GHRH Human

Growth Hormone Releasing Hormone Human
Shipped with Ice Packs
In Stock

Description

The human GHRH sequence is conserved across species and critical for receptor binding. Key residues include the N-terminal region (Tyr-Ala-Asp-Ala-Ile-Phe), which interacts with the extracellular domain of GHRHR .

Receptor Interaction

GHRH binds to GHRHR, a class B G protein-coupled receptor (GPCR) expressed in the pituitary and peripheral tissues. Cryo-EM studies reveal that GHRH adopts an α-helical conformation, forming extensive interactions with GHRHR’s extracellular loops and transmembrane helices .

GHRHR Activation PathwaysMechanism
cAMP-Dependent PathwayGHRH binding activates adenylyl cyclase via Gsα subunits, increasing cAMP and activating protein kinase A (PKA). PKA phosphorylates CREB, enhancing GH gene transcription
Phospholipase C PathwayGHRH stimulates PLC via Gβγ subunits, generating IP₃ (intracellular Ca²⁺ release) and DAG (protein kinase C activation), promoting GH secretion

Peripheral Tissue Effects

GHRH and its receptors are expressed in pancreatic β-cells, hepatocytes, epithelial cells, and tumors, enabling extrapituitary functions:

TissueEffect
PancreasPromotes β-cell survival and insulin secretion
LiverInhibits IGF-1 production, reducing tumor growth in cancer models
CardiovascularEnhances myocardial repair and reduces hypertrophy
Immune SystemModulates inflammation and cellular repair

Antagonism with Somatostatin

GHRH’s secretion is pulsatile and reciprocally regulated by somatostatin (GHIH), creating rhythmic GH release. Somatostatin inhibits GH secretion via distinct receptors, balancing metabolic and growth signals .

Agonists and Antagonists

GHRH analogs are used to modulate GH/IGF-1 axis activity:

CompoundMechanismApplication
MR-409Agonist; inhibits IGF-1 secretion in hepatocytes and tumorsCancer therapy, metabolic disorders
TesamorelinAgonist; approved for HIV-associated lipodystrophyGH deficiency treatment
GHRH AntagonistsBlock autocrine GHRH signaling in tumorsInhibit growth of lung, prostate, and breast cancers

Cancer Therapy

GHRH antagonists (e.g., MIA-690) suppress tumor growth by disrupting autocrine/paracrine signaling loops involving GHRH receptors (e.g., splice variant SV1). This approach reduces IGF-1 levels and cell cycle proteins (e.g., cyclin D1) in cancers .

Molecular Insights from Cryo-EM

The GHRH-GHRHR-Gs complex structure reveals:

  1. N-terminal interactions: Critical for receptor activation and conformational changes in TM6

  2. Disease Implications: Mutations in GHRHR (e.g., isolated GH deficiency) disrupt ligand binding and signaling

Peripheral Receptor Expression

GHRHR is detected in hepatocytes, tumor cells, and pancreatic islets, enabling direct modulation of IGF-1 and GH-independent effects .

