RND3 Human

Rho Family GTPase 3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Physiological Roles in Human Tissues

Cytoskeletal and Cellular Dynamics

  • Actin Reorganization: Induces stress fiber disassembly and filopodia formation in epithelial and immune cells, critical for migration and adhesion .

  • Mitochondrial Homeostasis: Maintains mitochondrial oxidative phosphorylation, ATP synthesis, and prevents glycolysis dependency. RND3 KO cells exhibit mitochondrial fragmentation and lipid droplet accumulation .

  • Neurodevelopment: Essential for neuronal migration along the rostral migratory stream (RMS) and olfactory bulb development .

Tissue-Specific Expression

TissueExpression LevelFunctional RoleSource
BrainHighNeuronal migration, mitochondrial function
PlacentaModerateTrophoblast proliferation and apoptosis
Adipose TissueLowMetabolic regulation

Pathological Roles and Disease Associations

Cardiovascular Diseases

  • Heart Failure: RND3 haploinsufficiency exacerbates cardiac apoptosis under hemodynamic stress (e.g., transverse aortic constriction). ROCK1 inhibition rescues cardiac function in Rnd3+/− mice .

  • Hypertension: ROCK1 hyperactivation in RND3-deficient hearts correlates with elevated myosin light chain phosphorylation and vascular remodeling .

Cancer

Cancer TypeRND3 ExpressionMechanismClinical ImpactSource
Lung CancerDownregulatedNICD stabilization via Notch signalingPromotes proliferation
GlioblastomaDownregulatedSnail1 degradation via proteasomal pathwayInhibits invasion
MelanomaUpregulatedRhoA/ROCK/Cofilin axis activationEnhances podosome dynamics

Reproductive Disorders

  • Recurrent Miscarriage (RM): Elevated RND3 in cytotrophoblasts (CTBs) correlates with reduced proliferation, increased apoptosis, and impaired migration via RhoA-ROCK1 and ERK1/2 pathways .

Neurodegenerative Diseases

  • Parkinson’s Disease: ROCK1-mediated mitochondrial fission and oxidative stress may involve RND3 dysregulation .

Therapeutic Potential and Challenges

Targeting RND3 in Diseases

  • Cancer Therapy: Overexpression of RND3 inhibits Notch signaling (lung cancer) and Snail1-mediated EMT (glioblastoma), suggesting RND3 as a tumor suppressor .

  • Heart Failure: ROCK1 inhibitors (e.g., fasudil) may mitigate RND3 deficiency-induced apoptosis .

  • Recurrent Miscarriage: Modulating RND3-FOXD3 axis to restore trophoblast function .

Challenges

  • Context-Dependent Effects: RND3 exhibits oncogenic or tumor-suppressive roles depending on cancer type (e.g., upregulated in melanoma vs. downregulated in lung cancer) .

  • Tissue-Specific Delivery: Diverse expression patterns necessitate targeted therapeutic strategies .

