RASD2 Human

RASD Family Member 2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RASD2 Human

RASD2 (Ras homolog enriched in striatum) is a gene located on chromosome 22q12.3 that encodes a GTP-binding protein critical for modulating dopaminergic signaling and striatal function. It belongs to the Ras superfamily of small GTPases and is developmentally regulated by thyroid hormone and dopamine innervation . The protein is highly expressed in the striatum and nucleus accumbens, with distinct patterns in humans compared to rodents .

Expression Patterns

Tissue-Specific Expression
RASD2 exhibits high expression in brain regions critical for reward and cognition:

Tissue/RegionExpression LevelKey Functions
StriatumEnrichedModulates cAMP/PKA signaling, DRD2 activity .
Nucleus accumbensModerateRegulates stress-induced depression-like behaviors .
Prefrontal cortexModerateLinked to schizophrenia-related phenotypes .
HippocampusLowImplicated in antidepressant responses .

Single-Cell RNA-Seq Data
Human and rodent brains show divergent RASD2 expression clusters. In humans, it is enriched in striatal GABAergic neurons and cortical glutamatergic neurons .

Genetic Variants and Clinical Associations

Key Genetic Findings

SNPLocationClinical AssociationSource
rs6518956IntronReduced prefrontal mRNA in schizophrenia .
rs57501125′ UTRPotential regulatory variant

Schizophrenia and Psychosis

  • Reduced RASD2 mRNA in prefrontal cortex of schizophrenia patients .

  • Genetic variation (rs6518956) correlates with prefrontal-striatal gray matter volume and working memory deficits .

Functional Roles in Signaling Pathways

Primary Pathways

PathwayMechanismOutcome
cAMP/PKAInhibits GPCR-mediated cAMP productionAlters striatal dopamine signaling .
DRD2 signalingModulates DRD2-dependent signalingRegulates antidepressant responses .
mTOR/AKTInteracts with mTOR and AKT cascadesImplicated in neurodegenerative diseases .

Behavioral Implications

  • Depression: Stress-induced Rasd2 downregulation in mice exacerbates depression-like behaviors, reversed by Rasd2 overexpression in DRD2+ neurons .

  • Schizophrenia: Rasd2 knockout mice show impaired sensorimotor gating and heightened sensitivity to psychotomimetics (e.g., amphetamine, PCP) .

Research Findings in Psychiatric Disorders

Schizophrenia

  • RASD2 mRNA reduction in prefrontal cortex correlates with cortical thinning and working memory deficits .

  • Genetic risk variants (e.g., rs6518956) linked to disrupted prefronto-striatal connectivity .

Depression

ModelInterventionOutcome
5-day UMS stressRasd2 overexpressionAlleviates anhedonia, improves sucrose preference .
OvariectomyRasd2 knockdownWorsens immobility in forced swim test .

Gender-Specific Effects

  • Rasd2 overexpression in hippocampus shows antidepressant-like effects in ovariectomized mice, with increased DRD2 and BDNF expression .

Therapeutic Implications

Targeting RASD2

  • Depression: DREADD activation of DRD2+ PrL-NAcc neurons enhances Rasd2 expression and mitigates stress-induced depression .

  • Schizophrenia: Modulating RASD2 may restore striatal cAMP/PKA balance, reducing psychosis risk .

Precision Medicine

  • RASD2 variants (e.g., rs6518956) could predict therapeutic responses to DRD2-targeted interventions .

Product Specs

Introduction
RASD2, a member of the Ras superfamily of small GTPases, binds to GTP and exhibits GTPase activity. It plays a crucial role in regulating signaling pathways involving G-protein-coupled receptors and heterotrimeric proteins. Notably, RASD2 is highly expressed in the striatum.
Description
Recombinant RASD2, expressed in E. coli, is a non-glycosylated polypeptide chain consisting of 289 amino acids (specifically, residues 1-266). With a molecular weight of 32.8 kDa, it features a 23 amino acid His-tag at the N-terminus. The protein undergoes purification using proprietary chromatographic techniques.
Physical Appearance
The product is a clear solution that has been sterilized by filtration.
Formulation
The RASD2 protein solution is provided at a concentration of 0.5 mg/ml. It is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure long-term stability, consider adding a carrier protein such as 0.1% HSA or BSA. It is crucial to avoid repeated freeze-thaw cycles.
Purity
The purity of the RASD2 protein is determined to be greater than 85.0% using SDS-PAGE analysis.
Synonyms
GTP-binding protein Rhes, Ras homolog enriched in striatum, TEM2, RASD2, Rhes, Tumor endothelial marker 2, RASD Family Member 2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMMKTLSS GNCTLSVPAK NSYRMVVLGA SRVGKSSIVS RFLNGRFEDQ YTPTIEDFHR KVYNIRGDMY QLDILDTSGN HPFPAMRRLS ILTGDVFILV FSLDNRESFD EVKRLQKQIL EVKSCLKNKT KEAAELPMVI CGNKNDHGEL CRQVPTTEAE LLVSGDENCA YFEVSAKKNT NVDEMFYVLF SMAKLPHEMS PALHRKISVQ YGDAFHPRPF CMRRVKEMDA YGMVSPFARR PSVNSDLKYI KAKVLREGQA RERDKCTIQ.

