SST

Somatostatin
Shipped with Ice Packs
In Stock

Description

Definition and Structure

Somatostatin (SST) is a peptide hormone existing in two bioactive isoforms:

  • SST-14: 14-amino-acid form (Ala-Gly-Cys-Lys-Asn-Phe-Phe-Trp-Lys-Thr-Phe-Thr-Ser-Cys\text{Ala-Gly-Cys-Lys-Asn-Phe-Phe-Trp-Lys-Thr-Phe-Thr-Ser-Cys}) .

  • SST-28: Extended 28-amino-acid form derived from prohormone processing .

Both isoforms bind to five G protein-coupled receptor subtypes (SSTR1–5) with nanomolar affinity (Table 1) .

Receptor Subtypes and Signaling Pathways

SSTRs mediate SST’s antisecretory, antiproliferative, and antiangiogenic effects via distinct pathways:

Table 1: SSTR Binding Affinities and Functional Roles

ReceptorKey Ligand Affinity (IC₅₀, nM)Primary Functions
SSTR1SST-14: 0.1–2.26; SST-28: 0.1–2.2Inhibits GH, prolactin, and calcitonin secretion; activates tyrosine phosphatase
SSTR2SST-14: 0.2–1.3; Octreotide: 0.4–2.1Dominates endocrine regulation; suppresses gastrin, histamine, and GH secretion
SSTR3SST-14: 0.3–1.6; Pasireotide: 1.5Induces apoptosis via p53/Bax activation; inhibits tumor angiogenesis
SSTR4SST-14: 0.3–1.8Anti-inflammatory and antinociceptive effects; poorly characterized in tumors
SSTR5SST-28: 0.05–0.4; Pasireotide: 0.16Inhibits insulin, glucagon, and amylase secretion; linked to metabolic regulation

Mechanistically, SSTR activation inhibits adenylyl cyclase, modulates MAPK pathways, and regulates ion channels (e.g., K⁺/Ca²⁺), leading to membrane hyperpolarization and reduced exocytosis .

Gene Family and Evolutionary Conservation

Four SST genes (SST1, SST3, SST5, SST6) were identified in Scatophagus argus (Table 2), revealing conserved SS-14 domains and tissue-specific expression:

  • Hypothalamus: Highest expression across all genes .

  • Gender-specific roles: SST5 is highly expressed in ovaries, while SST3 shows broader tissue activity .

Table 2: SST Gene Characteristics in S. argus

GeneProtein Length (aa)Key Expression SitesFunctional Impact
SST1123Liver, muscleInhibits Igf-1/2 expression
SST3127Hypothalamus, ovariesReduces Igf-1/2 at 3h
SST5106Ovaries (female), hypothalamus (male)Stimulates Igf-1 at 6h
SST6110Stomach, intestineDual regulation of Igf-1/2

Antitumor Effects

  • Neuroendocrine tumors: SST analogs (e.g., octreotide, lanreotide) target SSTR2/5 to inhibit hormone secretion and cell proliferation .

  • Angiogenesis suppression: SSTR3 activation reduces nitric oxide synthase (NOS) and MAPK activity, limiting tumor vascularization .

Novel Agonists for Chronic Pain

  • SSTR4-selective agonists (e.g., Compounds 1–4):

    • Potent G protein activation (EC50\text{EC}_{50}: 16–75 nM) without β-arrestin recruitment .

    • Oral administration (100–500 µg·kg⁻¹) achieves 65–80% anti-hyperalgesia in neuropathy models .

Research Advancements

  • Site-specific templates (SSTs): A computational method for retrosynthetic planning in drug design, enabling precise reaction center labeling .

  • High-temperature polymers: SST Thermal Solutions produces radiation-resistant compounds for aerospace and nuclear industries, though unrelated to the peptide hormone .

Challenges and Future Directions

  • Receptor specificity: Synthetic analogs like pasireotide show partial agonism at SSTR2, necessitating refined targeting .

  • Gene redundancy: Evolutionary conservation of SST genes across vertebrates suggests overlapping roles requiring further study .