Product Specs

Introduction
Belonging to the Ras superfamily, RND3 is a small GTPase that regulates the actin cytoskeleton's organization in response to extracellular growth factors. This activity leads to a reduction in stress fibers and promotes cell migration, partially by modulating the Rho-dependent kinase ROCK1. RND3 also plays a role in inhibiting DNA damage-induced apoptosis and cell cycle arrest. As a GTP-binding protein, RND3 exhibits resistance to Rho-specific GTPase-activating proteins.
Description
Produced in E. coli, Recombinant Human RND3 is a single, non-glycosylated polypeptide chain. This protein consists of 261 amino acids (specifically, amino acids 1-241) and has a molecular weight of 29.2 kDa. A 20 amino acid His-Tag is fused to the N-terminus of RND3, which is then purified using proprietary chromatographic methods.
Physical Appearance
A clear, sterile solution without any color.
Formulation
The RND3 Human solution is prepared with the following components: 20mM Tris-HCl (pH 8), 1mM DTT, 1mM EDTA, 0.1M NaCl, and 40% glycerol.
Stability
For optimal storage, keep the vial at 4°C if it will be used completely within 2-4 weeks. For longer storage periods, freeze the solution at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Repeated freezing and thawing cycles should be avoided.
Purity
Analysis by SDS-PAGE confirms that the purity is greater than 90.0%.
Synonyms
ARHE, memB, Rho8, RhoE, Rho-related GTP-binding protein RhoE, Protein MemB, Rho family GTPase 3, Rho-related GTP-binding protein Rho8, Rnd3.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MKERRASQKL SSKSIMDPNQ NVKCKIVVVG DSQCGKTALL HVFAKDCFPE NYVPTVFENY TASFEIDTQR IELSLWDTSG SPYYDNVRPL SYPDSDAVLI CFDISRPETL DSVLKKWKGE IQEFCPNTKM LLVGCKSDLR TDVSTLVELS NHRQTPVSYD QGANMAKQIG AATYIECSAL QSENSVRDIF HVATLACVNK TNKNVKRNKS QRATKRISHM PSRPELSAVA TDLRKDKAKS C.

Q&A

What is RND3 and what makes it unique among GTPases?

RND3 (also known as RhoE) is a small G-protein belonging to the Rnd family of Rho GTPases, which is part of the Ras superfamily. Unlike conventional Rho GTPases, RND3 is considered atypical because it lacks GTPase activity and remains constitutively in the GTP-bound active state. The human RND3 protein is approximately 27 kDa in size and 244 amino acids in length, with the mature chain extending from amino acids 1-241 . What distinguishes RND3 from other GTPases is its inability to hydrolyze GTP, which results in a constitutively active state that is primarily regulated through changes in expression level, protein stability, and subcellular localization rather than GTP/GDP cycling.

Where is RND3 expressed in human tissues?

RND3 is ubiquitously expressed across human tissues, with particularly notable expression in the brain, placenta, and various epithelial cells. In the brain, RND3 is predominantly expressed in the cytoplasm of cortical cells . In placental tissue, RND3 is expressed in both cytotrophoblasts (CTB) and syncytiotrophoblasts (STB) during the first trimester . RND3 can be detected in multiple human cell lines including HeLa cervical epithelial carcinoma cells, SH-SY5Y neuroblastoma cells, MCF-7 breast cancer cells, and Jurkat acute T cell leukemia cells . Immunohistochemistry and immunofluorescence studies have confirmed cytoplasmic localization of RND3 in normal tissues, with minimal or no nuclear staining .

What are the primary biological functions of RND3?

RND3 serves multiple critical biological functions:

  • Cell cycle regulation: RND3 inhibits cell proliferation by preventing S-phase entry and promoting G0/G1 phase accumulation .

  • Apoptosis regulation: RND3 promotes apoptosis in certain contexts through interaction with NF-κB P65 and by modulating the ERK1/2 signaling pathway .

  • Cell migration and invasion: RND3 regulates cytoskeletal dynamics, affecting cell motility and migration capabilities .

  • Differentiation: RND3 is involved in keratinocyte differentiation and stratification, where its overexpression results in cell size enlargement and an increase in the number of stratified cells .

  • Development: Genetic deletion of Rnd3 can result in developmental abnormalities including aqueductal stenosis leading to hydrocephalus and heart calcium leakage resulting in heart failure in mouse models .

How is RND3 expression altered in disease states?

RND3 expression shows significant alterations in various pathological conditions:

  • Recurrent miscarriage (RM): RND3 expression is significantly increased in the cytotrophoblasts of patients with RM compared to normal villi. This elevated expression correlates with decreased proliferation and increased apoptosis of trophoblast cells, potentially contributing to pregnancy complications .

  • Hepatocellular carcinoma (HCC): RND3 expression is downregulated in HCC cell lines and tissues compared to normal liver. Low expression of RND3 is associated with the presence of satellite nodules in HCC, suggesting a role in cancer progression .

