GHRL Protein

Ghrelin Human
Shipped with Ice Packs
In Stock

Description

Ghrelin Human contains 28 amino acids and a total molecular mass of 3370.9 Dalton and a molecular formula of C149H249N47O42.
The GHRL is purified by proprietary chromatographic techniques.

Product Specs

Introduction
Obestatin, found in the stomach and small intestine lining of mammals like humans, is believed to suppress appetite. This peptide hormone, a small protein, shares a gene with ghrelin, an appetite stimulant. This gene produces a protein that splits into ghrelin and obestatin. While ghrelin, an endogenous ligand for the growth hormone secretagogue receptor, regulates growth hormone release, obestatin is thought to counter ghrelin's effects. Ghrelin, originating from the preprohormone preproghrelin, also yields obestatin. Acting as an endogenous ligand for the orphan G protein-coupled receptor GPR39, ghrelin contributes to satiety and reduced food intake.
Description
Human Ghrelin, comprising 28 amino acids, has a molecular weight of 3370.9 Daltons and a molecular formula of C₁₄₉H₂₄₉N₄₇O₄₂. The purification of GHRL is achieved through specialized chromatographic methods.
Physical Appearance
Sterile Filtered Yellowish lyophilized (freeze-dried) powder, potentially exhibiting a gel-like consistency.
Formulation
GHRL is lyophilized without the addition of any extra substances.
Solubility
To prepare a working solution, add deionized water to achieve an approximate concentration of 0.5mg/ml. Allow the lyophilized pellet to fully dissolve.
Stability
For long-term storage, keep lyophilized Ghrelin at -20°C. After reconstitution, aliquot the product to minimize freeze-thaw cycles. Reconstituted GHRL can be stored at 4°C for a limited duration; it remains stable for up to two weeks at this temperature.
Purity
Purity exceeds 97%, as determined by: (a) RP-HPLC analysis. (b) SDS-PAGE analysis.
Synonyms
Appetite-regulating hormone precursor, Growth hormone secretagogue, Growth hormone-releasing peptide, GHRP, Motilin-related peptide, M46 protein, Ghrelin, Obestatin, MTLRP.
Amino Acid Sequence
Gly-Ser-Ser(n-octanoyl)-Phe-Leu-Ser-Pro-Glu-His-Gln-Arg-Val-Gln-Gln-Arg-Lys-Glu-Ser-Lys-Lys-Pro-Pro-Ala-Lys-Leu-Gln-Pro-Arg-OH.

Q&A

What is GHRL protein and what are its primary functions?

GHRL encodes ghrelin, a peptide hormone primarily secreted by the stomach. Ghrelin acts upon the growth hormone secretagogue receptor (GHSR1), a G protein-coupled receptor with multiple physiological functions including:

  • Growth hormone secretion stimulation

  • Appetite regulation

  • Energy expenditure modulation

  • Adiposity regulation

  • Insulin release control

How does GHRL protein expression differ between tissues?

GHRL expression varies significantly across different tissues, with primary expression in the stomach. When investigating tissue-specific expression, researchers should:

  • Employ qRT-PCR for quantitative analysis using appropriately designed primers. Example primers used in gastric cancer research:

    • GHRL: 5′-TACTACTCTCCACGCCC-3′ (F), 5′-AGGGGCCATCCACAGTCTTC-3′ (R)

  • Use immunohistochemistry (IHC) with specific antibodies (e.g., Anti-GHRL, Santa Cruz Biotechnology, sc-293422) to visualize tissue localization .

  • Analyze publicly available datasets from resources such as GEPIA, UALCAN, and GTEx to compare expression across tissues .

Research has shown that GHRL expression in gastric cancer is lower compared to healthy tissue samples (p = 0.0036), yet interestingly shows higher expression in middle and late-stage cancers than in early stages .

What biological pathways and protein interactions involve GHRL?

GHRL protein participates in multiple signaling pathways and protein interactions:

  • Heteromerization with other receptors: GHSR1a can form heteromers with other transmembrane proteins, most notably the dopamine receptor D1 (DRD1). These GHSR1a-DRD1 complexes promote synaptic plasticity and formation of hippocampal memory .

  • Immune system interactions: GHRL expression correlates with tumor-infiltrating lymphocytes (TILs) in gastric cancer, showing significant associations with markers of:

    • CD8+ T cells (CD8A, CD8B)

    • B cells (CD19)

    • Monocytes (CD86)

    • M2 macrophages (CD163)

    • Dendritic cells (BDCA-1/CD1C)

    • T-regulatory cells (FOXP3, CCR8)

    • T-cell exhaustion (CTLA4, LAG3)

  • Signaling cross-talk: Beyond direct binding, GHRL can influence other pathways through signaling cross-talk, which can modify GHSR1a signaling through:

    • Biased signaling (preferential coupling to one pathway)

    • G protein switching

    • Signaling suppression or enhancement

What are the most effective techniques for studying GHRL expression in clinical samples?