Q&A

What is RASD2 and what are its primary functions in human neural systems?

RASD2 is a GTP-binding protein that belongs to the RAS superfamily and is highly enriched in the striatum while being expressed at lower levels in the hippocampus, cerebral cortex, and olfactory bulb . It functions primarily as a negative regulator of G protein–coupled receptor-mediated cAMP production, with targeted deletion of RASD2 in mice significantly activating the cAMP/protein kinase A signaling pathway in the striatum . Recent research has revealed that RASD2 also regulates the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mechanistic target of rapamycin signaling pathway, suggesting its role in several neurological and psychiatric diseases, including schizophrenia and Huntington's disease . RASD2's function is closely tied to dopamine function, as depleting the striatum of dopaminergic input decreases RASD2 mRNA expression, and activation of dopamine D2 receptors (DRD2) produces exaggerated responses in RASD2 knockout mice .

How does RASD2 interact with dopamine receptors and related signaling pathways?

RASD2 exhibits a complex relationship with dopamine receptors, particularly the D2 receptor (DRD2). Co-immunoprecipitation studies have demonstrated that RASD2 directly interacts with DRD2 in the hippocampus of ovariectomized mice, suggesting a physical association between these proteins . RASD2 deficiency produces aberrant DRD2-dependent activity through abnormal Ca²⁺-dependent modulation of PI3K/Akt signaling, indicating that RASD2 serves as a critical regulator of dopamine signaling . RASD2 mRNA has been located in dopamine D1 receptor-medium spiny neurons, DRD2-medium spiny neurons, and cholinergic interneurons, showing its widespread influence on dopaminergic pathways . Additionally, RASD2 regulates dopamine-dependent neurotransmission by affecting the survival of nigrostriatal dopaminergic neurons, which further demonstrates its crucial role in maintaining proper dopamine signaling .

What are the primary experimental methods for studying RASD2 expression in human tissue samples?

When investigating RASD2 expression in human tissue samples, researchers typically employ a combination of molecular and biochemical techniques. RNA sequencing (RNA-seq) represents a powerful method for quantifying RASD2 gene expression changes across different experimental conditions, as demonstrated in studies examining acute fasting effects on gene expression in mouse prefrontal cortex . Western blot analysis provides a reliable approach for measuring RASD2 protein levels in tissue samples, allowing researchers to compare expression across different brain regions or between experimental and control groups . Enzyme-linked immunosorbent assays (ELISA) offer another quantitative method for detecting RASD2 protein levels in serum or tissue homogenates . Co-immunoprecipitation techniques are valuable for investigating protein-protein interactions involving RASD2, such as its association with DRD2 . For spatial localization studies, immunohistochemistry can visualize the distribution of RASD2 across different brain regions and cell types.

How does RASD2 regulate the PI3K/Akt/mTOR signaling pathway in neuronal cells?

RASD2 functions as a critical regulator of the PI3K/Akt/mTOR signaling pathway in neuronal cells through multiple mechanisms. Research has shown that RASD2 deficiency leads to abnormal excitatory responses of cholinergic interneurons to activation of DRD2 receptors, with PI3K inhibitors rescuing these abnormal DRD2 responses in RASD2 knockout mice . This suggests that RASD2 acts as a molecular bridge between PI3K and Akt signaling components . Experimental evidence from fasting studies revealed activation of the PI3K-Akt pathway (as shown in KEGG pathway analysis) along with changes in Akt expression, which were reversed by DRD2 antagonist treatment . The PI3K-Akt pathway represents one of the key DRD2-linked signaling pathways implicated in mood disorders and BDNF-mediated neuroprotection . RASD2 appears to produce antidepressant-like effects by regulating the expression of Akt and associated signaling molecules, thereby influencing neuroplasticity and cellular resilience mechanisms that protect against stress-induced neuronal damage .

