RGS2 Human

Regulator of G-Protein Signaling 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RGS2 Human

RGS2 (Regulator of G protein signaling 2) is a critical modulator of G protein-coupled receptor (GPCR) signaling pathways, primarily functioning as a GTPase-activating protein (GAP) to terminate heterotrimeric G protein signaling. It belongs to the B/R4 subfamily of RGS proteins and is widely expressed in cardiovascular, neuronal, and immune systems .

Molecular Structure

RGS2 is a 211-amino acid protein (molecular mass: 26.5 kDa) containing:

  • RGS domain: 120-amino acid region critical for GAP activity toward Gαq/11 subunits.

  • N-terminal domain: An amphipathic α-helix and hydrophobic motifs enabling plasma membrane targeting.

  • C-terminal tail: Short region involved in protein interactions .

FeatureDescriptionSource
Chromosomal locationChromosome 1 (1q31.2)
Key binding partnersGαq, Gαi/o, GNAQ, GNAI3, PRKG1, ADCY3, PPP1R9B
GAP selectivityPrimary: Gαq; Secondary: Gαi/o (context-dependent)

Functional Mechanisms

RGS2 regulates signaling by:

  1. Accelerating GTP hydrolysis on Gα subunits, driving them to inactive GDP-bound states .

  2. Directly inhibiting adenylyl cyclase (e.g., ADCY3) to suppress cAMP production .

  3. Modulating synaptic plasticity via interactions with β-arrestin and G protein receptor kinases .

Key Pathways Affected:

  • Gαq/11 signaling: Regulates vasoconstriction, blood pressure, and endothelial function .

  • Serotonergic signaling: Attenuates 5-HT2AR-mediated phospholipase C activation .

  • cGMP signaling: Interacts with PRKG1/2 to influence smooth muscle relaxation .

Cardiovascular and Metabolic Disorders

ConditionMechanismEvidence
HypertensionReduced RGS2 expression enhances Gαq signaling, increasing vascular toneRGS2-deficient mice exhibit elevated blood pressure .
PreeclampsiaHypoxia-induced suppression of RGS2 via HIF-1α/P300 complexAltered placental G protein signaling linked to disease pathogenesis .
ObesityRGS2 knockout mice show increased adipose tissue mass and insulin resistanceImpaired Gαq/11 signaling in adipocytes .

Neuropsychiatric Disorders

ConditionMechanismEvidence
Suicide riskPolymorphisms in RGS2 (e.g., SNP3) correlate with altered 5-HT2AR signaling and amygdala functionRGS2 immunoreactivity is elevated in suicide victims’ amygdala .
AnxietyRGS2 knockout mice exhibit reduced anxiety-like behaviorsGenetic studies link RGS2 variants to panic disorders and PTSD .
Cocaine addictionRGS2 regulates dopamine D2 receptor signaling in ventral tegmental areaRGS2 knockdown enhances D2R-mediated G protein activity .

Key Studies

Study FocusFindingsReference
Gαq Structural InteractionRGS2 binds Gαq’s α-helical domain, enhancing GAP activity and selectivity
Bronchoprotectionβ2-adrenoceptor agonists + glucocorticoids synergistically induce RGS2 in airway smooth muscle, reducing bronchoconstriction
Prostate CancerRGS2 downregulation promotes epithelial-to-mesenchymal transition (EMT) via TWIST1 upregulation and E-cadherin loss
Cancer DormancyHigh RGS2 expression correlates with quiescent non-small cell lung cancer (NSCLC) cells and poor prognosis