Product Specs

Introduction
Growth-hormone-releasing hormone (GHRH), also known as growth-hormone-releasing factor (GRF or GHRF) or somatocrinin, is a 44-amino acid hormone that is produced in the hypothalamus and stimulates the production and release of growth hormone (GH). GHRH is released from the arcuate nucleus of the hypothalamus and travels to the anterior pituitary gland through the hypothalamo-hypophysial portal circulation. Upon reaching the anterior pituitary gland, GHRH binds to specific receptors and triggers the release of GH. GHRH release is pulsatile, leading to a similar pulsatile release of GH. Besides its role in GH regulation, GHRH also directly promotes slow-wave sleep.
Description
Synthetic human growth hormone-releasing hormone is a single, non-glycosylated polypeptide chain composed of 29 amino acids, with a molecular weight of 3358.9 Daltons. This sequence corresponds to the amino-terminal segment of the naturally occurring human growth hormone-releasing hormone, which consists of 44 amino acid residues. The purification of GHRH is achieved through proprietary chromatographic techniques.
Physical Appearance
White, sterile, lyophilized powder.
Formulation
The GHRH peptide was lyophilized at a concentration of 1 mg/ml after extensive dialysis against 1.7 mg sodium phosphate buffer, consisting of 0.1 mg sodium phosphate monobasic and 1.6 mg sodium phosphate dibasic.
Solubility
To reconstitute the lyophilized GHRH, it is recommended to dissolve it in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/ml. This solution can then be further diluted in other aqueous solutions. GHRH is also soluble in 1% acetic acid at a concentration exceeding 1 mg/ml, resulting in a clear, colorless solution.
Stability
Lyophilized growth hormone-releasing hormone remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store it desiccated at a temperature below -18°C. Once reconstituted, GHRF should be stored at 4°C for 2-7 days. For extended storage, it should be kept at a temperature below -18°C. It is important to note that a carrier protein such as 0.1% HSA or BSA should be added for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of this product is greater than 98.0%, as determined by: (a) Reverse-phase high-performance liquid chromatography (RP-HPLC) analysis and (b) Sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) analysis.
Biological Activity
GHRH directly stimulates the anterior pituitary gland, leading to an increase in plasma growth hormone concentrations by promoting the release of natural human growth hormone.
Synonyms
Somatoliberin, Growth hormone-releasing factor, GRF, Growth hormone-releasing hormone, GHRH, Somatocrinin, Somatorelin, Sermorelin, GHRF, MGC119781.
Amino Acid Sequence
The free base of sermorelin has the empirical formula C149H246N 44O42S.
Tyr-Ala-Asp-Ala-Ile-Phe-Thr-Asn-Ser-Tyr-Arg-Lys-Val-Leu-Gly-Gln-Leu-Ser-Ala-Arg-Lys-Leu-Leu-Gln-Asp-Ile-Met-Ser-Arg-NH2.

Q&A

What is GHRH and how does it function in human physiology?

Growth Hormone-Releasing Hormone (GHRH) is a peptide hormone produced primarily in the hypothalamus that plays a fundamental role in regulating growth hormone (GH) secretion from the anterior pituitary gland. GHRH functions by binding to specific receptors on somatotroph cells in the anterior pituitary, stimulating both GH gene expression and the release of GH into the circulation. This mechanism represents a critical component of the hypothalamic-pituitary axis that governs normal human growth and metabolism .

The physiological activity of GHRH begins with its production in the hypothalamus, followed by its release into the hypophyseal portal circulation. Upon reaching the anterior pituitary, GHRH binds to its receptor (GHRHR) on somatotroph cells, activating intracellular signaling cascades that primarily involve the stimulation of adenylyl cyclase and subsequent increase in cyclic adenosine monophosphate (cAMP) production. This signaling pathway ultimately leads to enhanced GH synthesis and secretion, which then acts on various target tissues throughout the body to regulate growth, metabolism, and other physiological processes .

Beyond its classical endocrine role, GHRH has been identified in various extrapituitary tissues, where it may function in autocrine or paracrine signaling systems. This localized production and action of GHRH has been demonstrated in tissues including the skin, hair follicles, and various tumor types, suggesting broader physiological and pathophysiological roles for this hormone .

Research has also revealed that GHRH signaling intersects with multiple other physiological systems, including those involved in energy metabolism, immune function, and cell proliferation. This complex network of interactions underscores the multifaceted role of GHRH in human physiology beyond its namesake function in growth regulation .

What methodologies are used to study GHRH receptor function in human tissues?

Researchers employ multiple complementary approaches to investigate GHRH receptor function in human tissues. mRNA expression analysis using reverse transcription-polymerase chain reaction (RT-PCR) represents a primary method for detecting and quantifying GHRH receptor expression, including its splice variants, in both normal and pathological tissues. This technique has been instrumental in identifying splice variant isoforms (SV1, SV2) of GHRH receptors in various human cancer cell lines and primary tumors .

Functional studies of GHRH receptor signaling often utilize in vitro cell culture systems where cellular responses to GHRH stimulation or antagonism can be measured. These responses include changes in cAMP production, as demonstrated in gastroenteropancreatic cancer cells where GHRH stimulation increased cAMP levels and GHRH antagonists competitively inhibited this effect . Proliferation assays measuring cell growth in response to GHRH and GHRH antagonists provide additional insights into receptor functionality, particularly in cancer cell lines derived from pancreatic, colonic, and gastric origins .

