Hexarelin (C₄₇H₅₈N₁₂O₆), also known as Examorelin, is a synthetic hexapeptide growth hormone-releasing peptide (GHRP) with the sequence His-D-2-methyl-Trp-Ala-Trp-D-Phe-Lys-NH₂ . It belongs to the GHRP family, which mimics the action of ghrelin, the natural growth hormone secretagogue, but exhibits greater stability and potency . Hexarelin is not approved for clinical use but has been extensively studied for its therapeutic potential in endocrinology, cardiology, and metabolic disorders.
Hexarelin stimulates growth hormone (GH) secretion through two primary pathways:
Pituitary Activation: Binds to growth hormone secretagogue receptor (GHSR1a) in the pituitary gland, increasing GH release while suppressing somatostatin (GH-inhibiting hormone) .
Direct Cardiac Effects: Activates CD36 and GHSR1a receptors in cardiac tissue, promoting cardioprotection and improving myocardial function .
Parameter | Value | Source |
---|---|---|
Molecular Weight | 887.0 g/mol | |
GH Secretion (ED₅₀) | 0.50–0.64 µg/kg (IV) | |
Plasma Half-Life | ~55 minutes |
Hexarelin induces dose-dependent GH secretion, peaking at 30–60 minutes post-administration . In clinical trials:
Healthy Adults: A 2 µg/kg IV dose increased peak GH to 55.0 ng/mL, with an AUC₀–₁₈₀ of 3.695 µg·min/mL .
Children: Improved growth velocity (5.3 cm/year → 7.4 cm/year) despite partial pituitary desensitization after chronic use .
Hexarelin demonstrates cardioprotective effects in animal models:
In insulin-resistant MKR mice, twice-daily hexarelin (200 µg/kg IP) improved:
Glucose Tolerance: Reduced plasma/liver triglycerides.
Long-term use (16–20 weeks) in healthy adults caused partial attenuation of GH responses, with AUC₆₅₅₀ reducing from 19.1 µg/L·h (baseline) to 10.5 µg/L·h (week 16). Recovery occurred within 4 weeks post-treatment .
Hexarelin outperforms natural ghrelin in cardiovascular models:
Ischemia/Reperfusion: Greater reduction in LV end-diastolic pressure vs. ghrelin in hypophysectomized rats .
GHRP vs. MK-0677: Higher coronary vasoconstriction (30 µmol/L) vs. peptidomimetics like EP51389 .
Condition | Model | Hexarelin Dose | Key Outcome |
---|---|---|---|
Ischemic Injury | Rat hearts | 1 µmol/L | ↓ Infarct size, ↑ LV function |
Cardiac Fibrosis | Hypertensive rats | 30 days | ↓ Collagen I/III, ↓ TGF-β |
Atherosclerosis | Sprague-Dawley | Chronic | ↓ Plaque formation, ↑ HDL/LDL ratio |
Hexarelin is a synthetic hexapeptide comprising six amino acids that functions primarily as a growth hormone secretagogue. Its mechanism of action involves binding to the growth hormone secretagogue receptor (GHSR) in the hypothalamus and pituitary gland, initiating a signaling cascade that stimulates the release of growth hormone (GH) . This process mimics the action of ghrelin, an endogenous peptide hormone. Unlike some peptides that require adjunct compounds, Hexarelin can function independently to provide reliable GH stimulation with long-lasting effects due to its unique molecular configuration .
The molecular weight of the specifically labeled Hexarelin receptor in cardiac tissues has been identified as approximately 84,000 Da through photoaffinity labeling studies with [125I]Tyr-Bpa-Ala-hexarelin . This binding differs from typical GHRP interactions, suggesting tissue-specific receptor variants.
Hexarelin distinguishes itself from other GHRPs through several key characteristics:
Potency: Hexarelin demonstrates higher potency in stimulating GH release compared to older established peptides .
