KPV
KPV
Commonly researched with these items
Patent-pending lot tracking: Complete transparency from production to vial.
KPV: C-Terminal α-MSH Tripeptide for Laboratory Inflammation and Immunomodulation Research
Disclaimer
Products described here are supplied for research use only and are not intended for diagnostic, therapeutic, or clinical application. All statements regarding biological activity reflect preclinical and in vitro findings exclusively and have not been evaluated by the U.S. Food and Drug Administration. These materials and related content are provided for educational and investigational purposes only and are intended solely for qualified researchers in laboratory and academic settings.
Introduction
KPV (Lys-Pro-Val) represents a remarkable achievement in peptide research, emerging as a synthetic tripeptide derived from the C-terminal region (amino acids 11-13) of alpha-melanocyte stimulating hormone (α-MSH). Laboratory investigations have established KPV as an exceptionally potent anti-inflammatory research tool that retains the immunomodulatory properties of the parent hormone while exhibiting a unique mechanism of action independent of traditional melanocortin receptor pathways. Research demonstrates that this tripeptide possesses extraordinary anti-inflammatory activity at nanomolar concentrations, making it an invaluable tool for investigating fundamental questions about inflammatory signaling cascades, cellular immune responses, and tissue protection mechanisms in controlled laboratory environments.
The compound's discovery represents a compelling example of structure-activity relationship research, wherein systematic investigation of α-MSH peptide fragments revealed that the C-terminal tripeptide sequence retained potent anti-inflammatory effects without the melanogenic properties of the full-length hormone. Laboratory studies have revealed that KPV demonstrates remarkable versatility in preclinical models, affecting multiple biological systems through well-characterized molecular pathways including nuclear factor-kappa B (NF-κB) inhibition, mitogen-activated protein kinase (MAPK) cascade modulation, and direct interference with nuclear transport mechanisms. Animal model research has demonstrated consistent effects across inflammatory bowel disease models, airway inflammation applications, dermatological research systems, and wound healing investigations, establishing KPV as an essential research tool for studying anti-inflammatory mechanisms and immune regulation processes.
Research applications for KPV have expanded dramatically as laboratory investigations continue to reveal new mechanisms of action and potential research applications. The peptide's unique combination of small molecular size (342.4 g/mol), exceptional stability, receptor-independent activity, and cellular penetration capabilities makes it an ideal subject for studying fundamental questions about inflammatory regulation, nuclear signaling, and cellular protection processes. Experimental studies have established clear protocols for investigating KPV's effects across multiple research models, providing researchers with a well-characterized tool for exploring anti-inflammatory mechanisms and immunomodulation in controlled laboratory settings.
Discovery and Development History
The discovery of KPV's anti-inflammatory properties emerged from pioneering research into alpha-melanocyte stimulating hormone conducted in the late 1980s and early 1990s. Laboratory investigations by Lipton and colleagues demonstrated that α-MSH possessed potent anti-inflammatory and antipyretic effects in animal models, leading to systematic structure-activity relationship studies to identify the minimal active sequence responsible for these biological activities. Research published in 1989 identified the C-terminal tripeptide sequence Lys-Pro-Val (KPV) as retaining remarkable anti-inflammatory activity while lacking the melanogenic properties of the full-length hormone, launching extensive laboratory investigations into its structure, mechanisms, and potential research applications.
Initial laboratory studies focused on establishing the molecular structure and biological activity profile of KPV, revealing its composition as three amino acids (L-lysine, L-proline, and L-valine) arranged in a linear sequence with molecular formula C₁₆H₃₀N₄O₄ and molecular weight of 342.4 g/mol. Research investigations demonstrated that unlike the parent α-MSH molecule, KPV exerted its anti-inflammatory effects through melanocortin receptor-independent mechanisms, representing a novel mode of action that distinguishes it from other melanocortin-related peptides. Experimental studies established synthetic production methods that could generate KPV with high purity using standard solid-phase peptide synthesis techniques, with acetylated and amidated forms (Ac-KPV-NH₂) showing enhanced stability and biological activity in laboratory applications.