For comprehensive GHRL expression analysis in clinical samples, researchers should employ a multi-technique approach:

  • Quantitative Real-Time PCR (qRT-PCR):

    • Extract total RNA using specialized kits (e.g., Yeasen Biotechnology RNA extraction kit)

    • Synthesize cDNA using appropriate SuperMix (e.g., Hifair III 1st Strand cDNA Synthesis SuperMix)

    • Perform qRT-PCR using SYBR Primix systems on instruments like ABI-7500

    • Use the 2^(-△△Ct) method for expression comparison with appropriate housekeeping genes like Actin

  • Immunohistochemistry (IHC):

    • Use validated anti-GHRL antibodies

    • Follow standardized staining protocols for tissue specimens

    • Analyze staining patterns and intensity to determine protein localization and expression levels

  • Database Analysis:

    • Utilize databases like GEPIA2, UALCAN, and GTEx for comparative expression analysis

    • Apply appropriate statistical methods (e.g., log-rank test for survival analysis, Spearman's correlation for association studies)

  • Meta-analysis of Transcriptomics Data:

    • Apply quantile normalization to individual datasets

    • Use the ComBat procedure to address batch effects across datasets

    • Conduct Cochran's Q-test to assess statistical heterogeneity

    • Perform meta-analysis using a random-effects model

    • Apply appropriate false discovery rate methods (Benjamini–Hochberg)

This comprehensive approach provides more reliable results than any single method, reducing false positives and contradictions caused by heterogeneity in sampling and study design.

How can protein language models be applied to study GHRL structure and function?

Protein language models (pLMs) offer powerful approaches to study GHRL structure and function:

  • Sequence Analysis:

    • AMPLIFY 350M (Amgen-Mila Protein Language model for InFerence and discoverY) provides efficient analysis with 43× fewer parameters than ESM2 15B while achieving superior performance

    • Use sequence-only models to analyze the full distribution of natural proteins, including GHRL

  • Structure Prediction and Analysis:

    • Sequence-to-structure and structure-to-sequence models can be employed, though benchmarking shows they may not match the performance of pLMs on some protein design tasks

    • Note that structure-based models like AlphaFold2 may struggle to distinguish between non-proteins and disordered proteins

  • Methodological Considerations:

    • Training datasets should be derived from comprehensive databases like UniRef100 and supplemented with paired sequences from specialized databases

    • Validation sets should be selected using protein evidence level annotations

    • Proteome completeness scores ensure validation sets represent natural distributions

These computational approaches complement experimental methods and can provide insights into GHRL protein structure-function relationships that might be difficult to obtain through traditional laboratory techniques alone.

What experimental designs are most appropriate for studying GHRL heteromerization?

Studying GHRL heteromerization, particularly GHSR1a interactions with other transmembrane proteins, requires specialized experimental approaches:

  • Overexpression Systems in Cell Lines:

    • Co-express GHSR1a with potential interacting partners (e.g., DRD1) in appropriate cell lines

    • Include appropriate controls to verify specificity of interactions

    • Note that verification in native tissues is essential, as many interactions have been shown primarily in overexpression systems

  • Biophysical Techniques:

    • Förster/fluorescence resonance energy transfer (FRET) to detect protein-protein interactions

    • Bioluminescence resonance energy transfer (BRET) for real-time monitoring of interactions

    • Co-immunoprecipitation followed by Western blotting to confirm physical associations

  • Functional Assays:

    • G protein coupling assays to detect G protein switching

    • Second messenger production measurements to identify biased signaling

    • Receptor trafficking studies to monitor internalization and recycling dynamics

  • In vivo Models:

    • Develop transgenic models expressing tagged versions of GHSR1a and interacting partners

    • Use tissue-specific expression systems to study heteromerization in relevant physiological contexts

    • Apply proximity ligation assays in tissue sections to visualize heteromers

When designing these experiments, researchers should be aware that GHSR1a forms documented heteromers with DRD1, promoting synaptic plasticity and hippocampal memory formation, with implications for conditions like Alzheimer's disease where GHSR1a-DRD1 complexes are reduced in favor of GHSR1a-Aβ complexes .

How does GHRL expression correlate with immune infiltration in cancer progression?

GHRL expression shows significant correlations with immune infiltration in gastric cancer (GC), potentially influencing cancer progression:

This relationship between GHRL and immune infiltration provides potential therapeutic targets and prognostic indicators for gastric cancer treatment strategies.