Receptor sensitivity: It exhibits significantly lower rates of pituitary response desensitization than other GHRPs .
Half-life advantage: Its molecular configuration provides a longer half-life and more sustained activity than other GHRPs such as GHRP-2 or GHRP-6 .
Independent function: Unlike some peptides that require stacking or adjunct compounds, Hexarelin can function independently to provide reliable GH stimulation .
Hormonal profile: Hexarelin patients reportedly experience no appetite stimulation, cortisol spikes, or prolactin increases, which are common side effects with other GHRPs .
These distinctive properties make Hexarelin particularly valuable for research applications requiring consistent growth hormone stimulation with minimal secondary effects.
Hexarelin affects multiple physiological systems through both direct receptor binding and secondary growth hormone-mediated effects:
Endocrine system: Beyond GH release, Hexarelin influences cortisol, prolactin, and adrenocorticotropic hormone (ACTH) levels . These secondary hormonal effects contribute to its comprehensive physiological impact.
Cardiovascular system: Hexarelin has demonstrated protecting activity against postischemic ventricular dysfunction in hearts from GH-deficient and senescent rats . It induces dose-dependent increases in coronary perfusion pressure, partially mediated by L-type Ca2+ channels and protein kinase C .
Metabolic regulation: Research indicates that Hexarelin regulates PPARγ through interaction with scavenger receptor CD36, potentially influencing metabolic processes . Importantly, unlike traditional PPARγ agonists like troglitazone, Hexarelin does not increase CD36 expression, which may prevent proatherosclerotic events .
Musculoskeletal system: Administration leads to greater lean muscle mass, enhanced collagen production, increased bone density, and accelerated soft tissue repair .
Neurological effects: Emerging research suggests potential benefits for neurological conditions such as amyotrophic lateral sclerosis (ALS) .
This multisystem influence underscores the complexity of Hexarelin's physiological effects and its potential value in various research applications.
Hexarelin exhibits distinct mechanisms in cardiac tissue compared to its pituitary actions, suggesting tissue-specific pathways:
In cardiac tissue, Hexarelin binds to specific receptors that induce a dose-dependent increase in coronary perfusion pressure . This vasoconstriction effect is partially mediated by L-type Ca2+ channels and protein kinase C, as demonstrated by inhibition studies using nifedipine, chelerythrine, and bisindolylmaleimide . Notably, prostaglandins and thromboxanes do not appear to be involved in this process, as diclofenac and 1-(7-carboxyheptyl)imidazole had no inhibitory effect on Hexarelin-induced vasoconstriction .
The cardiac receptor for Hexarelin has been characterized through photoaffinity labeling studies using [125I]Tyr-Bpa-Ala-hexarelin. These studies revealed specific binding sites with distinct properties from those in pituitary tissue . Autoradiographic distribution studies have mapped these binding sites throughout the rat heart, providing evidence for direct cardiac actions independent of GH-mediated effects .
In contrast, pituitary actions primarily involve GHSR activation leading to GH release, with subsequent systemic effects. This dual-pathway action makes Hexarelin particularly valuable for cardiovascular research applications.
Hexarelin exhibits a unique interaction with PPARγ signaling that distinguishes it from traditional PPARγ agonists:
Unlike typical PPARγ agonists such as troglitazone, Hexarelin regulates PPARγ through interaction with scavenger receptor CD36 . This alternative pathway leads to a distinct pattern of gene regulation. While Hexarelin influences some PPARγ targets, it does not affect others typically modulated by troglitazone, including adipocyte fatty acid binding protein FABP4 (aP2) and adipophilin .
Importantly, Hexarelin does not increase CD36 expression at either the gene or protein level, contrasting with troglitazone which significantly induces CD36 in both adipocytes and macrophages . This selective modulation may prevent undesired increases in macrophage CD36, a condition associated with proatherosclerotic events .