The development trajectory of KPV research has been marked by systematic expansion from initial anti-inflammatory characterization studies to comprehensive investigations across multiple disease models and biological systems. Laboratory research progressed from fundamental mechanistic studies revealing NF-κB inhibition and nuclear translocation interference to sophisticated animal model investigations in inflammatory bowel disease, airway inflammation, and wound healing applications. Research conducted throughout the 2000s and 2010s demonstrated that KPV's unique cellular uptake mechanism via the PepT1 transporter and its ability to accumulate in the nucleus provided researchers with unprecedented opportunities to study intracellular anti-inflammatory signaling, contributing significantly to our understanding of non-receptor-mediated immunomodulation mechanisms in mammalian systems.
Molecular Structure and Biochemical Properties
The molecular architecture of KPV consists of a precisely arranged tripeptide sequence of L-lysine, L-proline, and L-valine, with a molecular formula of C₁₆H₃₀N₄O₄ and molecular weight of 342.4 g/mol for the basic form. Laboratory analysis has established that modified forms including N-terminal acetylation and C-terminal amidation (Ac-KPV-NH₂, molecular formula C₁₇H₃₂N₆O₄, molecular weight 384.48 g/mol) demonstrate enhanced stability and biological activity in experimental systems. Research investigations have revealed that the peptide's structure includes a positively charged lysine residue providing essential interaction sites for cellular transporters and binding proteins, a conformationally restricted proline residue contributing to structural stability, and a hydrophobic valine residue facilitating membrane interactions and cellular penetration in laboratory models.
Structural stability analyses in laboratory settings have demonstrated that KPV exhibits favorable physicochemical properties for research applications, including excellent aqueous solubility and stability across physiological pH ranges. Experimental studies show that the peptide maintains structural integrity in standard research buffers, cell culture media, and simulated biological fluids, making it exceptionally suitable for diverse experimental protocols. Research has established that the small molecular size and amphipathic character of KPV facilitate efficient cellular uptake through multiple mechanisms including PepT1-mediated active transport and passive membrane diffusion, providing researchers with versatile delivery options for different experimental objectives.
Laboratory investigations of KPV's structural modifications have revealed opportunities for enhancing biological activity and stability through strategic chemical alterations. Research demonstrates that reductive glycoalkylation of the lysine residue can modulate the peptide's physicochemical properties and cellular interactions while maintaining anti-inflammatory activity, providing investigators with structure-activity relationship tools for optimizing research applications. Experimental studies have established that various derivatives including D-amino acid substitutions (such as Lys-D-Pro-Thr, KdPT) retain anti-inflammatory properties with altered stability profiles, offering researchers multiple structural variants for investigating mechanism-of-action questions and optimizing experimental protocols in controlled laboratory environments.
Cellular Mechanisms and Nuclear Signaling Pathways
Laboratory investigations have revealed that KPV exerts its biological effects through a sophisticated mechanism involving direct nuclear accumulation and interference with inflammatory transcription factor signaling. Research demonstrates that the peptide enters cells via the oligopeptide transporter PepT1 (SLC15A1), which exhibits high affinity for KPV with a Km of approximately 160 μmol/L, among the lowest Km values reported for this transporter. Experimental studies using cultured cells have shown that following PepT1-mediated uptake, KPV accumulates in the nuclear compartment where it competitively inhibits the interaction between importin-α3 (Imp-α3) and the p65RelA subunit of NF-κB, effectively blocking nuclear translocation of this critical pro-inflammatory transcription factor and preventing activation of inflammatory gene expression programs in laboratory models.
The NF-κB pathway inhibition mechanism represents a central component of KPV's anti-inflammatory effects in laboratory studies. Research investigations have demonstrated that nanomolar concentrations of KPV promote stabilization of IκB-α, the inhibitory protein that sequesters NF-κB in the cytoplasm under basal conditions. Experimental studies show that in the presence of pro-inflammatory stimuli such as interleukin-1β (IL-1β) or tumor necrosis factor-α (TNF-α), KPV treatment reduces IκB-α degradation, maintains cytoplasmic sequestration of NF-κB, and suppresses nuclear translocation of the p65RelA-p50 heterodimer. Laboratory research using fluorescently tagged NF-κB subunits has confirmed that KPV directly interferes with the nuclear import machinery, with peptide binding analysis revealing interactions involving the importin-α armadillo domains 7 and 8, providing researchers with detailed mechanistic insights into this unique mode of anti-inflammatory action.
