Tesamorelin & Ipamorelin Blend: Studying their Potential in Growth Hormone Production

Tesamorelin & Ipamorelin Blend: Studying their Potential in Growth Hormone Production

Tesamorelin and Ipamorelin represent two distinct synthetic growth hormone-releasing peptides (GHRPs) that have garnered considerable attention in the realm of biochemical research. Both compounds, Tesamorelin and Ipamorelin, appear to possess the potential to stimulate the release of endogenous growth hormone (GH) through the activation of the ghrelin receptor. This unique action has spurred increasing scientific interest in these peptides, leading to numerous investigations aimed at unraveling their molecular mechanisms and potential impacts.

Research into Tesamorelin has principally centered on its possible utility within the context of metabolic disorders, particularly in the context of HIV-associated lipodystrophy.[1] On the other hand, Ipamorelin appears to have exhibited promise in a wider range of research fields, including obesity and growth promotion.[2]

This introductory exposition explores the scientific foundation underlying Tesamorelin and Ipamorelin, and underscores their potential synergy, highlighting the importance of further exploratory research in elucidating their combined pharmacological actions.

 

Tesamorelin

Tesamorelin is a synthetic peptide with a specific amino acid sequence, also known as a growth hormone-releasing peptide (GHRP). Its structure consists of a chain of 44 amino acids, and it appears to function by stimulating the release of growth hormone (GH) from the pituitary gland.[3]

Tesamorelin has been studied for its potential action in research concerning conditions like HIV-associated lipodystrophy and age-related decline in GH levels. Its speculated ability to enhance GH secretion makes it an intriguing subject of research within the context of various health-related issues, although further studies are needed to fully determine its potential.

 

Ipamorelin

Ipamorelin is a synthetic peptide belonging to the family of growth hormone-releasing peptides (GHRPs). Its chemical structure consists of five amino acids: alanine, glutamine, histidine, leucine, and arginine.[4]

Scientists suggest that this specific sequence appears to impart Ipamorelin with the ability to stimulate the release of growth hormone (GH) by activating the ghrelin receptor, a property that potentially makes it a potent GH secretagogue. Unlike some other GHRPs, Ipamorelin appears to exhibit selective action with minimal impact on other hormones, making it an attractive candidate for research related to GH regulation.

 

Mechanism of Action

 

Tesamorelin

Tesamorelin appears to exert physiological action through the activation of growth hormone releasing hormone (GHRH) receptors localized within the anterior pituitary gland. This activation, in turn, is said to elicit an augmented synthesis and release of growth hormone (GH) into the bloodstream. Subsequently, GH appears to exert its multifaceted actions on various cell types, including hepatocytes, thereby prompting the synthesis of insulin-like growth factor-1 (IGF-1). IGF-1, akin to GH, is considered to play a pivotal role in cellular growth promotion, apoptosis inhibition, glucose regulation, and lipolysis.[5]

Notably, study findings have indicated that research models of HIV-associated lipodystrophy often manifest diminished levels of GH and IGF-1. Tesamorelin may rectify this imbalance, possibly managing overall lipid metabolism and accumulation. 

An intriguing aspect of Tesamorelin lies in its structural modification at the N-terminus, diverging from natural GHRH. This structural alteration appears to enhance peptide stability and may confer heightened resistance to enzymatic deactivation.

 

Ipamorelin

Ipamorelin represents a pioneering synthetic growth hormone secretagogue that appears to operate by enhancing the endogenous production of growth hormone. It is speculated to elicit the release of growth hormone from the anterior pituitary gland, thereby sustaining physiological hormone levels. 

This peptide appears to exhibit a remarkable degree of specificity for the growth hormone-releasing peptide (GHRP) receptors situated within the pituitary gland. Analogous to other GHRP receptor agonists, Ipamorelin may engage these receptors to stimulate the secretion of growth hormone, hGH.[6]

 

Research and Scientific Studies

 

Tesamorelin and Ipamorelin Peptide Blend and Growth Hormone Deficiency

The Tesamorelin and Ipamorelin blend appears to exhibit a multifaceted mechanism of action that, when combined, may offer synergistic effects in the context of growth hormone deficiency. 

When introduced concurrently, the Tesamorelin and Ipamorelin blend has been suggested by researchers to manifest a dual-action approach. Tesamorelin appears to stimulate GH release via GHRH receptor activation, while Ipamorelin seems to directly stimulate GHSR, apparently leading to increased GH secretion. This combined action is presumed to amplify the overall GH response, potentially offering advantages in terms of optimizing mass composition, lipid profiles, insulin sensitivity, and overall metabolic functionality. 

By addressing different components of the growth hormone axis through their distinct mechanisms, Tesamorelin and Ipamorelin may possibly enhance, collectively, pituitary gland activity that ultimately facilitates the release of endogenous growth hormone.[7]

 

Tesamorelin and Ipamorelin Peptide Blend and Lipodystrophy

Lipodystrophy often accompanies insulin resistance, dyslipidemia, and an elevated susceptibility to cardiovascular complications. Clinical research has indicated that the introduction of Tesamorelin to lipodystrophic research subjects may engender a noteworthy reduction in visceral adipose tissue (VAT) alongside possible enhancements in insulin sensitivity and lipid profiles. 

Tesamorelin’s mechanism of action appears to involve the activation of the growth hormone-releasing hormone receptor (GHRHR), potentially fostering the secretion of endogenous growth hormone and facilitating lipolysis. As a result, Tesamorelin may contribute to the preservation of abdominal subcutaneous adipose tissue and favorable alterations in lipid metabolism.

In parallel, Ipamorelin has exhibited signs of promise in modulating adipose tissue metabolism. The conjecture surrounding the combined utilization of Tesamorelin and Ipamorelin postulates synergistic effects, which may further augment the reduction of VAT and ameliorate metabolic parameters in models of lipodystrophy.[8]

 

Tesamorelin and Ipamorelin Peptide Blend and Cognitive Improvement

Growing study data suggests that growth hormone and its secretagogues, including Tesamorelin and Ipamorelin, may exert modulatory influences on neuroplasticity, neuronal survival, and synaptic plasticity, all pivotal elements for optimal cognitive function. Preclinical investigations have revealed that Tesamorelin may potentially enhance memory and learning capacities, possibly via neurogenesis and synaptic plasticity. 

Concurrently, Ipamorelin appears to have some potential to augment spatial memory and cognitive function in animal models. The combined presentation of Tesamorelin and Ipamorelin may prospectively magnify these cognitive benefits through their proposed synergistic mechanisms of action. However, the validation of these findings and the elucidation of the blend’s biological effects on cognitive functioning necessitate future study.

 

Tesamorelin and Ipamorelin Peptide Blend and Type 2 Diabetes

The combination of Tesamorelin and Ipamorelin has garnered attention of researchers within the context of Type 2 Diabetes Mellitus (T2DM) due to promising outcomes observed in exploratory investigations. Both peptides appear to exhibit potential for enhancing glycemic control and ameliorating the metabolic irregularities associated with T2DM. 

Tesamorelin’s mechanism of action is speculated to involve the stimulation of endogenous growth hormone secretion, a process linked to improved insulin sensitivity and enhanced glucose utilization. Ipamorelin similarly has been suggested to influence glucose metabolism and insulin sensitivity. When presented together, Tesamorelin and Ipamorelin may confer complementary impacts, potentially encompassing reductions in hemoglobin A1c (HbA1c) levels, enhancements in insulin sensitivity, and reductions in visceral adiposity in the case of T2DM.[9]

 

Tesamorelin and Ipamorelin Peptide Blend and Pituitary Glad

The combination of Tesamorelin and Ipamorelin may have a notable influence on the pituitary gland, a pivotal endocrine organ responsible for the regulation of growth hormone secretion. 

