Fact-checked by Grok 2 weeks ago

Proteolysis

Proteolysis is the enzymatic of peptide bonds in proteins, resulting in their breakdown into smaller or individual , a process mediated by proteases that serves as a primary for protein and turnover in cells. This irreversible is ubiquitous across all living organisms and plays a crucial role in maintaining cellular by eliminating damaged, misfolded, or unnecessary proteins, thereby preventing their accumulation and associated . In humans, proteolysis involves over 600 proteases, classified into five major classes—aspartic, , metallo, serine, and —each employing distinct catalytic s to cleave specific peptide bonds with varying degrees of specificity. Beyond basic degradation, proteolysis regulates a wide array of biological processes, including progression, , , immune responses, and tissue remodeling, often through targeted activation or inactivation of key regulatory proteins. For instance, in eukaryotic cells, the ubiquitin-proteasome system dominates regulated proteolysis, where proteins are first tagged with chains by a cascade of E1, E2, and E3 enzymes before being degraded by the 26S complex, ensuring precise temporal control over protein levels during events like . Dysregulation of proteolysis is implicated in numerous diseases, such as cancer (via uncontrolled oncoprotein accumulation), neurodegenerative disorders (from protein aggregates), and inflammatory conditions, highlighting its therapeutic potential through inhibitors or modulators. In prokaryotes and other organisms, proteolysis also facilitates to environmental stresses and recycling, with energy-dependent AAA+ proteases unfolding and degrading substrates in a process often guided by specific degrons—short sequences that mark proteins for destruction. Examples include the caspase-mediated during , where over 1,700 sites have been identified, and lysosomal cathepsins contributing to and . Overall, proteolysis exemplifies a finely tuned that balances protein synthesis with degradation to sustain life at the molecular level.

Fundamentals

Definition and Overview

Proteolysis is the of proteins into smaller polypeptides or through the cleavage of bonds, a fundamental biochemical process that breaks the linkages between residues. This typically proceeds via a nucleophilic by on the carbonyl carbon of the , forming a tetrahedral and ultimately yielding a and an amine group, often accelerated by biological catalysts. The specificity of cleavage depends on the primary structure of the protein—the linear sequence of connected by these bonds—while secondary structures, such as alpha helices or sheets stabilized by hydrogen bonds, can influence the accessibility and susceptibility of target sites to . The recognition of proteolysis dates to the 19th century, emerging from investigations into digestive processes; in 1836, isolated from gastric juice, identifying it as an agent capable of dissolving proteins and marking an early milestone in understanding enzymatic protein breakdown. Building on such discoveries, proteolysis has since been established as a cornerstone of cellular , primarily mediated by specialized enzymes known as proteases. Biologically, proteolysis is indispensable for upholding cellular , including protein quality control through the removal of misfolded or damaged proteins to prevent aggregation and . It also facilitates nutrient recycling by liberating from degraded proteins, enabling their reuse in , and supports adaptive responses to environmental stresses by rapidly modulating protein levels. These functions ensure efficient and metabolic flexibility across organisms.

Types of Proteolytic Processes

Proteolysis encompasses a range of processes that break down proteins into smaller peptides or , primarily categorized into enzymatic and non-enzymatic types based on the involvement of catalysts. Enzymatic proteolysis is mediated by specialized enzymes known as proteases, which exhibit high specificity determined by the sequences surrounding the cleavage site. In contrast, non-enzymatic proteolysis occurs without enzymatic , relying instead on spontaneous chemical reactions that are often accelerated under extreme conditions. Within enzymatic proteolysis, proteases are subdivided into endopeptidases and exopeptidases according to the site of cleavage. Endopeptidases hydrolyze internal s within the protein chain, typically recognizing specific motifs in the substrate's interior for precise cuts. Exopeptidases, on the other hand, act at the termini of the polypeptide, removing one or a few from either the (aminopeptidases) or (carboxypeptidases), with specificity often based on the terminal residue's . These distinctions allow for targeted processing versus sequential trimming in protein maturation. Enzymatic processes can further be classified as limited or unlimited proteolysis depending on the extent of degradation. Limited proteolysis involves one or a few specific cleavages at defined sites, often to activate or modify a protein without full breakdown, as seen in regulatory cascades. Unlimited proteolysis, by comparison, entails complete degradation into individual or very short peptides through repeated cleavages, typically for recycling or clearance. Non-enzymatic proteolysis proceeds via spontaneous of bonds, which can be induced or enhanced by environmental factors such as extreme , elevated temperatures, or chemical agents, leading to random or less specific cleavages. At high temperatures (e.g., 95°C), the rate of these reactions increases significantly, while acidic or basic shifts promote mechanisms like direct scission or intramolecular aminolysis. The prevalence and nature of these proteolytic types are influenced by cellular conditions, including and , which modulate activity and in enzymatic cases or in non-enzymatic ones. For instance, lysosomal enzymes operate optimally at acidic (~4.5-5.0), while neutral cytosolic environments favor other proteases; deviations can shift toward non-enzymatic pathways. Temperature extremes beyond physiological ranges (~37°C) denature enzymes, thereby promoting spontaneous proteolysis.

Proteolytic Enzymes

Classification and Mechanism

Proteases are classified into six major classes based on the nature of their catalytic residues and the mechanism of hydrolysis: aspartic, , , metallo-, , and proteases. This classification, established by the MEROPS database, reflects the used in —either an activated water molecule (in aspartic, , and metalloproteases) or a specific (in , , and proteases). Aspartic proteases employ two residues to activate water for nucleophilic attack on the carbonyl. proteases utilize the group of a residue as the , often enhanced by a nearby . proteases, rare in eukaryotes, feature a and pair that polarizes the carbonyl. Metalloproteases coordinate a or other metal ion to activate water, facilitating . proteases rely on a serine hydroxyl group, while proteases use a , both typically within a or dyad. A prominent example of catalytic mechanism is found in serine proteases, where the consisting of serine (Ser), (His), and (Asp) residues orchestrates . The acts as a general base, deprotonating the serine hydroxyl to generate a potent that attacks the substrate's carbonyl carbon, forming a tetrahedral intermediate. This intermediate is stabilized by the oxyanion hole, formed by backbone hydrogens (e.g., from Gly193 and Ser195 in numbering), which hydrogen-bond to the negatively charged oxygen. The process proceeds in two stages: , yielding a covalent acyl-enzyme intermediate, and deacylation, where water hydrolyzes the bond to release the product and regenerate the . In the classic Ser-His-Asp triad, the aspartate orients the and stabilizes the positive charge developed during , enhancing the serine's nucleophilicity by up to 10^6-fold. Many proteases are synthesized as inactive zymogens to prevent premature activity, and activation occurs through limited proteolysis that cleaves an N-terminal prosegment, inducing conformational changes to expose the active site. This prosegment, varying from a few residues to over 100, sterically blocks the catalytic residues in the zymogen form and often aids in proper folding. For instance, in trypsinogen, cleavage at an internal site removes the prosegment, repositioning loops to form the oxyanion hole and substrate-binding pockets, thereby activating the enzyme autocatalytically or via another protease. This mechanism ensures spatial and temporal control, as seen in digestive and blood coagulation cascades. Protease activity is modulated by inhibitors that bind the or nearby regions, classified as competitive or non-competitive. Competitive inhibitors, such as canonical serpins or small-molecule analogs, occupy the cleft, mimicking the and preventing binding; for example, bovine pancreatic inhibitor (BPTI) forms a tight complex with trypsin's via its reactive loop. Non-competitive inhibitors bind exosites outside the , inducing allosteric changes that distort the catalytic machinery without directly competing with ; ecotin, for instance, uses a secondary to inhibit multiple serine proteases. These interactions often involve bonds, , or metal coordination, with affinities reaching picomolar levels. Evolutionarily, protease catalytic domains exhibit remarkable conservation across diverse species, reflecting ancient origins in . The Ser-His-Asp triad geometry, including nucleophilic elbow motifs and hole configurations, is preserved in over 23 independent folds, constraining stereochemistry (e.g., carbonyl attack angles near 90°) despite sequence divergence. reveals lineage-specific expansions but core catalytic residues remain invariant, underscoring their essential role in acyl-enzyme formation and efficiency.