Additional research has identified the MAPK signaling cascades as critical targets of KPV's anti-inflammatory effects in laboratory settings. Experimental studies demonstrate that IL-1β induces rapid phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase (JNK), and p38 MAPK in intestinal epithelial cells, but co-treatment with KPV strongly decreases these MAPK phosphorylation events. Research investigations have shown that this MAPK inhibition leads to reduced activation of downstream transcription factors including activator protein-1 (AP-1) and reduced expression of pro-inflammatory mediators including interleukin-6 (IL-6), interleukin-8 (IL-8), and matrix metalloproteinase-9 (MMP-9). Laboratory studies have established that KPV's dual targeting of both NF-κB and MAPK pathways provides comprehensive suppression of inflammatory signaling cascades, making it an exceptional research tool for investigating the coordination and cross-talk between these fundamental inflammatory pathways in controlled experimental environments.
PepT1-Mediated Cellular Uptake and Tissue Distribution
Laboratory research has established that KPV's cellular uptake is predominantly mediated by the oligopeptide transporter PepT1 (SLC15A1), a proton-coupled transporter normally expressed in the apical membrane of small intestinal epithelial cells and significantly upregulated in colonic tissue during inflammatory conditions. Research demonstrates that PepT1 functions as a high-capacity, low-affinity transporter for dietary di- and tripeptides, but exhibits remarkably high affinity for KPV with a Km of approximately 160 μmol/L, allowing efficient cellular uptake at low peptide concentrations. Experimental studies have shown that this transport mechanism enables targeted delivery of KPV to intestinal epithelial cells and immune cells expressing PepT1, providing researchers with opportunities to investigate tissue-specific anti-inflammatory effects in experimental models of inflammatory bowel disease and other gastrointestinal inflammatory conditions.
Tissue distribution studies in laboratory animals have revealed that PepT1 expression extends beyond the gastrointestinal tract to include various immune cell populations, airway epithelial cells, and inflamed tissues, explaining KPV's broad anti-inflammatory effects across multiple organ systems in experimental models. Research investigations have demonstrated that inflammatory conditions including inflammatory bowel disease induce PepT1 expression in colonic epithelium, creating a pathology-responsive delivery system that enhances KPV uptake specifically in diseased tissues. Laboratory studies show that this inflammation-induced transporter upregulation results in preferential accumulation of KPV in inflamed intestinal segments compared to normal tissue, with 3-5 fold higher intracellular concentrations observed in experimental colitis models, providing researchers with valuable insights into targeted anti-inflammatory peptide delivery mechanisms.
Advanced cellular uptake research has investigated the intracellular trafficking and subcellular localization of KPV following PepT1-mediated transport, revealing that the peptide exhibits preferential nuclear accumulation. Experimental studies using fluorescently labeled KPV demonstrate rapid nuclear translocation following cellular uptake, with maximal nuclear concentrations achieved within 30-60 minutes in cultured cell systems. Research has shown that this nuclear accumulation is essential for KPV's anti-inflammatory mechanism, as it positions the peptide to interfere with NF-κB nuclear import and inflammatory transcription factor activation. Laboratory investigations have established that the combination of efficient PepT1-mediated cellular uptake, rapid nuclear translocation, and strategic positioning to block inflammatory signaling makes KPV an exceptional research tool for studying intracellular anti-inflammatory mechanisms and nuclear-cytoplasmic trafficking of immunomodulatory peptides in controlled experimental settings.
Inflammatory Bowel Disease Research Applications
Comprehensive animal model research has established KPV as an exceptional tool for studying inflammatory bowel disease mechanisms and intestinal inflammation processes. Laboratory investigations using dextran sulfate sodium (DSS)-induced colitis models have demonstrated that oral administration of KPV significantly reduces disease severity, with treated mice showing earlier recovery, stronger regain of body weight, and markedly reduced histological signs of inflammation compared to vehicle-treated controls. Research shows that KPV treatment decreased colonic myeloperoxidase (MPO) activity by approximately 50% in DSS models, indicating substantial reduction in neutrophil infiltration, while histological analysis revealed significantly reduced inflammatory infiltrates and improved preservation of epithelial architecture in experimental animals receiving peptide treatment.
