamic behaviors, which collectively determine how these macromolecules maintain and respond to environmental cues. The thermodynamic of a folded protein is primarily characterized by the change upon folding, \Delta G_{\text{fold}}, which balances enthalpic and entropic contributions to favor the native state under physiological conditions. Typically, \Delta G_{\text{fold}} ranges from -5 to -15 kcal/mol for stable proteins, reflecting a delicate that can be disrupted by or ligands. changes, \Delta C_p, during unfolding further influence , as proteins in the unfolded state absorb more heat due to exposed hydrophobic groups, leading to a parabolic dependence of \Delta G on temperature. Denaturation curves, obtained via (), reveal the melting temperature T_m, the midpoint of thermal unfolding where half the protein is denatured. In , excess peaks at T_m, often between 40–80°C for globular proteins, providing a direct measure of unfolding \Delta H and confirming two-state transitions in many cases. For instance, hyperthermophilic proteins exhibit higher T_m values, up to 100°C, due to enhanced \Delta G from optimized bridges and hydrophobic packing. Optical properties of proteins arise from their residues and enable non-invasive structural probing. (CD) spectroscopy in the far-UV range (190–250 nm) assesses by measuring differential absorption of left- and right-circularly polarized , with characteristic spectra for \alpha-helices (strong negative bands at 208 and 222 nm) and \beta-sheets (negative at ~215 nm). This technique quantifies folding and ligand-induced changes, as helical content correlates with mean residue ellipticity at 222 nm. Intrinsic from residues, excited at ~280 nm, reports on local environment; occurs via collisional or static mechanisms when increases upon unfolding or binding, shifting emission from ~330 nm (buried) to ~350 nm (exposed). Protein dynamics involve conformational fluctuations on timescales from picoseconds to seconds, with (μs) to (ms) motions critical for . Residence times for intermediate states often fall in the μs–ms range, probed by techniques like NMR relaxation dispersion, revealing hidden states that modulate and . (MD) simulations provide an overview of these by evolving atomic trajectories under force fields, capturing bond vibrations (fs–ps), side-chain rotations (ns–μs), and loop motions (μs–ms) in explicit solvent. A classic example of dynamic regulation is the allosteric in , where proton binding at μs–ms timescales stabilizes the tense (T) state, reducing oxygen affinity and promoting cooperative release via quaternary shifts between tense and relaxed (R) conformations. In contrast, (IDPs) derive functional versatility from high conformational , with unfolded ensembles spanning diverse angles that oppose folding but enable rapid binding; losses upon disorder-to-order transitions can drive specificity in signaling. Recent advances in 2025 have illuminated dynamics through atomic-level simulations, revealing how modulates allosteric networks and channel gating on μs–ms scales, as seen in mechanosensors where membrane tension induces cooperative subunit rearrangements.

References

  1. [1]
    What are proteins and what do they do?: MedlinePlus Genetics
    Mar 26, 2021 · Proteins are complex molecules and do most of the work in cells. They are important to the structure, function, and regulation of the body.
  2. [2]
    Protein Structure | Learn Science at Scitable - Nature
    Proteins are the end products of the decoding process that starts with the information in cellular DNA. As workhorses of the cell, proteins compose structural ...
  3. [3]
    Physiology, Proteins - StatPearls - NCBI Bookshelf
    A protein is a chain of amino acids bound to one another via peptide bonds. Like a string of beads, these strings can twist and fold into a final protein shape.
  4. [4]
    Origin of the Word 'Protein' | Nature
    I claimed for Berzelius the authorship of the word 'protein'. This claim was based on the following passages in a letter to Mulder, the Dutch chemist.
  5. [5]
    Jöns Jacob Berzelius – A Father of Chemistry
    Recognizing the importance of this observation, Berzelius suggested the term “protein,” which is derived from the Greek adjective πρώτειος, meaning “primary” ...
  6. [6]
    [PDF] by Devin Molnau A creative component submitted to the graduate ...
    Proteins were first described by Gerardus Johannes Mulder in 1838 and named by Jons Jacob Berzelius. DNA was not discovered until 1869 by the young Swiss ...
  7. [7]
    The power of proteins – a brief history - VUMC News
    The term “protein” goes back to 1838, when Swedish chemist Jöns Berzelius coined it from the Greek proteios (primary) to emphasize the importance of this group ...
  8. [8]
    Chronology - Historical Developments - Biological Sciences
    Nov 24, 2005 · Gerardus Johannes Mulder carried out the first systematic studies of proteins. Mulder coined the term "protein". 1839: Jean-Baptiste-Joseph ...
  9. [9]
    Biochemistry, Primary Protein Structure - StatPearls - NCBI Bookshelf
    Oct 31, 2022 · To reiterate, the primary structure of a protein is defined as the sequence of amino acids linked together to form a polypeptide chain.
  10. [10]
    Biochemistry, Essential Amino Acids - StatPearls - NCBI Bookshelf
    Apr 30, 2024 · The 20 amino acids that comprise proteins include alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, ...Introduction · Fundamentals · Molecular Level · Mechanism
  11. [11]
    Peptides and Proteins - MSU chemistry
    The different amino acids that make up a peptide or protein, and the order in which they are joined together by peptide bonds is referred to as the primary ...