Notable Proteases and Sources

Proteases are found across diverse biological sources, exemplifying the wide distribution and specialized adaptations of proteolytic enzymes in nature. These enzymes vary in their catalytic classes and cellular or extracellular locales, contributing to fundamental processes in organisms from to pathogens. In digestive systems, serves as a prominent aspartic protease secreted by gastric chief cells in the lining of vertebrates, where it initiates protein breakdown in the acidic environment of gastric juice. , a produced as the inactive by pancreatic acinar cells in mammals, is activated in the to further degrade dietary proteins into peptides. Intracellular proteases include , a of proteases synthesized as zymogens in the of eukaryotic cells, pivotal in signaling pathways for . The 20S , a cylindrical multicatalytic complex composed of alpha and beta subunits, functions as the core proteolytic component of the ubiquitin-proteasome system within eukaryotic cells, targeting ubiquitinated proteins for degradation. Extracellular proteases encompass matrix metalloproteinases (MMPs), a group of zinc-dependent metalloendopeptidases secreted by various types including fibroblasts and macrophages, essential for remodeling the in tissues during development and repair. Venom-derived proteases, such as batroxobin from the snake, are serine proteases that exhibit effects by specifically cleaving fibrinogen to produce degradation products, aiding in prey immobilization. Microbial proteases include bacterial collagenases, metalloproteinases produced by pathogens like species and , which hydrolyze native in host tissues to facilitate invasion and tissue dissemination during infection. Plant and viral proteases feature , a abundant in the latex of Carica papaya () fruit, where it contributes to defense against herbivores by digesting ingested proteins. In viruses, protease is an aspartic protease encoded by the pol gene of human immunodeficiency virus type 1, responsible for cleaving viral polyproteins into functional mature proteins during particle assembly.

Biological Functions

Protein Maturation and Processing

Protein maturation often requires precise proteolytic processing to convert precursor proteins, such as preproteins or proproteins, into their functional forms, enabling proper folding, localization, and activity. This involves targeted cleavage events that remove inhibitory segments or signal sequences, which is essential for the assembly of mature proteins in cellular compartments like the and Golgi apparatus. In both prokaryotes and eukaryotes, these processes ensure that nascent polypeptides achieve their biologically active conformations without broad degradation. A fundamental step in protein maturation is the removal of the N-terminal residue, which is added during initiation. In prokaryotes, this occurs co-translationally via (MetAP), excising the formylmethionine from most nascent chains to expose the penultimate residue, which is critical for subsequent folding and stability. Eukaryotic cells employ similar MetAP enzymes, such as MetAP1 and MetAP2, to remove the initiator from approximately 50-70% of cytosolic proteins, with the depending on the second residue's and hydrophobicity; for instance, small residues like or serine facilitate cleavage. This excision prevents steric hindrance and influences protein and targeting. For secretory and membrane proteins, signal peptide cleavage is a key maturation event mediated by signal peptidases in the or Golgi. These enzymes recognize and hydrolyze the hydrophobic signal sequence at the of preproteins, typically after translocation across the via the Sec61 translocon, allowing the mature protein to engage in folding and . In eukaryotes, the signal peptidase complex (SPC), consisting of subunits like SPC12, SPC18, and SPC25, performs this endoproteolytic cleavage at specific -1/-3 rule sites (small residues at positions -1 and -3 relative to the cut), ensuring efficient release of proteins destined for or organelle integration. This processing is vital for proteins like or hormones, where uncleaved signals would impair export. Viral polyprotein processing exemplifies maturation through sequential proteolysis, as seen in , where the 3C protease autocatalytically cleaves the single translated polyprotein into functional units like proteins and replicases. The 3Cpro enzyme, a , specifically targets Gln-Gly dipeptide bonds within the polyprotein precursor, generating mature non-structural proteins essential for and ; this process begins intra-molecularly and proceeds inter-molecularly for efficiency. Such maturation is conserved in picornaviruses, enabling rapid production of viral components from a compact . Proprotein conversion to active hormones involves targeted cleavages by , as in the maturation of insulin from preproinsulin. In pancreatic cells, preproinsulin is first processed in the by signal peptidase to yield , which then folds into a structure with A, B, and C chains connected by bonds. Subsequently, in immature secretory granules, PC1/3 and PC2 endoproteases cleave at dibasic sites (Arg-Arg or Lys-Arg) flanking the , followed by carboxypeptidase E trimming of basic residues, resulting in mature insulin and free ; this stepwise proteolysis is pH-dependent and ensures proper storage and secretion. Disruptions in these convertases lead to impaired insulin production, highlighting their role in endocrine maturation. Autoproteolysis represents a self-catalyzed maturation mechanism in certain proteins, where internal residues act as proteases to excise segments. Inteins, in prokaryotes and lower eukaryotes, undergo via a multi-step process involving nucleophilic attacks by conserved motifs (e.g., Cys or Ser at the and Asn at the ), leading to self-excision and of flanking exteins into a mature protein; this is crucial for host protein functionality in organisms like . In viral maturation, such as in Nodamura , the alpha protein undergoes pH-triggered autoproteolysis at a specific site (e.g., after residue 363), releasing a C-terminal that facilitates conformational changes from procapsid to stable , essential for and infectivity. Proteolysis also contributes to the assembly of complex macromolecules like and by enabling subunit maturation and interactions. In assembly, endoproteolytic processing by furin-like convertases in the Golgi cleaves proforms of envelope (e.g., in viruses or cellular receptors), exposing fusion peptides or binding domains that promote oligomerization and membrane insertion during ER-to-plasma membrane trafficking. For , limited N-terminal proteolysis of (apoB) during ER translation and lipidation prevents aggregation and facilitates microsomal transfer protein (MTP)-mediated lipid loading, yielding nascent (VLDL) particles; this maturation step is rate-limiting for hepatic secretion. These cleavages ensure structural integrity and functional assembly in secretory pathways.

Protein Degradation and Turnover

Protein degradation and turnover represent essential processes in cellular , where proteolysis breaks down unnecessary, damaged, or short-lived proteins into for and reuse in protein . This turnover ensures the proteome remains dynamic, allowing cells to adapt to changing conditions by removing aberrant proteins and replenishing building blocks. Intracellularly, two primary pathways dominate: the ubiquitin-proteasome system () for selective of soluble proteins and the lysosomal pathway for bulk or selective clearance of cytoplasmic and components. Extracellularly, proteolysis facilitates acquisition through in the gastrointestinal tract. These mechanisms collectively regulate protein half-lives, which vary from minutes to days depending on structural features and environmental cues. The ubiquitin-proteasome system initiates degradation by tagging target proteins with ubiquitin, a small 76-amino-acid protein, through a involving E1 activating enzymes, E2 conjugating enzymes, and ligases that confer specificity. E1 enzymes activate in an ATP-dependent manner, transferring it to E2, which then collaborates with to form polyubiquitin chains on residues of the substrate protein, marking it for destruction. These ubiquitinated proteins are recognized and unfolded by the 19S regulatory particle of the 26S , a barrel-shaped complex comprising a catalytic core and 19S caps; the core's proteases cleave the protein into short peptides, while is recycled. This pathway handles the majority of intracellular , particularly for regulatory proteins with half-lives under 10 hours. Lysosomal degradation complements the by targeting larger structures and membrane-bound proteins via and . Macroautophagy engulfs cytoplasmic portions into double-membrane autophagosomes that fuse with lysosomes, where acid hydrolases degrade the contents; microautophagy involves direct invagination of the lysosomal membrane to sequester small cytoplasmic regions. delivers extracellular or plasma membrane proteins to lysosomes through endosomal maturation, enabling receptor-mediated uptake and degradation. These processes are crucial for clearing aggregated or long-lived proteins, such as those in organelles, and contribute to recycling during nutrient scarcity. Extracellular protein degradation primarily occurs in the digestive system, where in the acidic initiates of dietary proteins into peptides, followed by pancreatic enzymes like and in the alkaline that further cleave them into absorbable and small peptides. , activated from by enterokinase, preferentially cuts at and residues, while targets aromatic , ensuring efficient breakdown for nutrient absorption. This pathway recycles exogenous proteins into endogenous pools, supporting systemic turnover. Protein half-lives are tightly regulated by signals like the , which dictates degradation rates based on the N-terminal residue: stabilizing residues (e.g., ) confer longer half-lives, while destabilizing ones (e.g., ) accelerate ubiquitination via ligases like UBR1. Factors such as oxidative damage, which introduces carbonyl groups and unfolds proteins, or misfolding exposing hydrophobic regions, trigger rapid clearance to prevent ; oxidized or misfolded proteins are preferentially ubiquitinated or autophagized. Both and are energy-intensive, relying on : the proteasome's 19S subunit uses six ATPases for unfolding and translocation, while requires ATP for autophagosome formation and fusion. These dependencies ensure degradation occurs under favorable metabolic conditions, linking turnover to cellular energy status.30978-4)