Research indicates that Tesamorelin may possess a specific affinity for the growth hormone-releasing hormone receptor (GHRHR), potentially initiating the intracellular signaling cascade leading to the synthesis and subsequent release of growth hormone. As supported by previous studies, “Tesamorelin, as a synthetic growth hormone-releasing hormone, exerts its action on the anterior pituitary gland to stimulate endogenous growth hormone secretion”[10]

In contrast, Ipamorelin exhibits potent agonistic activity toward the growth hormone secretagogue receptor (GHSR), possibly facilitating the secretion of growth hormone. As documented in scientific literature, “Ipamorelin stands as the pioneering GHRP-receptor agonist, demonstrating a selectivity for GH release akin to that of GHRH. The specificity inherent in Ipamorelin positions it as a compelling candidate for prospective clinical development”[11]

When introduced together, these peptides appear to elicit a synergistic effect on the pituitary gland, potentially resulting in heightened production of growth hormone. This cooperative interplay between Tesamorelin and Ipamorelin holds promise as a valuable avenue for the optimization of growth hormone levels.[7]

 

In Summary

The combination of Tesamorelin and Ipamorelin peptides, when introduced concurrently, presents an intriguing prospect for magnifying biological benefits through their proposed complementary modes of action. By jointly stimulating the growth hormone axis and modulating neurotrophic factors, this peptide blend may hold promise as a catalyst for cognitive improvement. 

The majority of these findings stem from research conducted in animal models. Therefore, ongoing research remains imperative to fully harness the benefits of these peptides and explore their relevance in the realm of cognitive improvement across a wider variety of species.

Disclaimer: The products mentioned are not intended for human or animal consumption. Research chemicals are intended solely for laboratory experimentation and/or in-vitro testing.  Bodily introduction of any sort is strictly prohibited by law.  All purchases are limited to licensed researchers and/or qualified professionals. All information shared in this article is for educational purposes only.

 

References:

  1. Dhillon S. Tesamorelin: a review of its use in the management of HIV-associated lipodystrophy. Drugs. 2011 May 28;71(8):1071-91. doi: 10.2165/11202240-000000000-00000. PMID: 21668043. https://pubmed.ncbi.nlm.nih.gov/21668043/ 
  2. Gao Y, Yuan X, Zhu Z, Wang D, Liu Q, Gu W. Research and prospect of peptides for use in obesity treatment (Review). Exp Ther Med. 2020 Dec;20(6):234. doi: 10.3892/etm.2020.9364. Epub 2020 Oct 16. PMID: 33149788; PMCID: PMC7604735. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7604735/ 
  3. National Center for Biotechnology Information (2023). PubChem Compound Summary for , Tesamorelin. https://pubchem.ncbi.nlm.nih.gov/compound/Tesamorelin
  4. National Center for Biotechnology Information (2023). PubChem Compound Summary for CID 9831659, Ipamorelin. https://pubchem.ncbi.nlm.nih.gov/compound/Ipamorelin
  5. LiverTox: Clinical and Research Information on Drug-Induced Liver Injury [Internet]. Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases; 2012-. Tesamorelin. [Updated 2018 Oct 20]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK548730/ 
  6. K. Raun et al., Ipamorelin, the first selective growth hormone secretagogue, European Journal of Endocrinology, November 1998, 139 552-561.  https://pubmed.ncbi.nlm.nih.gov/9849822/ 
  7. Rogério G. Gondo et al, Growth Hormone-Releasing Peptide-2 Stimulates GH Secretion in GH-Deficient Patients with Mutated GH-Releasing Hormone Receptor, The Journal of Clinical Endocrinology & Metabolism, Volume 86, Issue 7, 1 July 2001, Pages 3279–3283, https://doi.org/10.1210/jcem.86.7.7694 
  8. Falutz J, Mamputu JC, Potvin D, Moyle G, Soulban G, Loughrey H, Marsolais C, Turner R, Grinspoon S. Effects of tesamorelin (TH9507), a growth hormone-releasing factor analog, in human immunodeficiency virus-infected patients with excess abdominal fat: a pooled analysis of two multicenter, double-blind placebo-controlled phase 3 trials with safety extension data. J Clin Endocrinol Metab. 2010 Sep;95(9):4291-304. doi: 10.1210/jc.2010-0490. Epub 2010 Jun 16. PMID: 20554713. https://pubmed.ncbi.nlm.nih.gov/20554713 
  9. Clemmons DR, Miller S, Mamputu JC. Safety and metabolic effects of tesamorelin, a growth hormone-releasing factor analogue, in patients with type 2 diabetes: A randomized, placebo-controlled trial. PLoS One. 2017 Jun 15;12(6):e0179538. doi: 10.1371/journal.pone.0179538. PMID: 28617838; PMCID: PMC5472315. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5472315/  
  10. Adrian S, Scherzinger A, Sanyal A, Lake JE, Falutz J, Dubé MP, Stanley T, Grinspoon S, Mamputu JC, Marsolais C, Brown TT, Erlandson KM. The Growth Hormone Releasing Hormone Analogue, Tesamorelin, Decreases Muscle Fat and Increases Muscle Area in Adults with HIV. J Frailty Aging. 2019;8(3):154-159. doi: 10.14283/jfa.2018.45. PMID: 31237318; PMCID: PMC6766405. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6766405/ 
  11. Raun K, Hansen BS, Johansen NL, Thøgersen H, Madsen K, Ankersen M, Andersen PH. Ipamorelin, the first selective growth hormone secretagogue. Eur J Endocrinol. 1998 Nov;139(5):552-61. doi: 10.1530/eje.0.1390552. PMID: 9849822. https://pubmed.ncbi.nlm.nih.gov/9849822/ 
GHK Basic (Tripeptide 1): Research in Skin Regeneration and Cancer Control

GHK Basic (Tripeptide 1): Research in Skin Regeneration and Cancer Control

GHK, a tripeptide constituting the amino acid sequence glycine-histidine-lysine, appears to be ubiquitously found in plasma, saliva, and urine, exhibiting an age-related decline. In the context of plasma, GHK concentrations are suggested to be present at levels of approximately 200 ng/mL during early adulthood; however, these concentrations are speculated to diminish to approximately 80 ng/mL over time. This age-related decline in GHK levels is concomitant with the reduction in the regenerative potential of the organism.[1]

It is postulated that GHK forms a copper 2+-complex, which may expedite the wound healing and skin repair processes. GHK is speculated to exert a multifaceted influence, possibly promoting collagen and glycosaminoglycan synthesis while also possibly regulating metalloproteinases and their inhibitors. Moreover, GHK appears to stimulate the production of collagen, dermatan sulfate, chondroitin sulfate, and decorin, potentially rejuvenating fibroblasts following radiation. 

The molecule is said to possess chemo-attractive properties, recruiting immune and endothelial cells to injury sites, thereby possibly accelerating wound healing in diverse anatomical regions, including the skin, hair follicles, gastrointestinal tract, bone tissue, and foot pads in canines. Additionally, studies suggest that the peptide may induce systemic wound healing in rodents and pigs.[1]

 

GHK Mechanism of Action

Research suggests that GHK tripeptide demonstrates a high affinity towards the copper ions (Cu2+). When bound to copper, GHK forms GHK-Cu, which appears to be the active form of the peptide. This copper complex appears to be extremely vital for many of GHK’s potential biological activities.[2]

GHK, both in its copper-bound (GHK-Cu) and copper-free forms, is speculated to exert a profound influence on the transcription of numerous genes central to an organism’s response to stress and injury. These transcriptional effects potentially encompass a wide spectrum of biological processes, including tissue remodeling, antioxidant defenses, anti-inflammatory responses, pain modulation, anxiety alleviation, angiogenesis, nerve outgrowth, and anti-cancer actions. Notably, the GHK sequence is speculated to be integral to the collagen molecule, and the secretion of GHK, in conjunction with the SPARC protein, appears to occur naturally following an injury because of protein degradation processes.[2]

Copper, as a transition metal, appears to play an indispensable role in the biology of eukaryotic organisms. GHK’s apparently pivotal role is attributed to its ability to transition between oxidized Cu(2+) and reduced Cu forms, rendering it a possible essential cofactor in a diverse array of biochemical reactions predicated on electron transfer mechanisms. Approximately a dozen enzymes are said to rely on the redox properties of copper to catalyze critical biochemical reactions encompassing cellular respiration, maintenance of antioxidant defenses, detoxification processes, blood clotting, and the formation of connective tissues.[1]

Beyond these functions, copper also appears integral to essential biological processes such as iron metabolism, oxygenation, neurotransmission, embryonic development, and numerous other facets of physiological functioning.