What epigenetic mechanisms influence RASD2 expression during neural development and in response to environmental factors?

Epigenetic regulation of RASD2 represents an emerging area of research that explains how environmental factors like stress, hormonal changes, and dietary interventions might modulate its expression. While the provided research does not directly address epigenetic mechanisms, the differential expression of RASD2 in response to acute fasting suggests potential epigenetic regulation . Fasting is known to induce significant metabolic changes that can influence epigenetic marks such as DNA methylation and histone modifications across the genome. The relationship between estrogen and RASD2 expression points to potential hormone-responsive elements in the RASD2 promoter region that may be epigenetically regulated . Research has shown that ovariectomy (which reduces estrogen levels) decreases RASD2 expression in the hippocampus, suggesting hormonal control of its transcription . Studies in related fields have demonstrated that nutritional interventions like fasting can trigger widespread epigenetic reprogramming through changes in metabolic cofactors required for chromatin-modifying enzymes.

How do RASD2 expression levels correlate with DRD2-CREB-BDNF signaling cascades in different brain regions?

RASD2 expression shows region-specific correlations with DRD2-CREB-BDNF signaling components across different brain areas. In the hippocampus, overexpression of RASD2 in ovariectomized mice significantly increased DRD2 expression, suggesting a positive regulatory relationship between these proteins in this brain region . Experimental evidence demonstrated that RASD2 overexpression also increased BDNF expression in the hippocampus, indicating that RASD2 positively influences the CREB-BDNF pathway downstream of DRD2 activation . In the prefrontal cortex and hippocampus, acute fasting increased CREB and BDNF expression, and these effects were antagonized by DRD2 antagonist treatment, confirming that the CREB-BDNF signaling pathway is involved in fasting-induced antidepressant effects mediated by DRD2 and RASD2 . The striatum, which contains the highest natural expression of RASD2, showed different patterns of regulation compared to the hippocampus and prefrontal cortex, suggesting that RASD2's influence on DRD2-CREB-BDNF signaling may be region-specific and possibly influenced by different cellular compositions and circuit connections .

What are the most effective animal models for studying RASD2 function in relation to neuropsychiatric disorders?

The ovariectomized mouse model has proven particularly valuable for studying RASD2 function in relation to depression. This model involves surgical removal of the ovaries to reduce estrogen levels, which induces depression-like behaviors that can be measured through standard behavioral tests . Ovariectomized mice show increased immobility time in forced swimming and tail suspension tests, decreased swimming time, and reduced sucrose consumption, all indicating depression-like phenotypes . These behavioral changes correlate with decreased RASD2 and DRD2 expression in the hippocampus, establishing a clear link between RASD2 levels and depressive behaviors . RASD2 knockout mice represent another powerful model that has revealed the protein's role in dopamine signaling, as these animals show exaggerated stereotypy in response to DRD2 activation and abnormal Ca²⁺-dependent modulation of PI3K/Akt signaling . Viral-mediated RASD2 overexpression models, as demonstrated in the provided research, allow region-specific manipulation of RASD2 levels to evaluate its direct effects on behavior and molecular pathways .

What are the optimal protocols for viral-mediated RASD2 overexpression or knockdown studies in specific brain regions?

For effective viral-mediated manipulation of RASD2 expression in specific brain regions, researchers should follow several critical considerations. Based on the research provided, lentiviral vectors represent an effective delivery system for achieving RASD2 overexpression in targeted brain areas such as the dorsal hippocampus . The experimental timeline should allow sufficient time for viral expression before behavioral testing; the cited study waited 14 days after virus injection before performing ovariectomy, followed by an additional 7 days before behavioral assessments and tissue collection . Precise stereotaxic injection techniques are essential for targeting specific brain regions, as demonstrated by the researchers' focus on the dorsal hippocampus due to its high density of DRD2 binding sites . For knockdown studies, which were not specifically detailed in the provided research, siRNA or shRNA approaches delivered through similar viral vectors would be appropriate. Post-experimental verification of successful RASD2 manipulation through Western blot or immunohistochemistry is crucial for confirming the efficacy of viral transduction and expression .

How can researchers effectively measure changes in RASD2-mediated signaling pathways following experimental interventions?