Regulatory Roles in Cellular Processes

Controlled proteolysis serves as a critical regulatory in cellular processes, enabling precise temporal control over signaling pathways and developmental events through the irreversible cleavage of key proteins. Unlike reversible modifications such as , proteolysis commits cells to specific outcomes, amplifying signals and preventing feedback loops that could disrupt . This irreversibility provides a robust switch for processes requiring commitment, such as progression and . In cell cycle regulation, the anaphase-promoting complex/cyclosome (APC/C) orchestrates the timed degradation of s, ensuring orderly transitions between phases. For instance, APC/C-mediated ubiquitination targets B1 for proteasomal degradation at the metaphase-anaphase transition, thereby inactivating (CDK1) and allowing mitotic exit. This process is checkpoint-dependent, with the checkpoint inhibiting APC/C until chromosomes align, preventing premature breakdown and genomic instability. Apoptosis relies on a proteolytic cascade initiated by caspases, where initiator caspases like caspase-9 activate effector caspases such as caspase-3 through specific cleavage events. This cascade amplifies the death signal, leading to the ordered dismantling of cellular components, including the cleavage of poly(ADP-ribose) polymerase (PARP) to halt DNA repair. The hierarchical activation ensures rapid, irreversible execution once triggered by stressors like DNA damage.00482-3.pdf) In signal transduction, proteolysis activates the Notch pathway via sequential cleavages: initial shedding of the extracellular domain by ADAM proteases, followed by intramembrane cleavage by γ-secretase, releasing the Notch intracellular domain (NICD) for nuclear translocation and gene transcription. This regulated proteolysis is essential for developmental decisions, such as cell fate determination in tissues like the nervous system, where NICD modulates target genes like Hes1.00382-1) Inflammatory responses are fine-tuned by ADAM proteases, which process membrane-bound cytokines into soluble forms. ADAM17, for example, cleaves pro-TNF-α to release active TNF-α, initiating downstream signaling that recruits immune cells and amplifies inflammation. This ectodomain shedding allows rapid cytokine dissemination, coordinating acute responses to infection without requiring new protein synthesis.00083-4) Circadian rhythms are maintained through rhythmic proteolysis of (PER) proteins, which form a negative feedback loop with CLOCK-BMAL1 transcription factors. marks PER for ubiquitination by SCF E3 ligases, leading to its and resetting the ~24-hour cycle; disruptions in this timing alter sleep-wake patterns and metabolic . The precise kinetics of PER ensure oscillatory stability across tissues.

Pathological Implications

Proteolysis in Disease Mechanisms

Aberrant proteolysis plays a central role in numerous pathologies by disrupting protein , leading to the accumulation of toxic aggregates, uncontrolled signaling, or insufficient degradation of harmful proteins. In neurodegenerative disorders, excessive proteolytic cleavage contributes to the formation of , while in cancer, dysregulated activity promotes tumor progression and . Similarly, overactivation of specific proteolytic pathways underlies cardiovascular conditions like , and viral proteases drive infectious replication. Genetic deficiencies in protease inhibitors result in unchecked proteolysis causing damage, and age-related declines in proteasomal function exacerbate cellular damage accumulation. These mechanisms highlight how imbalances in proteolytic processes can precipitate widespread tissue dysfunction and states. In neurodegenerative diseases such as , dysregulated proteolysis by β- and γ-secretases generates amyloid-β peptides from the amyloid precursor protein (), leading to plaque formation and neuronal toxicity. β-Secretase (BACE1) initiates the cleavage of to produce a C-terminal fragment, which is then processed by the γ-secretase complex—a multi-subunit including —to release amyloid-β42, the predominant toxic isoform that aggregates into extracellular plaques. This sequential proteolytic processing is central to Alzheimer's pathogenesis, as familial mutations in or enhance amyloid-β production and accelerate disease onset. Cancer often involves dysregulated proteasome activity that sustains pro-survival signaling and evades , allowing tumor cells to proliferate unchecked. The 26S , responsible for degrading ubiquitinated proteins, becomes upregulated in many malignancies, preventing the clearance of oncogenic regulators like cyclins and inhibitors, thereby promoting cell cycle progression and anti-apoptotic pathways. For instance, in and other solid tumors, proteasome hyperactivity stabilizes pro-tumorigenic proteins, contributing to chemoresistance and disease progression. Additionally, matrix metalloproteinases (MMPs), a family of zinc-dependent endopeptidases, facilitate cancer by degrading components such as and , enabling tumor and ; MMP-2 and MMP-9 are particularly implicated in remodeling the to support distant spread. Antibodies targeting MMP-9, such as andecaliximab, have been investigated for cancer therapy to suppress tumor and . However, a phase 3 trial in patients with advanced gastric or gastroesophageal junction cancer did not demonstrate improved overall survival or compared to . As of 2025, development for solid tumors has been discontinued, though it shows promise in other conditions like . In cardiovascular diseases, overactivation of the renin- system () through () drives by enhancing proteolytic generation of II, a potent vasoconstrictor. , a metalloprotease expressed in endothelial cells, cleaves angiotensin I to produce II, which binds AT1 receptors to induce vascular smooth muscle contraction, sodium retention, and , ultimately elevating blood pressure and promoting cardiac . Chronic overactivation, often due to genetic or environmental factors, sustains this proteolytic cascade, contributing to end-organ damage in hypertensive patients. Infectious diseases like rely on proteases for replication, with the main protease (Mpro, or 3CLpro) being essential for processing polyproteins into functional units. This cleaves at 11 specific sites within the replicase polyproteins pp1a and pp1ab, enabling the of the replication-transcription complex; without Mpro activity, genome replication and particle are halted. Structural studies confirm Mpro's homodimeric form and conserved catalytic dyad as critical for its role in , making it a key target in antiviral strategies. Genetic disorders such as (AAT) deficiency arise from mutations in the SERPINA1 gene, leading to misfolded AAT protein that accumulates in hepatocytes and fails to inhibit , resulting in unchecked proteolysis and damage to lungs and liver. The Z variant (Glu342Lys) of AAT polymerizes in the , causing liver through toxic retention and reducing circulating AAT levels, which allows excessive -mediated degradation of lung and development. This dual mechanism—protease inhibition failure in the lungs and in the liver—underlies the progressive organ pathology in affected individuals. Aging is marked by impaired proteostasis, where declining proteasome and chaperone activities lead to the accumulation of damaged, misfolded proteins, exacerbating and tissue dysfunction. The ubiquitin-proteasome system efficiency wanes with age, resulting in protein aggregates that trigger , , and organ decline; for example, reduced autophagic flux and proteasomal degradation contribute to and neurodegeneration. This progressive loss of proteolytic capacity underlies the increased vulnerability to age-related diseases, as unresolved protein damage propagates systemic proteotoxic stress.