  12. [12]
    Biochemistry, Peptide - StatPearls - NCBI Bookshelf - NIH
    A peptide is a short string of 2 to 50 amino acids, formed by a condensation reaction, joining together through a covalent bond.Introduction · Fundamentals · Cellular Level · Pathophysiology
  13. [13]
    On 'A method for the determination of amino acid sequence in ... - NIH
    Edman degradation, the first method to determine the amino acid sequence of a peptide, was published in 1949 in the Archives of Biochemistry.
  14. [14]
    II. Basic Elements of Protein Structure - Kinemage
    Disulfide bridges are, of course, true covalent bonds (between the sulfurs of two cysteine side chains) and are thus considered part of the primary structure ...
  15. [15]
    Protein domain identification methods and online resources - PMC
    Protein domains are the fundamental units of protein structure, folding, function, evolution and design. They are considered to be homologous portions of ...
  16. [16]
    Evolutionary history and functional implications of protein domains ...
    Protein domains are the basic building blocks that determine the structure and function of proteins, and they may be considered the units of protein evolution.
  17. [17]
    The structure of a typical antibody molecule - Immunobiology - NCBI
    Each domain is constructed from two β sheets, which are elements of protein structure made up of strands of the polypeptide chain (β strands) packed together; ...3-2. Immunoglobulin heavy... · 3-3. The antibody molecule...
  18. [18]
    The Pfam Protein Families Database - PMC - NIH
    Pfam is a database of protein domain families. Pfam contains curated multiple sequence alignments for each family, as well as profile hidden Markov models.Missing: identification | Show results with:identification
  19. [19]
    Chapter 2: Protein Structure - Chemistry - Western Oregon University
    Folding is a spontaneous process that is mainly guided by hydrophobic interactions, formation of intramolecular hydrogen bonds, van der Waals forces, and it is ...
  20. [20]
    [PDF] Proteins Are Polymers that Fold into Specific Structures
    Folded proteins fall into three classes: globular, fibrous, and membrane proteins. Globular proteins are compact and roughly spherical, with axial ratios ...
  21. [21]
    The Shape and Structure of Proteins - Molecular Biology of the Cell
    Proteins are therefore also known as polypeptides. Each type of protein has a unique sequence of amino acids, exactly the same from one molecule to the next.
  22. [22]
    CHEM 440 - Lysozyme
    Nov 13, 2016 · Further features of the lysozyme mechanism. Lysozyme, a 14.6-kD globular protein discovered by Alexander Fleming in 1922, provides a useful ...
  23. [23]
    Biochemistry, G Protein Coupled Receptors - StatPearls - NCBI - NIH
    GPCRs are integral membrane proteins containing an extracellular amino terminus, seven transmembrane α-helical domains, and an intracellular carboxy terminus.
  24. [24]
    A structural perspective on G protein‐coupled receptors - PMC
    GPCRs are integral membrane proteins that reside in the plasma membrane of eukaryotic cells. They are central to transmitting signals from the extracellular ...
  25. [25]
    Lecture 19. Biological membranes
    Apr 4, 2019 · Further examples of integral membrane proteins illustrate multiple membrane-spanning secondary structure segments of the polypeptide chain.
  26. [26]
    Intrinsically Disordered Proteins: An Overview - PubMed Central - NIH
    Nov 14, 2022 · The naturally occurring, biologically active proteins that appear to possess a high degree of conformational flexibility have been referred to ...
  27. [27]
    Intrinsically disordered tau protein in Alzheimer's tangles - PubMed
    Tau protein, the major constituent of neurofibrillary tangles in Alzheimer's disease (AD) and related tauopathies, is classified as intrinsically disordered ...
  28. [28]
    Functional Evolution of Proteins - PMC - NIH
    The functional evolution of proteins advances through gene duplication followed by functional drift, whereas molecular evolution occurs through random ...<|control11|><|separator|>
  29. [29]
    The status of the human gene catalogue - PubMed - NIH
    Oct 4, 2023 · In the years since, much progress has been made in identifying protein-coding genes, currently estimated to number fewer than 20,000, with an ...
  30. [30]
    Protein Targeting and Transport as a Necessary Consequence of ...
    In this article, we aim to summarize the current knowledge and concepts of the evolutionary development of protein trafficking as a necessary premise and ...
  31. [31]
    Cytosolic Ca2+ Buffers - PMC - PubMed Central - NIH
    Examples of Ca2+ buffers include parvalbumins (α and β isoforms), calbindin-D9k, calbindin-D28k, and calretinin. Besides their proven Ca2+ buffer function, some ...
  32. [32]
    [PDF] 2.8.1 Introduction - U.OSU
    Proteins synthesized in the cytosol can be divided into two general classes with regard to localization: those that are not associated with membranes; ...
  33. [33]
    Mitochondrial Respiratory Complex I: Structure, Function and ...
    These complexes are known as NADH: ubiquinone oxidoreductase (complex I), succinate dehydrogenase (complex II), ubiquinol–cytochrome c oxidoreductase (complex ...
  34. [34]
    Nuclear import of histones - PMC - PubMed Central - NIH
    Dec 10, 2020 · Nuclear import of histones is mediated by proteins of the karyopherin family of nuclear transport receptors. Karyopherins recognize their ...
  35. [35]
    The essential functions of molecular chaperones and folding ...
    The first ER chaperone discovered was the Ig heavy chain binding protein, or Binding Protein (BiP), which associates with incompletely assembled antibody ...