Product Specs

Introduction
RGS2, a member of the RGS protein family, functions as a GTPase-activating protein for G alpha subunits within heterotrimeric G proteins. It regulates signal transduction by accelerating the GTPase activity of G protein alpha subunits, converting them into their inactive GDP-bound state.
Description
Produced in E. coli, RGS2 is a single, non-glycosylated polypeptide chain with 231 amino acids (1-211a.a.) and a molecular weight of 26.5 kDa. This protein consists of amino acids 1-211 and includes a 20 amino acid His-tag fused at the N-terminus. Purification is achieved through proprietary chromatographic techniques.
Physical Appearance
Sterile filtered colorless solution.
Formulation
The RGS2 protein solution is supplied at a concentration of 0.25mg/ml in a buffer composed of 20mM Tris-HCl (pH 8.0), 100mM NaCl, 2mM DTT, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of RGS2 is greater than 90.0% as determined by SDS-PAGE analysis.
Synonyms
Regulator of G-protein Signaling 2 24kDa, G0S8, G0/G1 switch regulatory protein 8, Cell growth-inhibiting gene 31 protein, RGS2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MQSAMFLAVQ HDCRPMDKSA GSGHKSEEKR EKMKRTLLKD WKTRLSYFLQ NSSTPGKPKT GKKSKQQAFI KPSPEEAQLW SEAFDELLAS KYGLAAFRAF LKSEFCEENI EFWLACEDFK KTKSPQKLSS KARKIYTDFI EKEAPKEINI DFQTKTLIAQ NIQEATSGCF TTAQKRVYSL MENNSYPRFL ESEFYQDLCK KPQITTEPHA T

Q&A

What is the RGS2 gene and what is its primary function in humans?

RGS2 (regulator of G-protein signaling 2) is a member of a family of proteins that negatively modulate G-protein coupled receptor transmission . The gene encodes a protein that plays a crucial regulatory role in G-protein signaling pathways, which are fundamental to numerous cellular processes. In the central nervous system, RGS2 is particularly important for modulating neurotransmission related to anxiety and stress responses. The protein functions by accelerating the deactivation of G-proteins, effectively serving as a brake on specific signaling cascades.

Methodologically, researchers typically study RGS2 function through:

  • Gene knockout models in mice

  • Expression analysis in different brain regions

  • Protein interaction studies using co-immunoprecipitation

  • Functional assays measuring G-protein activity

Which polymorphisms in the RGS2 gene are most frequently studied in human anxiety research?

The single nucleotide polymorphism (SNP) rs4606 is among the most frequently studied variations in the RGS2 gene in relation to anxiety disorders and PTSD. This SNP has been consistently associated with anxiety-related phenotypes across multiple studies . Research has shown that rs4606 is of particular interest because it is associated with variation in RGS2 mRNA expression levels .

Other notable polymorphisms include:

  • rs3767488, which has been studied in combination with rs4606 in panic disorder research

  • Several polymorphisms identified by Leygraf and colleagues (2006) showing association with panic disorder

  • Variants examined by Smoller et al. (2008) in relation to anxiety-related temperament and brain function

When designing genetic association studies for RGS2, researchers should consider:

  • Population stratification issues

  • Hardy-Weinberg equilibrium testing (as was done in studies reporting genotype frequencies of GG: 7.7%, CG: 36.4%, and CC: 55.8%)

  • Haplotype analysis approaches rather than single SNP examination

  • Gene-environment interaction models

How does RGS2 gene expression correlate with anxiety and depression phenotypes?

Research demonstrates a clear inverse relationship between RGS2 expression levels and anxiety/depression phenotypes. Lower RGS2 expression correlates with increased anxiety-like behaviors and depressive features in animal models . This relationship has been demonstrated through multiple behavioral paradigms:

Behavioral TestFinding in Low RGS2 ExpressionSignificance
Elevated Plus MazeReduced time in open armsp<0.0001
Forced Swim TestIncreased immobilityp<0.03
Novelty Suppressed FeedingIncreased latency to feedSignificant
Social Novelty TestReduced interaction with new strangerp<0.0004

Methodologically, researchers examine this correlation through:

  • Quantitative PCR to measure expression levels in specific brain regions

  • Behavioral testing batteries in animals with genetic modifications altering RGS2 expression

  • Correlation analyses between expression levels and behavioral outcomes

  • Examination of related neurotransmitter systems (particularly serotonergic)

How does RGS2 moderate the relationship between environmental stressors and PTSD symptom development?