Therapeutic Targeting of Proteolysis

Therapeutic targeting of proteolysis involves the development of pharmacological agents that either inhibit or enhance proteolytic activities to treat various diseases, particularly those driven by dysregulated function such as cancer, infectious diseases, and disorders. Small-molecule inhibitors represent a cornerstone of this approach, with serving as a prototypical approved for treatment; it reversibly binds the 20S proteasome's threonine active sites, preventing ubiquitin-tagged protein degradation and inducing in malignant cells. Similarly, inhibitors like target the viral aspartyl protease essential for polyprotein maturation, blocking the enzyme's homodimer formation and halting ; ritonavir's dual role as both an antiretroviral and a pharmacokinetic booster enhances the efficacy of combination therapies. Monoclonal antibodies offer high specificity in modulating extracellular proteolysis, minimizing intracellular off-target effects. For instance, antibodies targeting matrix metalloproteinases (MMPs), such as those inhibiting MMP-9, have been explored in various therapies. In , anti-ADAMTS-5 monoclonal antibodies like GSK2394002 selectively block aggrecanase activity, preserving integrity in preclinical models and advancing toward disease-modifying applications. Strategies to enhance proteolysis include zymogen modulation, particularly in clotting disorders like hemophilia, where a zymogen-like factor Xa variant (FXa I16L) promotes thrombin generation without excessive activation, correcting coagulation defects in animal models and offering a bypass therapy for factor deficiencies. Proteolysis-targeting chimeras (PROTACs) represent an innovative class of heterobifunctional molecules that recruit E3 ubiquitin ligases to induce targeted degradation of disease-related proteins via the proteasome; these agents have entered clinical trials for oncology, achieving complete elimination of targets like androgen receptor in prostate cancer cells, surpassing traditional inhibition. As of 2025, several PROTACs, including ARV-471 for breast cancer, are in phase 2/3 clinical trials, demonstrating selective protein degradation in oncology. Despite these advances, therapeutic targeting faces significant challenges, including achieving specificity to avoid off-target inhibition of homologous enzymes, which can lead to , and overcoming resistance mechanisms such as mutational escape or compensatory pathway upregulation observed in cancer and treatments. As a clinical example of indirect modulation, statins like influence proteolysis in regulation by depleting isoprenoids, which disrupts geranylgeranylation and promotes proteasomal degradation of regulatory proteins like Skp2, contributing to their pleiotropic cardiovascular benefits beyond inhibition.

Non-Enzymatic Proteolysis

Chemical and Oxidative Pathways

Non-enzymatic proteolysis via chemical and oxidative pathways encompasses the spontaneous of bonds in proteins through abiotic reactions, distinct from protease-mediated processes, and typically proceeds at rates orders of magnitude slower with low specificity, often resulting in random fragmentation. These mechanisms are influenced by environmental factors such as , , oxidants, and , leading to protein denaturation, structural alterations, and eventual breakdown without the catalytic efficiency of enzymes. Acid and heat hydrolysis represent fundamental chemical pathways for peptide bond cleavage, where low pH or elevated temperatures destabilize the amide linkage, enabling water-mediated hydrolysis. Under acidic conditions, protonation of the carbonyl oxygen in the peptide bond increases its electrophilicity, facilitating nucleophilic attack by water to form a tetrahedral intermediate that collapses to yield carboxylic acid and amine products; this process is enhanced at pH below 2 and temperatures around 110°C, as commonly applied in laboratory protocols for total protein hydrolysis. Heat alone, without strong acids, promotes hydrolysis by increasing molecular motion and weakening hydrogen bonds, leading to unfolding and subsequent bond rupture, particularly in extreme environments like hydrothermal vents where proteins encounter temperatures exceeding 100°C. Cleavage sites are often biased toward bonds following aspartic or glutamic acid residues due to side-chain cyclization forming reactive succinimide intermediates, with an algorithm predicting ~90% accuracy for susceptible sites based on local secondary structure and solvent exposure. Oxidative damage arises from reactive oxygen species (ROS), such as (H₂O₂), which target sulfur-containing like and , initiating chain reactions that culminate in fragmentation. is oxidized to by H₂O₂, a process accelerated in the presence of , disrupting and exposing cleavage-prone regions; further oxidation can lead to formation and backbone scission via peroxyl radical intermediates. residues, when deprotonated, react rapidly with H₂O₂ to form sulfenic acids or disulfides, which propagate oxidation to nearby , resulting in and fragmentation, as observed in proteome-wide studies where ROS reversibly modify approximately 3% of cysteines under . These modifications often occur randomly but preferentially at surface-exposed residues, contributing to overall protein instability without enzymatic involvement. Glycation through the involves non-enzymatic condensation of reducing sugars, such as glucose, with protein amino groups (primarily and ), forming (AGEs) that rigidify and fragment proteins. The reaction proceeds in stages: initial formation, to ketoamines, and irreversible oxidation/dehydration yielding AGEs like Nε-carboxymethyllysine (CML) or pentosidine, which introduce cross-links that alter protein charge, conformation, and susceptibility to . These modifications accumulate in long-lived proteins, promoting backbone cleavage via enhanced ROS generation and metal-catalyzed reactions, with studies showing up to 3-fold increased levels of glycated proteins under hyperglycemic conditions. Photo-oxidation, driven by (UV) light, induces direct or sensitized oxidation in proteins, particularly in exposed tissues like , leading to cleavage of bonds through radical-mediated mechanisms. UVB (280–315 nm) absorption by aromatic residues (, ) generates excited states that produce ROS, while (315–400 nm) relies on photosensitizers like flavins to form , attacking and to yield peroxides and fragmented chains; in , chronic UV exposure leads to significant elevation of protein carbonyls in the compared to protected sites. This process is less specific, often resulting in multiple sites, and is exacerbated in the where depletion amplifies damage. Kinetically, these pathways exhibit slow rates compared to enzymatic proteolysis, with non-enzymatic displaying pH-dependent mechanisms: at neutral , peptide bond half-lives exceed 500 years (rate constants ~10⁻¹¹ s⁻¹), accelerating to minutes under acidic or heated conditions but remaining 10⁶–10⁸ times slower than enzyme-catalyzed reactions, which achieve turnover numbers up to 10³ s⁻¹. Oxidative and glycative processes follow second-order with respect to ROS or concentration, yielding half-lives of hours to days under physiological , emphasizing their as gradual, cumulative degraders rather than rapid turnover mechanisms. Specificity is low, with cleavage often , though local factors like residue proximity modulate rates. Representative examples include protein denaturation during , where heat-induced in or products breaks down caseins or myofibrils into peptides, enhancing digestibility but altering , as seen in Maillard-driven at 100–150°C. In cellular , oxidative pathways dominate, with H₂O₂ accumulation during ischemia fragmenting cytoskeletal proteins like , contributing to structural collapse without activation.

Biological and Environmental Contexts

In living organisms, non-enzymatic proteolysis mediated by (ROS) plays a significant role during and aging, where elevated ROS levels oxidize proteins, leading to structural damage and the formation of protein aggregates. This oxidative modification disrupts protein function and contributes to cellular dysfunction, as seen in age-related diseases where carbonylated proteins accumulate and impair . For instance, in neurodegenerative conditions, non-enzymatic posttranslational modifications like oxidation promote the aggregation of proteins such as α-synuclein and , exacerbating pathological outcomes. In extremophilic environments, non-enzymatic proteolysis arises from extreme physical conditions, such as high temperatures in thermophilic bacteria or low in acidic organelles like lysosomes. Thermophilic bacteria, thriving at temperatures above 60°C, experience accelerated non-enzymatic of peptide bonds due to , which challenges protein stability and requires specialized adaptations like hyperstable structures to mitigate degradation. Similarly, in acidic organelles with values below 5, facilitates non-enzymatic cleavage of proteins, aiding in the breakdown of internalized materials independent of enzymatic activity. In , non-enzymatic proteolysis contributes to Maillard reactions during cooking, where reducing sugars react with amino groups in proteins, resulting in and alterations to through cross-linking and fragmentation. These reactions modify protein digestibility and sensory properties, such as tenderness in meats, by inducing conformational changes and partial without enzymatic involvement. Environmentally, (UV) induces non-enzymatic proteolysis of extracellular proteins in aquatic and terrestrial settings, including oceans and . In ecosystems, UV exposure photodegrades dissolved , including proteins, by direct bond cleavage and oxidative damage, influencing nutrient cycling and microbial food webs. In , UV accelerates the breakdown of surface proteins in , enhancing rates through photolytic fragmentation that doubles mass loss compared to dark conditions. From an evolutionary perspective, non-enzymatic proteolysis likely served as a precursor to enzymatic systems, enabling early turnover of peptides in prebiotic environments before the of specialized proteases. Such spontaneous reactions facilitated the recycling of , laying the groundwork for more efficient enzymatic control in evolving cellular . To detect these modifications, particularly oxidative ones, carbonyl assays are widely employed, quantifying protein carbonyl content via derivatization with to assess damage levels in biological samples.