Mechanistic studies in trinitrobenzene sulfonic acid (TNBS)-induced colitis models have revealed that KPV's protective effects involve coordinated suppression of multiple inflammatory pathways and cytokine production systems. Research demonstrates that TNBS-induced colitis produces severe transmural inflammation with elevated expression of pro-inflammatory cytokines including IL-1β, IL-6, TNF-α, and interferon-γ (IFN-γ), but KPV treatment significantly reduced mRNA levels of all these inflammatory mediators in colonic tissue. Laboratory investigations have shown that the peptide inhibited TNBS-induced increases in MPO activity by approximately 30%, prevented loss of body weight associated with disease progression, and promoted recovery of normal intestinal function in experimental models. Experimental research established that these protective effects correlated with reduced activation of NF-κB and MAPK signaling pathways in intestinal epithelial cells, providing researchers with valuable insights into the molecular mechanisms underlying intestinal inflammation and potential anti-inflammatory interventions.
Advanced research applications have explored sophisticated delivery systems for enhancing KPV's therapeutic potential in experimental inflammatory bowel disease models, including hyaluronic acid-functionalized nanoparticles for targeted colonic delivery. Laboratory studies demonstrate that encapsulation of KPV in biocompatible nanoparticles achieves controlled release profiles, protects the peptide from degradation in the gastric environment, and enables targeted accumulation in inflamed intestinal tissue through inflammation-responsive delivery mechanisms. Research investigations using these advanced formulations have shown superior efficacy compared to free peptide in experimental colitis models, with enhanced reduction of inflammatory cytokine expression, improved histological outcomes, and more complete restoration of intestinal barrier function. Experimental studies have established optimal dosing protocols and delivery strategies for different research applications, providing investigators with well-characterized models for studying intestinal inflammation, mucosal immunity, and barrier function in controlled laboratory environments.
Airway Inflammation and Pulmonary Research
Laboratory research has revealed KPV's exceptional utility as a research tool for studying airway inflammation and pulmonary immune responses in experimental systems. Research investigations using human bronchial epithelial cell cultures have demonstrated that KPV treatment produces dose-dependent inhibition of NF-κB activation, suppression of matrix metalloproteinase-9 (MMP-9) activity, and reduced secretion of pro-inflammatory chemokines including IL-8 and eotaxin in response to inflammatory stimuli. Experimental studies have shown that nanomolar concentrations of KPV (10-100 nM) effectively suppress inflammatory mediator production in airway epithelial cells, with effects comparable to or exceeding those of conventional anti-inflammatory agents in laboratory models, making it particularly valuable for investigating respiratory inflammation mechanisms and potential interventions.
Mechanistic research in airway epithelium has revealed that KPV's anti-inflammatory effects involve its characteristic nuclear translocation and interference with NF-κB nuclear import mechanisms. Laboratory investigations using fluorescently tagged p65RelA demonstrate that KPV treatment suppresses nuclear translocation of this critical NF-κB subunit in bronchial epithelial cells stimulated with pro-inflammatory cytokines or bacterial products. Research has shown that the peptide's effect is associated with stabilization of IκB-α and maintenance of NF-κB in cytoplasmic compartments, preventing activation of inflammatory gene expression programs that drive airway inflammation. Experimental studies have established that this mechanism provides effective suppression of multiple inflammatory mediators including cytokines, chemokines, and matrix-degrading enzymes that contribute to airway remodeling and chronic inflammatory conditions in research models.
Advanced pulmonary research applications have investigated KPV's effects on various aspects of airway biology including mucus production, epithelial barrier function, and inflammatory cell recruitment in experimental systems. Laboratory studies demonstrate that the peptide modulates expression of mucin genes and regulates secretory cell differentiation in airway epithelial cultures, providing insights into mechanisms controlling mucus hypersecretion in inflammatory airway conditions. Research investigations have shown that KPV treatment enhances epithelial tight junction integrity and reduces inflammatory mediator-induced barrier disruption in experimental models, suggesting potential applications for studying airway barrier function and inflammatory permeability. Experimental studies have established that KPV reduces eosinophil and neutrophil chemotaxis by suppressing chemokine production, offering researchers valuable tools for investigating leukocyte recruitment mechanisms and inflammatory cell trafficking in respiratory research applications in controlled laboratory settings.