  36. [36]
    Seven-Transmembrane Receptors and Ubiquitination - PMC
    Examples of 7TMRs expressed in the heart, which play prominent roles in cardiac function include those that respond to epinephrine and norepinephrine (α and β ...
  37. [37]
    Current Methods for Identifying Plasma Membrane Proteins as ...
    Apr 5, 2023 · Human epidermal growth factor receptor 2 (HER2) [115,116,117,118,119,120,121,122] and epidermal growth factor receptor (EGFR) [123,124,125,126] ...
  38. [38]
    Membrane Proteins: Structure, Function and Motion - PMC - NIH
    An example is olfactory receptors (ORs) in vertebrate animals. ORs are members of the G protein-coupled receptor (GPCR) family.
  39. [39]
    SEPDB: a database of secreted proteins - PMC - NIH
    Feb 12, 2024 · (ii) Secretory proteins come in a variety of forms, including antibodies, digestive enzymes, exosomes and metabolic molecules. Navigating this ...
  40. [40]
    Cysteine proteases as digestive enzymes in parasitic helminths - PMC
    Aug 23, 2018 · We briefly review cysteine proteases (orthologs of mammalian cathepsins B, L, F, and C) that are expressed in flatworm and nematode parasites.
  41. [41]
    Signal Peptides: From Molecular Mechanisms to Applications in ...
    Jun 18, 2025 · Signal peptides (SPs) are short amino acid sequences located at the N-terminus of nascent proteins and are widely present across various life forms.
  42. [42]
    Types of nuclear localization signals and mechanisms of protein ...
    May 22, 2021 · Nuclear localization signals (NLS) are generally short peptides that act as a signal fragment that mediates the transport of proteins from the cytoplasm into ...
  43. [43]
    From RNA to Protein - Molecular Biology of the Cell - NCBI Bookshelf
    The initiation and termination of translation occur through variations on the translation elongation cycle described above. The site at which protein ...
  44. [44]
    RNA polymerase II transcription initiation: A structural view - PNAS
    In eukaryotes, RNA polymerase II transcribes messenger RNAs and several small nuclear RNAs. Like RNA polymerases I and III, polymerase II cannot act alone.<|separator|>
  45. [45]
    Eukaryotic core promoters and the functional basis of transcription ...
    Enhancers bind regulatory proteins known as transcription factors and recruit transcription cofactors (reviewed in REFS,), and can increase transcription from a ...
  46. [46]
    Genetics, Inducible Operon - StatPearls - NCBI Bookshelf
    Oct 17, 2022 · The lac operon ensures the sequential utilization of different carbon sources depending on the bacterial cells' nutritional environment. ...
  47. [47]
    The Information in DNA Determines Cellular Function via Translation
    Because there are only 20 different amino acids but 64 possible codons, most amino acids are indicated by more than one codon.
  48. [48]
    A Two-Step Chemical Mechanism for Ribosome-Catalyzed Peptide ...
    A peptide bond forms and the ester bond linking the peptide to the 3′-oxygen of peptidyl-tRNA breaks, leaving a deacylated tRNA and a peptide lengthened by one ...Figure 1 · Figure 3 · Methods
  49. [49]
    Enhancer–promoter specificity in gene transcription - Nature
    Apr 25, 2024 · In this review, we provide an overview of recent progress in the eukaryotic gene transcription field pertaining to enhancer–promoter specificity.
  50. [50]
    Regulation of mRNA translation and stability by microRNAs - PubMed
    MicroRNAs (miRNAs) regulate gene expression by base-pairing to mRNA, repressing protein synthesis and causing mRNA decay.
  51. [51]
    Introduction: Posttranslational Protein Modification | Chemical Reviews
    Feb 14, 2018 · At the same time, existing proteins can undergo certain chemical modifications, commonly referred to as protein post-translational modifications ...Author Information · Biographies<|control11|><|separator|>
  52. [52]
    Solid Phase Peptide Synthesis. I. The Synthesis of a Tetrapeptide
    R. B. Merrifield. ACS Legacy Archive. Open PDF. Journal of the American Chemical Society. Cite this: J. Am. Chem. Soc. 1963, 85, 14, 2149–2154. Click to copy ...
  53. [53]
    Challenges and Perspectives in Chemical Synthesis of Highly ... - NIH
    Mar 4, 2020 · Thirteen decades of peptide synthesis: key developments in solid phase peptide synthesis and amide bond formation utilized in peptide ligation.
  54. [54]
    Use of bacteriophage T7 RNA polymerase to direct selective high ...
    A gene expression system based on bacteriophage T7 RNA polymerase has been developed. T7 RNA polymerase is highly selective for its own promoters.
  55. [55]
    Recombinant protein expression in Escherichia coli: advances and ...
    Apr 16, 2014 · We review the different approaches for the synthesis of recombinant proteins in E. coli and discuss recent progress in this ever-growing field.
  56. [56]
    Genetic Approach to Facilitate Purification of Recombinant Proteins ...
    Nov 1, 1988 · We describe a general purification method for recombinant proteins based upon the selective interaction between a poly-histidine peptide, which is fused to the ...
  57. [57]
    Cell-free gene expression | Nature Reviews Methods Primers
    Jul 15, 2021 · Cell-free gene expression (CFE) emerged as an alternative approach to living cells for specific applications in protein synthesis and labelling.