For in vivo research, xenograft models where human cancer cells are implanted into immunodeficient mice allow for the evaluation of GHRH and its receptors in a more complex physiological environment. These models have proven valuable for testing the efficacy of GHRH antagonists against various human cancers and for studying the expression patterns of GHRH and its receptor variants in tumor tissues .

More recently, computational approaches have emerged as powerful tools for studying GHRH receptor structure, function, and interactions with potential antagonists. Various computational tools have been utilized to identify small-molecule compounds that could potentially target GHRH receptors, particularly in therapeutic development efforts .

How do GHRH and GH signaling impact human hair follicle physiology?

GHRH and GH signaling exert complex and sometimes counterintuitive effects on human hair follicle physiology. Research has revealed that human hair follicles express functional elements of the somatotropic axis, including GH receptors on outer root sheath keratinocytes. This suggests that hair follicles can respond directly to circulating or locally produced GH, making them important peripheral targets of this hormone system .

Clinical observations provide compelling evidence for GH's role in hair growth regulation. Acromegaly, characterized by excessive GH serum levels, is associated with hypertrichosis (excessive androgen-independent growth of body hair) and hirsutism in females. Conversely, Laron syndrome, which involves dysfunctional GH receptor-mediated signaling, manifests with alopecia and prominent hair follicle defects. These clinical phenotypes demonstrate that appropriate GH signaling is essential for normal hair follicle function and that both excessive and insufficient GH activity can lead to hair growth abnormalities .

Interestingly, experimental studies have shown that recombinant human GH can inhibit female human scalp hair follicles' growth ex vivo, contrary to what its name might suggest. This inhibitory effect is likely mediated through stimulation of transforming growth factor-β2 (TGF-β2) expression, which is known to induce catagen (the regression phase of the hair cycle). This finding illustrates the complexity of GH actions on hair follicles and suggests that GH may have tissue-specific effects that differ from its classical growth-promoting role .

Methodologically, researchers study these interactions using ex vivo human hair follicle organ culture systems, immunohistochemical analyses to detect receptor expression patterns, and gene expression studies to identify downstream molecular targets of GHRH and GH signaling in hair follicle compartments. These approaches have helped elucidate the nonclassical functions of these neurohormones in human skin physiology .

How do GHRH receptor splice variants influence cancer pathophysiology?

GHRH receptor splice variants, particularly SV1 and SV2, have emerged as critical mediators in cancer pathophysiology, establishing autocrine/paracrine stimulatory loops that promote tumor growth. Research has demonstrated that mRNA for both GHRH and the SV1 isoform of GHRH receptors is expressed in a wide range of human cancers, including pancreatic (SW1990, PANC-1, MIA PaCa-2, Capan-1, Capan-2, CFPAC1), colonic (COLO 320DM, HT-29), and gastric (NCI-N87, HS746T, AGS) cancer cell lines . Additionally, mRNA for the SV2 isoform has been detected in several of these tumor types, indicating a complex receptor expression profile in cancer tissues .

Functionally, these splice variants appear to mediate direct proliferative effects of locally produced GHRH on cancer cells. In vitro studies have demonstrated that GHRH(1-29)NH₂ stimulates the growth of pancreatic, colonic, and gastric cancer cells, while GHRH antagonists like JV-1-38 inhibit this proliferation. The stimulatory effect of GHRH on some gastroenteropancreatic cancer cells is accompanied by increased cAMP production, suggesting that these splice variants can activate similar signaling pathways as the full-length receptor .

Recent research has revealed that SV1 of the human GHRHR can transduce biased signals, adding another layer of complexity to its role in cancer pathophysiology. This constitutive signal bias mediated by SV1 may contribute to altered cellular responses in cancer cells expressing this variant, potentially influencing tumor growth, invasion, and resistance to therapy .

Methodologically, researchers investigating GHRH receptor splice variants in cancer typically employ a combination of molecular approaches, including RT-PCR for detection of variant-specific mRNA expression, in vitro proliferation assays to assess functional responses to GHRH and its antagonists, and signaling studies measuring second messengers like cAMP. These techniques, combined with xenograft models, provide comprehensive insights into how these receptor variants contribute to cancer development and progression .