 

Research Studies

 

GHK Basic Peptide and Fibrinogen Suppression

Fibrinogen comprises of three distinct polypeptide chains: alpha, beta, and gamma. GHK appears to exert a pronounced inhibitory effect on the genetic expression of the beta chain (FGB) of fibrinogen. 

This inhibitory action is said to be particularly noteworthy as the synthesis of fibrinogen necessitates the presence of equimolar quantities of all three polypeptide chains, rendering an insufficiency in FGB detrimental to the overall fibrinogen production process.

Furthermore, GHK appears to extend its influence to the regulation of interleukin-6 (IL-6), a pivotal pro-inflammatory cytokine known for its role as a prominent positive modulator of fibrinogen synthesis through interactions with fibrinogen genes.[3]

In cellular models, GHK appears to demonstrate a capacity to downregulate IL-6 secretion in skin fibroblasts and curtail IL-6 gene expression within SZ95 sebocytes. In summation, the dual impact of GHK on the FGB gene, coupled with its potential ability to mitigate IL-6 production, signifies an overarching suppression of the overall fibrinogen production cascade.

 

GHK Basic Peptide and Skin Regeneration

In a recent scientific investigation, it was indicated that the pre-treatment of mesenchymal stem/stromal cells (MSC) with GHK, delivered through a biodegradable carrier in the form of alginate gel, possibly yielded a dose-dependent augmentation in the secretion of proangiogenic factors. Notably, this suggested increase encompassed key factors such as vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF).

Moreover, a pivotal observation emerged when MSC were pre-exposed to antibodies targeting integrins alpha 1 and beta 1. Under such circumstances, MSC possibly exhibited a marked incapacity to elicit the enhanced secretion of VEGF. 

This compelling finding strongly implies that the influence of GHK on the secretion of trophic factors by MSC is probably entwined with the cellular pathway mediated by integrins alpha 1 and beta 1. 

Reports state, “GHK-Cu, in concentrations of 0.1–10 micromolar, increased expression of epidermal stem cell markers such as integrins and p63 in basal keratinocytes in dermal skin equivalents, which according to the authors indicate increased stemness and proliferative potential of basal keratinocytes. Therefore, restoration of gene pattern characteristic of healthy stem cells, which leads to activation of integrins and p63 cellular pathways may be another target of GHK’s gene modulatory activity relevant to skin regeneration.”[4] [5]

 

GHK Basic peptide and the Ubiquitin Proteasome System (UPS)

The Ubiquitin Proteasome System (UPS) is considered to be a crucial mechanism responsible for regulating and removing damaged proteins within the cell. It is said to play a pivotal role in maintaining protein quality and overall cellular health. Dysfunction in the Ubiquitin system may accelerate aging-related symptoms.

In a noteworthy study[6], researchers introduced a specific peptide to research test models. Interestingly, this peptide appeared to exhibit potential to modulate the expression of UPS-related genes. Notably, it appeared to upregulate the expression of 41 UPS genes while concurrently possibly suppressing the activity of a single UPS gene. This outcome suggests a positive impact on the Ubiquitin Proteasome System.

 

GHK Basic peptide and Insulin-like Gene Suppression

The insulin family of proteins has been postulated to play a role as a negative regulator of longevity. Elevated levels of insulin and insulin-like proteins have been associated with a reduction in lifespan. GHK, in its biological capacity, is speculated to exert an influence on this system by upregulating the expression of three genes while simultaneously downregulating six genes within this insulin-related framework.

The insulin/IGF-1-like receptor pathway appears to stand as a significant contributor to the biological aging process across various organisms. A closer examination of gene expression data reveals that GHK possibly exerts a suppressive effect on this system, as suggested by the downregulation of six out of the nine-affected insulin/IGF-1 genes.[7]

In vitro experiments have provided compelling insights, indicating that mutations that curtail insulin/IGF-1 signaling may decelerate the degenerative aging process and possibly extend the lifespan of various organisms, including mice. Furthermore, it is worth noting that reduced IGF-1 signaling may be considered a contributing factor to the “anti-aging” benefits associated with calorie restriction.

 

GHK Basic Peptide and Cancer Control

In the context of cancer suppression, genes responsible for caspase activity, growth regulation, and DNA repair play pivotal roles. 

In a noteworthy study conducted in 2010, Hong et al. identified a probable comprehensive set of 54 genes closely associated with the aggressive and metastatic attributes of colon cancer. To explore potential avenues for intervention, researchers harnessed the resources of the Broad Institute’s Connectivity Map, seeking compounds that may be possibly capable of counteracting the distinct gene expression patterns linked to aggressive cancer phenotypes.

The outcomes of this investigation revealed compelling insights. Specifically, two molecules, GHK and Securinine, appeared to exhibit a remarkable potential to reverse the differential gene expression observed in these cancer-related genes. 

 

Conclusion

In conclusion, GHK Basic (Tripeptide 1) emerges as a possible compelling agent in the realm of skin and regenerative research. Its apparent multifaceted potential encompasses wound healing, collagen synthesis, metalloproteinase modulation, and gene expression regulation. The intricate mechanisms through which GHK likely operates, particularly when complexed with copper, may underscore its potential in promoting skin regeneration and tissue repair.

However, while the prospects are promising, it is essential to emphasize the need for continued rigorous research to unravel the full spectrum of GHK’s capabilities and optimize its efficacy.

Disclaimer: The products mentioned are not intended for human or animal consumption. Research chemicals are intended solely for laboratory experimentation and/or in-vitro testing.  Bodily introduction of any sort is strictly prohibited by law.  All purchases are limited to licensed researchers and/or qualified professionals. All information shared in this article is for educational purposes only.

 

References


  1. Pickart L, Vasquez-Soltero JM, Margolina A. GHK Peptide as a Natural Modulator of Multiple Cellular Pathways in Skin Regeneration. Biomed Res Int. 2015;2015:648108. doi: 10.1155/2015/648108. Epub 2015 Jul 7. PMID: 26236730; PMCID: PMC4508379. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4508379/ 
  2. Pickart L, Freedman JH, Loker WJ, Peisach J, Perkins CM, Stenkamp RE, Weinstein B. Growth-modulating plasma tripeptide may function by facilitating copper uptake into cells. Nature. 1980 Dec 25;288(5792):715-7. doi: 10.1038/288715a0. PMID: 7453802. https://pubmed.ncbi.nlm.nih.gov/7453802/ 
  3. Pickart L, Vasquez-Soltero JM, Margolina A. GHK and DNA: resetting the human genome to health. Biomed Res Int. 2014;2014:151479. doi: 10.1155/2014/151479. Epub 2014 Sep 11. PMID: 25302294; PMCID: PMC4180391. https://pubmed.ncbi.nlm.nih.gov/25302294/ 
  4. Kang YA, Choi HR, Na JI, Huh CH, Kim MJ, Youn SW, Kim KH, Park KC. Copper-GHK increases integrin expression and p63 positivity by keratinocytes. Arch Dermatol Res. 2009 Apr;301(4):301-6. doi: 10.1007/s00403-009-0942-x. Epub 2009 Mar 25. PMID: 19319546. https://pubmed.ncbi.nlm.nih.gov/19319546/
  5. Choi HR, Kang YA, Ryoo SJ, Shin JW, Na JI, Huh CH, Park KC. Stem cell recovering effect of copper-free GHK in skin. J Pept Sci. 2012 Nov;18(11):685-90. doi: 10.1002/psc.2455. Epub 2012 Sep 28. PMID: 23019153. https://pubmed.ncbi.nlm.nih.gov/23019153/ 
  6. Pickart L, Vasquez-Soltero JM, Margolina A. The human tripeptide GHK-Cu in prevention of oxidative stress and degenerative conditions of aging: implications for cognitive health. Oxid Med Cell Longev. 2012;2012:324832. doi: 10.1155/2012/324832. Epub 2012 May 10. PMID: 22666519; PMCID: PMC3359723. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3359723/ 
  7. Pickart L, Vasquez-Soltero JM, Margolina A. GHK and DNA: resetting the human genome to health. Biomed Res Int. 2014;2014:151479. doi: 10.1155/2014/151479. Epub 2014 Sep 11. PMID: 25302294; PMCID: PMC4180391. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4180391/
Thymosin Beta 4 and Cardiac Regeneration