  58. [58]
    Codon Optimization Using a Recurrent Neural Network
    Jun 21, 2022 · Codon optimization of a DNA sequence can significantly increase efficiency of protein expression, reducing the cost to manufacture biologic ...<|control11|><|separator|>
  59. [59]
    Enzymes: principles and biotechnological applications - PMC
    The simple answer is that it acts to stabilize the active site and provide an appropriate environment for interaction of the site with the substrate molecule.Missing: seminal | Show results with:seminal
  60. [60]
    Looking Back: A Short History of the Discovery of Enzymes and How ...
    ... enzymes worked. Through the famous lock‐and‐key model, proposed by Emil Fischer in 1894, as well as the Michaelis‐Menten model of enzyme kinetics ...
  61. [61]
    Lock-Key Model - an overview | ScienceDirect Topics
    The textbook lock-and-key model for enzyme catalysis was first introduced by the Nobel laureate organic chemist Emil Fischer in 1894 (Fischer, 1894). The ...
  62. [62]
    Translation of the 1913 Michaelis-Menten Paper - PMC - NIH
    Nearly 100 years ago Michaelis and Menten published their now classic paper (Michaelis, L., and Menten, M. L. (1913) Die Kinetik der Invertinwirkung, ...
  63. [63]
    Enzyme Nomenclature
    Oct 20, 2025 · In consultation with the IUPAC-IUBMB Joint Commission ... The common names of all listed enzymes are listed below, along with their EC numbers.
  64. [64]
    [PDF] Vitamins, Cofactors, Coenzymes and Prosthetic Groups - vscht.cz
    NAD+ or NADP+ are typical coenzymes of oxidoreductases (interestingly, it can be a prosthetic group of a hydrolase – S-adenosylhomocystein hydrolase). Niacin is ...
  65. [65]
    ON THE NATURE OF ALLOSTERIC TRANSITIONS: A PLAUSIBLE ...
    ON THE NATURE OF ALLOSTERIC TRANSITIONS: A PLAUSIBLE MODEL. J Mol Biol. 1965 May:12:88-118. doi: 10.1016/s0022-2836(65)80285-6.Missing: Jacques regulation
  66. [66]
    Physiology, Noncompetitive Inhibitor - StatPearls - NCBI Bookshelf
    Sep 12, 2022 · Noncompetitive inhibition, a type of allosteric regulation, is a specific type of enzyme inhibition characterized by an inhibitor binding to an allosteric site.
  67. [67]
    Ion Channels and the Electrical Properties of Membranes - NCBI - NIH
    Vertebrate neurons, for example, have acetylcholine-gated ion channels that differ from those of muscle cells in that they are usually formed from two subunits ...
  68. [68]
    G Protein-Coupled Receptors: A Century of Research and Discovery
    Mar 28, 2025 · G protein-coupled receptors (GPCRs), also known as 7 transmembrane domain receptors, are the largest receptor family in the human genome, ...Missing: seminal | Show results with:seminal
  69. [69]
    GPCR | Learn Science at Scitable - Nature
    GPCRs are a large family of cell surface receptors that respond to a variety of external signals. Binding of a signaling molecule to a GPCR results in G protein ...Missing: seminal | Show results with:seminal
  70. [70]
    Signal transduction pathway | Cell signaling (article) - Khan Academy
    Although proteins are important in signal transduction pathways, other types of molecules can participate as well. Many pathways involve second messengers, ...Missing: seminal | Show results with:seminal
  71. [71]
    Phosphorylation-independent desensitization of G protein ... - PubMed
    The activity of GPCRs is subject to three principal modes of regulation: desensitization, trafficking, and down-regulation. Desensitization is defined as a loss ...
  72. [72]
    GPCR Signaling Regulation: The Role of GRKs and Arrestins
    Here we focus on the mechanisms of GRK- and arrestin-mediated regulation of GPCR signaling, which includes homologous desensitization and redirection of ...
  73. [73]
    The complement system and innate immunity - Immunobiology - NCBI
    The complement system is one of the major mechanisms by which pathogen recognition is converted into an effective host defense against initial infection.2-5. Complement is a system... · 2-11. Phagocyte ingestion of...
  74. [74]
    Type I interferons in infectious disease | Nature Reviews Immunology
    Jan 23, 2015 · Type I interferons (IFNs) have diverse effects on innate and adaptive immune cells during infection with viruses, bacteria, parasites and fungi.
  75. [75]
    Major Histocompatibility Complex (MHC) Class I and MHC Class II ...
    Mar 17, 2017 · Antigen presentation by major histocompatibility complex (MHC) proteins is essential for adaptive immunity. Prior to presentation, peptides need ...Missing: source | Show results with:source
  76. [76]
    antigen processing and presentation by the class i major ...
    Apr 1, 1996 · Major histocompatibility complex (MHC) class I molecules bind peptides derived from cellular proteins and display them for surveillance by the immune system.
  77. [77]
    Transcriptional Regulation by P53 - PMC - PubMed Central - NIH
    The p53 transcription factor regulates several hundred genes. It tailors responses to stimuli by employing various different cofactors, posttranslational ...
  78. [78]
    Targeted protein degradation: mechanisms, strategies and application
    Apr 4, 2022 · Protein degradation via the ubiquitin-proteasome system (UPS). Proteins undergo ubiquitin-dependent degradation by a suite of three enzymes.