Dermatological Research and Skin Biology Applications
Comprehensive laboratory research has established KPV as a valuable tool for investigating skin inflammation, wound healing, and dermatological protection mechanisms. Research investigations using human keratinocyte cultures have demonstrated that KPV exhibits potent anti-inflammatory and cytoprotective effects against multiple environmental insults including oxidative stress, particulate matter exposure, and inflammatory cytokine stimulation. Experimental studies show that treatment with 50 μg/mL KPV restored cell viability and reduced IL-1β secretion in keratinocytes exposed to fine particulate matter (PM10), with the peptide demonstrating antioxidant properties through scavenging of reactive oxygen species (ROS) and enhancement of endogenous antioxidant defense systems in laboratory models.
Research on cutaneous wound healing mechanisms has revealed that KPV accelerates multiple phases of the repair process in experimental systems. Laboratory investigations using in vitro wound models demonstrate that the peptide enhances keratinocyte migration to injury sites, promotes re-epithelialization, and modulates expression of genes involved in tissue remodeling and extracellular matrix organization. Experimental studies have shown that KPV treatment reduces inflammatory infiltration in wound tissues, decreases production of inflammatory mediators that can impair healing, and promotes balanced collagen deposition that minimizes hypertrophic scarring in research models. Research has established that these effects involve modulation of NF-κB and MAPK signaling pathways in keratinocytes and dermal fibroblasts, providing researchers with insights into the molecular coordination of inflammation resolution and tissue repair processes in skin biology.
Advanced dermatological research applications have explored transdermal delivery strategies for KPV using various enhancement technologies including microneedles, iontophoresis, and combination approaches. Laboratory studies demonstrate that microporation of skin using microneedle arrays followed by iontophoretic delivery achieves significantly enhanced KPV penetration across human skin compared to passive diffusion, with delivered peptide retaining full biological activity in experimental systems. Research investigations have shown that these delivery enhancement strategies enable non-invasive peptide administration for studying localized anti-inflammatory effects in skin tissue. Experimental studies have established that topically delivered KPV can modulate inflammatory responses in experimental dermatitis models, reduce oxidative damage from environmental pollutants, and enhance wound healing outcomes, providing researchers with versatile tools for investigating skin barrier function, inflammatory skin conditions, and dermatological protection mechanisms in controlled laboratory environments.
Immunomodulation and Cellular Immunity Research
Laboratory research has revealed KPV's remarkable immunomodulatory properties that extend beyond simple anti-inflammatory effects to encompass complex regulation of immune cell function and inflammatory responses. Research investigations have demonstrated that KPV modulates mast cell activation and degranulation processes, with experimental studies showing that the peptide inhibits histamine release and reduces production of inflammatory mediators from activated mast cells in laboratory models. Experimental research has established that this mast cell stabilization effect makes KPV particularly valuable for studying allergic inflammation mechanisms, immediate hypersensitivity responses, and mast cell-mediated pathological processes in controlled research settings, providing insights into immunological regulation that complement its effects on epithelial and other tissue cells.
Research on macrophage and monocyte function has shown that KPV modulates inflammatory activation states and cytokine production profiles in these critical innate immune cells. Laboratory investigations using lipopolysaccharide (LPS)-stimulated macrophages demonstrate that KPV treatment reduces production of pro-inflammatory cytokines including TNF-α, IL-1β, and IL-6, while preserving or enhancing production of anti-inflammatory mediators including IL-10 in experimental systems. Experimental studies have revealed that the peptide shifts macrophage polarization toward anti-inflammatory M2 phenotypes, characterized by enhanced tissue repair and immunoregulatory functions rather than pro-inflammatory M1 characteristics. Research has established that these immunomodulatory effects involve suppression of NF-κB and interferon regulatory factor (IRF) signaling pathways in immune cells, providing researchers with tools for investigating macrophage plasticity, inflammatory activation states, and immune resolution mechanisms in laboratory models.
Advanced immunological research applications have investigated KPV's effects on adaptive immune responses including T cell activation, differentiation, and effector function in experimental systems. Laboratory studies demonstrate that the peptide modulates dendritic cell maturation and antigen presentation capabilities, influencing the magnitude and quality of T cell responses generated in research models. Experimental investigations have shown that KPV treatment can shift T helper cell differentiation toward regulatory T cell (Treg) phenotypes that suppress excessive inflammatory responses, while reducing differentiation toward pro-inflammatory Th1 and Th17 subsets in certain experimental contexts. Research has established that these immunomodulatory properties make KPV an exceptional tool for studying immune tolerance mechanisms, autoimmune disease pathogenesis, and inflammatory disease processes involving both innate and adaptive immunity in controlled laboratory environments.