Thymosin Beta 4 and Cardiac Regeneration

Thymosin Beta 4 (TB4) is a naturally occurring peptide. It consists of 43 amino acids and research suggests that it may appear in high concentrations in various cell lines, including blood platelets and lymphocytes. Synthetic Thymosin Beta 4 is also known as TB-500. [1]

Research studies suggest that Thymosin Beta 4 may exhibit some potential to modulate actin dynamics, a process that is potentially responsible for cell movement and maintaining structural integrity. This property seems to be particularly relevant in cellular processes such as cell migration and tissue repair. Moreover, Thymosin Beta 4 is hypothesized to inhibit inflammatory cytokines and possibly reduce oxidative stress, which may contribute to its protective potential at a cellular level of cardiac cells.[1]

Research also posits that Thymosin Beta 4 might stimulate stem cell differentiation and possibly angiogenesis (the formation of new blood vessels), which are thought to be crucial processes in the life cycle of cells including cardiac cells. More specifically, it’s been proposed that Thymosin Beta 4 might interact with cardiac progenitor cells, and stimulate their migration. Hypothetically, this peptide might also affect processes such as cardiac cell proliferation or survival, which are also key aspects of potentially facilitating faster cardiac regeneration.[2]

 

Thymosin Beta 4 Peptide Potential Mechanism of Action

The mechanisms of action for Thymosin Beta 4 are still being explored. Nevertheless, there are some preliminary insights into its potential activity. For example, it seems to function as an actin-binding protein that may inhibit the polymerization of globular actin (G-actin) into filamentous actin (F-actin). This process is known as actin sequestration, which could potentially cause in elevated G-actin levels.[3] [4] [5] Actin, which appears to be a major component of the cellular cytoskeleton including cardiac cells, may provide structural support to cells and is involved in various cellular processes, including cardiac cell motility to facilitate regeneration. Thymosin Beta 4 is thought to bind with actin primarily (but not exclusively) via its central actin-binding domain (aa 17-23), also known as Ac-LKKTETQ.[6] Researchers posit that “thymosin beta(4) has the potential for significant roles in tissue development, maintenance, repair, and pathology” including cardiac regeneration. The potential prevention of F-actin polymerization by Thymosin Beta 4 might alter the cellular cytoskeleton, which could affect the ability of cardiac cells to move and change shape. This process could have implications in various physiological and pathological processes where cardiac cell motility is crucial, such as cardiac regeneration.[7] It’s important to note that these mechanisms are hypothesized to explain the potential action of intracellular Thymosin Beta 4.

Additionally, TB4 may also hold potential when present outside of cardiac cells (extracellularly).[8] Some research in blood vessel cells suggests that the influence of extracellular Thymosin Beta 4 might regulate processes such as cardiac cell motility and angiogenesis. Scientists posit that the peptide might regulate these processes by interacting with cell surface-located ATP synthase enzymes. These are cellular enzymes considered to be involved in the energy production of the cardiac and other cells.[9] Extracellular Thymosin Beta 4 might also potentially become oxidized in sites of inflammation to Thymosin Beta 4 sulfoxide, and the latter is thought to have potent anti-inflammatory properties.[10] Furthermore, Thymosin Beta 4 might also reduce inflammation by possibly increasing the expression of microRNA-146a (miR-146a). This could potentially decrease the expression of two pro-inflammatory cytokines, called L-1 receptor-associated kinase 1 (IRAK1) and tumor necrosis factor receptor-associated factor 6 (TRAF6).[11] Apparently reducing inflammation and potentially stimulating cardiac cell motility is thought to play a major role in tissue regeneration, including cardiac recovery.

 

Thymosin Beta 4 Peptide and Regeneration of Cardiac Cells

Experimental studies suggest that endothelial progenitor cells (EPC) may have a potential for inducing cardiac regeneration, and the addition of TB4 appears to enhance this process. The researchers posited that EPC with addition of Thymosin Beta 4 peptide may have approved the apparent cardiac function in damaged myocardium and facilitated cardiac repair.[12] This was likely due to its potential action on cardiac cell motility and apparent stimulation of cardiac cell progenitors. Thymosin Beta 4 may also have potentially contributed to cardiac regeneration by apparently reducing inflammation. The peptide has been suggested to potentially decrease reactive oxygen species (ROS) and lipid peroxidation while possibly increasing antioxidant levels. It may also inhibit the activation of nuclear factor kappa B, thus apparently suppressing pro-inflammatory cytokine production, and preventing fibrosis – a potentially undesired regeneration process known to impede tissue function.[13] These findings suggest that Thymosin β4 and cardiac reprogramming technology might work synergistically to limit damage to the heart and promote cardiac regeneration, possibly also through the stimulation of endogenous cells within the heart. Thymosin Beta 4 might also promote myocardial survival in hypoxic conditions and apparently stimulates angiogenesis, which could potentially lead to cardiac repair. The researchers also propose a potential mechanism involving the reprogramming of cardiac fibroblasts to cardiomyocyte-like cells[14] Ultimately, the authors commented that “thymosin β4 and cardiac reprogramming technology may synergistically limit damage to the heart and promote cardiac regeneration through the stimulation of endogenous cells within the heart.” A study in murine models of coronary artery ligation also reported that Thymosin Beta 4 application may upregulate integrin-linked kinase (ILK) and protein kinase B activity in the heart, potentially enhancing early myocyte survival and apparently improving cardiac function.[15] The scientists also posited that “Thymosin Beta 4 peptide promotes myocardial and endothelial cell migration in the embryonic heart and retains this property in postnatal cardiomyocytes.

 

Conclusion

In conclusion, Thymosin Beta 4 appears to have a complex and multifaceted role in cellular processes related to regeneration. That role appears to be particularly in relation to Thymosin Beta 4’s apparent potential on actin dynamics and cell motility including cardiac cells. Its potential influence on cardiac cells and its possible involvement in processes such as inflammation reduction and angiogenesis also suggest that it could play a significant role in cardiac regeneration. Most importantly, Thymosin Beta 4 may also upregulate cardiac cell progenitors, and via its apparent effects on cell motility, it may aid their relocation to injury sites for recovery and regeneration.

However, these are preliminary findings and the exact mechanisms of Thymosin Beta 4’s action, both intracellularly and extracellularly, remain to be fully elucidated. The apparent action of TB4 in the context of cardiac cell regeneration is particularly intriguing, but further research is needed to substantiate these claims and to explore the full range of Thymosin Beta 4’s potential.

Disclaimer: The products mentioned are not intended for human or animal consumption. Research chemicals are intended solely for laboratory experimentation and/or in-vitro testing.  Bodily introduction of any sort is strictly prohibited by law.  All purchases are limited to licensed researchers and/or qualified professionals. All information shared in this article is for educational purposes only.