What is the role of GHRL in the Correa cascade of gastric cancer progression?

The Correa cascade describes the progression from normal gastric mucosa to gastric cancer through sequential stages. GHRL shows dynamic expression changes throughout this cascade:

Understanding these expression patterns provides insights into the molecular mechanisms of gastric cancer progression and identifies potential intervention points in the Correa cascade.

How do GHRL-receptor interactions influence neurodegenerative diseases?

GHRL-receptor interactions, particularly GHSR1a heteromerization, have emerging roles in neurodegenerative conditions:

  • Alzheimer's Disease (AD) Associations:

    • GHSR1a-DRD1 complexes promote synaptic plasticity and hippocampal memory formation

    • A reduction in GHSR1a-DRD1 complexes in favor of GHSR1a-Aβ complexes correlates with Alzheimer's disease development

    • This shift in heteromerization partners may contribute to cognitive decline in AD

  • Molecular Mechanisms:

    • GHSR1a and DRD1 heteromers likely activate unique signaling pathways that support neuronal function

    • When GHSR1a binds to Aβ instead, these neuroprotective pathways may be disrupted

    • This represents a potential pathological function of GHSR1a heteromers in neurodegeneration

  • Therapeutic Implications:

    • Targeting GHSR1a heteromerization represents a novel approach for treating Alzheimer's disease

    • Compounds that stabilize GHSR1a-DRD1 interactions or prevent GHSR1a-Aβ formation could be therapeutically valuable

    • Understanding the structural basis of these interactions is crucial for drug development

This area represents an exciting frontier in GHRL research, connecting appetite regulation with cognitive function and neurodegeneration through receptor heteromerization mechanisms.

Why have GHSR1a antagonists failed to produce expected effects on appetite?

Despite the established role of GHSR1a in appetite stimulation, receptor antagonists have not produced the expected reductions in food intake, creating an apparent contradiction:

  • The Contradiction:

    • Following the discovery that GHSR1a stimulates food intake, receptor antagonists were developed as potential therapies to regulate appetite

    • Despite reducing GHSR1a signaling, these antagonists often failed to produce the desired effects on appetite

  • Explanatory Mechanisms:

    • Heteromerization complexity: GHSR1a forms heteromers with multiple partners, creating signaling dynamics not addressed by simple antagonism

    • Signaling cross-talk: Even when GHSR1a is blocked, parallel pathways may compensate

    • G protein switching: In certain heteromeric contexts, GHSR1a may couple to different G proteins, changing the signaling outcome in ways not affected by traditional antagonists

    • Biased signaling: Some antagonists may block certain pathways while leaving others intact

  • Research Implications:

    • Future therapeutic approaches should consider designing compounds that target specific heteromeric complexes

    • Understanding the full spectrum of GHSR1a interactions is essential for effective intervention in appetite regulation

    • Agents that modify heteromer formation may prove more effective than simple receptor antagonists

This contradiction highlights the importance of considering protein interactions beyond simple ligand-receptor binding in drug development.

How can researchers reconcile contradictory GHRL expression patterns across different cancer studies?

Researchers studying GHRL in cancer face contradictory findings regarding its expression and prognostic value:

  • Observed Contradictions:

    • Some studies report decreased GHRL expression in gastric cancer compared to healthy tissue

    • Other studies suggest increased GHRL expression correlates with advanced cancer stages and poor prognosis

    • These seemingly contradictory findings create challenges in establishing GHRL's role in cancer

  • Methodological Approaches to Reconcile Contradictions:

    • Meta-analysis of transcriptomics data: Integrating multiple datasets using random effects models and appropriate statistical methods

    • Standardization of sampling techniques: Ensuring consistent collection and processing of samples

    • Stratification by cancer subtypes and stages: Analyzing expression patterns within well-defined cancer subgroups

    • Integration of multiple techniques: Combining qRT-PCR, IHC, and database analysis to create more robust findings

  • Best Practices for Addressing Contradictions:

    • Clearly define the cancer stage and subtype being studied

    • Implement rigorous normalization and batch effect correction when comparing datasets

    • Account for tumor heterogeneity in sampling design

    • Consider GHRL's dynamic expression changes throughout cancer progression rather than static measurements

Meta-analysis approaches that integrate multiple datasets can significantly decrease false positives and contradictions caused by alterations in homogeneity, sampling, and study design .

What technical challenges exist in studying GHRL protein-protein interactions?

Studying GHRL protein-protein interactions, particularly heteromerization, presents several technical challenges:

Addressing these challenges requires integration of advanced experimental techniques with computational modeling approaches to build a complete picture of GHRL interaction networks in different physiological and pathological contexts.

