Vesugen 20mg (Bioregulator Peptide)


Price:
120
Stock:
In stock

Description

Research Dossier on Vesugen

(KED; Lys–Glu–Asp)

(Bioregulator Peptide)


Classification & Molecular Identity

Amino acid sequence, molecular weight, structural motifs

Vesugen is a synthetic ultrashort tripeptide with the sequence Lys–Glu–Asp (KED). Analytical entries list a molecular formula of C₁₅H₂₆N₄O₈ and molecular weight390.39 g·mol⁻¹ (free acid). The tripeptide is strongly polar/ionic (one basic and two acidic residues), which influences aqueous solubility and favors electrostatic interactions with polyanionic DNA and acidic protein surfaces in docking models (see Mechanisms). Commercial research-grade listings corroborate the sequence and formula and cross-reference PubChem CID 87571363. canlabintl.com+1

Although Vesugen is frequently described as a vascular wall–derived motif, it is not a direct fragment of a single endogenous protein; rather, it belongs to the family of organ-specific “peptide bioregulators” developed by the Russian school of short-peptide research (Khavinson and colleagues). Within that taxonomy, KED is attributed to the vascularsystem and is often grouped with other 2–4-mer peptides such as AEDG (Epitalon), EDR (Pinealon), KE (Thymogen), and KED (Vilon), each investigated for tissue-directed regulatory activity in vitro/in vivo. PMC

Discovery history (lab, year, species)

Work on organ-specific short peptides dates to the late 20th century in the USSR/Russia, where peptide fractions were refined, and defined short sequences were synthesized to probe gene-regulatory and cell-protective effects. In English-language, peer-reviewed sources, Vesugen/KED appears in mechanistic and small translational studies from the 2010s onward, focused on vascular endothelium and cell-aging paradigms. PubMed

Endogenous vs synthetic origin

  • Endogenous: There is no evidence that Lys–Glu–Asp circulates as a discrete endogenous hormone.

  • Synthetic (research-grade): Vesugen is manufactured by solid-phase peptide synthesis; most bench studies use H-Lys-Glu-Asp-OH with ≥95% purity (HPLC/MS identity typically reported in methods). canlabintl.com

Homologs, analogs, derivatives

  • Vilon (Lys–Glu)—a dipeptide frequently used as a vascular/immune comparator.

  • Pinealon (Glu–Asp–Arg; EDR)—a tripeptide investigated in neuronal and oxidative-stress models.

  • Epitalon (Ala–Glu–Asp–Gly; AEDG)—a tetrapeptide linked to chromatin and telomere pathways in cell/invertebrate models.
    These homologs often appear alongside KED in comparative studies on dendritogenesis, antihypoxic effects, and gene-promoter docking. PMC+1


Historical Development & Research Trajectory

Key milestones in discovery and study

  1. Epigenetic/gene-promoter studies (2014): In vascular endothelial cell cultures from young and old animals, KEDincreased Ki-67 expression (a proliferation marker), and molecular docking suggested direct interaction with the MKI67 (Ki-67) promoter region—one of the earliest English-abstracted mechanistic links between Vesugen and cell-cycle gene regulation. PubMed

  2. Antihypoxic stress (2008): In a hypobaric hypoxia model, Vesugen, Pinealon (EDR), Epitalon (AEDG) and Vilon (KE) all displayed antihypoxic activity; EDR was strongest, but KED showed protective trends, supporting a shared stress-response motif among short peptides. PubMed

  3. Cellular aging/induced neurons (2024): In human fibroblast-derived induced neurons, panels of short peptides—including Vesugen/KED—partially protected against age-associated phenotypes and stimulated dendritogenesis (spine metrics, neurite outgrowth), extending prior AD-model spine data to a human-derivedcellular system. PMC

  4. Human geropreventive pilot (2015): An open-label study (n=32, age 41–83) reported that Pinealon and Vesugencourses improved biological-age indices (CNS-related composite metrics), with Vesugen showing somewhat larger effects; however, the same study noted pro-oxidant chemiluminescence signals and decreases in CD34⁺ hematopoietic cells—an ambiguous safety/PD signature requiring caution and replication. PubMed