 

References


  1. Goldstein, A. L., Hannappel, E., Sosne, G., & Kleinman, H. K. (2012). Thymosin β4: a multi-functional regenerative peptide. Basic properties and clinical applications. Expert opinion on biological therapy, 12(1), 37–51. https://doi.org/10.1517/14712598.2012.634793
  2. Choudry, F. A., Yeo, C., Mozid, A., Martin, J. F., & Mathur, A. (2015). Increases in plasma Tβ4 after intracardiac cell therapy in chronic ischemic heart failure is associated with symptomatic improvement. Regenerative medicine, 10(4), 403–410. https://doi.org/10.2217/rme.15.9
  3. Sanders, M. C., Goldstein, A. L., & Wang, Y. L. (1992). Thymosin Beta 4 (Fx peptide) is a potent regulator of actin polymerization in living cells. Proceedings of the National Academy of Sciences of the United States of America, 89(10), 4678–4682. https://doi.org/10.1073/pnas.89.10.4678
  4. Irobi, E., Aguda, A. H., Larsson, M., Guerin, C., Yin, H. L., Burtnick, L. D., Blanchoin, L., & Robinson, R. C. (2004). Structural basis of actin sequestration by thymosin-beta4: implications for WH2 proteins. The EMBO journal, 23(18), 3599–3608. https://doi.org/10.1038/sj.emboj.7600372
  5. Belsky, J. B., Rivers, E. P., Filbin, M. R., Lee, P. J., & Morris, D. C. (2018). Thymosin Beta 4 regulation of actin in sepsis. Expert opinion on biological therapy, 18(sup1), 193-197.
  6. Sosne, G., Qiu, P., Goldstein, A. L., & Wheater, M. (2010). Biological activities of thymosin beta4 defined by active sites in short peptide sequences. FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 24(7), 2144–2151. https://doi.org/10.1096/fj.09-142307
  7. Yadav, T., Gau, D., & Roy, P. (2022). Mitochondria-actin cytoskeleton crosstalk in cell migration. Journal of cellular physiology, 237(5), 2387–2403. https://doi.org/10.1002/jcp.30729
  8. Huff, T., Müller, C. S., Otto, A. M., Netzker, R., & Hannappel, E. (2001). beta-Thymosins, small acidic peptides with multiple functions. The international journal of biochemistry & cell biology, 33(3), 205–220. https://doi.org/10.1016/s1357-2725(00)00087-x
  9. Freeman, K. W., Bowman, B. R., & Zetter, B. R. (2011). Regenerative protein thymosin beta-4 is a novel regulator of purinergic signaling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology, 25(3), 907–915. https://doi.org/10.1096/fj.10-169417
  10. Young, J. D., Lawrence, A. J., MacLean, A. G., Leung, B. P., McInnes, I. B., Canas, B., Pappin, D. J., & Stevenson, R. D. (1999). Thymosin Beta 4 sulfoxide is an anti-inflammatory agent generated by monocytes in the presence of glucocorticoids. Nature medicine, 5(12), 1424–1427. https://doi.org/10.1038/71002
  11. Santra, M., Zhang, Z. G., Yang, J., Santra, S., Santra, S., Chopp, M., & Morris, D. C. (2014). Thymosin β4 up-regulation of microRNA-146a promotes oligodendrocyte differentiation and suppression of the Toll-like proinflammatory pathway. The Journal of biological chemistry, 289(28), 19508–19518. https://doi.org/10.1074/jbc.M113.529966
  12. Zhu, J., Song, J., Yu, L., Zheng, H., Zhou, B., Weng, S., & Fu, G. (2016). Safety and efficacy of autologous thymosin β4 pre-treated endothelial progenitor cell transplantation in patients with acute ST segment elevation myocardial infarction: A pilot study. Cytotherapy, 18(8), 1037–1042. https://doi.org/10.1016/j.jcyt.2016.05.006
  13. Shah, R., Reyes-Gordillo, K., Cheng, Y., Varatharajalu, R., Ibrahim, J., & Lakshman, M. R. (2018). Thymosin β4 Prevents Oxidative Stress, Inflammation, and Fibrosis in Ethanol- and LPS-Induced Liver Injury in Mice. Oxidative medicine and cellular longevity, 2018, 9630175. https://doi.org/10.1155/2018/9630175
  14. Srivastava, D., Ieda, M., Fu, J., & Qian, L. (2012). Cardiac repair with thymosin β4 and cardiac reprogramming factors. Annals of the New York Academy of Sciences, 1270, 66–72. https://doi.org/10.1111/j.1749-6632.2012.06696.x
  15. Bock-Marquette, I., Saxena, A., White, M. D., Dimaio, J. M., & Srivastava, D. (2004). Thymosin beta4 activates integrin-linked kinase and promotes cardiac cell migration, survival and cardiac repair. Nature, 432(7016), 466–472. https://doi.org/10.1038/nature03000
Chonluten (T-34) and its Potential in Gene Expression

Chonluten (T-34) and its Potential in Gene Expression

Chonluten, also known as EDG tripeptide or T-34, is a short peptide made of 3 amino acids that may act as a potential bioregulator of gene expression. More specifically, it is made of the amino acids glycine, glutamine, and asparagine with the following configuration: Glu-Asp-Gly.

According to laboratory research, Chonluten (T-34) may be derived from respiratory lung tissue, more specifically mucosa.[1] There, Chonluten is considered to normalize the performance of bronchial mucous membrane cells by potentially regulating genes related to inflammation, antioxidant activity, and proliferation. The peptide may also potentially improve the regeneration of cells in the gastrointestinal system.[2]

 

Chonluten (T-34) Mechanisms of Action

The peptide likely achieves its potential for regulating gene expression thanks to its small size, which may allow it to penetrate the cell’s nuclei. Researchers report that “Short peptides, consisting of 2-7 amino acid residues, can penetrate into the nuclei and nucleoli of cells and interact with the nucleosome, the histone proteins, and both single- and double-stranded DNA. DNA-peptide interactions, including sequence recognition in gene promoters, are important for template-directed synthetic reactions, replication, transcription, and reparation. Peptides can regulate the status of DNA methylation, which is an epigenetic mechanism for the activation or repression of genes“.[3] [4] [5] However, it is important to note that the aforementioned researchers have shared this information for short-peptides in general and not for Chonluten per se.

 

Chonluten and Anti-Inflammatory Mechanisms

Chonluten may potentially activate the phosphorylation of STAT molecules, particularly STAT1, in macrophage cells. This activation appears to occur independently of receptor-associated kinases. STAT1, a transcription factor involved in signal transduction and gene expression regulation, potentially cooperates with receptor-associated kinases to facilitate the transfer of cytokine-mediated biological responses into the nuclei.[1] 

It is hypothesized that Chonluten may potentially downregulate the phosphorylation of STAT3, another transcription factor involved in cellular signaling. STAT3 is considered to be associated with acute inflammatory stimuli and may play a role in the transcription of IL-6, a key cytokine in the acute phase response during inflammation and infectious diseases. By modulating STAT3 phosphorylation, Chonluten could affect the transcriptional activity of IL-6 and influence the inflammatory response.[1]

Moreover, Chonluten (T-34) research has posited its possible efficacy in decreasing the levels of IL-6, TNF (tumor necrosis factor), and IL-17 in macrophages activated by lipopolysaccharide (LPS), a component of bacterial cell walls. IL-6 is a pro-inflammatory cytokine involved in immune responses, while TNF and IL-17 are also pro-inflammatory cytokines that contribute to inflammation and immune system regulation. Chonluten’s potential ability to downregulate the production of these cytokines suggests its possible role in dampening pro-inflammatory processes in activated macrophages. More specifically, the researchers comment that “tripeptide, derived from bronchial epithelial cells, inhibited in vitro tumor necrosis factor (TNF) production of monocytes exposed to pro-inflammatory bacterial lipopolysaccharide (LPS). The low TNF release by monocytes is linked to a documented mechanism of TNF tolerance, promoting attenuation of inflammatory action.[1] 

Additionally, Chonluten may attenuate the adhesion mechanism between endothelium (the inner lining of blood vessels) and immune cells. This observation was made in co-incubation experiments with LPS-activated endothelial cells. By modulating this adhesion mechanism, Chonluten might influence immune cell migration and trafficking, essential in inflammation and immune responses.[1]

 

Chonluten (T-34) and Gastric Cell Regeneration

Chonluten appears to have the potential to enhance gastrointestinal cell regeneration by potentially regulating the expression of genes associated with antioxidant enzymes, such as superoxide dismutase (SOD). Research conducted on the peptide has suggested this ability through the compound apparently normalizing the expression of these genes, through which Chonluten seemingly aids in restoring the balance of antioxidant defense mechanisms in the gastric mucosa. This regulatory action on antioxidant systems may contribute to a potential reduction in oxidative stress and promote cell regeneration.[2]