RGS2 demonstrates a classic gene-environment interaction pattern in relation to PTSD development. The rs4606 polymorphism in RGS2 has been shown to modify risk of post-disaster PTSD symptoms under conditions of high hurricane exposure and low social support . Similar interactions were observed for lifetime PTSD symptoms under conditions of lifetime exposure to potentially traumatic events with low social support.

The interaction follows a specific pattern:

  • Under low stress/high support conditions: minimal effect of RGS2 genotype

  • Under high stress/low support conditions: significant effect of genotype on PTSD symptoms

This interaction remained significant after adjustment for sex, ancestry, and age, demonstrating the robustness of the finding .

Methodologically, researchers investigating these interactions should:

  • Use validated measures of both stressor exposure and social support

  • Employ statistical approaches designed for gene-environment interaction testing

  • Consider multiple environmental factors simultaneously

  • Control for relevant demographic and clinical covariates

  • Use longitudinal designs when possible to establish temporal relationships

What is the relationship between RGS2 and serotonergic receptor expression in anxiety disorders?

Research indicates a significant relationship between RGS2 expression levels and serotonergic receptor expression, particularly 5-HT1A and 5-HT1B receptors in the raphe nucleus. Animals with reduced RGS2 expression (both heterozygous and homozygous for expression-reducing mutations) show significantly lower 5-HT1A and 5-HT1B receptor expression in the raphe nucleus compared to wild-type animals .

The functional implication is a stronger serotonergic inhibitory tone in animals with reduced RGS2 expression, resulting from:

  • Absence of RGS2 post-receptor shutdown of raphe inhibitory 5-HT1A/B receptor signaling

  • Compensatory down-regulation of inhibitory 5-HT1A and 5-HT1B receptors

  • Altered serotonergic transmission contributing to anxiety/depression phenotypes

This relationship has been demonstrated through:

  • Gene expression studies in specific brain regions (cortex, raphe nucleus, hypothalamus)

  • 8-OH-DPAT-induced hypothermia assays showing altered response in RGS2-reduced animals

  • Correlation of these molecular changes with behavioral phenotypes

Researchers investigating this relationship should employ:

  • Region-specific brain expression analysis

  • Functional assays of receptor activity

  • Pharmacological challenges to assess receptor sensitivity

  • Integration of molecular and behavioral data

How do different experimental approaches to modifying RGS2 expression compare in modeling anxiety disorders?

Multiple approaches to modifying RGS2 expression have been employed in research, each with distinct advantages for modeling different aspects of anxiety disorders:

ApproachAdvantagesLimitationsKey Findings
Complete knockoutComplete elimination of gene functionMay have developmental compensationsIncreased anxiety, hypertension, and neuronal excitability
Heterozygous mutationModels partial reduction in expressionVariable expressivityIntermediate anxiety phenotype
Expression-reducing mutationsMore subtle modulationRequires careful quantificationDose-dependent relationship with anxiety behaviors
Pharmacological modulationTemporal controlLess specificityPotential for therapeutic development

Research indicates that even partial reduction in RGS2 expression (heterozygous condition) can produce significant anxiety-like behaviors, while homozygous reduction may be required for more pronounced depression-like phenotypes . This suggests different thresholds of RGS2 reduction for anxiety versus depression manifestations.

Methodologically, researchers should consider:

  • Carefully quantifying expression levels in their model systems

  • Employing multiple behavioral paradigms to assess both anxiety and depression phenotypes

  • Comparing results across different model systems

  • Controlling for potential developmental compensations in genetic models

What are the molecular mechanisms through which RGS2 variants influence neural circuit function in anxiety?