Product Specs

Introduction
Somatostatin, also known as growth hormone inhibiting hormone (GHIH) or somatotropin release-inhibiting hormone (SRIF), is a peptide hormone that regulates the endocrine system. It influences neurotransmission and cell proliferation by interacting with G-protein-coupled somatostatin receptors and inhibiting the release of several secondary hormones. Somatostatin exists in two active forms, generated by alternative cleavage of a single preproprotein: one consisting of 14 amino acids and the other of 28 amino acids.
Description
Synthetic Somatostatin is a single, non-glycosylated polypeptide chain composed of 14 amino acids. It has a molecular weight of 1637.9 Daltons and a molecular formula of C76H104N18O19S2. The CAS registry number for this compound is 38916-34-6.
Physical Appearance
Sterile Filtered White Lyophilized Powder
Formulation
The protein has been lyophilized without the addition of any other substances.
Solubility
To reconstitute the lyophilized Somatostatin, it is recommended to dissolve it in sterile 18MΩ-cm H2O at a concentration not lower than 100 µg/ml. This solution can be further diluted with other aqueous solutions as needed.
Stability
Lyophilized Somatostatin (SST) remains stable at room temperature for up to 3 weeks. However, for long-term storage, it is recommended to store it in a desiccated state below -18°C. Once reconstituted, Somatostatin should be stored at 4°C for 2-7 days. For extended storage periods, it is advisable to store it below -18°C. To ensure optimal stability during long-term storage, consider adding a carrier protein like 0.1% HSA or BSA. It's important to minimize freeze-thaw cycles.
Purity
The purity of this product is greater than 98.0%, as determined by the following methods: (a) Analysis by RP-HPLC. (b) Analysis by SDS-PAGE.
Synonyms
Growth hormone release-inhibiting factor, SST, SMS, SMST, GHIH.
Amino Acid Sequence

Ala-Gly-Cys-Lys-Asn-Phe- Phe-Trp-Lys-Thr-Phe-Thr-Ser-Cys-OH.

Q&A

What are somatostatin receptor subtypes and how are they classified in current research?

Somatostatin receptor subtypes (sst) are a family of G protein-coupled receptors that mediate the diverse physiological effects of somatostatin. Research has identified five distinct receptor subtypes, designated sst1 through sst5, each encoded by separate genes. These subtypes belong to the G protein-coupled receptor superfamily and demonstrate unique tissue distribution patterns, binding affinities, and signaling pathways .

The classification of these receptors has evolved through extensive pharmacological and molecular characterization, with each subtype showing distinct functional properties. For example, studies in porcine somatotropes have revealed that sst1 and sst2 primarily mediate inhibitory effects on growth hormone release, while sst5 can actually stimulate GH secretion under specific conditions . This functional diversity highlights the complexity of the somatostatin signaling system and emphasizes the importance of subtype-specific approaches in research.

Methodologically, researchers should approach subtype classification through multiple complementary techniques, including:

  • Receptor binding studies with subtype-selective ligands

  • mRNA quantification using qRT-PCR or in situ hybridization

  • Protein detection through immunohistochemistry with validated antibodies

  • Functional assays measuring distinct signaling outcomes

How does receptor subtype distribution vary across tissue types and species?

The distribution pattern of somatostatin receptor subtypes shows remarkable tissue specificity and species variation, which has significant implications for experimental design and data interpretation. In pituitary tissue, research has demonstrated that somatotrope subpopulations can be separated by density gradient centrifugation into low-density (LD) and high-density (HD) cells with distinct receptor expression profiles . These subpopulations show differential expression of sst subtypes, with sst5 being more abundant in HD somatotropes, while sst1 and sst2 mRNA predominate in LD cells .

This heterogeneous distribution pattern extends across other tissues and varies significantly between species. When designing experiments, researchers must consider these variations, particularly when translating findings between animal models and human applications. Methodologically, tissue-specific expression mapping should incorporate:

  • Comprehensive receptor profiling across multiple tissues

  • Comparison of expression at both mRNA and protein levels

  • Functional correlation with receptor-mediated responses

  • Careful documentation of species-specific distribution patterns

The experimental design should include appropriate controls to account for this heterogeneity, such as parallel testing in multiple species or cell types when feasible.

What are the standard experimental models for studying SST function?

The selection of appropriate experimental models is critical for valid investigations of somatostatin receptor function. Several established models offer distinct advantages depending on the research question:

Experimental ModelAdvantagesLimitationsBest Applications
Primary cell culturesPhysiological receptor levels; native signaling machineryLimited lifespan; heterogeneous populationsStudying endogenous receptor function
Recombinant cell systemsControlled expression; isogenic backgroundPotential artifacts from overexpressionPharmacological characterization
Transgenic animal modelsIn vivo relevance; systemic effectsComplex phenotypes; compensatory mechanismsPhysiological function studies
Ex vivo tissue preparationsPreserved tissue architecture; acute responsesLimited viability; technical challengesIntegrative physiology

For investigating differential subtype expression and function, primary pituitary cell cultures separated by density gradient centrifugation have provided valuable insights into how receptor distribution correlates with functional outcomes . This approach has revealed that low-density and high-density somatotrope populations exhibit distinct responses to subtype-selective agonists, correlating with their different receptor expression patterns .