Additionally, Hexarelin helps maintain PPARγ expression and steady-state levels in adipocytes and macrophages, whereas troglitazone decreases PPARγ expression . This differential regulation suggests that Hexarelin may offer metabolic benefits without some of the undesirable effects associated with traditional PPARγ agonists.
Parameter | Hexarelin Effect | Troglitazone Effect |
---|---|---|
FABP4/aP2 expression | Minimal change | Significant increase |
Adipophilin | Minimal change | Significant increase |
CD36 expression | No change | Significant increase |
PPARγ expression | Maintained | Decreased |
These findings indicate that Hexarelin represents a novel class of selective PPARγ modulators with potentially advantageous tissue-specific effects.
The Langendorff isolated perfused heart model has proven particularly valuable for studying Hexarelin's direct cardiovascular effects:
This experimental setup allows for precise measurement of coronary perfusion pressure changes in response to Hexarelin administration . The model effectively isolates the heart from systemic influences, enabling researchers to distinguish direct cardiac effects from those mediated by secondary hormonal changes. Implementation involves:
Animal selection: Male Sprague-Dawley rats (300-325g) have been successfully used, though other rodent models may be appropriate depending on research questions .
Heart isolation and mounting: Following animal euthanasia, hearts must be quickly excised and mounted on the Langendorff apparatus to maintain tissue viability .
Experimental protocol: Dose-response curves can be established through successive infusions of increasing Hexarelin concentrations (10 nmol/L to 30 μmol/L), with sufficient time at each concentration to reach steady state (typically 5-10 minutes) . Washout periods between experiments prevent carryover effects.
Intervention studies: The influence of various signaling pathways can be assessed by administering specific inhibitors (e.g., nifedipine for L-type Ca2+ channels, chelerythrine for protein kinase C) before repeating the dose-response curve .
Data analysis: Changes in coronary resistance are evaluated by computing the percentage change in coronary perfusion pressure before and after reaching steady state with each Hexarelin dose .
This methodology has successfully demonstrated Hexarelin's dose-dependent vasoconstrictor effects and identified key mediating pathways .
Several complementary techniques have proven effective for identifying and characterizing Hexarelin binding sites:
Photoaffinity labeling with radiolabeled ligands: Using [125I]Tyr-Bpa-Ala-hexarelin as a photoactivatable radioligand enables specific labeling of Hexarelin receptors . The protocol involves:
Incubating membrane preparations with the radioligand (1.5-45 nmol/L for saturation studies)
Defining nonspecific binding through displacement with excess unlabeled Hexarelin (10 μmol/L)
UV exposure to activate cross-linking
SDS-PAGE separation and autoradiography detection
Competition binding assays: These assess the affinity of various ligands by measuring their ability to displace the radiolabeled photoactivatable hexarelin derivative .
Deglycosylation studies: Treatment with N-Glycosidase F can identify glycosylation patterns of Hexarelin receptors, helping distinguish receptor subtypes .
Autoradiographic distribution studies: This technique maps binding sites in tissue sections:
These methodologies have successfully characterized Hexarelin receptors in various tissues, revealing tissue-specific binding profiles and receptor properties.
Effective protocols for evaluating Hexarelin's effects on growth hormone secretion should address several key methodological considerations:
Subject preparation: Standardize testing conditions to minimize confounding variables:
Study design: Implement controlled, randomized designs:
Administration protocol: Standardize administration methods:
Comparative analysis: Include reference compounds:
Sampling strategy: Implement appropriate blood sampling protocols:
Collect multiple post-administration samples to capture the complete response curve
Use validated GH assays with appropriate sensitivity and specificity
This methodological approach allows for rigorous assessment of Hexarelin's GH-secreting properties and the factors that modulate its efficacy.
Hexarelin demonstrates significant therapeutic potential for cardiovascular conditions through several mechanisms:
The peptide features protecting activity against postischemic ventricular dysfunction, particularly in hearts from GH-deficient and senescent rats . This cardioprotective effect suggests potential applications in conditions involving ischemia-reperfusion injury, such as myocardial infarction or cardiac surgery.