  5. Urologic microvascular pilot (2014): In men with vasculogenic erectile dysfunction, a Russian-language study (English abstract) examined Vezugen as a vasoactive tripeptide adjunct; methodology details are sparse, but the report underscores Vesugen’s vascular positioning. PubMed

Paradigm shifts & controversies

  • From vasoprotective “bioregulator” to gene-targeting micro-ligand. The research language evolved from broad vascular bioregulation to promoter-level hypotheses where short peptides dock to DNA/RNA motifs and modulate transcription/translation of cell-cycle and stress genes—provocative, but not yet proven by gold-standard in vivo chromatin assays for KED. PubMed

  • Evidence quality & generalizability. A substantial fraction of Vesugen work originates from a limited set of laboratories; many clinical-style reports are small, open-label, or non-randomized. Independent multicenter replication under contemporary CONSORT standards remains limited. PubMed

Evolution of scientific interest

The thematic arc has moved from vascular endothelium (Ki-67/proliferation) → hypoxia toleranceneuroplasticity/dendritic spines (aging and AD-adjacent cell models) → small pilot human studies in aging, with exploratory clinical signals counter-balanced by safety/hematopoietic questions that remain open. PubMed+2PubMed+2


Mechanisms of Action

Primary and secondary receptor interactions

No canonical cell-surface receptor has been identified for KED. The working hypotheses in peer-reviewed reports include:

  1. Nucleic-acid docking (promoter-level “micro-ligand” action). Molecular docking suggested that KED can bind the MKI67 promoter and enhance expression of Ki-67, a proliferation index suppressed with vascular aging. While in silico and in-vitro correlations are intriguing, direct proof of in vivo promoter occupancy (e.g., peptide-ChIP with orthogonal validation) is Not established. PubMed

  2. Chromatin & epigenetic modulation (class-level). Reviews of short peptides (including KED) posit sequence-specific interactions with DNA/RNA and histone or transcription factor complexes, enabling tissue-selectivechanges in gene programs (e.g., proliferation, cytoprotection). Such class claims are supported by several peptide exemplars, but mechanistic granularity for KED per se is incomplete. PMC

  3. Membrane/cytoplasmic signaling interfaces. Given the peptide’s size/charge, endocytosis or transporter-mediated uptake into endothelial or neuronal cells is plausible; downstream effects (ERK/MAPK, redox enzymes) are inferred, not mapped comprehensively for KED.

Evidence grading (mechanism):

  • A: KED increases Ki-67 expression and proliferation metrics in vascular endothelial cultures; docking supports proximity to MKI67 promoter. PubMed

  • B: Class-level peptide-DNA interaction and neuronal dendritogenesis effects (human iNeurons) provide biological plausibility. PMC

  • C: Direct identification of primary targets, binding constants, and in vivo promoter engagement for KED are Not established.

Intracellular signaling pathways

  • Cell-cycle/proliferation: Up-regulation of Ki-67 (MKI67), a marker critical for cycling cells (G1, S, G2, M), suggests that KED can re-activate proliferation programs in senescent or aged vascular endothelial cells. PubMed

  • Antihypoxic defenses: In hypobaric models, short peptides (including KED) reduced hypoxia-induced damage; downstream redox and mitochondrial pathways (e.g., Nrf2, ROS scavenging) are postulated but not fully delineated for KED. PubMed

  • Neuroplasticity: In AD-relevant and aging models, short peptides (including KED) increased dendritic spine density/dendritogenesis, implying effects on cytoskeletal dynamics, synaptic gene sets (e.g., calmodulin/CaMKII, PSD-95 networks), or CREB signaling; precise KED-specific pathway maps remain incomplete. PMC

CNS vs peripheral effects

  • Peripheral (vascular/endothelial)—the strongest mechanistic footprint (Ki-67, endothelial proliferation).

  • CNS—emerges from aging/AD cell models and induced neuron work; direct in vivo CNS pharmacology for KED is limited.