RGS2 variants influence neural circuit function through several interconnected molecular mechanisms:

  • Altered G-protein signaling kinetics: RGS2 accelerates the hydrolysis of GTP to GDP on Gα subunits, thereby limiting the duration of G-protein signaling. Variants affecting this function can lead to prolonged signaling of various neurotransmitter receptors.

  • Modulation of neuronal excitability: RGS2 knockout mice show increased excitability in CA1 neurons in the hippocampus , suggesting a role in regulating neuronal firing patterns crucial for anxiety-related behaviors.

  • Serotonergic system interactions: Reduced RGS2 expression leads to altered expression of 5-HT1A and 5-HT1B receptors in the raphe nucleus , affecting serotonergic tone throughout connected brain regions.

  • Stress response pathway regulation: RGS2 appears to modulate the relationship between environmental stressors and neurobiological responses, particularly under conditions of high stress and low social support .

Researchers investigating these mechanisms should employ:

  • Electrophysiological recordings to assess neuronal excitability

  • Calcium imaging to examine signaling dynamics

  • Circuit-specific manipulations using optogenetics or chemogenetics

  • In vivo recordings during anxiety-provoking behavioral paradigms

  • Molecular studies of downstream signaling cascades

What are the most effective behavioral paradigms for assessing RGS2-related anxiety phenotypes?

Research indicates that multiple behavioral paradigms can effectively capture RGS2-related anxiety phenotypes, with varying sensitivity to different aspects of anxiety-like behavior:

Behavioral TestWhat It MeasuresSensitivity to RGS2 Variation
Elevated Plus MazeApproach-avoidance conflictHigh - shows dose-dependent relationship
Novelty Suppressed FeedingHyponeophagia/conflict behaviorHigh - particularly for homozygous RGS2 reduction
Forced Swim TestBehavioral despairModerate - more sensitive in homozygous condition
Three-chamber Social TestSocial anxiety/novelty seekingHigh - particularly for social novelty phase
Light-Dark BoxApproach-avoidance conflictHigh - demonstrated in previous RGS2 studies

When designing behavioral testing protocols, researchers should:

  • Include multiple tests that capture different aspects of anxiety

  • Consider the order of testing (from least to most stressful)

  • Control for time of day and other environmental factors

  • Include appropriate control groups (wild-type littermates ideal)

  • Examine both anxiety and depression-related behaviors given their relationship with RGS2

How should researchers approach gene-environment interaction studies involving RGS2?

Gene-environment interaction studies for RGS2 require careful methodological considerations:

  • Environmental measurement:

    • Use validated, quantitative measures of stressor exposure

    • Consider both acute and chronic stressors

    • Measure potential buffering factors (e.g., social support)

    • Collect timing information to establish temporal relationships

  • Genetic analysis:

    • Examine multiple polymorphisms or haplotypes rather than single SNPs

    • Consider pathways analysis including other genes in related systems

    • Verify Hardy-Weinberg equilibrium (as done in studies showing GG: 7.7%, CG: 36.4%, CC: 55.8%)

    • Control for population stratification

  • Statistical approaches:

    • Use models specifically designed for gene-environment interactions

    • Consider multiple testing corrections for examining various stressors

    • Include relevant covariates (sex, age, ancestry, etc.)

    • Consider curvilinear relationships between variables

  • Measurement of outcomes:

    • Use validated, continuous measures of anxiety or PTSD symptoms

    • Consider multiple informants when possible

    • Examine potential mediators of the gene-environment relationship

The Florida Hurricanes study provides an exemplar design, examining both hurricane exposure and social support as environmental factors interacting with rs4606 genotype .

What are the recommended approaches for measuring RGS2 expression in human studies?