  79. [79]
    Heat Shock Response and Heat Shock Proteins - PubMed Central
    This review highlights the significant features and functions of various HSPs in cellular homeostasis and human diseases, with a primary focus on the structure ...Missing: source | Show results with:source
  80. [80]
    Apoptosis: A Review of Programmed Cell Death - PMC
    This proteolytic cascade, in which one caspase can activate other caspases, amplifies the apoptotic signaling pathway and thus leads to rapid cell death.
  81. [81]
    Growth Hormone(s), Testosterone, Insulin-Like Growth Factors, and ...
    Hormones are largely responsible for the integrated communication of several physiological systems responsible for modulating cellular growth and development.
  82. [82]
    Review - The Evolution of Adaptive Immune Systems - Cell Press
    May 24, 2005 · Very recently, we have learned that vertebrates without jaws (agnathans) also have an adaptive immune system that is based on recombinatorial ...
  83. [83]
    The origins of vertebrate adaptive immunity - PMC - PubMed Central
    Adaptive immunity is mediated through numerous genetic and cellular processes that generate favourable somatic variants of antigen-binding receptors.
  84. [84]
    Proteome Turnover in the Spotlight: Approaches, Applications, and ...
    At a steady state, protein turnover is constant to maintain protein homeostasis, but in dynamic responses, proteins change their rates of synthesis and ...
  85. [85]
    Determinants and Regulation of Protein Turnover in Yeast
    Sep 27, 2017 · Protein turnover maintains the recycling needs of the proteome, and its malfunction has been linked to aging and age-related diseases.
  86. [86]
    Ubiquitin-like protein conjugation and the ubiquitin–proteasome ...
    Dec 10, 2010 · Ubiquitin is a highly conserved 76 amino-acid protein that covalently attaches to protein substrates targeted for degradation by the 26S ...
  87. [87]
    The ubiquitin system: from cell signalling to disease biology and ...
    Jan 14, 2021 · Forty years ago, ubiquitin was first discovered as a post-translational modification earmarking proteins for degradation [1].
  88. [88]
    Lysosome biology in autophagy | Cell Discovery - Nature
    Feb 11, 2020 · Autophagy is a major intracellular degradation system that derives its degradative abilities from the lysosome. The most well-studied form ...
  89. [89]
    Lysosomal microautophagy: an emerging dimension in mammalian ...
    Dec 15, 2023 · Autophagy is a self-catabolic process through which cellular components are delivered to lysosomes for degradation.
  90. [90]
    In Vivo Half-Life of a Protein Is a Function of Its Amino-Terminal ...
    The function of the previously described posttranslational addition of single amino acids to protein amino-termini may also be accounted for by the N-end rule.
  91. [91]
    The N-end rule pathway of protein degradation - PubMed
    The N-end rule relates the in vivo half-life of a protein to the identity of its N-terminal residue. Similar but distinct versions of the N-end rule operate ...
  92. [92]
    The N-end rule - PNAS
    Contributed by Alexander Varshavsky, August 6, 1996. ABSTRACT. The N-end rule relates the in vivo half-life of a protein to the identity of its N-terminal ...
  93. [93]
    Protein Quality Control and Metabolism - NIH
    Protein quality control is vital to cellular homeostasis and integrity. Loss of protein homeostasis leads to accumulation of misfolded proteins and protein ...
  94. [94]
    Autophagy: for better or for worse | Cell Research - Nature
    Sep 13, 2011 · Autophagy is a lysosomal degradation pathway that degrades damaged or superfluous cell components into basic biomolecules, which are then ...
  95. [95]
    Alpha-synuclein in Parkinson's disease and other synucleinopathies
    Mar 1, 2023 · Abnormal forms of α-syn trigger selective and progressive neuronal death through mitochondrial impairment, lysosomal dysfunction, and alteration of calcium ...
  96. [96]
    Expanding the role of proteasome homeostasis in Parkinson's disease
    Feb 4, 2021 · The compromise of proteasomal degradation leads to aberrant protein aggregates, such as α-synuclein, which in turn binds to proteasome ...
  97. [97]
    Alpha-Synuclein defects autophagy by impairing SNAP29-mediated ...
    Sep 17, 2021 · We found α-Syn overexpression to reduce autophagy turnover by compromising the fusion of autophagosomes with lysosomes, thus leading to a decrease in the ...
  98. [98]
    Physiology, Pepsin - StatPearls - NCBI Bookshelf - NIH
    1 Mei 2023 · As mentioned earlier, the stomach provides pepsin with an ideal environment for protein digestion. ... pH of the stomach. A low pH (1.5 to ...
  99. [99]
    Pepsinogens and Pepsins
    Optimal activity of pepsins is at pH of 1.8 to 3.5, depending on the isoform. They are reversibly inactivated at about pH 5 and irreversibly inactivated at pH 7 ...
  100. [100]
    Physiology, Digestion - StatPearls - NCBI Bookshelf
    Sep 12, 2022 · The pancreas produces many digestive enzymes, including pancreatic amylase, pancreatic lipase, trypsinogen, chymotrypsinogen, ...
  101. [101]
    Exocrine Secretions of the Pancreas
    Trypsin and chymotrypsin digest proteins into peptides and peptides into smaller peptides, but they cannot digest proteins and peptides to single amino acids.