  • Brain disorders: Altered RND3 expression affects neuronal apoptosis, with RND3 knockout mice showing decreased rates of brain apoptosis, suggesting its regulatory role in neural development and potentially in neurodegenerative conditions .

What molecular mechanisms underlie RND3's regulation of cell cycle progression?

RND3 exerts complex control over cell cycle progression through several interrelated mechanisms:

  • G1/S transition inhibition: Overexpression of RND3 leads to accumulation of cells in the G0/G1 phase (51.23% ± 0.2569 vs. 43.41% ± 0.332 in controls) and reduction in S phase cells (37.32% ± 3.705 vs. 50.5% ± 1.637 in controls) . This cell cycle arrest is mediated through:

    • Downregulation of cyclin D1, a critical G1-S phase cycling protein, as demonstrated in RND3-overexpressing cells compared to control groups .

    • Reduced expression of Ki-67, a proliferation marker expressed only in active phases of the cell cycle (G2/M and S phases) .

  • ROCK1 pathway modulation: RND3 regulates the RhoA-ROCK1 signaling pathway, which has established roles in cytoskeletal reorganization necessary for mitosis. Inhibition of this pathway contributes to RND3's anti-proliferative effects .

  • Telomerase regulation: RND3 knockdown decreases human telomerase reverse transcriptase (hTERT) expression, inducing senescence in hepatocellular carcinoma cells. Interestingly, re-expression of RND3 allows cells to bypass senescence and regain proliferative capacity with restored hTERT expression .

These mechanisms collectively explain how RND3 serves as a checkpoint regulator for cell proliferation, with potential implications for cancer research and therapeutic development.

How does RND3 influence apoptotic pathways in different cellular contexts?

RND3's role in apoptosis is context-dependent and involves multiple signaling pathways:

  • NF-κB P65 interaction: RND3 directly interacts with NF-κB P65 as demonstrated by immunoprecipitation analysis. This interaction blocks the anti-apoptotic action of NF-κB P65, thereby promoting apoptotic signaling . In Rnd3-knockout mice, decreased rates of brain apoptosis are observed through immunofluorescence and TUNEL assays, confirming RND3's pro-apoptotic function in neural tissue .

  • ERK1/2 pathway regulation: In trophoblast cells, RND3 inhibits apoptosis through modulation of the ERK1/2 signaling pathway. This appears contradictory to its effects in neural tissue, highlighting the cell type-specific nature of RND3's function .

  • Balance with proliferative signals: The apoptotic effect of RND3 must be understood in balance with its anti-proliferative activities. In hepatocellular carcinoma, RND3 silencing induces growth arrest not through increased cell death but through induction of senescence, suggesting a complex relationship between proliferation inhibition and apoptosis promotion .

The dual and sometimes opposing roles of RND3 in apoptosis across different tissues underscores the importance of cellular context in RND3 research and the need for tissue-specific experimental designs when investigating its function.

What is the relationship between RND3 and transcription factors in development and disease?

RND3 exhibits important interactions with transcription factors that influence developmental processes and disease progression:

  • FOXD3 regulatory axis: FOXD3 (Forkhead Box D3) has been identified as a key transcription factor that binds to the RND3 core promoter region and regulates its expression. In recurrent miscarriage tissues, both FOXD3 and RND3 expression are significantly increased in cytotrophoblasts . This transcriptional regulation is functionally significant as FOXD3-mediated upregulation of RND3 inhibits proliferation and migration while promoting apoptosis in trophoblast cells .

  • hTERT expression correlation: RND3 knockdown decreases human telomerase reverse transcriptase (hTERT) expression, though the exact transcriptional mechanism remains to be fully elucidated. This relationship is reversible, as re-expression of RND3 restores hTERT levels, suggesting dynamic transcriptional control .