To comprehensively assess RASD2-mediated signaling changes following experimental interventions, researchers should employ a multi-level analytical approach. Western blot analysis represents a fundamental technique for measuring protein expression changes in RASD2 and its downstream effectors like DRD2, CREB, BDNF, and Akt in dissected brain regions . Co-immunoprecipitation assays are valuable for detecting protein-protein interactions between RASD2 and its binding partners, such as DRD2, providing insights into how experimental manipulations affect these molecular associations . RNA sequencing offers a broader perspective by identifying genome-wide transcriptional changes associated with RASD2 manipulation, as demonstrated in the fasting studies where RNA-seq revealed significant RASD2 expression changes . Pharmacological challenge experiments using receptor-specific agonists or antagonists (such as the DRD2 antagonist sulpiride) can help dissect the specific signaling pathways influenced by RASD2 . Behavioral assays (forced swimming test, tail suspension test, open field test, and sucrose preference test) provide functional readouts of how RASD2-mediated signaling changes translate to behavioral phenotypes relevant to neuropsychiatric disorders .

What statistical approaches are most appropriate for analyzing complex RASD2 interaction data across multiple signaling pathways?

When analyzing complex RASD2 interaction data across multiple signaling pathways, researchers should employ robust statistical approaches that account for the multidimensional nature of these datasets. Two-way analysis of variance (ANOVA) represents an effective statistical method for examining the interaction effects between two independent variables (such as treatment with sulpiride and fasting) on RASD2 and related signaling proteins . Post-hoc tests such as Tukey's Honestly Significant Difference (HSD) test allow for specific comparisons between experimental groups after significant ANOVA results are obtained . Effect size calculations using Cohen's d provide valuable information about the magnitude of experimental effects beyond mere statistical significance, as demonstrated in the behavioral test analyses . For complex protein interaction networks, pathway enrichment analysis (such as KEGG pathway analysis mentioned in the research) can identify significantly affected biological processes and place RASD2 functions within broader cellular contexts . When examining correlations between RASD2 expression and other variables, regression analyses with appropriate controls for confounding factors should be employed to establish meaningful relationships.

What is the current evidence linking RASD2 dysfunction to depression and how does this compare to other neuropsychiatric conditions?

Current evidence strongly supports a role for RASD2 in depression pathophysiology through several key observations. Ovariectomized mice exhibiting depression-like behaviors show significantly decreased RASD2 expression in the hippocampus, establishing a correlation between RASD2 reduction and depressive phenotypes . Direct overexpression of RASD2 in the hippocampus of ovariectomized mice produces clear antidepressant-like effects, including decreased immobility in forced swimming and tail suspension tests and increased sucrose consumption, demonstrating a causal relationship between RASD2 levels and depressive behaviors . RASD2's regulation of DRD2 signaling provides a mechanistic link to depression, as dopamine dysregulation has been implicated in depression pathophysiology, and RASD2 overexpression increases DRD2 expression in the hippocampus . The involvement of RASD2 in the PI3K/Akt and CREB-BDNF pathways, which are known to be dysregulated in depression, further supports its role in this disorder . Regarding other neuropsychiatric conditions, previous research has implicated RASD2 in schizophrenia and Huntington's disease, suggesting that this protein may serve as a common molecular node across multiple brain disorders with distinct clinical presentations .

How do sex differences influence RASD2 expression and function in neuropsychiatric disorders?

Sex differences significantly impact RASD2 expression and function, with estrogen playing a particularly important regulatory role. Research has demonstrated that ovariectomy, which reduces estrogen levels, leads to decreased RASD2 expression in the hippocampus alongside depression-like behaviors, suggesting estrogen positively regulates RASD2 expression . RNA-seq data indicates that estrogen and acute fasting exert antidepressant-like effects through a common gene, RASD2, pointing to convergent hormonal and metabolic regulation of this protein . Previous studies have found that RASD2 regulates striatal-dependent behaviors in a gender-specific manner, indicating that its functional effects may differ between males and females . The interaction between RASD2 and estrogen receptor beta (ERβ) in the context of fasting-induced antidepressant effects further supports sex-specific regulation mechanisms . These findings collectively suggest that therapeutic approaches targeting RASD2 may need to be tailored differently for male and female patients with neuropsychiatric disorders, and future research should systematically investigate sex differences in RASD2 function across various brain regions and conditions.

What are the potential mechanisms by which acute fasting produces antidepressant effects through RASD2-mediated pathways?