Research indicates that Hexarelin may reduce recovery time from cardiac bypass surgery , possibly through its protective effects on cardiac function during ischemia-reperfusion sequences. This application could significantly improve outcomes for patients undergoing cardiac procedures.
Importantly, Hexarelin's cardiac effects appear to involve direct receptor-mediated mechanisms rather than exclusively resulting from systemic GH increases . The identification and characterization of specific Hexarelin binding sites in cardiac tissue provide a molecular basis for these direct cardiac actions .
The unique interaction of Hexarelin with PPARγ signaling pathways, particularly its failure to increase CD36 expression (unlike traditional PPARγ agonists), may help prevent proatherosclerotic events . This selective modulation could offer metabolic and cardiovascular benefits without promoting atherosclerosis.
These properties position Hexarelin as a promising candidate for treating various cardiovascular conditions, particularly those involving ischemic damage or requiring myocardial protection during procedures.
The development of Hexarelin-based therapies for metabolic diseases requires careful consideration of several factors:
Distinct PPARγ modulation: Hexarelin interacts with PPARγ through the scavenger receptor CD36, resulting in a unique pattern of gene regulation different from traditional PPARγ agonists . Development strategies should leverage this selective modulation to maximize metabolic benefits while minimizing adverse effects.
CD36 expression control: Unlike traditional PPARγ agonists that increase CD36 expression (potentially promoting atherosclerosis), Hexarelin maintains stable CD36 levels . This property may be particularly valuable for diabetic patients who already face elevated cardiovascular risk.
PPARγ expression maintenance: Hexarelin helps maintain PPARγ expression and steady-state levels in adipocytes and macrophages, contrasting with traditional agonists like troglitazone which decrease PPARγ expression . This effect should be preserved in therapeutic development.
Insulin response improvement: Evidence suggests Hexarelin may improve the insulin response in diabetics , though the mechanism remains to be fully elucidated. This effect could make it particularly valuable for type 2 diabetes therapy.
Hormonal considerations: When developing metabolic therapies, researchers must account for Hexarelin's effects on multiple hormonal systems, including growth hormone, cortisol, prolactin, and ACTH . The ideal therapeutic approach would maximize beneficial metabolic effects while minimizing unwanted hormonal perturbations.
Tissue specificity: Receptor distribution studies using autoradiography can inform targeted delivery approaches to maximize effects in metabolically relevant tissues while minimizing systemic exposure.
These considerations should guide both preclinical development and clinical trial design for Hexarelin-based metabolic disease therapies.
Several areas show particular promise for advancing Hexarelin research:
Receptor subtype characterization: Further investigation of tissue-specific Hexarelin receptor variants identified through photoaffinity labeling could reveal new therapeutic targets and explain tissue-specific responses. Structural biology approaches could elucidate binding domain differences.
Metabolic pathway modulation: Deeper exploration of Hexarelin's unique PPARγ modulation through CD36 could yield insights into selective PPARγ modulation without atherogenic effects. Comprehensive transcriptomic and proteomic analyses would help identify the full spectrum of regulated genes.
Cardioprotective mechanisms: Building on observations of protection against postischemic ventricular dysfunction , research should identify precise cellular and molecular mechanisms. Long-term studies could assess potential for preventing heart failure progression.
Neurological applications: Given reported benefits for ALS patients , mechanistic studies of Hexarelin's neuroprotective effects could reveal novel therapeutic approaches for neurodegenerative diseases. Animal models of neurodegeneration would provide valuable insights.
Aging and longevity effects: As a 3rd generation HGH therapy with enhanced anti-aging benefits , Hexarelin's impact on cellular senescence markers, telomere maintenance, and age-related disease outcomes warrants investigation through longitudinal studies.
Combinatorial approaches: Evaluating synergistic effects of Hexarelin with other therapeutic agents could optimize clinical applications across multiple conditions.