Furthermore, it is hypothesized that the Chonluten peptide may have an anti-inflammatory potential by influencing the expression of genes involved in the inflammatory response, such as TNF-α and cyclooxygenase-2 (Cox-2). By potentially reducing the expression of these inflammatory mediators, the Chonluten peptide supposedly assists in controlling inflammation in the gastric mucosa, thus facilitating the cell regeneration process.[2] 

Moreover, the Chonluten may possibly stimulate granulation tissue formation, which is crucial for tissue repair. It may stimulate the proliferation of fibroblasts and the growth of blood vessels within the granulation tissue and also may facilitate the regeneration of damaged gastric mucosa. Additionally, Chonluten may promote the epithelialization of the ulcer by the apparent stimulation of the proliferation of epithelial cells, potentially leading to the closure of the ulcer defect.[2]

It is also posited that Chonluten (T-34) may exhibit a reparative action by potentially reducing excessive apoptosis (programmed cell death) in the gastric mucosa. It has been suggested by researchers to possibly regulate the expression of heat shock protein 70 (HSP70), which is believed to play a role in protecting cells from apoptotic stimuli. By modulating HSP70 expression, the peptide might help prevent excessive cell apoptosis and potentially promote tissue survival and repair.[2]

In contrast, additional studies have reported that Chonluten may have insignificant potential for the proliferation of other cell lines, such as skin cell lines.[6] Therefore, more research is needed to delve into the potential mechanisms and actions of Chonluten.

 

Conclusion

In conclusion, Chonluten, a short peptide consisting of three amino acids, appears to hold promising potential as a bioregulator of gene expression. The mechanisms of action of Chonluten suggest that it may have the ability to penetrate cell nuclei and potentially interact with DNA, hypothetically regulating gene activation or repression. Further, Chonluten (T-34) seemingly exhibits anti-inflammatory properties by modulating the phosphorylation of transcription factors involved in cytokine-mediated responses and potentially reducing the production of pro-inflammatory cytokines. Some studies also suggest that Chonluten might promote gastric cell regeneration by regulating antioxidant enzyme expression, controlling inflammatory mediators, stimulating tissue formation and epithelialization, and preventing excessive apoptosis, but more research is needed. Overall, further investigation is required to fully understand the potential of Chonluten on other cell lines and cell cultures. 

Disclaimer: The products mentioned are not intended for human or animal consumption. Research chemicals are intended solely for laboratory experimentation and/or in-vitro testing.  Bodily introduction of any sort is strictly prohibited by law.  All purchases are limited to licensed researchers and/or qualified professionals. All information shared in this article is for educational purposes only.

 

References


  1. Avolio, F., Martinotti, S., Khavinson, V. K., Esposito, J. E., Giambuzzi, G., Marino, A., Mironova, E., Pulcini, R., Robuffo, I., Bologna, G., Simeone, P., Lanuti, P., Guarnieri, S., Trofimova, S., Procopio, A. D., & Toniato, E. (2022). Peptides Regulating Proliferative Activity and Inflammatory Pathways in the Monocyte/Macrophage THP-1 Cell Line. International journal of molecular sciences, 23(7), 3607. https://doi.org/10.3390/ijms23073607
  2. Khavinson, V. K.h, Lin’kova, N. S., Dudkov, A. V., Polyakova, V. O., & Kvetnoi, I. M. (2012). Peptidergic regulation of expression of genes encoding antioxidant and anti-inflammatory proteins. Bulletin of experimental biology and medicine, 152(5), 615–618. https://doi.org/10.1007/s10517-012-1590-2
  3. Khavinson, V. K., Popovich, I. G., Linkova, N. S., Mironova, E. S., & Ilina, A. R. (2021). Peptide Regulation of Gene Expression: A Systematic Review. Molecules (Basel, Switzerland), 26(22), 7053. https://doi.org/10.3390/molecules26227053
  4. Khavinson, V. K., Lin’kova, N. S., & Tarnovskaya, S. I. (2016). Short Peptides Regulate Gene Expression. Bulletin of experimental biology and medicine, 162(2), 288–292. https://doi.org/10.1007/s10517-016-3596-7
  5. Fedoreyeva, L. I., Kireev, I. I., Khavinson, V. K.h, & Vanyushin, B. F. (2011). Penetration of short fluorescence-labeled peptides into the nucleus in HeLa cells and in vitro specific interaction of the peptides with deoxyribooligonucleotides and DNA. Biochemistry. Biokhimiia, 76(11), 1210–1219. https://doi.org/10.1134/S0006297911110022
  6. Voicekhovskaya, M. A., Chalisova, N. I., Kontsevaya, E. A., & Ryzhak, G. A. (2012). Effect of bioregulatory tripeptides on the culture of skin cells from young and old rats. Bulletin of experimental biology and medicine, 152(3), 357–359. https://doi.org/10.1007/s10517-012-1527-9
CJC-1295 & GHRP-6 Blend and the Central Nervous System

CJC-1295 & GHRP-6 Blend and the Central Nervous System

The CJC-1295 and GHRP-6 blend combines two peptides that may interact with calls in the pituitary gland and the hypothalamus called somatotrophs. These cells are thought to play a role in growth hormone synthesis and regulation. More specifically, the blend consists of CJC-1295, an apparent growth hormone-releasing hormone (GHRH) analog, and GHRP-6 (Growth Hormone Releasing Peptide 2), an apparent mimetic of the receptors of the hunger hormone ghrelin.

The CJC-1295 and GHRP-6 blend may exhibit a potentially synergistic action on cells involved in growth hormone release. Combining these peptides may enhance both the amplitude and the frequency of the growth hormone pulses by somatotroph cells. This synergistic action is believed to be mediated through complementary mechanisms of action, as CJC-1295 appears to target the GHRH receptor pathway, while GHRP-6 may act on the ghrelin receptor pathway.

 

CJC-1295 & GHRP-6 Blend Structure and Affinity

CJC-1295, also known as tetra-substituted GRF (1-29), appears to be a synthetic peptide analog of the endogenous GHRH hormone. It comprises the shortest amino-acid chain that may possess an affinity to the GHRH receptors, and consists of the first 29 amino acids of GHRH. CJC-1295 appears to be modified as it has 4 of the original 29 amino acids replaced to potentially make the peptide more resistant to rapid cleavage by the enzyme dipeptidyl peptidase-4 and other peptides that appear to result in peptide inactivation[1]. More specifically, the amino acids that appear to be modified and replaced are the 2nd, 8th, 15th, and 27th amino acids. In addition, CJC-1295 has undergone modification by including a drug affinity complex (DAC) component, which may potentially bind to plasma proteins. The DAC component specifically refers to the attachment of a derivative of lysine called N-epsilon-3-maleimidopropionamide at the C terminus of CJC-1295. This combination of modified amino acid sequence and a DAC component may potentially enhance the pharmacokinetics of CJC-1295, while still maintaining an apparent affinity to the GHRH receptors. Indeed, scientists have commended that the peptide “was found to be present in plasma beyond 72 h.[2]

On the other hand, GHRP-6 is a hexapeptide that appears to belong to the class of synthetic growth hormone-releasing peptides, which may act by potentially binding to the ghrelin receptor on the surface of pituitary cells and certain hypothalamic neurons. Upon binding, GHRP-6 appears to stimulate the so-called growth hormone secretagogue receptor (GHS-R1a). These receptors are also sometimes termed the ghrelin receptors, as ghrelin appears to be their main natural ligand. GHRP-6 appears to bind to these receptors, and may induce an intracellular calcium response and protein kinase C activity.[3] By activating them, GHRP-6 appears to stimulate the production of growth hormones in pituitary cells. In addition, GHRP-6 may also have an affinity for the CD36 receptors.[4] These CD36 receptors may serve multiple roles, including a potential action in lipid metabolism, acting as a scavenger receptor for lipids, facilitating their uptake, and potentially modulating immune responses, regulating phagocytosis and inflammation. CD36 pathways may also play a potential role in angiogenesis regulation.