  102. [102]
    Protein Digestion and Absorption – Nutrition - VCU Pressbooks
    Protein digestion in the stomach. Because of the hydrochloric acid in the stomach, it has a very low pH of 1.5-3.5. The acidity of the stomach causes food ...
  103. [103]
    Absorption of Amino Acids and Peptides
    Dietary proteins are, with very few exceptions, not absorbed. Rather, they must be digested into amino acids or di- and tripeptides first.Missing: jejunum | Show results with:jejunum
  104. [104]
    Clinical relevance of intestinal peptide uptake - PMC - NIH
    Protein digestion and absorption in humans depends on initial enzymatic hydrolysis in the stomach and proximal small intestine. The hydrolytic products include ...
  105. [105]
    How the Liver Works - University of Rochester Medical Center
    Oxygenated blood flows into the liver through the hepatic artery. Nutrient-rich blood flows into the liver from the intestines through the hepatic portal vein.
  106. [106]
    Physiology, Gastrointestinal Hormonal Control - StatPearls - NCBI
    The five GI hormones that qualify as endocrines are gastrin, cholecystokinin (CCK), secretin, glucose-dependent insulinotropic peptide (GIP), and motilin.
  107. [107]
    Biochemistry, Cholecystokinin - StatPearls - NCBI Bookshelf - NIH
    Mediates digestion by regulating the release of pancreatic exocrine enzymes which plays a role in the digestion of fats, proteins, and carbohydrates. Causes ...
  108. [108]
    Oral proteases: a new approach to managing coeliac disease - PMC
    Incomplete luminal hydrolysis of proline‐rich peptides may thus be compensated by epithelial processing in controls and patients treated for coeliac disease.
  109. [109]
    Amino acids: MedlinePlus Medical Encyclopedia
    Jan 21, 2025 · The 9 essential amino acids are: histidine, isoleucine, leucine, lysine, methionine, phenylalanine, threonine, tryptophan, and valine.
  110. [110]
    l-Lysine supplementation affects dietary protein quality and growth ...
    Nov 15, 2023 · l-Lysine (Lys) is an essential amino acid (EAA) in mammals, and must be obtained from foods or dietary supplements since it cannot be ...
  111. [111]
    A Simple Outline of Methods for Protein Isolation and Purification - NIH
    Methods for purifying target proteins from dirty mixtures vary widely, but preparation-grade purification is most commonly achieved using chromatography [1,2].
  112. [112]
    Protein production and purification - PMC - NIH
    This review presents methods that could be applied at the outset of any project, a prioritized list of alternate strategies and a list of pitfalls that trip ...
  113. [113]
    Challenges and solutions for the downstream purification of ... - NIH
    In this review, we outline the purification strategies in order to obtain products of high purity, highlighting the corresponding solutions to circumvent the ...
  114. [114]
    1Progress, applications, challenges and prospects of protein ...
    Dec 6, 2022 · Conventional protein purification generally includes three steps: pretreatment, rough fractionation, and fine fractionation. Each of the steps ...
  115. [115]
    Density Gradient Ultracentrifugation to Isolate Endogenous Protein ...
    Jul 1, 2016 · This protocol describes the isolation of native protein complexes by density gradient ultracentrifugation.
  116. [116]
    Selective enzyme purification by affinity chromatography. - PNAS
    Selective enzyme purification by affinity chromatography. P Cuatrecasas, M Wilchek, and C B AnfinsenAuthors Info & Affiliations. October 15, 1968.
  117. [117]
    Isoelectric Point Separations of Peptides and Proteins - PMC - NIH
    Jan 25, 2017 · One such electrophoretic technique is isoelectric focusing (IEF) which provides separation of ampholytic components, molecules that act as ...1. Introduction · 2. 2de And Ipg Strips · 4. Applications Of...
  118. [118]
    High resolution two-dimensional electrophoresis of proteins - PubMed
    A technique has been developed for the separation of proteins by two-dimensional polyacrylamide gel electrophoresis.
  119. [119]
    A rapid and sensitive method for the quantitation of ... - PubMed
    A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. ... Author. M M Bradford.
  120. [120]
    Measurement of protein using bicinchoninic acid - ScienceDirect.com
    This reagent forms the basis of an analytical method capable of monitoring cuprous ion produced in the reaction of protein with alkaline Cu2+ (biuret reaction).
  121. [121]
    Quality assessment and optimization of purified protein samples
    In this review, we aim at proposing a simple-to-follow workflow based on an ensemble of widely available physico-chemical technologies, to assess sequentially ...
  122. [122]
    Challenges and opportunities in the purification of recombinant ...
    The purification of recombinant proteins by affinity chromatography is one of the most efficient strategies due to the high recovery yields and purity achieved.
  123. [123]
    Homology modeling in the time of collective and artificial intelligence
    Homology modeling is a method for building protein 3D structures using protein primary sequence and utilizing prior knowledge gained from structural ...
  124. [124]
    Highly accurate protein structure prediction with AlphaFold - Nature
    Jul 15, 2021 · Here we provide the first computational method that can regularly predict protein structures with atomic accuracy even in cases in which no similar structure ...
  125. [125]
    [PDF] Computational Protein Design and Protein Structure Prediction
    Oct 9, 2024 · Hassabis and John Jumper the Nobel Prize in Chemistry 2024, for computational protein design and protein structure prediction. Introduction.