When designing such experiments, researchers should carefully control for variables that might affect receptor function, including culture conditions, passage number, and the presence of endogenous ligands. The experimental design should also incorporate appropriate validation of the model system, such as characterization of receptor expression levels and functional coupling to downstream signaling pathways.

How should experiments be designed to differentiate the effects of multiple somatostatin receptor subtypes?

Designing experiments to differentiate between the effects of multiple somatostatin receptor subtypes requires sophisticated methodological approaches that account for potential overlapping functions and interactions. Research has demonstrated that different receptor subtypes can exert opposing effects on the same biological process, as seen with sst1/sst2 (inhibitory) versus sst5 (stimulatory) effects on growth hormone release in porcine somatotropes .

A comprehensive experimental design should incorporate:

What methodological approaches are most effective for investigating receptor-ligand interactions?

Investigating receptor-ligand interactions for somatostatin receptor subtypes requires methodology that captures both binding parameters and functional outcomes. The selection of appropriate techniques depends on the specific research question and available resources.

For binding characterization, researchers should consider:

  • Radioligand binding assays: Provide quantitative binding parameters (Kd, Bmax) and can be used for competition studies with unlabeled ligands to determine relative affinities.

  • Functional readouts: Measuring downstream signaling events such as cAMP inhibition, calcium mobilization, or ERK phosphorylation provides information about receptor activation and efficacy.

  • Real-time kinetic measurements: Techniques such as bioluminescence resonance energy transfer (BRET) allow monitoring of receptor-ligand interactions and conformational changes in living cells.

When conducting these studies, researchers must consider potential confounding factors:

  • Receptor expression levels: Variations in expression can significantly impact measured responses

  • G protein availability: Different cell types may have varying levels of G protein subtypes

  • Signal amplification: Downstream pathways may show different degrees of amplification

Data from receptor-ligand studies should be analyzed using appropriate mathematical models, such as the law of mass action for binding data or operational models for functional responses that account for receptor reserve.

Research has shown that different somatostatin receptor subtypes exhibit distinct pharmacological profiles. For example, studies with porcine somatotropes demonstrated that sst5 selective agonists stimulate GH release at specific dose ranges, while sst1 and sst2 agonists show inhibitory effects at different concentration thresholds .

How can researchers effectively control variables in SST experiments?

Controlling variables is critical for valid scientific inquiry in somatostatin receptor research. Experimental design literature emphasizes that any variable that might affect the outcome, other than the treatment being tested, must be controlled to establish causality . For somatostatin receptor studies, this presents particular challenges due to the complexity of receptor signaling and the heterogeneity of experimental systems.

Key variables requiring control include:

  • Receptor expression levels: Quantify and standardize expression across experimental groups using techniques such as qRT-PCR, flow cytometry, or radioligand binding.

  • Cell culture conditions: Standardize media composition, serum concentration, cell density, and passage number, as these factors can influence receptor expression and signaling.

  • Treatment parameters: Carefully control concentration, timing, and duration of agonist/antagonist applications. Research has shown that responses to sst-specific agonists vary based on dose and cell type .

  • Environmental factors: Control temperature, pH, and CO2 levels, which can affect receptor conformation and function.

  • Experimental timing: Standardize time points for measurements, as receptor desensitization and trafficking can introduce time-dependent variations.

The experimental design should incorporate randomization and appropriate control groups. As described in experimental design literature, this includes a control group receiving no treatment and experimental groups receiving different treatments, with all other variables held constant . When comparing multiple treatments, such as different receptor subtype agonists, each experimental group should differ from others only in the specific treatment being tested .

How should researchers interpret contradictory findings in SST subtype function across different experimental systems?

Contradictory findings are common in somatostatin receptor research, often resulting from methodological differences or biological complexity. Researchers should approach such contradictions systematically:

  • Examine methodological differences: Experimental design principles emphasize that variations in protocols, reagents, or analysis methods can significantly impact outcomes . Document and compare all methodological details between studies showing contradictory results.