What are the current approaches for developing GHRH antagonists as cancer therapeutics?

The development of GHRH antagonists as cancer therapeutics involves multiple strategic approaches targeting different aspects of GHRH signaling. Peptide-based GHRH antagonists represent the historical foundation of this field, with compounds like JV-1-38 demonstrating significant antiproliferative effects against various experimental human cancers both in vitro and in xenograft models. These peptide antagonists competitively inhibit GHRH binding to its receptors, preventing the activation of downstream signaling pathways that promote cancer cell proliferation .

More recently, computational approaches have gained prominence in the search for novel small-molecule GHRH antagonists. Researchers have utilized various computational tools to identify potential compounds that could target the GHRH receptor, particularly focusing on non-peptide molecules that might overcome the pharmacokinetic limitations of peptide-based antagonists . These in silico methods often involve virtual screening of chemical libraries, molecular docking simulations, and structure-activity relationship analyses to identify and optimize lead compounds.

The discovery that GHRH receptor splice variants, particularly SV1, are expressed in various human cancers has provided a more targeted approach for antagonist development. Since these variants appear to mediate direct proliferative effects of GHRH on cancer cells, antagonists specifically designed to block SV1 signaling represent a promising therapeutic strategy. This approach benefits from the relatively restricted expression pattern of SV1 in cancer tissues compared to the full-length GHRHR, potentially reducing off-target effects .

Methodologically, the evaluation of GHRH antagonists typically proceeds through a pipeline that includes in vitro assessments of binding affinity and functional inhibition, followed by cell-based proliferation assays and ultimately in vivo testing in xenograft models. This multifaceted approach allows researchers to comprehensively characterize the pharmacological properties and anticancer efficacy of candidate antagonists before advancing them toward clinical development .

How does the experimental investigation of autocrine/paracrine GHRH signaling differ from systemic GHRH effects?

Investigating autocrine/paracrine GHRH signaling requires distinct methodological approaches compared to studying systemic GHRH effects. Autocrine/paracrine signaling involves locally produced GHRH acting on nearby cells within the same tissue, necessitating techniques that can detect and manipulate GHRH production and signaling at the tissue level. In contrast, systemic effects involve hypothalamic GHRH regulating pituitary GH secretion with subsequent effects throughout the body .

For autocrine/paracrine signaling, researchers typically begin by establishing the co-expression of both GHRH and its receptors within the same tissue or cell population. RT-PCR and immunohistochemistry are commonly employed to detect mRNA and protein expression, respectively, for both the ligand and receptor components. This co-localization provides initial evidence for potential autocrine/paracrine activity . Functional studies of autocrine/paracrine signaling often utilize in vitro cell culture systems where the effects of locally produced GHRH can be isolated from systemic influences. These may include conditioned media experiments, where GHRH secreted by cells into culture media affects neighboring cells, or co-culture systems where GHRH-producing and GHRH-responsive cells interact directly .

Demonstration of an autocrine/paracrine loop requires showing that blocking either GHRH production or its receptor binding affects cellular function in the absence of exogenous GHRH. This is typically accomplished using techniques such as RNA interference to suppress GHRH expression, neutralizing antibodies to sequester secreted GHRH, or receptor antagonists to block signaling .

In contrast, studying systemic GHRH effects often involves whole-organism approaches such as measuring serum GH levels in response to GHRH administration or antagonism, assessing downstream effects on IGF-1 production, or evaluating physiological outcomes like growth rates. These studies typically require in vivo models or clinical investigations in human subjects .

What are the key methodological considerations in studying GHRH receptor signal transduction bias?

Studying GHRH receptor signal transduction bias requires sophisticated methodological approaches that can detect and quantify differential activation of downstream signaling pathways. Signal bias refers to the phenomenon where receptors preferentially activate certain signaling pathways over others, often in a ligand-dependent manner. Recent research has identified constitutive signal bias mediated by the human GHRHR splice variant 1 (SV1), highlighting the importance of investigating this aspect of receptor function .

A fundamental methodological consideration involves the simultaneous measurement of multiple signaling outputs from a single receptor. While traditional approaches often focus on canonical pathways like cAMP production for GHRHR signaling, comprehensive evaluation of bias requires monitoring additional pathways such as calcium mobilization, ERK activation, β-arrestin recruitment, and G protein subtype engagement. Employing multiplexed assay systems or parallel experimental setups allows researchers to generate signaling signatures that can reveal biased activation patterns .