Measuring RGS2 expression in human studies presents unique challenges compared to animal models. Recommended approaches include:

  • Tissue-specific considerations:

    • Post-mortem brain tissue analysis for region-specific expression

    • Peripheral blood mononuclear cells (PBMCs) as a more accessible proxy

    • Induced pluripotent stem cells (iPSCs) differentiated into neurons

    • Fibroblasts or lymphoblastoid cell lines from subjects

  • Expression analysis methods:

    • Quantitative RT-PCR for mRNA quantification

    • RNA sequencing for comprehensive transcriptomic analysis

    • Protein quantification via Western blot or ELISA

    • In situ hybridization in post-mortem tissue

  • Genetic influences on expression:

    • Expression quantitative trait loci (eQTL) analysis

    • Analysis of rs4606 and other variants known to affect RGS2 expression

    • Consideration of epigenetic modifications (methylation, histone modifications)

  • Clinical correlations:

    • Relating expression levels to anxiety/depression symptom scales

    • Examining expression changes in response to stress challenges

    • Longitudinal assessment where possible

When designing human expression studies, researchers should account for factors known to influence gene expression, including medication status, time of sampling, recent stress exposure, and demographic variables.

How do findings on RGS2 from animal models translate to human anxiety disorders?

The translation of RGS2 findings from animal models to human anxiety disorders shows remarkable consistency across several domains:

  • Genetic associations:

    • Animal models: RGS2 knockout or reduced expression leads to increased anxiety behaviors

    • Human studies: RGS2 polymorphisms (particularly rs4606) associated with anxiety disorders, PTSD, and panic disorder

  • Environmental interactions:

    • Animal models: RGS2 effects more pronounced under stress conditions

    • Human studies: RGS2 variants moderate the relationship between stress exposure and PTSD symptoms

  • Neurobiological mechanisms:

    • Animal models: Altered serotonergic receptor expression in raphe nucleus

    • Human studies: Similar serotonergic system involvement inferred from imaging and pharmacological studies

  • Response to interventions:

    • Animal models: Potential for RGS2 modulators as novel anxiolytics

    • Human studies: Pharmacogenetic studies beginning to examine RGS2 variation and treatment response

Researchers should consider:

  • Cross-species validation of findings

  • Focusing on conserved aspects of RGS2 function

  • Using translational approaches in study design

  • Incorporating dimensional measures of anxiety symptoms rather than categorical diagnoses

What are the potential therapeutic implications of targeting RGS2 signaling in anxiety disorders?

Research on RGS2 suggests several promising therapeutic directions:

  • Direct RGS2 modulators:

    • Small molecules that increase RGS2 expression or activity could have anxiolytic effects

    • Compounds enhancing RGS2's GTPase-accelerating activity might reduce excessive G-protein signaling

    • Target-specific delivery to anxiety-relevant brain circuits could minimize side effects

  • Serotonergic system interactions:

    • RGS2-based therapies might complement or enhance the efficacy of SSRIs

    • Combined targeting of RGS2 and 5-HT1A/B receptors could provide synergistic effects

    • Understanding RGS2's role in serotonergic signaling could help address SSRI resistance

  • Personalized medicine approaches:

    • Genetic testing for RGS2 variants might predict treatment response

    • RGS2 genotype could inform environmental manipulation recommendations (e.g., stress reduction, social support enhancement)

    • Expression analysis might serve as a biomarker for treatment monitoring

  • Resilience enhancement:

    • Understanding RGS2's role in stress response might inform preventive interventions

    • Particularly relevant for high-risk populations (e.g., military, first responders)

As noted by researchers, "From a pharmacological standpoint, RGS2 modulators may afford innovative pharmacological avenues for treatment of anxiety disorders" . These approaches represent a potential advance beyond current anxiolytics by targeting upstream signaling mechanisms.

What emerging technologies might advance our understanding of RGS2 function in anxiety disorders?