 

CJC-1295 & GHRP-6 Blend and Somatotroph Cells

CJC-1295 appears to target somatotroph cells by potentially interacting with the GHRH receptor they express. More specifically, CJC-1295 may bind to specific binding sites on the receptor protein, leading to conformational changes in the receptor structure and potentially initiating a cascade of molecular events. These events appear initiated by intracellular signaling proteins acting as potential molecular switches.[5] These are the so-called G-proteins, which, once activated, may stimulate the potential production of second messengers like cyclic adenosine monophosphate (cAMP) or inositol trisphosphate (IP3).[6] Second messengers like cAMP may activate protein kinases, enzymes that are considered to modify specific proteins. These kinases have a regulatory potential towards cellular processes and may phosphorylate transcription factors, or proteins controlling gene expression. Phosphorylated transcription factors may enter the nucleus of somatotroph cells, potentially influencing genes related to growth hormone synthesis.[7] Ultimately, CJC-1295 binding appears to trigger events leading to the release of growth hormone from vesicles by the somatotroph cells. Researchers comment that the peptide may upregulate growth hormone production by somatotrophs, thus apparently contributing “to an overall increase in GH secretion … by 46%.” In turn, growth hormone appears to have a major anabolic mediator called insulin-like growth factor-1 (IGF-1). IGF-1 levels also apparently increased by 45%.[7] Another trial suggests that CJC-1295 may potentially upregulate “GH concentrations by 2- to 10-fold.[8]

In contrast, GHRP-6 appears to interact with somatotroph cells via the  GHS-R1a. More specifically, scientists posit that the peptide may apparently prefer an intracellular calcium response and protein kinase C activity instead of cAMP production.[9] The GHS-R1a activation appears to activate a specific unit of G-proteins inside somatotrophs. Gαq/11. This potentially preferred pathway by GHRP-6 apparently involves the enzyme phospholipase C. Gαq/11 may bind to phospholipase C, which may cleave a phospholipid called PIP2 into two messengers: the aforementioned IP3 and diacylglycerol (DAG).[10] IP3 appears to bind to receptors on the endoplasmic reticulum, releasing calcium ions, while DAG may activate protein kinase C, phosphorylating signaling molecules and amplifying the cascade. This potentially leads to the release of growth hormones by somatotroph cells.

 

Conclusion

In conclusion, the CJC-1295 & GHRP-6 blend may have a synergistic potential as they appear to interact with receptors in the central nervous system, particularly in the pituitary gland cells called somatotrophs. These peptides are hypothesized to have neuroendocrine actions and stimulate the release of growth hormones from the aforementioned somatotrophs. CJC-1295 appears to bind the GHRH receptors in the pituitary gland, potentially leading to conformational changes and the activation of intracellular signaling pathways for growth hormone production. On the other hand, GHRP-6 may bind to the GHS-R1a receptors, also known as ghrelin receptors, with a potential preference for an intracellular calcium response and protein kinase C activity instead of cAMP production. This may also initiate a cascade of molecular events, potentially resulting in the release of growth hormones. 

Disclaimer: The products mentioned are not intended for human or animal consumption. Research chemicals are intended solely for laboratory experimentation and/or in-vitro testing.  Bodily introduction of any sort is strictly prohibited by law.  All purchases are limited to licensed researchers and/or qualified professionals. All information shared in this article is for educational purposes only.

 

References


  1. Scarborough, R., Gulyas, J., Schally, A. V., & Reeves, J. J. (1988). Analogs of growth hormone-releasing hormone induce release of growth hormone in the bovine. Journal of animal science, 66(6), 1386–1392. https://doi.org/10.2527/jas1988.6661386x
  2. Jetté, L., Léger, R., Thibaudeau, K., Benquet, C., Robitaille, M., Pellerin, I., Paradis, V., van Wyk, P., Pham, K., & Bridon, D. P. (2005). Human growth hormone-releasing factor (hGRF)1-29-albumin bioconjugates activate the GRF receptor on the anterior pituitary in rats: identification of CJC-1295 as a long-lasting GRF analog. Endocrinology, 146(7), 3052–3058. https://doi.org/10.1210/en.2004-1286
  3. Sun, Q., Ma, Y., Zhang, L., Zhao, Y. F., Zang, W. J., & Chen, C. (2010). Effects of GH secretagogues on contractility and Ca2+ homeostasis of isolated adult rat ventricular myocytes. Endocrinology, 151(9), 4446–4454. https://doi.org/10.1210/en.2009-1432
  4. Demers, A., McNicoll, N., Febbraio, M., Servant, M., Marleau, S., Silverstein, R., & Ong, H. (2004). Identification of the growth hormone-releasing peptide binding site in CD36: a photoaffinity cross-linking study. The Biochemical journal, 382(Pt 2), 417–424. https://doi.org/10.1042/BJ20040036
  5. Martin, B., Lopez de Maturana, R., Brenneman, R., Walent, T., Mattson, M. P., & Maudsley, S. (2005). Class II G protein-coupled receptors and their ligands in neuronal function and protection. Neuromolecular medicine, 7(1-2), 3–36. https://doi.org/10.1385/nmm:7:1-2:003
  6. Newton, A. C., Bootman, M. D., & Scott, J. D. (2016). Second Messengers. Cold Spring Harbor perspectives in biology, 8(8), a005926. https://doi.org/10.1101/cshperspect.a005926
  7. Ionescu, M., & Frohman, L. A. (2006). Pulsatile secretion of growth hormone (GH) persists during continuous stimulation by CJC-1295, a long-acting GH-releasing hormone analog. The Journal of clinical endocrinology and metabolism, 91(12), 4792–4797. https://doi.org/10.1210/jc.2006-1702
  8. Teichman, S. L., Neale, A., Lawrence, B., Gagnon, C., Castaigne, J. P., & Frohman, L. A. (2006). Prolonged stimulation of growth hormone (GH) and insulin-like growth factor I secretion by CJC-1295, a long-acting analog of GH-releasing hormone, in healthy adults. The Journal of clinical endocrinology and metabolism, 91(3), 799–805. https://doi.org/10.1210/jc.2005-1536
  9. Sinha, D. K., Balasubramanian, A., Tatem, A. J., Rivera-Mirabal, J., Yu, J., Kovac, J., Pastuszak, A. W., & Lipshultz, L. I. (2020). Beyond the androgen receptor: the role of growth hormone secretagogues in the modern management of body composition in hypogonadal males. Translational andrology and urology, 9(Suppl 2), S149–S159. https://doi.org/10.21037/tau.2019.11.30
  10. Bill, C. A., & Vines, C. M. (2020). Phospholipase C. Advances in experimental medicine and biology, 1131, 215–242. https://doi.org/10.1007/978-3-030-12457-1_9
BPC-157 & TB-500 Blend: An Exploration in Cell Proliferation and Regeneration

BPC-157 & TB-500 Blend: An Exploration in Cell Proliferation and Regeneration

The BPC-157 & TB-500 blend incorporates two synthetic peptides with very different structures but similar research potential. TB-500 is a synthetic version of the thymosin beta-4 (Tβ4) peptide that naturally occurs in the cells of the thymus gland. It is made of 43 amino acids encoded by the TMSB4X gene. The peptide has the following sequence: SDKPDMAEI EKFDKSKLKK TETQEKNPLP SKETIEQEKQ AGES, and its molecular weight is 4921 g/mol. Based on thymosin beta-4 research, TB-500 may regulate cell migration, differentiation, and tissue repair.[1] It is thought to interact with different signaling pathways within cells to exert its action. Studies also suggest that TB-500 may potentially promote angiogenesis, which is the formation of new blood vessels. Additionally, it has been suggested by experimental models that TB-500 might contribute to cellular and tissue regeneration.

BPC-157, also known as L 14736, PL-10, and bepecin, is a much shorter peptide comprising 15 amino acids. This pentadecapeptide is fully synthetic, but its structure is derived from the biologically active sequence of another peptide naturally found in gastric juices. BPC-157 may be associated with various cellular processes and is believed to play a role in tissue repair. It is thought to interact with signaling pathways within cells, potentially influencing factors involved in regeneration and recovery. Some studies suggest that BPC-157 may also have the ability to promote angiogenesis and modulate inflammatory processes.[2] Additionally, it has been suggested by experimental models that BPC-157 might contribute to the protection and regeneration of different cells and tissues.