These research directions have significant potential to expand the therapeutic applications of Hexarelin and deepen our understanding of its molecular and physiological effects.
Several methodological advances could significantly enhance Hexarelin research:
Advanced receptor characterization techniques:
Cryo-electron microscopy to determine the three-dimensional structure of Hexarelin-receptor complexes
Single-molecule fluorescence resonance energy transfer (FRET) to visualize real-time binding dynamics
CRISPR-Cas9 receptor editing to create models for studying receptor subtype functions
Improved tissue-specific delivery systems:
Nanoparticle formulations targeting specific tissues (cardiac, neurological, or metabolic)
Controlled-release systems optimizing pharmacokinetic profiles
Tissue-specific promoters for genetic expression systems in animal models
Comprehensive physiological monitoring:
Multimodal in vivo imaging techniques for tracking tissue responses
Continuous glucose monitoring systems for metabolic studies
Telemetric systems for long-term cardiovascular monitoring in animal models
Systems biology approaches:
Integration of transcriptomic, proteomic, and metabolomic data to map pathway effects
Computational modeling of Hexarelin's effects across multiple physiological systems
Machine learning algorithms to identify predictive biomarkers of response
Translational research tools:
Patient-derived organoids for personalized response testing
Humanized animal models better reflecting human physiology
Improved clinical trial designs with adaptive protocols and biomarker stratification
These methodological advances would address current limitations in understanding Hexarelin's complex mechanisms and accelerate translation to clinical applications.
Combination therapies incorporating Hexarelin hold significant potential for managing complex conditions through synergistic mechanisms:
For metabolic syndrome and type 2 diabetes, combining Hexarelin with traditional antidiabetic agents could yield superior outcomes. Hexarelin's improvement of insulin response paired with its unique PPARγ modulation that maintains stable CD36 levels could complement medications targeting other pathways. This approach might achieve better glycemic control while reducing cardiovascular risk, a significant concern with some current diabetes therapies.
In cardiovascular applications, combining Hexarelin's cardioprotective effects with standard heart failure therapies could provide enhanced myocardial protection. The combination might be particularly valuable in settings of acute ischemia-reperfusion injury (e.g., following myocardial infarction or cardiac surgery) where Hexarelin has shown promise in reducing recovery time .
For neurological conditions like ALS, integrating Hexarelin with disease-modifying therapies could address multiple pathophysiological aspects simultaneously. Hexarelin's potential neuroprotective effects might complement treatments targeting excitotoxicity, protein aggregation, or neuroinflammation, providing more comprehensive disease management.
Age-related sarcopenia might be more effectively addressed by combining Hexarelin's effects on lean muscle mass and tissue repair with targeted resistance exercise protocols. This combination could synergistically enhance muscle protein synthesis and functional outcomes.
Hexarelin was originally developed in the 1980s by a team of researchers at Tulane Medical School. The goal was to create a growth hormone secretagogue that did not cause incidental elevation of sex hormones, insulin, or glucagon . This peptide was designed to mimic the action of ghrelin, a naturally occurring hormone that stimulates the release of growth hormone .
Hexarelin works by binding to the growth hormone secretagogue receptor (GHS-R1a) located in the pituitary gland and hypothalamus. This binding triggers a cascade of physiological pathways that lead to the release of growth hormone. Hexarelin also inhibits the release of somatostatin, a hormone that inhibits growth hormone secretion, thereby increasing the overall levels of growth hormone in the body .
Hexarelin has been studied for its potential benefits in various areas, including:
Hexarelin has been found to be generally safe when used appropriately. However, like any compound, it may have side effects. Some studies have reported slight increases in prolactin, adrenocorticotropic hormone (ACTH), and cortisol levels in humans . There are conflicting reports on its ability to elevate insulin-like growth factor 1 (IGF-1) and insulin-like growth factor-binding protein 1 (IGFBP-1) levels .