  126. [126]
    Accurate structure prediction of biomolecular interactions ... - Nature
    May 8, 2024 · The introduction of AlphaFold 2 has spurred a revolution in modelling the structure of proteins and their interactions, enabling a huge ...Nature Machine Intelligence · Nobel Prize in Chemistry 2024
  127. [127]
    Evolutionary-scale prediction of atomic-level protein structure with a ...
    Mar 16, 2023 · We demonstrate direct inference of full atomic-level protein structure from primary sequence using a large language model.
  128. [128]
    Accurate prediction of protein structures and interactions using a ...
    Aug 19, 2021 · We investigated the utility of the RoseTTAFold to facilitate experimental structure determination by x-ray crystallography and cryo–electron ...
  129. [129]
    Design of a Novel Globular Protein Fold with Atomic-Level Accuracy
    Top7 was found experimentally to be folded and extremely stable, and the x-ray crystal structure of Top7 is similar (root mean square deviation equals 1.2 ...
  130. [130]
    De novo design of protein structure and function with RFdiffusion
    Jul 11, 2023 · We obtain a generative model of protein backbones that achieves outstanding performance on unconditional and topology-constrained protein monomer design.
  131. [131]
    AlphaFold2 and its applications in the fields of biology and medicine
    Mar 14, 2023 · AlphaFold2 (AF2) is an artificial intelligence (AI) system developed by DeepMind that can predict three-dimensional (3D) structures of proteins from amino acid ...
  132. [132]
    protein dynamic conformations modeling in the post-AlphaFold era
    Jul 15, 2025 · This review outlines the fundamental concepts of protein dynamic conformations, surveys recent computational advances in modeling these dynamics ...
  133. [133]
    The power and pitfalls of AlphaFold2 for structure prediction beyond ...
    IDPs represents a challenge for AF2 since it is difficult to identify evolutionary constraints from MSAs of IDPs and IDRs due to sequence hypervariability.
  134. [134]
    Artificial intelligence-based methods for protein structure prediction
    Jul 31, 2025 · Although AlphaFold2 can predict transmembrane regions of membrane proteins, it faces major challenges in modeling full-length membrane ...
  135. [135]
    How AI Is Transforming De Novo Protein Design in Drug Development
    Apr 15, 2025 · AI is driving a fundamental transformation in drug development by shifting from the prediction of natural proteins to the design of entirely new ones.Early Milestones And Missed... · What Ai Is Making Possible... · Ongoing Developments To...
  136. [136]
    Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS)
    Jun 11, 2020 · LC-MS/MS-based proteomics is a powerful tool for identifying and quantifying proteins in biological samples, outperforming conventional antibody-based methods ...
  137. [137]
    Liquid Chromatography Mass Spectrometry-Based Proteomics - NIH
    We present an overview of bottom-up proteomics and outline the key statistical issues that arise in protein identification and quantification.
  138. [138]
    Status of Large-scale Analysis of Post-translational Modifications by ...
    PTM Analysis via MS. Mass spectrometry has long been used to map modifications on purified proteins; in fact, this was one of the first applications of MS in ...
  139. [139]
    Characterization of Phosphorylated Proteins Using Mass Spectrometry
    In this article, we describe MS methods for characterizing phosphorylation sites within individual proteins as well as entire proteome samples.
  140. [140]
    Recent advances in quantitative and chemical proteomics for ...
    SILAC is often considered the “gold standard” of isotope labeling methods as labeling and sample mixing are performed at the earliest possible steps, ...
  141. [141]
    iTRAQ-Based and Label-Free Proteomics Approaches for Studies of ...
    Both isobaric tags for relative and absolute quantitation (iTRAQ) and label-free methods are widely used for quantitative proteomics.
  142. [142]
    Protein-Protein Interaction Detection: Methods and Analysis - NIH
    The in vivo methods in PPI detection are yeast two-hybrid (Y2H, Y3H) and synthetic lethality. In silico techniques are performed on a computer (or) via ...Missing: interactomics | Show results with:interactomics
  143. [143]
    Mass spectrometry‐based protein–protein interaction networks ... - NIH
    Jan 12, 2021 · This review discusses mass spectrometry techniques that have been instrumental for identifying protein‐protein interactions.
  144. [144]
    The STRING database in 2023: protein-protein association networks ...
    Jan 6, 2023 · The STRING database (https://string-db.org/) systematically collects and integrates protein-protein interactions-both physical interactions as well as ...
  145. [145]
    Proteomics Discovery of Disease Biomarkers - PMC - NIH
    Recent technological developments in proteomics have shown promising initiatives in identifying novel biomarkers of various diseases.
  146. [146]
    Applications of targeted proteomics in systems biology and ...
    Targeted proteomics can be used to validate biomarkers ... disease applications with many putative biomarkers such as cardiovascular disease and cancer.
  147. [147]
    Proteomics and Systems Biology: Current and Future Applications in ...
    This review will define systems biology, outline current proteomic methodologies, highlight successful applications of proteomics in nutrition research,
  148. [148]
    Deep Visual Proteomics maps proteotoxicity in a genetic liver disease
    Apr 16, 2025 · We use spatial proteomics by mass spectrometry and machine learning to map AATD in human liver tissue. Combining Deep Visual Proteomics (DVP) with single-cell ...<|control11|><|separator|>
  149. [149]
    Post-translational modification prediction via prompt-based fine ...