Practical Applications

Laboratory Methods and Techniques

Laboratory methods for studying proteolysis encompass a range of assays and techniques designed to measure activity, identify sites, and evaluate regulatory mechanisms in controlled experimental settings. These approaches enable researchers to quantify enzymatic , screen potential inhibitors, and model proteolytic processing events with high precision. assays utilizing fluorogenic substrates represent a for measuring proteolytic activity. In these assays, synthetic substrates conjugated to a , such as 7-amino-4-methylcoumarin (), are cleaved by s, releasing free that emits upon excitation, typically at wavelengths around 380 nm excitation and 460 nm emission. This method allows for sensitive detection of enzyme , with limits as low as picomolar concentrations, and is widely used for both purified enzymes and complex biological samples. A seminal approach involves configuring libraries with at the P1' position to profile specificity across diverse substrates, enabling rapid identification of preferences. Gel-based techniques, particularly zymography combined with , facilitate the detection and characterization of protease activity directly in gels. Samples are electrophoresed into gels copolymerized with substrates like or ; after renaturation, active proteases digest the substrate, resulting in clear bands of against a stained background, allowing of molecular weights and activity profiles. This method is especially valuable for identifying metalloproteinases and other extracellular proteases, with sensitivity enhanced by incorporating metal ions or inhibitors to distinguish classes. Reviews highlight its utility in microbial and eukaryotic samples, where it resolves activities from 20 to 200 kDa with minimal . Mass spectrometry (MS)-based methods, including peptide mapping, provide detailed identification of proteolysis cleavage sites at the sequence level. Proteins are digested with proteases, and the resulting peptides are analyzed by liquid chromatography-tandem MS (LC-MS/MS) to detect neo-N-termini or mass shifts indicative of specific cuts, often using labeling strategies like TAILS (terminal amine isotopic labeling of substrates) for quantitative enrichment. This approach maps cleavage motifs with single-residue resolution, revealing substrate preferences and processing dynamics in cellular lysates. Proteomic workflows have advanced to monitor proteolytic events in response to stimuli, such as , by comparing pre- and post-cleavage peptide profiles across thousands of sites. High-throughput screening platforms for protease inhibitors accelerate by evaluating compound libraries against target enzymes. Fluorescence-based or assays in 96- or 384-well formats measure inhibition of fluorogenic cleavage, often integrating cellular reporters for physiological relevance, such as fused to protease-cleavable linkers. These systems have identified hits against viral proteases like 3CLpro, with Z' factors above 0.7 indicating robust performance for screening millions of compounds. Automated platforms, including or microfluidic arrays, enable multiplexed testing of inhibitor potency and selectivity. In vitro models employing recombinant protein expression systems allow controlled studies of proteolytic processing. in hosts like E. coli or produces isotopically labeled substrates, which are then incubated with purified proteases to mimic maturation events, such as removal. Techniques like circular permutation of recombinant proteins reduce susceptibility to unintended degradation, enabling kinetic analysis of specific cleavages via or follow-up. These models have elucidated proteolysis in pathways, with yields improved by co-expression of chaperone fusions to stabilize intermediates. Quantitative real-time monitoring of proteolysis is achieved through (FRET)-based sensors, which report cleavage events via changes in ratios. These genetically encoded probes consist of donor-acceptor pairs flanking a protease-specific linker; upon cleavage, spatial separation disrupts energy transfer, increasing donor emission for dynamic tracking in live cells or lysates. Sensors targeting or mammalian proteases have demonstrated spatiotemporal resolution, detecting activities within minutes at sub-micromolar sensitivities. Advances include super-silent variants that minimize background, facilitating and imaging of proteolytic cascades.

Industrial and Biomedical Uses

Proteolysis plays a pivotal role in the , particularly through the use of specific proteases for tenderization and processing. , derived from , and , extracted from , are proteases widely employed to hydrolyze tough connective tissues in , enhancing tenderness without significantly affecting nutritional value. In cheese production, —a mixture containing , an aspartic protease—facilitates the of by cleaving kappa-casein, leading to formation essential for cheese yield and texture. In the detergent industry, , a produced by species, is incorporated into formulations to break down protein-based stains such as blood, egg, and grass, improving cleaning efficiency under alkaline conditions typical of wash cycles. Engineered variants of subtilisin enhance stability and activity at lower temperatures, reducing in modern washing machines while maintaining efficacy against diverse stains. Biomedical applications leverage proteolysis for diagnostics, wound care, and targeted therapies. Protease-based biosensors detect biomarkers by exploiting substrate cleavage to generate measurable signals, such as or electrochemical changes, enabling for conditions like through activity. In wound healing, from histolyticum serves as a debriding agent, selectively digesting denatured and necrotic tissue to promote and epithelialization without harming viable cells. An emerging application involves proteolysis-targeting chimeras (PROTACs), bifunctional molecules that recruit ubiquitin ligases to tag -related proteins for degradation via the , offering a novel strategy for treating cancers and neurodegenerative disorders; as of 2025, several PROTACs have advanced to Phase III clinical trials. In , controlled proteolysis in cell-free protein synthesis (CFPS) systems minimizes unwanted degradation by using protease-deficient extracts, allowing high-yield production of recombinant proteins for applications like development. Environmentally, microbial proteases facilitate by hydrolyzing protein-rich wastes, such as from animal byproducts or sludge in , converting them into biodegradable components and reducing in industrial effluents.