How might emerging protein language models advance GHRL structure-function research?

Protein language models (pLMs) offer exciting opportunities to advance GHRL research:

  • Advantages of Modern pLMs:

    • Models like AMPLIFY 350M demonstrate that well-designed smaller models can outperform larger models (43× fewer parameters than ESM2 15B)

    • These efficient models achieve 400 to 2,000× higher throughput at inference time, enabling more extensive analyses

  • Applications to GHRL Research:

    • Predicting structural features of GHRL and GHSR1a in different interaction contexts

    • Identifying potential heteromerization interfaces with partners like DRD1

    • Modeling the impact of mutations on GHRL function and interactions

    • Designing modified GHRL variants with enhanced or altered functionality

  • Integration with Experimental Approaches:

    • Using pLM predictions to guide experimental design

    • Validating computational models with targeted experiments

    • Creating iterative workflows where experimental results inform refined models

The careful curation of training data for these models often competes well with scale, suggesting that field-specific models trained on GHRL-related proteins could yield valuable insights even without massive computational resources .

What innovative methodologies might advance understanding of GHRL's role in disease?

Innovative methodologies that could advance understanding of GHRL's role in disease include:

  • Single-cell Transcriptomics and Proteomics:

    • Analyzing GHRL expression at single-cell resolution in healthy and diseased tissues

    • Identifying cell populations with unique GHRL expression or response patterns

    • Mapping cellular trajectories during disease progression

  • Spatial Transcriptomics:

    • Visualizing GHRL expression patterns within tissue architecture

    • Correlating spatial expression with microenvironmental features

    • Identifying regional heterogeneity in GHRL function

  • CRISPR-based Functional Genomics:

    • Systematic perturbation of GHRL pathway components

    • Creation of cell and animal models with precise modifications to GHRL interaction domains

    • Screens to identify novel GHRL regulators and effectors

  • Alternative Protein Research Approaches:

    • Applying emerging techniques from the alternative protein field

    • Engaging faculty with expertise in protein engineering to explore GHRL modifications

    • Building research ecosystems that foster interdisciplinary collaboration

  • Meta-analysis Frameworks:

    • Implementing standardized meta-analysis protocols for GHRL-related transcriptomics data

    • Using random effects models and appropriate statistical methods to integrate diverse datasets

    • Applying rigorous normalization and batch effect correction techniques

These innovative methodologies, particularly when combined, promise to provide more comprehensive understanding of GHRL's role in various diseases and could identify novel therapeutic targets.

Product Science Overview

Introduction

Ghrelin, often referred to as the “hunger hormone,” is a peptide hormone primarily produced by enteroendocrine cells in the gastrointestinal tract, especially the stomach . It plays a crucial role in regulating appetite and energy balance by signaling the brain to induce hunger . Ghrelin levels rise before meals and fall after eating, making it a key player in meal initiation and satiety .

Discovery and Nomenclature

Ghrelin was discovered in 1999 following the identification of its receptor, the growth hormone secretagogue receptor (GHS-R) . The name “ghrelin” is derived from the Proto-Indo-European root “gʰre-” meaning “to grow,” reflecting its role in stimulating growth hormone release .

Gene and Structure

The human ghrelin gene, located on chromosome 3 (3p25-26), spans 7.2 kb and consists of six exons . It is transcribed and translated into a precursor protein called pre-proghrelin, which is then cleaved to produce a 94-amino-acid intermediate known as proghrelin . Further processing yields the active 28-amino-acid ghrelin peptide .

Function and Mechanism

Ghrelin’s primary function is to regulate appetite by stimulating the hypothalamus, the brain region responsible for hunger and energy homeostasis . It binds to the GHS-R1A receptor, activating neuropeptide Y (NPY) and agouti-related peptide (AgRP) neurons, which promote food intake . Additionally, ghrelin increases gastric motility and stimulates the secretion of gastric acid, preparing the body for food intake .

Other Roles

Beyond its role in hunger regulation, ghrelin has several other physiological functions:

  • Growth Hormone Release: Ghrelin stimulates the release of growth hormone from the anterior pituitary gland .
  • Glucose Metabolism: It plays a role in glucose homeostasis by influencing insulin secretion and sensitivity .
  • Cardiovascular Health: Ghrelin has cardioprotective effects, including vasodilation and anti-inflammatory properties .
  • Cognitive Functions: It is involved in learning, memory, and reward-based behaviors .
Clinical Implications

Due to its role in hunger and energy balance, ghrelin is a target for obesity and weight management therapies. Modulating ghrelin levels could potentially help control appetite and reduce food intake . Additionally, its involvement in growth hormone release makes it relevant in conditions related to growth hormone deficiencies .

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.