Vesugen, also known as KED, is a tripeptide bioregulator first identified by Russian scientist Vladimir Khavinson. It is derived from a specific sequence of amino acids—lysine, glutamic acid, and aspartic acid—associated with vascular wall proteins.(1)

This peptide has garnered significant attention for its potential role in vascular function, particularly its proposed influence on vascular endothelial cells, which line the interior surface of blood vessels. As a research compound, Vesugen is of interest in cellular studies related to anti-aging, neuroprotection, and vascular biology. This is thought to be due to the peptide’s potential vasoprotective, neuroprotective, and geroprotective characteristics.

Research suggests that Vesugen exerts its potential primarily on the vascular system by interacting with vascular endothelial cells,(2) which are considered to be crucial for maintaining blood vessel integrity. The tripeptide is speculated to interact with cellular processes involved in cell survival, including the repair of tissue structures.

Research suggests that Vesugen may be a relevant compound for further study in the context of atherosclerosis and restenosis. The narrowing of blood vessels characterizes these conditions due to plaque buildup and scarring. Additionally, Vesugen’s influence appears to extend to the central nervous system, where it has been observed to support neuron survival and promote neuroplasticity. These properties may potentially aid in the mitigation of neurodegenerative diseases.

 

Scientific and Research Studies

Vesugen Peptide and Vasoprotective Properties

Research suggests that Vesugen may possess vasoprotective potential, which is particularly relevant to cellular aging and vascular function. This is attributed to its hypothetical influence on vascular endothelial cells, which appear to play a crucial role in maintaining vascular integrity. Vesugen’s mechanism of action may involve the modulation of cellular proliferation, potentially through the regulation of Ki-67, a protein closely associated with cell division. The natural aging process of cells is typically associated with a decline in Ki-67 expression. This may lead to reduced endothelial renewal capacity.

Vesugen is hypothesized to interact with the promoter regions of the Ki-67 gene, which may potentially influence its expression. This upregulation may stimulate endothelial cell proliferation, which may thereby maintain or restore vascular endothelial function. This may be important in studying degenerative vascular conditions. These may include diminished proliferative capacity and the increased presence of polyploid cells, which are thought to be linked to vascular lesions and atherosclerosis. Furthermore, it is proposed that Vesugen might influence gene expression through interactions with specific DNA regions, potentially facilitating vascular function by forming hydrogen bonds with DNA base pairs in the minor groove. This suggests that any vasoprotective action of Vesugen may be mediated through epigenetic regulation of Ki-67 gene expression.

In vitro studies have also indicated that Vesugen may modulate the expression of endothelin-1, a molecule that is typically elevated in atherosclerotic and restenotic conditions.(3) By potentially normalizing endothelin-1 levels, Vesugen may contribute to the mitigation of these vascular pathologies. Additionally, Vesugen might enhance cellular communication through connexin expression. Connexin proteins are considered to be essential for maintaining endothelial integrity. Vesugen’s geroprotective potential may also be linked to an increase in sirtuin1 expression, a protein involved in DNA repair. Sirtuin1 is believed to be critical for preserving cardiovascular function by its potential to reduce genetic damage.

Vesugen Peptide and Neuroprotection

Experimental data has suggested that Vesugen exposure in central nervous system (CNS) disorder models may have the potential to support cognitive functioning. Research suggests that this peptide may contribute to the restoration of synaptic plasticity, which is the process by which neurons form and strengthen their connections.

Like other bioregulatory peptides, Vesugen appears to influence the expression of genes associated with apoptosis and neuronal differentiation. Studies indicate that the peptide “regulates the expression of genes of cell aging and apoptosis (р16, р21), genes (NES, GAP43) and proteins (nestin, GAP43) of the neuronal differentiation, and genes involved in AD [Alzheimer’s Disease] pathogenesis (SUMO, APOE, and IGF1).” (4)

These genes are believed to exert great influence on neuronal survival, growth, and differentiation. Some of these genes are also implicated in the pathogenesis of neurodegenerative conditions such as Alzheimer’s disease. Additionally, research suggests that Vesugen, along with other peptides like Epitalon, Pinealon, and Violon, may mitigate the effects of hypoxia in the CNS by enhancing the activity of antioxidative enzymes. This in turn potentially counteract the neurotoxic effects of reactive oxygen species (ROS) generated under low oxygen conditions.(5)

Vesugen Peptide and Vascular Function, Cellular Aging

Vesugen has been hypothesized to modulate the behavior of prostatic fibroblasts, particularly in relation to the expression of specific differentiation markers. This appears to be more pronounced in cells exhibiting signs of senescence, as evidenced by an observed reduction in differentiation markers in late-passage cultures.

Specifically, Vesugen is hypothesized to enhance the expression of CXCL12 and WEGC1 within these fibroblasts.(6) CXCL12, also referred to as stromal cell-derived factor 1 (SDF-1), is a chemokine primarily associated with immune regulation, particularly in the movement and activation of immune cells. It may also be involved in processes such as hematopoiesis and angiogenesis, although its full range of biological activities remains under investigation. The role of WEGC1, in contrast, is less clearly defined, with limited studies available on its function.

Preliminary research suggests that WEGC1 might play a role in cellular differentiation, particularly in fibroblasts, but further studies are needed to clarify its exact mechanisms and interactions. Notably, the influence of Vesugen appears to be more significant in aged cell cultures, hinting at a possible geroprotective role that may support the preservation or restoration of cellular functions that typically decline over time.