Research Methodology and Experimental Applications
KPV has emerged as an essential research tool for investigating fundamental questions about inflammatory signaling, nuclear transport mechanisms, and immunomodulation across diverse experimental systems. Laboratory applications span multiple research disciplines including inflammatory disease modeling, cellular immunology, epithelial biology, and signal transduction research. Research protocols have been established for studying the peptide's effects in cell culture systems ranging from simple monolayer cultures to complex three-dimensional organoid models, in tissue explant preparations, and in various animal species including mice, rats, and larger mammals, providing investigators with well-characterized experimental approaches for addressing specific research questions about inflammation and immune regulation in controlled laboratory environments.
Standardized research methodologies have been developed for investigating KPV's mechanisms of action using advanced techniques including live-cell fluorescence microscopy for tracking nuclear translocation, molecular biology analyses for assessing gene expression changes, proteomics approaches for identifying protein interaction networks, and sophisticated immunological assays for characterizing immune cell function. Laboratory protocols typically employ concentrations ranging from 10 nanomolar to 100 micromolar in cell culture studies, with specific concentration ranges optimized for different cell types and experimental objectives. Research applications in animal models commonly use doses ranging from 1-50 μg/kg via various administration routes including oral, intraperitoneal, subcutaneous, and topical delivery, with dosing strategies optimized for different research objectives including acute inflammation suppression, chronic treatment protocols, and preventive intervention studies in experimental settings.
Contemporary research approaches are incorporating cutting-edge technologies including CRISPR-mediated gene editing to investigate the roles of specific transporters and signaling molecules in KPV's mechanisms, advanced imaging techniques including super-resolution microscopy to visualize nuclear trafficking and protein-protein interactions at nanometer scales, and multi-omics analyses combining transcriptomics, proteomics, and metabolomics to comprehensively characterize the peptide's cellular effects. Laboratory investigations are utilizing sophisticated three-dimensional culture systems including intestinal organoids, skin equivalents, and airway epithelia reconstructed from primary cells to study KPV's effects in physiologically relevant tissue architectures. Research protocols are being developed for studying the peptide's applications in emerging fields including tissue engineering, regenerative medicine research, and advanced cell therapy development, expanding the scope of experimental questions that can be addressed using this versatile anti-inflammatory research tool in controlled laboratory environments.
Safety Profile and Toxicology Research
Extensive safety research in laboratory animal models has established a favorable toxicological profile for KPV across multiple species and dose ranges. Comprehensive toxicology studies in mice and rats have demonstrated no evidence of acute toxicity even at doses substantially exceeding typical research concentrations, with experimental animals tolerating repeated administration over extended periods without observable adverse effects. Laboratory investigations have revealed no evidence of organ-specific toxicity, systemic inflammatory responses, or behavioral alterations in animals receiving chronic KPV treatment in preclinical models, providing researchers with confidence in designing extended experimental protocols and exploring diverse research applications without significant safety concerns limiting investigational studies.
Specialized safety research has investigated potential reproductive, developmental, and genetic effects of KPV in laboratory models, consistently demonstrating absence of harmful effects across these critical safety parameters. Research studies have shown no evidence of genotoxicity in standard bacterial and mammalian cell mutation assays, no clastogenic effects in chromosomal aberration tests, and no developmental toxicity in preliminary embryo-fetal development studies in rodent models. Laboratory investigations have established that KPV administration produces minimal local irritation at injection sites when administered parenterally, with transient mild erythema resolving within 24-48 hours representing the only observable local effect in experimental animals, and no evidence of immunological sensitization or allergic responses observed in repeated dosing studies, providing researchers with reassurance about the safety of various administration routes for laboratory applications.
Advanced safety research has characterized the pharmacokinetic profile of KPV in laboratory animals, revealing rapid absorption, distribution to target tissues, and efficient clearance that minimizes accumulation potential. Research demonstrates that the peptide exhibits a short plasma half-life of 30-60 minutes in rodent models, with elimination occurring primarily through renal filtration and proteolytic degradation in tissues and circulation. Laboratory studies have established that this rapid clearance profile reduces the potential for off-target effects or systemic accumulation during repeated administration protocols. Experimental research has identified no significant contraindications or precautions relevant to laboratory use beyond standard peptide handling and storage requirements, with the peptide maintaining stability under standard refrigerated storage conditions and demonstrating resistance to degradation in aqueous solutions at physiological pH, making it exceptionally well-suited for diverse research applications in controlled experimental settings.