References

  1. [1]
    Proteolysis of Proteins - Wiley Online Library
    Mar 15, 2019 · Proteolysis is a hydrolysis reaction of peptide bonds in which proteins breakdown into smaller peptides and/or into individual amino acid residues.
  2. [2]
    Biochemical Tools for Tracking Proteolysis | Journal of Proteome ...
    Proteolysis is an important irreversible post-translational modification that regulates most, if not all, cellular processes. Proteases are a large family of ...
  3. [3]
    How Cells Use Proteolysis to Control Their Growth
    Protein destruction—proteolysis— plays a pivotal role in controlling the activity of these proteins. Proteolysis is particularly suited to controlling the ...<|control11|><|separator|>
  4. [4]
    Proteolysis - an overview | ScienceDirect Topics
    Proteolysis is an important cellular process that, through the removal of unwanted or damaged proteins, plays a key role in the overall maintenance of the cell.
  5. [5]
    Towards a Quantitative Description of Proteolysis - NIH
    Jan 2, 2025 · Proteolysis is a complex reaction that includes a large number of elementary reactions involving various polypeptide chains, designated R1 and R ...
  6. [6]
    Biochemistry, Primary Protein Structure - StatPearls - NCBI Bookshelf
    Oct 31, 2022 · The secondary structure contains regions of amino acid chains that are stabilized by hydrogen bonds from the polypeptide backbone.
  7. [7]
    Pepsinogen - an overview | ScienceDirect Topics
    Pepsin was discovered in 1836 by Theodor Schwann and was amongst the first enzymes to be crystallized (by John Northrop in 1929). Pepstatin, a low molecular ...
  8. [8]
    Proteolysis - Latest research and news - Nature
    Proteolysis is the enzymatic process by which proteins are degraded into their component polypeptide or amino acid parts.
  9. [9]
    Protein Homeostasis and Aging: the importance of exquisite quality ...
    Molecular chaperones and the proteolytic systems are responsible for this cellular quality control by assuring continuous renewal of intracellular proteins.
  10. [10]
    Regulated Proteolysis in Bacteria - PMC - PubMed Central
    Quality control through regulated proteolysis requires bacteria to discriminate between fatally misfolded or damaged proteins and proteins that simply share ...
  11. [11]
    Classification - MEROPS - the Peptidase Database
    Jan 9, 2017 · The families of peptidases are constructed by comparisons of amino acid sequences. Every member of a MEROPS family shows a statistically ...
  12. [12]
    The pH dependent mechanisms of non-enzymatic peptide bond ...
    The non-enzymatic cleavage rates of amide bonds located in peptides in aqueous solution is pH-dependent and involves two distinct mechanisms.Missing: spontaneous heat
  13. [13]
    Peptidases: structure, function and modulation of peptide‐mediated ...
    Endopeptidases recognize specific amino acids in the middle of the peptide, whereas exopeptidases recognize one or two terminal amino acids.Aminopeptidase N · Dipeptidyl Peptidase Iv · Other Peptidases<|control11|><|separator|>
  14. [14]
    Role of proteolytic enzymes in biological regulation (a review)
    The ultimate agency of activating enzymatic function is limited proteolysis, either in a single activation step or in a consecutive series (cascade).
  15. [15]
    Complete and limited proteolysis in cell cycle progression - PubMed
    Aug 16, 2004 · An important mechanism of regulation that controls progression through the cell cycle involves the timely degradation of specific regulatoryMissing: unlimited single cuts
  16. [16]
    Mechanisms of Protein Degradation | Thermo Fisher Scientific - ES
    Proteolysis is the enzymatic breakdown of proteins into peptides and amino acids that can then be recycled by the cell for future protein synthesis.
  17. [17]
    Factors Influencing Proteolysis and Protein Utilization in the Intestine ...
    In this review, we summarize and discuss factors influencing protein degradation and thereby affecting the efficiency of the animals.
  18. [18]
    Proteases: Multifunctional Enzymes in Life and Disease - PMC - NIH
    Proteases are the efficient executioners of a common chemical reaction: the hydrolysis of peptide bonds (16). Most proteolytic enzymes cleave α-peptide bonds ...<|control11|><|separator|>
  19. [19]
    Twenty-five years of nomenclature and classification of proteolytic ...
    Proteolytic enzymes are classified by tertiary structure similarity into clans and by sequence similarity into families. •. Criteria have been devised to ...
  20. [20]
    Serine Protease Mechanism and Specificity | Chemical Reviews
    Many experiments confirm the importance of the catalytic triad residues in serine protease catalysis. Chemical modification experiments were first used to ...Missing: catalytic triad
  21. [21]
    Unconventional serine proteases: Variations on the catalytic Ser/His ...
    Two of the best-known serine proteases that utilize the Ser/His/Asp triad are chymotrypsin and trypsin. They share the same protein fold and have their ...
  22. [22]
    Molecular mechanisms for the conversion of zymogens to active ...
    A common form of the activation segment is an N-terminal extension of the mature enzyme, or "prosegment," that sterically blocks the active site, and thereby ...
  23. [23]
    Mechanisms Of Macromolecular Protease Inhibitors - PMC - NIH
    A number of protease inhibitors are competitive, and bind in the protease active site, but also have secondary binding sites outside the active site, which are ...
  24. [24]
    Intrinsic evolutionary constraints on protease structure, enzyme ...
    Feb 4, 2013 · We examined the structures of these proteases, using a new framework to characterize the geometric relationships within each active site.
  25. [25]
    Pepsin properties, structure, and its accurate measurement
    Pepsins are aspartate proteases active in acidic conditions and are the major proteases in human gastric juice. Pepsins are therefore important in normal ...
  26. [26]
    5644 - Gene ResultPRSS1 serine protease 1 [ (human)] - NCBI
    Aug 19, 2025 · This gene encodes a trypsinogen, which is a member of the trypsin family of serine proteases. This enzyme is secreted by the pancreas and cleaved to its active ...
  27. [27]
    Caspases: the proteases of the apoptotic pathway - PubMed - NIH
    Dec 24, 1998 · Caspases are cysteine proteases that are the executionary arm of apoptosis, a form of cell death, and are part of a proteolytic cascade.
  28. [28]
    The ubiquitin-proteasome pathway: The complexity and ... - PNAS
    It is a “dumbshell-shaped” symmetric structure composed of a core catalytic unit, a barrel-shaped 20S proteasome complex, which is flanked at both sides by ...
  29. [29]
    Extracellular matrix remodelling: the role of matrix metalloproteinases
    Matrix metalloproteinases (MMPs) cleave extracellular matrix proteins, playing a central role in tissue remodeling and regulating cellular interactions.
  30. [30]
    The chemistry of snake venom and its medicinal potential - Nature
    Jun 10, 2022 · Batroxobin (Defibrase) is a thrombin-like serine protease ... From snake venom's disintegrins and C-type lectins to anti-platelet drugs.
  31. [31]
  32. [32]
    Papain protects papaya trees from herbivorous insects - PubMed
    We found that papain, a cysteine protease in latex of the Papaya tree (Carica papaya, Caricaceae), is a crucial factor in the defense of the papaya tree ...
  33. [33]
    Cleavage of Human Immunodeficiency Virus Type 1 Proteinase from ...
    Maturation of infectious human immunodeficiency virus (HIV) particles requires proteolytic cleavage of the structural polyproteins by the viral proteinase (PR), ...
  34. [34]
    Insulin Biosynthesis, Secretion, Structure, and Structure-Activity ...
    Feb 1, 2014 · Proinsulin is cleaved at those dibasic links by a trypsin-like enzyme to release the mature hormone and a free C-peptide (which lacks the ...
  35. [35]
    N-Terminal Methionine Processing - PMC - NIH
    In eukaryotes, methionine is removed either by cleavage of N-terminal signal peptide used for secretion etc., or by MAP. In prokaryotes, formylmethionine is ...Missing: review | Show results with:review
  36. [36]
    Control of protein life‐span by N‐terminal methionine excision
    N‐terminal methionine excision (NME) is the major pathway causing diversity of N‐terminal amino acids. As a result of NME, Gly, Ala, Pro, Cys, Ser, Thr or Val ...
  37. [37]
    The Proteomics of N-terminal Methionine Cleavage - ScienceDirect
    Methionine aminopeptidase (MAP) is a ubiquitous, essential enzyme involved in protein N-terminal methionine excision. According to the generally accepted ...
  38. [38]
    Structure of the human signal peptidase complex reveals the ...
    Oct 7, 2021 · The signal peptidase complex (SPC) is an essential membrane complex in the endoplasmic reticulum (ER), where it removes signal peptides (SPs) from a large ...
  39. [39]
    Signal Peptides: From Molecular Mechanisms to Applications in ...
    Jun 18, 2025 · The SP traverses a membrane channel into the ER lumen, where it reaches a specific stage before being hydrolyzed by signal peptidase (SPase) at ...
  40. [40]
    Proteolytic processing of poliovirus polyprotein - PubMed - NIH
    The viral proteinase 3C and the viral polymerase 3D result from cleavage at a Gln-Gly pair by proteinase 3C, whereas cleavage at a Tyr-Gly pair by proteinase 2A ...
  41. [41]
    Article A second virus-encoded proteinase involved in proteolytic ...
    The poliovirus polyprotein is cleaved at three different amino acid pairs. Viral polypeptide 3C is responsible for processing at the most common pair ...