Time-course analyses represent another critical consideration, as signaling bias may manifest not only in pathway selectivity but also in the kinetics of pathway activation. Some pathways might show rapid and transient activation while others exhibit delayed but sustained responses. Collecting data at multiple time points provides a more complete picture of the dynamic signaling landscape and can reveal temporal biases that might be missed in single time-point measurements .

Comparing the signaling profiles of different GHRH receptor variants (full-length GHRHR vs. SV1, SV2, etc.) under identical conditions is essential for characterizing variant-specific biases. This requires expression systems where receptor levels can be carefully controlled and normalized, as differences in expression levels can confound interpretations of signaling efficiency. Stable cell lines expressing individual receptor variants at comparable levels provide a reliable platform for such comparisons .

To establish the physiological or pathophysiological relevance of observed signaling biases, researchers must link biased signaling profiles to functional outcomes such as cell proliferation, migration, or hormone secretion. This translation from signaling events to cellular behaviors often requires integrative approaches combining molecular signaling assays with functional readouts in relevant cell types .

How are GHRH antagonists being evaluated for potential treatment of non-cancer diseases?

Methodologically, evaluating GHRH antagonists for non-cancer indications requires disease-specific models and outcome measures. For diabetes-related applications, researchers typically employ models of diabetic complications such as retinopathy, nephropathy, or neuropathy, and assess whether GHRH antagonism can prevent or reverse pathological changes in these tissues. This evaluation process includes histological analyses, functional tests of the affected organs, and molecular markers of tissue damage and repair .

GHRH antagonists are also being explored for potential applications in conditions characterized by excessive GH production, such as acromegaly. While current medical therapies for acromegaly include strategies that block GH release or inhibit GHR activation, GHRH antagonists could provide an alternative approach by targeting the upstream stimulus for GH secretion. Evaluating this application involves measuring the ability of GHRH antagonists to normalize GH and IGF-1 levels in models of GH hypersecretion and assessing improvements in acromegaly-related symptoms and complications .

The discovery of GHRH and its receptors in hair follicles has opened another potential application area for GHRH antagonists in treating certain hair growth disorders. Since GH excess is associated with hypertrichosis and hirsutism, while GH receptor dysfunction leads to alopecia, modulating GHRH signaling could potentially help manage these conditions. Evaluation methods include ex vivo human hair follicle culture systems, where the effects of GHRH antagonists on hair growth, cycling, and follicular gene expression can be assessed .

What challenges exist in translating preclinical findings on GHRH antagonism to human clinical trials?

Translating preclinical findings on GHRH antagonism to human clinical trials faces several significant challenges. Pharmacokinetic considerations represent a primary hurdle, particularly for peptide-based GHRH antagonists. These compounds typically exhibit poor oral bioavailability, short half-lives in circulation, and limited penetration across physiological barriers like the blood-brain barrier. Developing formulations or delivery systems that can overcome these limitations is essential for successful clinical translation .

The potential for off-target effects stemming from the complex interplay between GHRH, GH, and IGF-1 signaling presents another challenge. Since GHRH antagonism ultimately affects GH and IGF-1 levels, which have widespread physiological roles, clinical trials must carefully monitor for unintended consequences in multiple organ systems. This requires comprehensive safety assessments and biomarker analyses to detect both anticipated and unanticipated effects of GHRH antagonism .

Patient selection and stratification represent critical methodological considerations for clinical trials of GHRH antagonists. The heterogeneous expression patterns of GHRH receptors and their splice variants across different tumor types and individual patients suggest that not all patients might respond equally to GHRH antagonism. Developing companion diagnostic tests to identify patients with GHRH-dependent disease processes could enhance the probability of clinical trial success .

Determining appropriate clinical endpoints presents another significant challenge. For cancer indications, traditional endpoints like tumor response rates or progression-free survival may be applicable, but for metabolic or endocrine applications, the relevant endpoints might be less straightforward. Establishing validated biomarkers that can serve as surrogate endpoints for clinical benefit would facilitate the evaluation of GHRH antagonists in early-phase clinical trials .