  • Consider biological variables: Different cell types or tissues may exhibit distinct receptor coupling preferences or downstream signaling machinery. For example, research has shown that low-density and high-density somatotropes respond differently to the same sst agonists .

  • Evaluate receptor expression patterns: Contradictions may arise from differences in relative expression levels of receptor subtypes. Studies have demonstrated that sst subtype expression correlates with functional outcomes, with sst5 being more abundant in HD somatotropes while sst1 and sst2 mRNA predominate in LD cells .

  • Assess species differences: Significant species variations exist in receptor pharmacology and signaling. When comparing results across studies, carefully consider the species source of the experimental system.

  • Design bridging experiments: When facing contradictory findings, design experiments specifically aimed at identifying the source of discrepancy. This might involve side-by-side comparison of different protocols or parallel testing in multiple systems.

  • Consider receptor interactions: Somatostatin receptors can form homo- and heterodimers with altered signaling properties, potentially explaining seemingly contradictory results.

  • Apply statistical rigor: Ensure adequate statistical power and appropriate statistical tests for comparing results across studies, following experimental design best practices .

What statistical approaches are most appropriate for analyzing dose-dependent responses to SST agonists?

Analyzing dose-dependent responses to somatostatin receptor agonists requires statistical approaches that capture the complexity of receptor pharmacology. Studies have shown that somatostatin receptor subtypes demonstrate distinct dose-response relationships, with some exhibiting stimulatory effects at specific concentration ranges .

Appropriate statistical approaches include:

  • Nonlinear regression for dose-response analysis: Standard sigmoidal models (four-parameter logistic equation) provide parameters such as EC50, maximum effect, and Hill coefficient. For complex responses, consider:

    • Biphasic models for bell-shaped dose-response curves

    • Models incorporating receptor reserve concepts

    • Operational models accounting for efficacy and affinity

  • ANOVA with appropriate post-hoc tests: For comparing responses across multiple doses, receptor subtypes, or experimental conditions. When designing such experiments, consider:

    • Factorial designs to assess interactions between variables

    • Repeated measures approaches for within-subject comparisons

    • Appropriate multiple comparison corrections

  • Statistical power considerations: Ensure adequate sample size through a priori power analysis, particularly important when expecting subtle differences between receptor subtypes.

  • Statistical validation of experimental controls: Proper experimental design requires statistical comparison between treatment and control groups to establish that observed effects are due to the treatment variable and not confounding factors .

Statistical ApproachBest ApplicationAdvantagesConsiderations
Four-parameter logisticSimple monotonic dose-responsesProvides standard pharmacological parametersMay not fit complex responses
Biphasic modelsBell-shaped responsesCaptures dual effects at different concentrationsRequires more data points
Two-way ANOVAComparing multiple subtypes across dosesTests for interaction effectsRequires assumptions of normality
Mixed-effects modelsRepeated measures with missing dataAccounts for within-subject correlationMore complex interpretation

How can researchers effectively analyze receptor subtype expression data across different experimental conditions?

Analyzing somatostatin receptor subtype expression data presents unique challenges due to the potential for co-expression of multiple subtypes and regulation of expression under different conditions. Research has shown that receptor subtype expression patterns correlate with functional responses, highlighting the importance of accurate expression analysis .

Effective analytical approaches include:

  • Normalization strategies: For qRT-PCR data, carefully select stable reference genes validated for the specific experimental conditions. Consider:

    • Multiple reference genes rather than a single housekeeping gene

    • Geometric averaging of multiple reference genes

    • Validation of reference gene stability across experimental conditions

  • Relative vs. absolute quantification: Consider absolute quantification when comparing expression levels between different receptor subtypes, as PCR efficiency may vary between primer sets.

  • Correlation analysis: Examine correlations between receptor expression and functional outcomes. Studies have demonstrated correlation between receptor distribution patterns and functional responses to subtype-selective agonists .

  • Multivariate analysis: When measuring multiple receptor subtypes across different conditions, consider principal component analysis (PCA) or cluster analysis to identify patterns.

  • Visualization techniques: Effectively communicate complex expression patterns through:

    • Heat maps showing expression across subtypes and conditions

    • Radar plots for comparative expression profiles

    • Correlation networks linking expression with functional outcomes

When interpreting expression data, researchers should consider:

  • Post-transcriptional regulation affecting the relationship between mRNA and protein levels

  • Subcellular localization of receptors, which may affect functionality

  • Potential for receptor heteromerization altering signaling properties

What are the methodological challenges in studying SST receptor trafficking and internalization?