Acute fasting produces antidepressant effects through several RASD2-mediated mechanisms that converge on dopamine and neurotrophic signaling. RNA sequencing studies have demonstrated that 9-hour fasting induces significant upregulation of RASD2 gene expression, particularly in the prefrontal cortex . Fasting increases RASD2 protein levels in the hippocampus, prefrontal cortex, and striatum, with these effects being most pronounced in the hippocampus and mediated through DRD2, as evidenced by the ability of the DRD2 antagonist sulpiride to block these changes . The antidepressant effects of fasting are associated with activation of the CREB-BDNF signaling pathway, as shown by increased CREB and BDNF expression in the prefrontal cortex and hippocampus following fasting, effects that are dependent on RASD2 and DRD2 function . Fasting also activates the PI3K-Akt pathway and increases Akt expression, representing another DRD2-linked signaling pathway that has been implicated in mood disorders and BDNF-mediated neuroprotection . Additionally, fasting upregulates estrogen receptor beta expression through RASD2, providing a potential link between metabolic state, hormone signaling, and mood regulation that may be particularly relevant for female depression models .

What are the most promising therapeutic targets within the RASD2 signaling network for treating depression?

The RASD2 signaling network offers several promising therapeutic targets for depression treatment based on current research findings. DRD2 represents a primary target within this network, as RASD2 directly interacts with and regulates DRD2, and DRD2 antagonism blocks the antidepressant effects of both RASD2 overexpression and acute fasting . The CREB-BDNF pathway downstream of RASD2 activation presents another valuable target, as increased CREB and BDNF expression correlates with antidepressant effects, and these proteins have established roles in neuroplasticity and resilience mechanisms relevant to depression . The PI3K/Akt signaling pathway regulated by RASD2 offers additional therapeutic possibilities, particularly since PI3K inhibitors have been shown to rescue abnormal DRD2 responses in RASD2 knockout mice . Estrogen receptor beta (ERβ) emerges as a sex-specific target within the RASD2 network, given that fasting upregulates ERβ expression through RASD2-dependent mechanisms, suggesting potential for tailored approaches in female depression patients . RASD2 itself represents a direct therapeutic target, as viral-mediated overexpression of RASD2 in the hippocampus produced robust antidepressant effects in ovariectomized mice, suggesting that compounds enhancing RASD2 expression or function could have therapeutic potential .

How might dietary interventions that mimic fasting be optimized to modulate RASD2 expression for therapeutic benefit?

Optimizing dietary interventions to modulate RASD2 expression for therapeutic benefit requires careful consideration of several factors. Duration appears critical, as the research specifically examined 9-hour fasting periods, suggesting that precise timing may be important for achieving optimal RASD2 upregulation without inducing stress responses that could counteract beneficial effects . The temporal dynamics of RASD2 expression changes following fasting should be characterized in detail to determine whether shorter fasting periods might be effective or if intermittent fasting regimens could maintain elevated RASD2 levels over extended periods . Brain region specificity must be considered, as fasting increased RASD2 expression in the hippocampus, prefrontal cortex, and striatum, but with varying magnitudes across these regions . Mechanistic understanding of how fasting increases RASD2 expression could help identify specific dietary components or metabolic pathways that might be targeted without requiring complete food restriction . Individual factors including sex, age, and baseline metabolic status likely influence the effectiveness of fasting for modulating RASD2, particularly given the interactions between estrogen signaling and RASD2 function demonstrated in the research .

What novel biomarkers related to RASD2 function could be developed to improve depression diagnosis and treatment monitoring?

Several promising biomarker approaches related to RASD2 function could enhance depression diagnosis and treatment monitoring. Blood-based RASD2 protein levels represent a potential peripheral biomarker, as the research states that "RASD2 can be postulated to be a potential predictive marker for depression," though further studies are needed to determine whether peripheral RASD2 levels correlate with brain expression . The RASD2-DRD2 interaction profile could serve as a more sophisticated biomarker, potentially measurable through advanced molecular techniques applied to blood cells or through neuroimaging approaches that can assess receptor binding potential . Downstream signaling molecules in the RASD2 pathway, such as BDNF, CREB, and Akt phosphorylation states, could function as surrogate biomarkers that reflect RASD2 activity and correlate with treatment response . Functional readouts from neuroimaging studies examining regions with high RASD2 expression (striatum, hippocampus, prefrontal cortex) might provide non-invasive biomarkers of RASD2-related circuit activity . Genetic or epigenetic markers related to RASD2 expression or regulation could serve as predictive biomarkers for identifying patients most likely to respond to treatments targeting RASD2 pathways, allowing for personalized therapeutic approaches .