    Aug 7, 2024 · Their method involves intricate neural network architectures optimized for analyzing protein sequences, signifying a refined integration of deep ...
  150. [150]
    The micro-mechanics of single molecules studied with atomic force ...
    Measurement of force-induced elongation with the AFM can determine the mechanical properties of proteins and polysaccharides with exquisite sensitivity.
  151. [151]
    Atomic force microscopy reveals the mechanical design of a ... - PNAS
    We examine the mechanical properties of the resulting tandem modular proteins by using single protein atomic force microscopy.
  152. [152]
    Protein fibers with self-recoverable mechanical properties via ...
    Sep 2, 2023 · The mechanical properties of as-spun and post-stretched DIF fibers were systematically evaluated regarding ultimate tensile strength, Young's ...
  153. [153]
    A Look through the Perspective of Molecular Viscoelastic Properties
    Feb 1, 2013 · The viscoelastic transition is the starting point of our analysis. We consider the “stress-strain” diagram (actually a force-deformation diagram) ...
  154. [154]
    Stepwise unfolding of titin under force-clamp atomic force microscopy
    As shown, stepwise unfolding events occur over a narrow range of forces (in this case ≈170–250 pN) and a much shorter time (≈2 s) than those observed under ...
  155. [155]
    Chemistry and Enzymology of Disulfide Cross-Linking in Proteins
    In uPAR, disulfide bonds affix loops to the β-strands in the three copies of the β-sheet fold that constitute the receptor. In phospholipase A2, disulfides ...
  156. [156]
    The elaborate structure of spider silk - NIH
    Spider silk is one outstanding fibrous biomaterial which consists almost entirely of large proteins. Silk fibers have tensile strengths comparable to steel.
  157. [157]
    Mechanical Properties of Collagen Fibrils: Biophysical Journal
    Determining the mechanical properties of single collagen fibrils (diameter 50–200nm) provides new insights into collagen structure. In this work, the reduced ...
  158. [158]
    Bioinspired and biomimetic protein-based fibers and their applications
    Apr 18, 2024 · Protein-based fibers combine unique mechanical properties with biocompatibility and biodegradability, and often outperform polymer-based fibers.
  159. [159]
    Engineering Applications of Biomolecular Motors - Annual Reviews
    Aug 15, 2011 · Biomolecular motors, in particular motor proteins from the kinesin and myosin families, can be used to explore engineering applications of ...
  160. [160]
    Mega-scale experimental analysis of protein folding stability ... - Nature
    Jul 19, 2023 · To infer each sequence's thermodynamic folding stability (for unfolding at pH 7.4 and 298 K (room temperature); hereafter referred to as ΔG), ...Missing: seminal | Show results with:seminal
  161. [161]
    Protein Stability Analysis of Heat and Cold Denaturation without ...
    Apr 11, 2023 · Here we demonstrate a straightforward model-independent evaluation of heat capacity experiments in terms of protein unfolding enthalpy ΔH(T), entropy ΔS(T), ...
  162. [162]
    Differential Scanning Calorimetry — A Method for Assessing the ...
    Differential scanning calorimetry (DSC) is an analytical technique that measures the molar heat capacity of samples as a function of temperature.
  163. [163]
    Thermodynamic stability and folding of proteins from ... - FEBS Press
    Aug 3, 2007 · Here, we summarize the current data on thermodynamic and kinetic folding/unfolding behaviors of proteins from hyperthermophilic microorganisms.Introduction · Nonheme Iron Proteins · Dna-Binding ProteinsMissing: seminal | Show results with:seminal
  164. [164]
    Using circular dichroism spectra to estimate protein secondary ... - NIH
    Circular dichroism (CD) is an excellent method for rapidly evaluating the secondary structure, folding and binding properties of proteins. Briefly, circular ...
  165. [165]
    Tryptophan Fluorescence Quenching Assays for Measuring Protein ...
    Jun 5, 2019 · Tryptophan fluorescence quenching is a binding assay where tryptophan fluorescence is quenched by changes in environment polarity upon ligand ...
  166. [166]
    Connecting Protein Millisecond Conformational Dynamics to ... - NIH
    Protein conformational fluctuations span a vast range of time scales, from picoseconds to minutes, with microsecond to millisecond (μs–ms) dynamics being ...Missing: residence | Show results with:residence
  167. [167]
    Applications of Molecular Dynamics Simulation in Protein Study - NIH
    MD Simulations has been used to study protein function, protein stability, protein–protein interaction, enzymatic reactions and drug–protein interactions, and ...
  168. [168]
    Half a Century of Hemoglobin's Allostery - PMC - PubMed Central
    The model postulates that the hemoglobin tetramer populates two conformational states, different in quaternary and tertiary structure, called R for relaxed and ...
  169. [169]
    Conformational Entropy of Intrinsically Disordered Proteins ... - Nature
    Jul 3, 2015 · This work quantitatively characterizes intrinsic disorder in proteins in terms of sequence composition and backbone conformational entropy.
  170. [170]
    The lipid bilayer strengthens the cooperative network of membrane ...
    Jul 2, 2025 · This study demonstrates the pivotal role of lipid solvation in modulating the stability of membrane proteins and their responses to external stimuli.