Regulatory considerations also impact the translation of GHRH antagonists to clinical applications. The novelty of targeting GHRH receptors, particularly their splice variants, may require additional preclinical data to satisfy regulatory requirements for first-in-human studies. Providing robust evidence of the mechanism of action, target engagement, and safety profile is essential for advancing these agents through the regulatory pathway .

How might genomic and proteomic approaches enhance our understanding of GHRH signaling in human diseases?

Genomic and proteomic approaches offer powerful tools for advancing our understanding of GHRH signaling in human diseases. Transcriptomic profiling via RNA sequencing (RNA-seq) enables comprehensive analysis of gene expression changes in response to GHRH stimulation or antagonism, revealing downstream molecular targets and signaling networks that mediate GHRH effects in different tissues and disease states. This approach can identify novel genes and pathways involved in GHRH signaling that might serve as additional therapeutic targets or biomarkers .

Single-cell RNA sequencing represents a particularly valuable methodology for studying heterogeneous tissues where GHRH might affect specific cell populations differently. By providing cell-type-specific transcriptomic profiles, this approach can identify which cells within a tissue express GHRH receptors and respond to GHRH signaling, offering insights into the cellular basis of GHRH effects in complex tissues like tumors or the pituitary gland .

Proteomic approaches complement genomic analyses by capturing post-transcriptional and post-translational regulatory events that influence protein expression, modification, and function. Mass spectrometry-based proteomics can identify changes in protein abundance, phosphorylation status, and protein-protein interactions following GHRH receptor activation or inhibition, providing a more complete picture of GHRH signaling dynamics than transcriptomic analysis alone .

Phosphoproteomics specifically focuses on protein phosphorylation events, which are crucial for signal transduction. This approach can map the phosphorylation cascades initiated by GHRH receptor activation, identifying key regulatory nodes and feedback mechanisms within the signaling network. Such information is valuable for understanding how GHRH receptor splice variants might differentially activate downstream pathways and how GHRH antagonists disrupt these signaling events .

Integrative multi-omics approaches that combine genomic, transcriptomic, proteomic, and metabolomic data offer the most comprehensive view of GHRH signaling systems. By correlating changes across these different molecular levels, researchers can construct detailed models of GHRH signaling networks and their dysregulation in disease states. These models can guide rational drug design efforts for developing more specific and effective GHRH-targeting therapeutics .

Product Science Overview

Introduction

Growth Hormone-Releasing Hormone (GHRH), also known as somatocrinin or somatorelin, is a peptide hormone that plays a crucial role in regulating the secretion of growth hormone (GH) from the anterior pituitary gland. GHRH is produced in the hypothalamus and is essential for normal growth, metabolism, and overall body structure.

Discovery and Nomenclature

GHRH was first identified in the human hypothalamus between 18 and 29 weeks of gestation, which corresponds to the onset of growth hormone production in fetuses . The hormone is known by several names, including somatocrinin, somatoliberin, and growth hormone-releasing factor (GHRF) .

Structure and Function

GHRH is a 44-amino acid peptide hormone . It is released from neurosecretory nerve terminals in the hypothalamus and carried by the hypothalamo-hypophyseal portal system to the anterior pituitary gland. Here, it binds to the growth hormone-releasing hormone receptor (GHRHR) on somatotroph cells, stimulating the secretion of growth hormone .

Mechanism of Action

The binding of GHRH to its receptor activates the cAMP-dependent pathway, leading to increased intracellular cyclic adenosine monophosphate (cAMP) levels . This activation results in the phosphorylation of the transcription factor cAMP response element-binding protein (CREB), which enhances the transcription of the GH gene . Additionally, GHRH promotes slow-wave sleep directly .

Regulation

The secretion of GHRH is regulated by various factors, including somatostatin, a hormone that inhibits GH release . Ghrelin, known as the “hunger hormone,” also regulates GHRH and GH release . Physiological conditions such as sleep, stress, exercise, and food intake influence the release of GHRH and somatostatin .

Clinical Significance

Excessive production of GHRH can lead to conditions such as acromegaly, characterized by the enlargement of hands, feet, and facial features . Conversely, insufficient GHRH production can result in growth hormone deficiency, affecting normal growth and metabolism .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.