Studying somatostatin receptor trafficking and internalization presents several methodological challenges that researchers must address through careful experimental design. The dynamic nature of receptor movement between cellular compartments requires specialized techniques and controls.

Key methodological considerations include:

  • Temporal resolution: Receptor trafficking occurs on timescales ranging from seconds to hours. Experimental designs must incorporate appropriate time points to capture:

    • Rapid internalization events (minutes)

    • Recycling to the plasma membrane (30-60 minutes)

    • Downregulation through degradation (hours)

  • Spatial resolution: Distinguishing between membrane-bound and internalized receptors requires techniques with sufficient spatial discrimination. Options include:

    • Confocal microscopy for visual confirmation of localization

    • Biochemical fractionation to separate membrane and intracellular compartments

    • Flow cytometry with antibodies recognizing extracellular epitopes

  • Receptor tagging strategies: While tags facilitate detection, they may interfere with trafficking. Consider:

    • Small epitope tags versus fluorescent protein fusions

    • C-terminal versus N-terminal tagging based on receptor topology

    • Validation that tagged receptors retain normal trafficking properties

  • Quantification methods: Converting visual observations to quantitative data requires rigorous approaches:

    • Automated image analysis with defined parameters

    • Ratiometric measurements of surface/total receptor

    • Internal standards for normalization across experiments

  • Experimental controls: Essential controls include:

    • Positive controls using receptors with well-characterized trafficking

    • Validation with multiple complementary techniques

    • Pharmacological tools to block specific trafficking steps

The experimental design should also consider the potential impact of receptor density on trafficking kinetics, as overexpression systems may not accurately reflect physiological trafficking patterns. Research has shown that different somatostatin receptor subtypes exhibit distinct trafficking properties, which may contribute to their differential signaling characteristics .

How can researchers effectively investigate signaling pathway selectivity among SST subtypes?

Investigating signaling pathway selectivity among somatostatin receptor subtypes requires sophisticated experimental approaches that can distinguish between different downstream cascades. Research has demonstrated that receptor subtypes can couple preferentially to distinct signaling pathways, contributing to their diverse physiological effects .

Effective methodological approaches include:

  • Pathway-specific readouts: Measure multiple signaling outputs simultaneously to capture the full signaling profile:

    • cAMP levels for Gi/o-mediated pathways

    • Calcium mobilization for Gq-coupled responses

    • ERK1/2 phosphorylation for MAPK pathway activation

    • β-arrestin recruitment for G protein-independent signaling

  • Temporal profiling: Different pathways may show distinct activation kinetics. Design time-course experiments with appropriate resolution to capture:

    • Rapid and transient responses (seconds to minutes)

    • Sustained signaling events (minutes to hours)

    • Adaptive responses and feedback regulation

  • Pharmacological dissection: Use pathway-specific inhibitors to delineate signaling cascade components:

    • G protein subtype-specific inhibitors (e.g., pertussis toxin for Gi/o)

    • Kinase inhibitors for specific branches of signaling networks

    • Scaffolding protein disruptors to target specific signaling complexes

  • Genetic manipulation: Employ RNA interference or CRISPR-based approaches to selectively modulate signaling components:

    • Knockdown/knockout of specific G protein subunits

    • Modification of scaffold proteins or adaptor molecules

    • Expression of dominant-negative signaling mediators

  • Biased agonism assessment: Design experiments to detect ligand bias:

    • Calculation of bias factors using operational models

    • Comparison of concentration-response curves across different pathways

    • Assessment of pathway-specific kinetics and desensitization

When analyzing pathway selectivity data, researchers should consider:

  • Potential for pathway crosstalk influencing observed responses

  • Cell type-specific variations in signaling machinery

  • Effects of receptor expression levels on coupling efficiency

Research has shown that somatostatin receptor subtypes exhibit distinct signaling profiles that correlate with their physiological effects. For example, in pig somatotropes, sst1 and sst2 primarily mediate inhibitory effects on growth hormone release, while sst5 can stimulate GH secretion under specific conditions , highlighting the importance of subtype-specific signaling.

What are the best practices for developing and validating subtype-selective pharmacological tools?

Developing and validating subtype-selective pharmacological tools for somatostatin receptor research requires rigorous methodology to ensure specificity and reliability. These tools are essential for dissecting the roles of individual receptor subtypes in complex biological systems.