  42. [42]
    The making of insulin in health and disease | Diabetologia
    Sep 7, 2020 · Following proinsulin folding in the ER and the removal of the C-peptide, mature insulin is formed in secretory granules (SGs). Environmental ...
  43. [43]
    Evidence That Human β-Cells Process Proinsulin With Prohormone ...
    Apr 14, 2020 · ... C-peptide to liberate mature insulin. Current theory posits that the B-chain–C-peptide junction is cleaved by prohormone convertase 1/3 (PC1 ...
  44. [44]
    Structural and Dynamical Features of Inteins and Implications on ...
    Apr 2, 2014 · Protein splicing is a posttranslational modification where intervening proteins (inteins) cleave themselves from larger precursor proteins ...
  45. [45]
    Protein Splicing of Inteins: A Powerful Tool in Synthetic Biology
    Feb 20, 2022 · Inteins are protein segments that are capable of ligating the flanking exteins (external proteins) into a new protein, a process known as protein splicing.
  46. [46]
    Characterization of Large Conformational Changes and ...
    Procapsids transition to capsids when pH is lowered from 7.6 to 5.0. Capsids undergo autoproteolysis at residue 570, generating the 74-residue C-terminal ...
  47. [47]
    Identification of host cell surface proteins inhibiting furin dependent ...
    Jul 15, 2025 · Proteolytic cleavage by furin-like proteases is a crucial first step in the posttranslational modification of various glycoproteins found in ...
  48. [48]
    Reconstituting Initial Events during the Assembly of ApoB-containing ...
    In this study, we use a prokaryotic cell-free expression system to reconstitute early events in the assembly of apoB-containing lipoprotein that involve the N- ...
  49. [49]
    Lipid transfer proteins in the assembly of apoB-containing lipoproteins
    MTTP is the major and essential lipid transfer protein that transfers phospholipids and triacylglycerols to nascent apoB for the assembly of lipoproteins.
  50. [50]
    Perilous journey: a tour of the ubiquitin–proteasome system - NIH
    Jan 20, 2014 · Proteins become degraded upon their conjugation to chains of ubiquitin where they are then directed to the 26S proteasome, a macromolecular protease.Missing: paper | Show results with:paper
  51. [51]
    Ubiquitin-like protein conjugation and the ubiquitin–proteasome ...
    Dec 10, 2010 · The ubiquitin–proteasome system (UPS) and ubiquitin-like protein (UBL) conjugation pathways are integral to cellular protein homeostasis.Missing: paper | Show results with:paper
  52. [52]
    Autophagy: process and function - Genes & Development
    This process is quite distinct from endocytosis-mediated lysosomal degradation of extracellular and plasma membrane proteins. There are three types of autophagy ...Missing: micro | Show results with:micro
  53. [53]
    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 ...Missing: micro | Show results with:micro
  54. [54]
    Biochemistry, Protein Catabolism - StatPearls - NCBI Bookshelf - NIH
    Dec 11, 2022 · One particular protease, trypsin, is the most important to the digestive process because it cleaves proteins and activates other proteases from ...Missing: extracellular | Show results with:extracellular
  55. [55]
    The N-end rule pathway of protein degradation - PubMed
    The N-end rule pathway of protein degradation. Genes Cells. 1997 Jan;2(1):13-28. doi: 10.1046/j.1365-2443.1997.1020301.x. Author. A Varshavsky. Affiliation. 1 ...
  56. [56]
    N-degron and C-degron pathways of protein degradation - PNAS
    Jan 7, 2019 · N-degrons and C-degrons are degradation signals whose main determinants are, respectively, the N-terminal and C-terminal residues of cellular proteins.
  57. [57]
    Toward Understanding the Biochemical Determinants of Protein ...
    Feb 17, 2021 · This oxidative stress leads to unfolding of oxidized proteins, which compromises their function and exposes buried hydrophobic patches. This ...Introduction · Protein Degradation Pathways · Determinants of Protein...
  58. [58]
    Physiological levels of ATP negatively regulate proteasome function
    Aug 31, 2010 · Autophagy inhibition compromises degradation of ubiquitin-proteasome pathway substrates. ... ATP binding and ATP hydrolysis play distinct ...
  59. [59]
    Phosphorylation Meets Proteolysis - ScienceDirect.com
    Apr 4, 2012 · Although the two events are fundamentally different—phosphorylation is a reversible modification, whereas proteolysis is irreversible—there is ...
  60. [60]
    The APC/C recruits cyclin B1–Cdk1–Cks in prometaphase before D ...
    Aug 23, 2010 · This initiates cyclin A degradation, whereas cyclin B1 is stabilized by the spindle checkpoint. Upon checkpoint release, the RXXL destruction ...
  61. [61]
    Caspase activation cascades in apoptosis - PubMed
    In this review, we discuss current perspectives concerning the diverse pathways to caspase activation. Publication types. Lecture; Research Support, Non-U.S. ...
  62. [62]
    The period of the circadian oscillator is primarily determined ... - PNAS
    Mounting evidence suggests that PERIOD (PER) proteins play a central role in setting the speed (period) and phase of the circadian clock.
  63. [63]
    The Biology of Proteostasis in Aging and Disease - PMC
    Loss of protein homeostasis (proteostasis) is a common feature of aging and disease that is characterized by the appearance of nonnative protein aggregates ...
  64. [64]
    Alzheimer's β-secretase, β-site amyloid precursor protein-cleaving ...
    Because β- and γ-secretases play a critical role for Aβ formation, inhibitors of these proteases are promising therapeutics for Alzheimer's disease (7, 8). β- ...Missing: paper | Show results with:paper
  65. [65]
    γ-Secretase in Alzheimer's disease - PMC - PubMed Central - NIH
    Aβ is produced from amyloid precursor protein (APP) by sequential proteolytic cleavages by β-secretase and γ-secretase, and the polymerization of Aβ into ...App Processing And Aβ · Gsi And Gsm-Binding Sites · Gsmp In Neuroinflammation
  66. [66]
    Dysregulation of the Ubiquitin Proteasome System in Human ...
    Dysregulation of the UPS results in loss of ability to maintain protein quality through proteolysis, and is closely related to the development of various ...
  67. [67]
    Regulation of proteasome activity in health and disease
    In this review we discuss the contributions of the proteasome to human pathology, describe mechanisms that regulate the proteolytic capacity of the proteasome,
  68. [68]
    Hypertension: Renin–Angiotensin–Aldosterone System Alterations
    Mar 13, 2015 · This review provides an update about all these aspects, critically discussing the many controversies and allowing the reader to obtain a full understanding.
  69. [69]
    Coronavirus biology and replication: implications for SARS-CoV-2
    Oct 28, 2020 · Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved a-ketoamide inhibitors. Science 368, 409–412 (2020) ...
  70. [70]
    A Review of α 1 -Antitrypsin Deficiency - ATS Journals
    Aug 9, 2011 · AAT is mainly produced in the liver and functions to protect the lung against proteolytic damage (e.g., from neutrophil elastase). Among the ...
  71. [71]
    Bortezomib, a newly approved proteasome inhibitor for the treatment ...
    Bortezomib (Velcade, Millennium Pharmaceuticals, Inc., Cambridge, MA), a first-in-class proteasome inhibitor, has been approved for patients with MM who have ...
  72. [72]
    Ritonavir - PubMed
    Aug 8, 2023 · Although ritonavir was initially designed to inhibit HIV protease, studies have found that it also inhibits cytochrome P450-3A4. Because of its ...
  73. [73]
    Monoclonal Antibody Targeting the Matrix Metalloproteinase 9 ...
    Here, we demonstrate that a monoclonal antibody targeting MMP9 alleviates neuropathic pain and several mechanisms linked to CIPN. This study is particularly ...
  74. [74]
    Translational Development of an ADAMTS-5 Antibody for ... - PubMed
    Mar 20, 2015 · A humanized ADAMTS-5-selective monoclonal antibody (GSK2394002) was progressed as a potential OA disease modifying therapeutic.
  75. [75]
    A zymogen-like factor Xa variant corrects the coagulation defect in ...
    Oct 23, 2011 · FXa(I16L) may provide an effective strategy to enhance blood clot formation and act as a rapid pan-hemostatic agent for the treatment of ...Missing: disorders | Show results with:disorders<|separator|>
  76. [76]
    Targeted protein degradation by PROTACs - PubMed - NIH
    Herein, we highlight the advantages of protein degradation using PROTACs, and provide specific examples where degradation offers therapeutic benefit over ...Missing: heterobifunctional | Show results with:heterobifunctional<|separator|>
  77. [77]
    Statin-induced depletion of geranylgeranyl pyrophosphate inhibits ...
    Feb 20, 2015 · Statin-induced depletion of geranylgeranyl pyrophosphate inhibits cell proliferation by a novel pathway of Skp2 degradation.
  78. [78]
    The pH-Rate Profile for the Hydrolysis of a Peptide Bond
    Except at extremes of pH and temperature, nonenzymatic peptide bond hydrolysis is a slow process that is difficult to quantify accurately.
  79. [79]
    Peptide Bond Hydrolysis: Enzymatic and Non-Enzymatic Pathways in
    Hydrolysis of peptide bonds is the reverse process, where the bond between two amino acids is broken through the addition of water.
  80. [80]
    Molecular Investigation of the Mechanism of Non-Enzymatic ...
    May 19, 2016 · This algorithm measures four key properties during a molecular dynamics simulation, which relate to the key steps of the hydrolysis mechanism, ...