Several cutting-edge technologies hold promise for deepening our understanding of RGS2's role in anxiety:

  • CRISPR-Cas9 gene editing:

    • Creation of precise human cellular models with specific RGS2 variants

    • Development of novel animal models with human-specific variants

    • Temporal and region-specific manipulation of RGS2 expression

  • Single-cell transcriptomics:

    • Cell-type specific analysis of RGS2 expression in anxiety-relevant circuits

    • Identification of co-expression networks and novel interaction partners

    • Examination of cell-specific responses to stress

  • Optogenetics and chemogenetics:

    • Circuit-specific manipulation of RGS2-expressing neurons

    • Real-time correlation of RGS2 activity with anxiety-related behaviors

    • Testing causal relationships between RGS2 signaling and anxiety states

  • Advanced neuroimaging:

    • Functional connectivity in humans with different RGS2 genotypes

    • PET imaging with novel ligands to examine downstream effects of RGS2 variation

    • Combined genetic-imaging approaches to map brain-wide effects of RGS2 variants

These technologies will allow researchers to move beyond correlational findings to establish causal mechanisms and circuit-specific effects of RGS2 in anxiety disorders.

How might integrating RGS2 research with other anxiety-related genes advance understanding of anxiety disorders?

Integrating RGS2 research with other anxiety-related genes presents opportunities for a systems-level understanding of anxiety disorders:

  • Pathway analysis approaches:

    • Examining interactions between RGS2 and other G-protein signaling components

    • Integrating RGS2 with serotonergic system genes (based on findings showing altered 5-HT1A/B expression)

    • Constructing regulatory networks involving multiple anxiety-related genes

  • Polygenic risk approaches:

    • Including RGS2 variants in polygenic risk scores for anxiety disorders

    • Examining how RGS2 risk combines with other genetic risks

    • Investigating potential epistatic interactions between RGS2 and other genes

  • Cross-disorder analyses:

    • Understanding shared genetic architecture across anxiety, depression, and PTSD

    • Examining RGS2's role in comorbidity patterns

    • Identifying transdiagnostic mechanisms involving RGS2

  • Developmental perspectives:

    • Investigating how RGS2 interacts with genes involved in neurodevelopment

    • Examining changing patterns of interaction across the lifespan

    • Identifying sensitive periods for gene-environment interactions

This integrated approach recognizes that "PTSD is a complex phenotype, and that most likely there are many genetic and environmental factors that confer risk for the disorder" , with RGS2 representing one important piece of a larger puzzle.

Product Science Overview

Introduction

The Regulator of G-Protein Signaling 2 (RGS2) is a protein encoded by the RGS2 gene in humans. It is a member of the Regulator of G-Protein Signaling (RGS) family, which plays a crucial role in modulating the signaling pathways mediated by G-protein-coupled receptors (GPCRs). RGS2 acts as a GTPase-activating protein (GAP) for G alpha subunits of heterotrimeric G proteins, thereby accelerating the deactivation of G proteins and terminating the signal transduction .

Structure and Function

RGS2 is composed of 211 amino acids and has a molecular weight of approximately 24 kDa. The protein contains a conserved RGS domain, which is responsible for its GAP activity. This domain interacts with the G alpha subunits, enhancing their intrinsic GTPase activity and promoting the hydrolysis of GTP to GDP, thus inactivating the G protein .

RGS2 is involved in various physiological processes, including the regulation of blood pressure, vascular tone, and cardiac function. It plays a significant role in the negative regulation of the angiotensin-activated signaling pathway and the modulation of vascular smooth muscle contraction and relaxation .

Clinical Significance

RGS2 has been implicated in several pathological conditions. Mutations or dysregulation of RGS2 expression have been associated with hypertension, heart failure, and other cardiovascular diseases. Additionally, RGS2 has been shown to have protective effects against myocardial hypertrophy and atrial arrhythmias .

Therapeutic Potential

Given its critical role in GPCR signaling and its involvement in various diseases, RGS2 has emerged as a potential therapeutic target. Modulating RGS2 activity could provide new avenues for the treatment of hypertension, heart failure, and other related conditions. Research is ongoing to develop small molecules or other therapeutic agents that can specifically target RGS2 and modulate its activity .

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.