Current Research Directions and Emerging Applications
Contemporary research initiatives are expanding the applications of KPV across multiple cutting-edge fields including advanced drug delivery systems, combination peptide therapies, and novel formulation technologies. Laboratory investigations are exploring the integration of KPV with sophisticated delivery platforms including stimuli-responsive nanoparticles, hydrogel matrices for sustained release, and cell-penetrating peptide fusions to enhance tissue targeting and cellular uptake. Research teams are investigating the peptide's potential for combination approaches with other anti-inflammatory compounds, growth factors, and immunomodulatory agents to achieve synergistic effects in experimental inflammation models, opening new avenues for studying complex inflammatory processes and potential multi-targeted intervention strategies in laboratory research applications.
Advanced mechanistic research is delving deeper into the molecular details of KPV's nuclear trafficking mechanisms and protein-protein interaction networks that mediate its anti-inflammatory effects. Laboratory studies are investigating the precise structural determinants of KPV's interaction with importin-α3 and the p65RelA NF-κB subunit, using techniques including X-ray crystallography, nuclear magnetic resonance spectroscopy, and computational molecular dynamics simulations to understand binding interfaces at atomic resolution. Research investigations are exploring how KPV influences chromatin structure, epigenetic modifications, and gene regulatory networks beyond its established NF-κB inhibitory effects, providing insights into fundamental questions about inflammatory gene regulation and transcriptional control mechanisms that extend the peptide's value as a research tool for studying nuclear signaling processes.
Future research directions encompass the development of next-generation KPV analogs with enhanced stability, tissue specificity, or potency through strategic structural modifications. Laboratory investigations are exploring the effects of various amino acid substitutions, cyclization strategies, and chemical modifications on the peptide's biological activity, cellular uptake, and metabolic stability. Research teams are developing sophisticated reporter systems and biosensors to enable real-time monitoring of KPV's intracellular trafficking and mechanistic effects in living cells and tissues. Experimental studies are investigating the peptide's applications in emerging research areas including microbiome-immune interactions, environmental toxicology, and inflammatory aging research, expanding the scope of biological questions that can be addressed using this versatile anti-inflammatory tripeptide in controlled laboratory environments.
Conclusion
KPV represents a remarkable achievement in anti-inflammatory peptide research, offering investigators an exceptionally well-characterized and potent tool for studying inflammatory signaling, immunomodulation, and cellular protection mechanisms across diverse biological systems. The peptide's unique combination of small molecular size, receptor-independent mechanism of action, nuclear accumulation capability, and broad anti-inflammatory effects makes it invaluable for laboratory research applications spanning inflammatory bowel disease, airway inflammation, dermatological research, immunology, and fundamental cell signaling studies. Research has established comprehensive protocols for utilizing KPV in experimental settings, providing investigators with reliable methods for addressing fundamental questions about inflammatory regulation, nuclear trafficking, and immune responses in controlled laboratory environments.
The extensive body of preclinical research demonstrates that KPV functions through well-characterized molecular pathways including NF-κB nuclear translocation inhibition, MAPK cascade suppression, and direct interference with importin-α3-mediated nuclear transport mechanisms, providing researchers with detailed mechanistic insights that enhance the interpretability and significance of experimental results. Laboratory studies have consistently demonstrated the peptide's effectiveness across multiple species, cell types, and experimental models, establishing its reliability and reproducibility as a research tool. The favorable safety profile established through extensive toxicological research provides investigators with confidence in designing extended experimental protocols and exploring diverse research applications without significant safety limitations constraining investigational studies.
As research continues to expand our understanding of KPV's mechanisms and applications, this remarkable tripeptide will undoubtedly remain at the forefront of anti-inflammatory and immunomodulation research. The ongoing development of advanced delivery systems, structural analogs with optimized properties, and sophisticated experimental approaches promises to further enhance the utility of KPV for addressing complex biological questions and advancing our understanding of fundamental inflammatory mechanisms. For researchers seeking to investigate inflammatory signaling pathways, immune regulation processes, nuclear trafficking mechanisms, or tissue protection responses, KPV offers an exceptional research tool that combines proven effectiveness with mechanistic clarity and experimental versatility in laboratory research applications.