 

BPC-157 & TB-500 Blend Mechanisms of Action

BPC-157 & TB-500 appear to have different mechanisms, despite their similar potential. For example, TB-500 (Tβ4) appears to regulate the cellular actin-cytoskeleton and cellular migration by sequestering G-actin.[3] [4] Furthermore, studies suggest that there appears to be a specific signaling pathway involved with damaged tissues that TB-500 may regulate. For instance, researchers have conducted experiments using cell models and observed that TB-500 potentially increases the expression of microRNA-146a (miR-146a), which might be a negative regulator of specific signaling pathways in cells. It appeared that this resulted in TB-500 possibly decreasing the expression of two proinflammatory cytokines associated with the aforementioned signaling pathways – L-1 receptor-associated kinase 1 (IRAK1) and tumor necrosis factor receptor-associated factor 6 (TRAF6). As a further suggestion that the increased expression of miR-146a may be TB-500’s main mechanism of action, the researchers commented that “transfection of anti-miR-146a nucleotides reversed the inhibitory effect of Tβ4 on IRAK1 and TRAF6.[5]  

On the other hand, BPC-157 appears to exert its actions via multiple mechanisms, which may include nitric oxide synthesis, regulating cells involved in tissue repair, growth factors, and inflammation. There is a possibility that BPC-157 interacts with the NO system, potentially offering protection to the endothelium and possibly inducing angiogenic action by promoting the formation of new blood vessels.[6] It may have the potential to stimulate the expression of the early growth response 1 gene, which might be responsible for generating cytokines and growth factors, and perhaps facilitating the early formation of the extracellular matrix, including collagen. It should be noted that BPC-157’s interaction with nerve growth factor 1-A binding protein-2 might have repressive effects on certain factors. However, further research is needed to fully understand and confirm these potential mechanisms of BPC-157.

 

BPC-157 & TB-500 Blend and the Musculoskeletal System

The BPC-157 & TB-500 appear to both have the potential for speeding up the regeneration of connective tissue, such as the one found in tendons and ligaments. In one murine study, TB-500 was investigated for its potential on ligament recovery and regeneration.[7] The scientists performed histological analysis to compare TB-500 against a placebo in a model of ligament injury. They commended that TB-500 may have induced the formation of a more uniform and evenly spaced bundles of collagen fibers within the granulation tissue that also have larger diameters compared to the control. Furthermore, the mechanical properties of the regenerating tissues, including the femur-medial collateral ligament-tibia complexes, appeared to be improved in the TB-500 group compared to the control.

Another study explored the potential impact of BPC 157 on the outgrowth of tendon fibroblasts from cultured tendon explants.[8] The findings suggested that BPC 157 possibly enhanced the outgrowth of tendon explants. BPC 157 potentially increased the survival of these cells under H(2)O(2) stress. Furthermore, the researchers commented that “BPC 157 markedly increased the in vitro migration of tendon fibroblasts in a dose-dependent manner as revealed by transwell filter migration assay. BPC 157 also dose-dependently accelerated the spreading of tendon fibroblasts on culture dishes.” This effect was potentially associated with the induction of F-actin formation, as evidenced by FITC-phalloidin staining. The study also investigated the potential involvement of the FAK-paxillin (two focal adhesion–associated proteins that transmit signals downstream of integrins) pathway in mediating the potential of BPC 157. Western blot analysis suggested that the phosphorylation levels of both FAK and paxillin were apparently increased by BPC 157, while the total amounts of protein remained unchanged.

 

Conclusion

In summary, the BPC-157 & TB-500 blend shows a synergistic research potential. TB-500 may be involved in regulating cell migration, differentiation, and repair, potentially promoting angiogenesis and connective tissue regeneration. Furthermore, BPC-157 is also believed to have a potential in cellular repair, possibly by influencing factors involved in the normal recovery process. These peptides appear to have different mechanisms of action, with TB-500 possibly regulating cellular actin-cytoskeleton and migration, while BPC-157 may interact with various processes such as nitric oxide synthesis, growth factors, and inflammation regulation. Both peptides show potential in speeding up the regeneration of connective tissue, such as tendons and ligaments, with TB-500 potentially improving collagen fiber formation and regeneration and BPC-157 potentially enhancing tendon fibroblast outgrowth and migration. Further research is needed to confirm these mechanisms.

Disclaimer: The products mentioned are not intended for human or animal consumption. Research chemicals are intended solely for laboratory experimentation and/or in-vitro testing.  Bodily introduction of any sort is strictly prohibited by law.  All purchases are limited to licensed researchers and/or qualified professionals. All information shared in this article is for educational purposes only.

 

References


  1. Maar, K., Hetenyi, R., Maar, S., Faskerti, G., Hanna, D., Lippai, B., Takatsy, A., & Bock-Marquette, I. (2021). Utilizing Developmentally Essential Secreted Peptides Such as Thymosin Beta-4 to Remind the Adult Organs of Their Embryonic State-New Directions in Anti-Aging Regenerative Therapies. Cells, 10(6), 1343. https://doi.org/10.3390/cells10061343
  2. Seiwerth, S., Milavic, M., Vukojevic, J., Gojkovic, S., Krezic, I., Vuletic, L. B., Pavlov, K. H., Petrovic, A., Sikiric, S., Vranes, H., Prtoric, A., Zizek, H., Durasin, T., Dobric, I., Staresinic, M., Strbe, S., Knezevic, M., Sola, M., Kokot, A., Sever, M., … Sikiric, P. (2021). Stable Gastric Pentadecapeptide BPC 157 and Wound Healing. Frontiers in pharmacology, 12, 627533. https://doi.org/10.3389/fphar.2021.627533
  3. Huff, T., Müller, C. S., Otto, A. M., Netzker, R., & Hannappel, E. (2001). beta-Thymosins, small acidic peptides with multiple functions. The international journal of biochemistry & cell biology, 33(3), 205–220. https://doi.org/10.1016/s1357-2725(00)00087-x
  4. Sanders, M. C., Goldstein, A. L., & Wang, Y. L. (1992). Thymosin beta 4 (Fx peptide) is a potent regulator of actin polymerization in living cells. Proceedings of the National Academy of Sciences of the United States of America, 89(10), 4678–4682. https://doi.org/10.1073/pnas.89.10.4678
  5. Santra, M., Zhang, Z. G., Yang, J., Santra, S., Santra, S., Chopp, M., & Morris, D. C. (2014). Thymosin β4 up-regulation of microRNA-146a promotes oligodendrocyte differentiation and suppression of the Toll-like proinflammatory pathway. The Journal of biological chemistry, 289(28), 19508–19518. https://doi.org/10.1074/jbc.M113.529966
  6. Sikiric, P., Seiwerth, S., Rucman, R., Turkovic, B., Rokotov, D. S., Brcic, L., Sever, M., Klicek, R., Radic, B., Drmic, D., Ilic, S., Kolenc, D., Stambolija, V., Zoricic, Z., Vrcic, H., & Sebecic, B. (2012). Focus on ulcerative colitis: stable gastric pentadecapeptide BPC 157. Current medicinal chemistry, 19(1), 126–132. https://doi.org/10.2174/092986712803414015
  7. Xu, B., Yang, M., Li, Z., Zhang, Y., Jiang, Z., Guan, S., & Jiang, D. (2013). Thymosin β4 enhances the healing of medial collateral ligament injury in rat. Regulatory peptides, 184, 1–5. https://doi.org/10.1016/j.regpep.2013.03.026
  8. Chang, C. H., Tsai, W. C., Lin, M. S., Hsu, Y. H., & Pang, J. H. (2011). The promoting effect of pentadecapeptide BPC 157 on tendon healing involves tendon outgrowth, cell survival, and cell migration. Journal of applied physiology (Bethesda, Md. : 1985), 110(3), 774–780. https://doi.org/10.1152/japplphysiol.00945.2010