  81. [81]
    Methionine Oxidation and Reduction in Proteins - PMC - NIH
    Methionine in proteins acts as an antioxidant, protein stabilizer, and regulatory switch, and can be oxidized to methionine sulfoxide (MetO) and then reduced ...
  82. [82]
    Proteome-wide analysis of cysteine oxidation reveals metabolic ...
    Apr 20, 2018 · Reactive oxygen species (ROS) are increasingly recognised as important signalling molecules through oxidation of protein cysteine residues.
  83. [83]
    Protein oxidation and peroxidation | Biochemical Journal
    Protein peroxides can oxidize both proteins and other targets. One-electron reduction results in additional radicals and chain reactions with alcohols and ...
  84. [84]
    The Development of Maillard Reaction, and Advanced Glycation ...
    Advanced glycation end products (AGEs) are generated by nonenzymatic modifications of macromolecules (proteins, lipids, and nucleic acids) by saccharides ...
  85. [85]
    Advanced Glycation End-Products (AGEs): Formation, Chemistry ...
    ... proteins that, during thermal processing, undergo the Maillard reaction, leading to rapid AGE formation. It was initially thought that dAGEs were poorly ...
  86. [86]
    Photo-induced protein oxidation: mechanisms, consequences and ...
    Dec 17, 2019 · Absorption of incident UVB radiation by proteins is mainly related to the presence of the chromophoric side chains of tryptophan (Trp), tyrosine ...
  87. [87]
    Photoaging is Associated with Protein Oxidation in Human Skin In Vivo
    Chronic UV exposure induces accumulation of protein oxidation in human skin – investigation by immunoblotting. In tissue extracts prepared from separated ...
  88. [88]
    Impact of various factors on the kinetics of non-enzymatic ...
    A peptide bond is highly stable to non-enzymatic hydrolysis with a half-life of hundreds of years [4], [11], but certain sites are more prone to fragmentation.
  89. [89]
    (PDF) The Impact of Non-Enzymatic Reactions and Enzyme ...
    Apr 16, 2025 · In this review we discuss the impact of oxidative and other cellular stressors on the formation of metabolic side products, which originate as a ...
  90. [90]
    Proteolysis dysfunction in the process of aging and age-related ...
    Jul 22, 2022 · Protein quality control systems (proteostasis) are responsible for the functionality and turnover of cellular proteins. • During aging, the ...
  91. [91]
    [PDF] Protein Oxidation in Aging and Alzheimer's Disease Brain
    May 7, 2024 · Protein carbonylation occurs as a prominent oxidative modification as a result of elevated ROS in the aging brain, which is further supported by ...
  92. [92]
    Non-enzymatic posttranslational protein modifications in protein ...
    Dec 19, 2024 · This review focuses on major nPTMs found on proteins with central roles in neurodegenerative diseases such as α-synuclein, β-amyloid, and tau.α-Synuclein · β-Amyloid · Tau
  93. [93]
    Proteins from Thermophilic Thermus thermophilus Often Do Not Fold ...
    Oct 14, 2022 · Misfolding of thermophilic proteins grown using mesophilic hosts can lead to loss or change of protein functions which will harm both research ...Missing: enzymatic organelles
  94. [94]
    Intracellular alkaline proteases produced by thermoacidophiles
    In this study, we present data on screening for the alkaline proteolytic activities in 24 strains of thermophilic archea and bacteria which were previously ...Missing: organelles | Show results with:organelles
  95. [95]
    Control of Maillard Reactions: Strategies & Mechanisms
    May 23, 2017 · Maillard reactions lead to changes in food color, organoleptic properties, protein functionality, and protein digestibility.
  96. [96]
    Food Processing & Maillard Reaction Products: Health & Nutrition
    It is often called nonenzymatic browning reaction since it takes place in the absence of enzyme.
  97. [97]
    Temperature and UV light affect the activity of marine cell-free ... - BG
    Sep 13, 2017 · Abstract. Microbial extracellular enzymatic activity (EEA) is the rate-limiting step in the degradation of organic matter in the oceans.Missing: non- protein
  98. [98]
    UV radiation doubles microbial degradation of standing litter in a ...
    Jun 2, 2022 · However, these studies did not clearly distinguish the contributions of photodegradation and soil microbial degradation to mass loss ...Missing: oceans | Show results with:oceans
  99. [99]
    The widespread role of non-enzymatic reactions in cellular metabolism
    Jan 22, 2015 · Non-enzymatic reactions resemble catalytic mechanisms as found in all major enzyme classes and occur spontaneously, small molecule (e.g. metal-) ...
  100. [100]
    [37] Carbonyl assays for determination of oxidatively modified proteins
    Assay of carbonyl groups in proteins provides a convenient technique for detecting and quantifying oxidative modification of proteins.
  101. [101]
    Rapid and general profiling of protease specificity by using ... - PNAS
    Abstract. A method is presented for the preparation and use of fluorogenic peptide substrates that allows for the configuration of general substrate libraries ...
  102. [102]
    Measurement of Protease Activities Using Fluorogenic Substrates
    They are endopeptidases that require several defined amino acid residues in both N-terminal and C-terminal sides of the scissile bond. Fluorogenic Förster ...Missing: AMC seminal paper
  103. [103]
    Zymographic techniques for detection and characterization of ...
    We have presented a variety of zymographic techniques for identification and characterization of microbial proteases, using SDS-PAGE and PAGE in nondissociating
  104. [104]
    Zymography as a Research Tool in the Study of Matrix ... - NIH
    Jan 1, 2018 · The zymography technique is based on separation of proteins by nonreducing sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS–PAGE).
  105. [105]
    Monitoring proteolytic processing events by quantitative mass ...
    The powerful resolution of this technique also allows the researcher to map peptide cleavage sites upon external stimuli with high confidence and perform ...
  106. [106]
    Mass spectrometry-based proteomics strategies for protease ...
    The present review focuses on mass spectrometry-based proteomic methods for the analysis of protease cleavage sites, including the identification of the ...Missing: mapping | Show results with:mapping
  107. [107]
    High throughput screening for drugs that inhibit 3C-like protease in ...
    Jan 14, 2023 · We developed a high throughput cell-based screen that incorporates the essential viral 3C-like protease and its peptide cleavage site into a luciferase ...
  108. [108]
    A High-Throughput Screening Platform Identifies FDA-Approved ...
    Aug 6, 2024 · This high-throughput screening platform enables screening of large compound collections to discover novel small molecule SREBP inhibitors.
  109. [109]
    Tying up the loose ends: circular permutation decreases the ...
    Recombinant proteins often suffer from poor expression because of proteolysis. Existing genetic engineering or fermentation strategies work for only a subset of ...Protein Production · In Vitro Proteolysis · Results
  110. [110]
    Tackling Unwanted Proteolysis in Plant Production Hosts Used for ...
    Mar 7, 2016 · In vitro studies using recombinant proteolytic enzymes confirmed that two serine proteases, subtilisin (S8 family) and chymotrypsin (S1 ...
  111. [111]
    Super-silent FRET Sensor Enables Live Cell Imaging and Flow ...
    Sep 10, 2018 · This offered a single auto-quenching fluorophore based approach for the analysis of proteolytic activity, where amplification of signal upon ...
  112. [112]
    Enzyme Technology in the Food Industry: Molecular Mechanisms ...
    Sep 27, 2025 · Since bromelain can efficiently hydrolyze meat myofibril proteins, it is primarily utilized in the food industry for meat tenderization (Arshad ...
  113. [113]
    Molecular and Biotechnological Aspects of Microbial Proteases - PMC
    Papain, bromelain, keratinases, and ficin represent some of ... The major application of proteases in the dairy industry is in the manufacture of cheese.
  114. [114]
    Advances in protease engineering for laundry detergents
    Dec 25, 2015 · This comprehensive review discusses recent success stories in subtilisin protease engineering. Advances in protease engineering for laundry detergents
  115. [115]
    Laundry performance of subtilisin proteases - PubMed
    Effective laundry protease performance against susceptible stains depends upon both the enzyme itself and the environment in which it must work.
  116. [116]
    Protease detection in the biosensor era: A review - ScienceDirect.com
    Jan 15, 2024 · Protease biosensors may allow better comprehension of protease overexperession and potentially enable novel devices for point-of-care testing.
  117. [117]
    Enzymatic debridement with collagenase in wounds and ulcers
    Collagenase appears beneficial for wound healing and for its ability to remove necrotic or devitalised tissues. Even though studies have partially included ...
  118. [118]
    Reduction of protein degradation by use of protease-deficient ...
    In an Escherichia coli in vitro transcription/translation system, the degradation of produced proteins is often caused by endogenous proteases from E. coli ...
  119. [119]
    Microbial proteases and their applications - PMC - NIH
    Waste recycling/environmental protection. To process various forms of protein-rich waste, proteases can be used for liquid, solid, and hazardous waste.