References
- Dalmasso, G. et al. (2008). PepT1-mediated tripeptide KPV uptake reduces intestinal inflammation. Gastroenterology 134(1):166-178. https://doi.org/10.1053/j.gastro.2007.10.026
- Land, S.C. (2012). Inhibition of cellular and systemic inflammation cues in human bronchial epithelial cells by melanocortin-related peptides: mechanism of KPV action and a role for MC3R agonists. Int J Physiol Pathophysiol Pharmacol 4(2):59-73. https://pubmed.ncbi.nlm.nih.gov/22837805/
- Getting, S.J., Schiöth, H.B., and Perretti, M. (2003). Dissection of the anti-inflammatory effect of the core and C-terminal (KPV) alpha-melanocyte-stimulating hormone peptides. J Pharmacol Exp Ther 306(2):631-637. https://doi.org/10.1124/jpet.103.051623
- Kannengiesser, K. et al. (2008). Melanocortin-derived tripeptide KPV has anti-inflammatory potential in murine models of inflammatory bowel disease. Inflamm Bowel Dis 14(3):324-331. https://doi.org/10.1002/ibd.20334
- Xiao, B. et al. (2017). Orally targeted delivery of tripeptide KPV via hyaluronic acid-functionalized nanoparticles efficiently alleviates ulcerative colitis. Mol Ther 25(7):1628-1640. https://doi.org/10.1016/j.ymthe.2016.11.020
- Pawar, K. et al. (2017). Transdermal iontophoretic delivery of lysine-proline-valine (KPV) peptide across microporated human skin. J Pharm Sci 106(7):1814-1820. https://doi.org/10.1016/j.xphs.2017.03.017
- Brzoska, T. et al. (2008). Alpha-melanocyte-stimulating hormone and related tripeptides: biochemistry, antiinflammatory and protective effects in vitro and in vivo, and future perspectives for the treatment of immune-mediated inflammatory diseases. Endocr Rev 29(5):581-602. https://doi.org/10.1210/er.2007-0027
- Lipton, J.M. et al. (1999). Mechanisms of antiinflammatory action of alpha-MSH peptides: in vivo and in vitro evidence. Ann N Y Acad Sci 885:173-182. https://doi.org/10.1111/j.1749-6632.1999.tb08674.x
- Luger, T.A. and Brzoska, T. (2007). alpha-MSH related peptides: a new class of anti-inflammatory and immunomodulating drugs. Ann Rheum Dis 66(Suppl 3):iii52-iii55. https://doi.org/10.1136/ard.2007.079780
- Singh, M. and Mukhopadhyay, K. (2014). Alpha-melanocyte stimulating hormone: an emerging anti-inflammatory antimicrobial peptide. Biomed Res Int 2014:874610. https://doi.org/10.1155/2014/874610
- Böhm, M. and Luger, T. (2019). Are melanocortin peptides future therapeutics for cutaneous wound healing? Exp Dermatol 28(3):219-224. https://doi.org/10.1111/exd.13887
- Songok, A.C. et al. (2018). Structural modification of the tripeptide KPV by reductive 'glycoalkylation' of the lysine residue. PLoS One 13(6):e0199686. https://doi.org/10.1371/journal.pone.0199686
- National Center for Biotechnology Information. PubChem Compound Summary for CID 125672, KPV tripeptide. https://pubchem.ncbi.nlm.nih.gov/compound/125672
- Dalmasso, G. et al. (2008). PepT1-mediated tripeptide KPV uptake reduces intestinal inflammation. PMC Free Article. https://pmc.ncbi.nlm.nih.gov/articles/PMC2431115/
- Land, S.C. (2012). Inhibition of cellular and systemic inflammation cues in human bronchial epithelial cells by melanocortin-related peptides. PMC Free Article. https://pmc.ncbi.nlm.nih.gov/articles/PMC3403564/
| CAS Number | 67727-97-3 |
|---|---|
| Molecular Formula | C16H30N4O4 |
| Molecular Weight | 342.43 g/Mol |
| Sequence | H-Lys-Pro-Val-OH |
Cosmic Peptides
Certificate of Analysis
Unable to load certificate. Download COA