Hormonal, metabolic, immune interactions

Published studies do not demonstrate direct endocrine receptor activity for KED. In limited immune-cell work from the short-peptide literature, KED has been associated with proliferation of specific cell subsets or no effect in context-specific assays (e.g., pineal immune cells: increased proliferation but not differentiation). PubMed


Pharmacokinetics & Stability

ADME profile

Human PK for Lys–Glu–Asp is not reported in peer-reviewed English-language sources. By analogy to other 2–4-merpeptides:

  • Absorption: Parenteral administration is expected to yield rapid systemic exposure; oral bioavailability is typically low for free peptides of this size (unless protected). Intranasal transport has not been documented for KED. Status: Not established.

  • Distribution: With Mr ≈ 390 g·mol⁻¹, KED likely partitions to extracellular fluid and is filtered readily by the kidney. Tissue uptake (endocytosis/transporter) is plausible but unquantified. Status: Not established.

  • Metabolism: Exopeptidases and endopeptidases degrade short peptides to constituent amino acids; half-life is typically minutes to tens of minutes unless stabilized. Status: Not established for KED specifically.

  • Elimination: Expected renal clearance of intact/fragment peptides; no human t½ reported. Status: Not established.

Plasma half-life & degradation pathways

No validated for KED in mammals has been published in English; the short-peptide class is generally short-livedunless encapsulated/modified. Status: Not established.

Stability in vitro & in vivo

In cell studies, KED remains bioactive across typical incubation windows (hours). In vivo stability has not been quantified in peer-reviewed English sources. Status: Not established.

Storage/reconstitution considerations

Peer-reviewed CMC data are sparse. Research suppliers typically provide HPLC/MS certificates and recommend −20 °Cdry storage; solution stability depends on pH, buffer, and microbial control (vendor-specific).


Preclinical Evidence

Vascular endothelial biology & epigenetic regulation

  • Ki-67 restoration in aging endothelium. In tissue-specific cell cultures and dissociated vascular endothelialcultures from young and old animals, Vesugen/KED increased Ki-67 protein—reduced with age—implicating partial rejuvenation of proliferative capacity. Docking suggested KED–DNA contacts at the MKI67 promoter, consistent with transcriptional activation. Concentration/time details follow cell-biology norms; doses varied by experiment. PubMed

  • Gene-regulatory review (systematic). A comprehensive review of peptide regulation of gene expressioncatalogs short peptides, including KED, as putative sequence-specific DNA/RNA ligands that fine-tune transcription/translation in tissue-specific fashion; while the review compiles many datasets, it also acknowledges remaining gaps in target validation. PMC

Antihypoxic properties

  • In a hypobaric hypoxia model, KED (with EDR, AEDG, KE) showed antihypoxic activity; EDR appeared strongest, but KED was protective vs controls—supporting a stress-response role across the short-peptide panel. Investigational dosing used in study: the paper reports experimental injection schedules for each peptide in rodents; exact numeric details are in the methods. PubMed

Neuroplasticity/dendritic architecture (aging & AD models)

  • A 2024 study using human iNeurons reported that short peptides—including KED—partially protected against age-related changes (e.g., nuclear morphology, mitochondrial potential) and stimulated dendritogenesis; the authors propose epigenetic/transcriptional mechanisms. Investigational concentrations: low-to-mid micromolar in vitro for 24–72 h. PMC

Pineal immune cells—proliferation vs differentiation

  • In rat pineal-derived immune cell cultures, KED did not change differentiation patterns toward T-helper, CTL, or B lineage (unlike Vilon and Epitalon), but increased proliferative potential—a useful negative/positive control indicating context-specific effects across organs. PubMed

Urologic microvascular context (vasculogenic ED)

  • A PubMed-indexed Russian clinical paper (English abstract) investigated Vezugen in atherosclerosis-linkedvasculogenic erectile dysfunction; the study sought improvement in microvascular parameters. Methodological granularity is limited in the abstract, but it underscores KED’s vascular positioning. PubMed

Dose ranges tested (illustrative; all investigational)

  • Cell culture: KED typically 1–50 µM, 24–72 h, depending on endpoint (proliferation, gene expression). (Ranges inferred from general short-peptide methods; specific KED concentrations should be checked in the original endothelial/iNeuron protocols.)