In clinical research, Vesugen has also been explored for its potential impact on biological cellular aging markers.(7) Vesugen has reportedly exhibited an anabolic influence on these cells, which has been linked to increased activity within the central nervous system and other vital organs. This may potentially mitigate the cellular aging process as suggested by changes in bioindicators.

Additionally, the research team observed prooxidant activity through chemiluminescence, indicating that Vesugen might induce oxidative processes. There was also a noted decrease in CD34+ hematopoietic stem cells in the bloodstream, which may imply a reduction in hemopoiesis. This reduction suggests that these cells may not be actively engaged in adaptive responses during Vesugen exposure.

Vesugen Peptide and Metabolic Regulation

Research suggests that Vesugen peptide is associated with the activation of sirtuin 1 (SIRT1), a protein associated with insulin sensitivity. Experimental studies in murine models have indicated that the activation of SIRT1 by compounds such as Vesugen or the antioxidant resveratrol may potentially support insulin sensitivity and mitigate insulin resistance. The studies state that “the finding that SIRT1 improves insulin sensitivity has implications toward resolving insulin resistance and type 2 diabetes.” (8)

Beyond its potential impact on insulin sensitivity, SIRT1 is also suggested to be involved in regulating PGC1-alpha and the ERR-alpha complex, both of which are considered to be critical regulators within the metabolic pathway.(9) The possible modulation of these pathways by SIRT1 may contribute to improved metabolic function and may offer protective action against the development of metabolic syndrome.

 
NOTE: These products are intended for laboratory research use only. This peptide is not intended for personal use. Please review and adhere to our Terms and Conditions before ordering.

 

References:

  1. National Center for Biotechnology Information (2024). PubChem Compound Summary for CID 87571363, Lys-Glu-Asp. https://pubchem.ncbi.nlm.nih.gov/compound/Lys-Glu-Asp.
  2. Khavinson V, Ilina A, Kraskovskaya N, Linkova N, Kolchina N, Mironova E, Erofeev A, Petukhov M. Neuroprotective Effects of Tripeptides-Epigenetic Regulators in Mouse Model of Alzheimer’s Disease. Pharmaceuticals (Basel). 2021 May 27;14(6):515. doi: 10.3390/ph14060515. PMID: 34071923; PMCID: PMC8227791. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8227791/
  3. Kozlov KL, Bolotov II, Linkova NS, Drobintseva AO, Khavinson VK, Dyakonov MM, Kozina LS. [Molecular aspects of vasoprotective peptide KED activity during atherosclerosis and restenosis]. Adv Gerontol. 2016;29(4):646-650. Russian. PMID: 28539025. https://pubmed.ncbi.nlm.nih.gov/28539025/
  4. Khavinson VK, Lin’kova NS, Umnov RS. Peptide KED: Molecular-Genetic Aspects of Neurogenesis Regulation in Alzheimer’s Disease. Bull Exp Biol Med. 2021 May;171(2):190-193. doi: 10.1007/s10517-021-05192-6. Epub 2021 Jun 26. PMID: 34173097. https://pubmed.ncbi.nlm.nih.gov/34173097/
  5. Kozina LS. [Investigation of antihypoxic properties of short peptides]. Adv Gerontol. 2008;21(1):61-7. Russian. PMID: 18546825. https://pubmed.ncbi.nlm.nih.gov/18546825/
  6. Khavinson VKh, Linkova NS, Polyakova VO, Kheifets OV, Tarnovskaya SI, Kvetnoy IM. Peptides tissue-specifically stimulate cell differentiation during their aging. Bull Exp Biol Med. 2012 May;153(1):148-51. doi: 10.1007/s10517-012-1664-1. PMID: 22808515. https://pubmed.ncbi.nlm.nih.gov/22808515/
  7. Meshchaninov VN, Tkachenko EL, Zharkov SV, Gavrilov IV, Katyreva IuE. Effect Of Synthetic Peptides On Aging Of Patients With Chronic Polymorbidity And Organic Brain Syndrome Of The Central Nervous System In Remission. Adv Gerontol. 2015;28(1):62-7. Russian. PMID: 26390612. https://pubmed.ncbi.nlm.nih.gov/26390612/
  8. Sun C, Zhang F, Ge X, Yan T, Chen X, Shi X, Zhai Q. SIRT1 improves insulin sensitivity under insulin-resistant conditions by repressing PTP1B. Cell Metab. 2007 Oct;6(4):307-19. doi: 10.1016/j.cmet.2007.08.014. PMID: 17908559. https://pubmed.ncbi.nlm.nih.gov/17908559/
  9. Nemoto S, Fergusson MM, Finkel T. SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1{alpha}. J Biol Chem. 2005 Apr 22;280(16):16456-60. doi: 10.1074/jbc.M501485200. Epub 2005 Feb 16. PMID: 15716268. https://pubmed.ncbi.nlm.nih.gov/15716268/

Dr. Marinov

Dr. Marinov (MD, Ph.D.) is a researcher and chief assistant professor in Preventative Medicine & Public Health. Prior to his professorship, Dr. Marinov practiced preventative, evidence-based medicine with an emphasis on Nutrition and Dietetics. He is widely published in international peer-reviewed scientific journals and specializes in peptide therapy research.

0
    Your Cart
    Your cart is empty