How do behavioral test results correlate with RASD2 expression levels across different experimental models?

Behavioral test results demonstrate consistent correlations with RASD2 expression levels across multiple experimental paradigms. The forced swimming test (FST) shows a robust inverse relationship with RASD2 levels, where decreased RASD2 expression in ovariectomized mice coincides with increased immobility time, while RASD2 overexpression or fasting-induced RASD2 upregulation reduces immobility time . Similarly, the tail suspension test (TST) results mirror FST findings, with RASD2 overexpression significantly reducing immobility time in ovariectomized mice, suggesting consistent effects across different behavioral assessments of depression-like behavior . Sucrose preference test results, which measure anhedonia (a core symptom of depression), positively correlate with RASD2 expression, as demonstrated by decreased sucrose consumption in ovariectomized mice with reduced RASD2 levels and increased consumption following RASD2 overexpression . Notably, open field test measures of general locomotor activity and exploratory behavior show no significant correlation with RASD2 expression, indicating that RASD2's effects are specific to depression-related behaviors rather than general activity levels . These behavioral correlations are mechanistically linked to RASD2's regulation of DRD2 and downstream CREB-BDNF signaling, as demonstrated by the ability of DRD2 antagonists to block both the behavioral and molecular effects of RASD2 manipulation .

What experimental contradictions or inconsistencies have been observed in RASD2 research, and how might they be resolved?

Several experimental nuances and potential contradictions in RASD2 research warrant careful consideration and resolution through further study. Region-specific effects represent one area of complexity, as RASD2 shows differential expression and potentially different functions across brain regions, with the highest expression in the striatum but clearest antidepressant effects demonstrated through hippocampal manipulation . The relationship between RASD2 and estrogen signaling shows some complexity, as the research demonstrates that both estrogen and fasting upregulate RASD2, yet ovariectomy (which decreases estrogen) produces depression-like behavior, suggesting context-dependent regulation that requires further characterization . Molecular pathway integration presents another challenge, as RASD2 influences multiple signaling cascades (CREB-BDNF, PI3K/Akt, dopamine signaling), and how these pathways interact and potentially compensate for each other remains incompletely understood . Temporal dynamics of RASD2 effects need clarification, as acute (9-hour) fasting increases RASD2 expression and produces antidepressant effects, but the duration of these effects and whether they persist with prolonged fasting remains unclear . Resolving these questions will require comprehensive studies integrating multiple brain regions, signaling pathways, and temporal analyses to build a more complete understanding of RASD2 function.

Product Science Overview

Gene and Protein Structure

The RASD2 gene is located on chromosome 22q12.3 and encodes a protein that is approximately 30.2 kDa in size . The protein consists of several domains that are crucial for its function, including a GTP-binding domain and regions involved in protein-protein interactions .

Function and Mechanism

RASD2 functions as a GTPase signaling protein, which means it binds to and hydrolyzes GTP. This activity is essential for regulating signaling pathways involving G-protein-coupled receptors and heterotrimeric proteins such as GNB1, GNB2, and GNB3 . The protein is enriched in the striatum, a region of the brain involved in motor control and cognitive functions .

One of the critical roles of RASD2 is its function as an E3 ligase for the attachment of small ubiquitin-like modifier (SUMO) proteins. This modification process, known as sumoylation, is vital for various cellular processes, including nuclear-cytosolic transport, transcriptional regulation, apoptosis, and protein stability .

Clinical Significance

RASD2 has been implicated in several neurological disorders. For instance, it binds to mutant huntingtin (mHtt), the protein mutated in Huntington’s disease (HD). Sumoylation of mHtt by RASD2 may contribute to the degeneration of the striatum observed in HD patients . Additionally, reduced levels of RASD2 mRNA have been found in HD patients, suggesting a potential role in the disease’s pathology .

Moreover, RASD2 is involved in the activation of the mechanistic target of rapamycin 1 (mTOR1), which plays a role in myelination, axon growth, and regeneration . This function is crucial for maintaining neuronal health and function.

Recombinant Protein

Human recombinant RASD2 is produced using various expression systems, including HEK293T cells. The recombinant protein is used in research to study its function, interactions, and role in diseases. It is available in different quantities and is typically stored at -80°C to maintain stability .

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 2025 TheBiotek. All Rights Reserved.