  • Rodent hypoxia: The antihypoxic paper describes peptide injection schedules in rodent models; numeric translation to human dosing is not appropriate (species-specific PK). PubMed

Comparative efficacy/safety (preclinical)

  • Efficacy: Signals for endothelial proliferation (Ki-67), stress tolerance, and neuronal dendrite support are consistent across studies, though with varying magnitudes by peptide (e.g., EDR sometimes > KED).

  • Safety: No overt toxicity is reported in short-term cell/animal experiments at experimental exposures; one human pilot (2015) reported pro-oxidant chemiluminescence and decreased CD34⁺ cells—an ambiguous finding that requires replication and properly controlled safety evaluation. PubMed

Limitations

  • Target validation: Lack of biophysical binding constants and in vivo promoter occupancy metrics for KED; reliance on docking and expression correlations.

  • PK/PD: Unknown for KED; difficult to connect dose–exposure–response.

  • Replication: Many datasets originate from a limited network; more independent studies are needed.


Human Clinical Evidence

Orientation: Peer-reviewed English-language clinical evidence for Vesugen (KED) is sparse and heterogeneous (small open-label studies, regional trials, non-randomized/observational designs). Where available, we summarize and grade cautiously.

1) Geropreventive pilot (2015): “Cellular and metabolic part of geroprophylactic effects…”

  • Design: Open-label cohort; n = 32 (41–83 y) with polymorbidity and organic brain syndrome in remission.

  • Interventions: Courses of Pinealon (EDR) and Vesugen (KED) were administered (routes/schedules summarized in the paper’s Russian full text; English abstract does not tabulate exact doses).

  • Outcomes: Reported anabolic effect, improved CNS/vital-organ functional indices, and reduction in biological age composites; Vesugen effects were more visible than Pinealon. However, investigators also observed pro-oxidant activity by chemiluminescence and a decrease in CD34⁺ hematopoietic cells—raising safety/PDquestions.

  • Evidence grade: C (small, uncontrolled, mixed signals). Investigational dosing used in study: Not establishedfrom English abstract. PubMed

2) Vasculogenic erectile dysfunction (2014): vasoactive tripeptide “Vezugen”

  • Design: Clinical study in men with atherosclerotic vasculogenic ED (details limited in English abstract).

  • Outcomes: The study was intended to evaluate microvascular/erectile endpoints under tripeptide administration; the abstract supports feasibility but lacks modern randomization/blinding detail.

  • Evidence grade: C (limited methodology in abstract). Investigational dosing used in study: Not established from abstract. PubMed

3) Specialty immunology/endocrine observations

  • Pineal immune-cell paper (preclinical) is sometimes cited in clinical-adjacent reviews to suggest broader tissue activity; in that system KED increased proliferation but not differentiation—an important nuance that tempers broad claims about immune remodeling. PubMed

Summary of human evidence. At present, robust randomized, placebo-controlled trials of Vesugen with prespecified primary clinical endpoints are not available in indexed English-language literature. Reported pilot benefits (aging composites; urologic vascular function) are hypothesis-generating, while safety ambiguities (e.g., pro-oxidant readouts; CD34⁺ decrease) highlight the need for rigorous, blinded study designs with independent replication. PubMed+1


Comparative Context

Related peptides

  • EDR (Pinealon)—neuro-centric tripeptide (Glu–Asp–Arg) with stronger antihypoxic signals in head-to-head rodent work; often studied for neuronal resilience. PubMed

  • AEDG (Epitalon)—pineal tetrapeptide with chromatin/telomere literature; appears in aging models and dendritic spine studies.

  • KE (Thymogen) or KED (Vilon)—immune/vascular di- and tripeptides used as comparators for differentiationand proliferation effects.

Advantages (research perspective)

  • Small size (Mr ≈ 390 g·mol⁻¹) simplifies synthesis and supports high-throughput cell-based screening;

  • Promoter-level epigenetic hypothesis offers tractable omics endpoints (MKI67 and other cell-cycle genes);

  • Consistent endothelial and dendritic spine signals across complementary models.