Best practices include:

  • Comprehensive selectivity profiling:

    • Test compounds against all five somatostatin receptor subtypes

    • Evaluate binding affinity through competition binding assays

    • Assess functional activity across multiple signaling pathways

    • Determine selectivity ratios (affinity for target subtype / affinity for other subtypes)

  • Validation across multiple assay systems:

    • Confirm selectivity in different cell backgrounds

    • Test in recombinant systems and native tissues

    • Verify activity in different species if relevant

    • Use complementary techniques (binding, signaling, trafficking)

  • Structure-activity relationship studies:

    • Systematically modify chemical structure to enhance selectivity

    • Correlate structural features with binding and functional data

    • Use computational modeling to guide rational design

    • Develop analogs with varying efficacy profiles (full agonists, partial agonists, antagonists)

  • In vivo validation:

    • Assess pharmacokinetic properties

    • Confirm target engagement in relevant tissues

    • Evaluate off-target effects

    • Correlate in vitro selectivity with in vivo effects

  • Control experiments:

    • Use receptor knockout/knockdown systems as negative controls

    • Include non-selective reference compounds

    • Test against closely related GPCRs outside the somatostatin family

    • Validate tools under the specific experimental conditions where they will be used

Research has demonstrated the utility of subtype-selective agonists in dissecting the roles of different somatostatin receptor subtypes. For example, studies with porcine somatotropes showed that an sst5 selective agonist stimulated spontaneous GH secretion at doses ranging from 10^-13 to 10^-9 M, whereas sst1-, sst2-, sst3-, and sst4-specific agonists inhibited GHRH-evoked GH release . These selective tools enabled researchers to identify the distinct roles of sst5 versus sst1/sst2 in regulating growth hormone secretion.

Integrating findings across diverse experimental approaches in SST research

The field of somatostatin receptor subtype research benefits from the integration of diverse experimental approaches to build a comprehensive understanding of receptor function. By synthesizing findings from molecular, cellular, and physiological studies, researchers can develop more robust models of somatostatin signaling and its biological implications.

Key considerations for integrative approaches include:

The complexity of the somatostatin receptor system, with its five subtypes exhibiting distinct and sometimes opposing functions, necessitates such integrative approaches. Research has demonstrated that in pigs, sst1 and sst2 primarily mediate inhibitory effects of somatostatin, whereas sst5 mediates stimulatory actions on growth hormone release . Understanding such nuanced functional differences requires multiple complementary experimental strategies, ranging from receptor binding studies to signaling pathway analysis and physiological readouts.

Product Science Overview

Structure and Forms

Somatostatin exists in two active forms, produced by the alternative cleavage of a single preproprotein:

  1. Somatostatin-14: Consists of 14 amino acids.
  2. Somatostatin-28: Consists of 28 amino acids .
Production and Distribution

Somatostatin is produced by various tissues in the body, primarily in the nervous and digestive systems. Key production sites include:

  • Hypothalamus: Regulates the release of growth hormone from the pituitary gland.
  • Pancreas: Inhibits the secretion of insulin and glucagon.
  • Gastrointestinal Tract: Regulates the activity of the gastrointestinal system .
Functions

The primary function of somatostatin is to inhibit the release of other hormones and regulate various physiological processes. Some of its key functions include:

  • Inhibition of Hormone Secretion: Somatostatin inhibits the secretion of several hormones, including growth hormone, thyroid-stimulating hormone (TSH), and insulin .
  • Regulation of Gastrointestinal Activity: It slows down the digestive process by inhibiting the secretion of gastric acid and digestive enzymes .
  • Cell Proliferation: Somatostatin prevents the rapid reproduction of normal and tumor cells .
Receptors

Somatostatin exerts its effects through interaction with G protein-coupled somatostatin receptors. There are five known somatostatin receptors (SSTR1 to SSTR5), each with distinct tissue distribution and functional roles .

Clinical Significance

Somatostatin and its synthetic analogs are used in the treatment of various medical conditions, including:

  • Acromegaly: A condition characterized by excessive growth hormone production.
  • Neuroendocrine Tumors: Tumors that arise from hormone-producing cells.
  • Gastrointestinal Disorders: Conditions such as peptic ulcers and diarrhea .
Evolutionary Perspective

Among vertebrates, there are six different somatostatin genes (SS1 to SS6), allowing for a wide range of functions. Humans, however, have only one somatostatin gene (SST) .

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.