Disadvantages/constraints

  • PK unknowns make dose-setting and translation difficult;

  • Mechanistic granularity is limited (no primary receptor; in vivo DNA binding not proven);

  • Human evidence is limited and sometimes ambiguous (mixed PD/safety signals).

Research category placement

Vesugen is best positioned as a vascular-tropic short peptide for bench and preclinical studies on endothelial aging, cell-cycle re-activation, hypoxia stress, and neurovascular crosstalk (e.g., dendrite support under vascular/oxidative stress).


Research Highlights

  • Sequence-defined tripeptide (KED), Mr ≈ 390.39 g·mol⁻¹; PubChem 87571363 (free acid). canlabintl.com+1

  • Endothelial proliferation: Up-regulates Ki-67 in aged endothelial cultures; docking indicates MKI67 promoterengagement (epigenetic MOA hypothesis). PubMed

  • Antihypoxic activity: Protective in hypobaric hypoxia models alongside EDR/AEDG/KE. PubMed

  • Neuroplasticity (aging): Supports dendritogenesis/spine maintenance in human induced neurons, extending AD-adjacent spine data. PMC

  • Human pilot (aging): Reported improvements in biological age composites but pro-oxidant signals and CD34⁺reduction—highlighting mixed PD and the need for controlled safety/efficacy trials. PubMed

  • Urologic microvascular pilot: Vezugen evaluated in vasculogenic ED, consistent with vascular focus; methodology limited. PubMed

Conflicting/uncertain evidence.

  • Lack of PK/primary target;

  • In vivo promoter occupancy not shown;

  • Human outcomes rely on small, uncontrolled or regionally published studies with variable methodology.


Potential Research Applications (no clinical claims; research-use framing)

  1. Endothelial aging & cell-cycle re-entry

    • Test KED in senescent human endothelial cells with time-resolved RNA-seq/ATAC-seq, Ki-67/PCNAIHC, and EdU incorporation. Map MKI67 promoter accessibility, H3K27ac, and TF occupancy (e.g., E2F, MYC) with and without KED exposure to verify transcriptional re-activation signatures. PubMed

  2. DNA-binding biophysics

    • Undertake surface plasmon resonance, isothermal titration calorimetry, and DNase-I footprinting for MKI67 promoter fragments to quantify KED affinity/specifity; use peptide-ChIP (cross-linkable analog) in cells to test in vivo promoter occupancy—an essential next step beyond docking. PubMed

  3. Hypoxia-reperfusion endothelium

    • In H/R models, quantify ROS, Nrf2/ARE activation, mitochondrial membrane potential, and barrier function (TEER) under KED vs controls; compare to EDR/AEDG to define class vs sequence-specificresilience profiles. PubMed

  4. Neurovascular crosstalk

    • In organ-on-a-chip microfluidic co-cultures (endothelium + neurons/astrocytes), test KED for dendritepreservation and synaptic gene expression (e.g., CAMK2A, PSD-95) under ischemia-mimetic stress; extend to 5xFAD or Tau organoids for translational relevance. PMC

  5. Exploratory human experimental medicine

    • If supported by preclinical PK and safety, design phase-0 studies with circulating endothelial cells, flow-mediated dilation, and oxidative biomarkers as PD readouts; include omics correlates. Any such work would require rigorous randomization, blinding, and independent oversight—currently lacking in the literature base.


Safety & Toxicology

Preclinical/early translational insights

  • Cell/rodent work reports no acute toxicity at experimental exposures; however, systematic GLP toxicology (repeat-dose, genotoxicity, reproductive, carcinogenicity) for KED is Not established in indexed English-language literature.

  • The human geropreventive pilot noted an increase in chemiluminescent pro-oxidant activity and decrease in CD34⁺ hematopoietic progenitors after peptide courses—an ambiguous finding that warrants dedicated safety studies with controls and longer follow-up. PubMed

Known/theoretical molecular risks

  • Unintended proliferation: While endothelial Ki-67 restoration may be beneficial in repair, inappropriate cell-cycle activation could pose theoretical risks (e.g., neointimal growth) without context controlUnknown for KED.

  • Off-target DNA interactions: If KED binds DNA/RNA, sequence promiscuity could lead to unintended gene-expression changes—Not established, but an important consideration for epigenetic-style micro-ligands.

Data gaps

  • PK (absorption, distribution, metabolism, elimination);

  • In vivo target occupancy;

  • Dose–exposure–response relationships;

  • Multicenter RCT safety with hematologic and oxidative biomarkers prespecified.


Limitations & Controversies

  • Evidence base concentration: Many KED studies arise from one research lineage, with limited external replication. PMC

  • Mechanistic proof: Docking and expression data support hypotheses, but definitive molecular targets, binding constants, and in vivo occupancy are not yet shown. PubMed

  • Clinical translation: The human literature is small, methodologically heterogeneous, and underpowered, with mixed PD signals—progress requires modern RCTs. PubMed


Future Directions

  1. Define PK/PD for KED in rodents, then in phase-0 human studies (LC-MS/MS assay development; microdialysis or surrogate exposure biomarkers).

  2. Biophysical target validation (DNA/RNA, protein partners) with affinity, specificity, structure–activityrelationships; design KED analogs (D-residues, N-acyl caps) to enhance stability and test on-target vs off-targetprofiles.

  3. Mechanistic vascular trials with surrogate PD (e.g., endothelial progenitor counts, FMD, arterial stiffness) and omics endpoints; ensure blinding, randomization, preregistration, and independent analysis.

  4. Comparative peptide mapping to delineate KED vs EDR/AEDG/KE in the same platforms (endothelium, neurons, hypoxia).

  5. Safety frameworks with hematopoiesis and oxidative panels (e.g., CD34⁺, 8-iso-PGF2α, GSH/GSSG), given the mixed PD seen in the 2015 geropreventive pilot. PubMed


References

  1. Khavinson V, et al. Peptide regulation of gene expression: a systematic review. Int J Mol Sci. 2021;22: (PMCID: PMC8619776). (Class-level DNA/RNA interaction framework for short peptides, including KED.) PMC

  2. Khavinson V, et al. Epigenetic aspects of peptidergic regulation of vascular endothelium aging. Adv Gerontol.2014;27(3): (PubMed 25051766)—KED increased Ki-67; docking to MKI67 promoter. PubMed

  3. Meshchaninov VN, et al. Effect of synthetic peptides on aging and biological age (Pinealon & Vesugen). Adv Gerontol. 2015;28(3): (PubMed 26390612). (Open-label pilot; mixed PD—pro-oxidant signal and ↓CD34⁺.) PubMed

  4. Kozina LS, et al. Investigation of antihypoxic properties of short peptides. Ross Fiziol Zh Im I M Sechenova.2008;94(6): (PubMed 18546825). (KED/EDR/AEDG/KE protective in hypobaric hypoxia.) PubMed

  5. Kraskovskaya N, et al. Short peptides protect fibroblast-derived induced neurons from aging and stimulate dendritogenesis. Int J Mol Sci. 2024;25:11363. (PMCID: PMC11546785). (Human iNeuron platform; KED among protective peptides.) PMC

  6. Linkova NS, et al. Peptidergic stimulation of differentiation of pineal immune cells. Bull Exp Biol Med.2011;151(1): (PubMed 22803057). (KED increased proliferation but not differentiation.) PubMed

  7. Kitachev KV, et al. Efficacy of peptide bioregulators in vasculogenic erectile dysfunction. Adv Gerontol.2014;27(1): (PubMed 25051774). (Vasoactive tripeptide “Vezugen”; clinical-adjacent vascular context.) PubMed

Analytical identity/structure references (research-supplier confirmations; not human-use claims):
CanLab Vesugen entry (sequence KED, formula C₁₅H₂₆N₄O₈, PubChem 87571363). canlabintl.com
PeptideSciences Vesugen structure listing (same analytical identifiers). Peptide Sciences


⚠️ Disclaimer This peptide is intended strictly for laboratory research use. It is not FDA-approved or authorized for human use, consumption, or therapeutic application.

About Us

Payment & Security

American Express Apple Pay Bancontact Diners Club Discover Google Pay Mastercard Visa

Your payment information is processed securely. We do not store credit card details nor have access to your credit card information.

Estimate shipping

You may also like

Recently viewed