  • Cell-type specific transcriptional programs: The varied effects of RND3 across different tissues suggest interaction with tissue-specific transcriptional programs. For example, in keratinocytes, RND3 influences differentiation and stratification, which involves distinct transcriptional cascades compared to its role in neural tissue .

Understanding these transcriptional networks is crucial for developing targeted therapeutic approaches that modulate RND3 expression in a tissue-specific manner.

How do post-translational modifications affect RND3 function?

While the search results don't provide comprehensive details on post-translational modifications of RND3, we can infer from the general understanding of Rho GTPases that post-translational modifications likely play crucial roles in regulating RND3's activity, localization, and interactions.

Unlike conventional Rho GTPases that cycle between active GTP-bound and inactive GDP-bound states, RND3 remains constitutively GTP-bound due to its lack of intrinsic GTPase activity. Therefore, its regulation must occur through alternative mechanisms, including post-translational modifications that affect:

  • Protein stability and turnover: Modifications like ubiquitination and phosphorylation can influence RND3 protein half-life and degradation pathways.

  • Subcellular localization: Prenylation, phosphorylation, or other modifications may direct RND3 to specific cellular compartments, affecting its access to effector proteins.

  • Protein-protein interactions: Modifications may create or disrupt binding sites for interacting partners like ROCK1 or NF-κB P65, altering downstream signaling.

Further research specifically targeting the post-translational modification landscape of RND3 would provide valuable insights into these regulatory mechanisms.

What antibodies and detection methods are most effective for studying RND3 expression?

Based on the search results, several validated antibodies and detection methods have proven effective for RND3 research:

  • Western Blot:

    • Human/Mouse RND3 (RD1 Peptide) Monoclonal Antibody (R&D Systems, Catalog # MAB6618) at 1 μg/mL concentration has effectively detected RND3 as a specific band at approximately 29 kDa in multiple human cell lines including HeLa, SH-SY5Y, MCF-7, and Jurkat cells .

    • Anti-RhoE/Rnd3 Antibody, clone 4 (Sigma-Aldrich, product 05-723) has been validated for western blot applications at 1:500-1:2000 dilution in 3T3/A31 cell lysates .

    • Western blot analysis should be conducted under reducing conditions using appropriate immunoblot buffer systems .

  • Immunocytochemistry/Immunofluorescence:

    • Anti-RhoE/Rnd3 Antibody, clone 4 (Sigma-Aldrich) has been reported to show positive immunostaining for RhoE/Rnd3 at 1:50 dilution in 3T3/A31 cells fixed with paraformaldehyde and permeabilized with Triton X-100 .

    • For tissue sections, both immunofluorescence (IF) and immunohistochemistry (IHC) staining have successfully detected Rnd3 signals primarily in the cytoplasm of cortical brain cells in wild-type mice .

  • Detection recommendations:

    • For optimal results, cells should be fixed with paraformaldehyde and permeabilized with Triton X-100 for immunofluorescence applications.

    • Laboratory-specific optimization of antibody dilutions is recommended for each application and cell type.

    • For western blot analysis, RIPA lysates have been effectively used to extract RND3 protein .

What experimental models are best suited for studying RND3 function?

Several experimental models have proven valuable for investigating RND3 functions:

  • Cell line models:

    • HTR-8 cells (human trophoblast cell line) have been successfully used to study RND3's role in trophoblast proliferation, apoptosis, and migration through siRNA knockdown and overexpression approaches .

    • HCC cell lines have been utilized to investigate RND3's involvement in hepatocellular carcinoma, revealing its role in senescence and telomerase regulation .

    • 3T3/A31 cells serve as a positive antigen control for RND3 detection methods .

  • Genetic knockout models:

    • Rnd3-knockout mice generated from gene trap embryonic stem (ES) cell lines provide a valuable in vivo model. These mice exhibit phenotypes including aqueductal stenosis leading to hydrocephalus and heart calcium leakage resulting in heart failure .

    • For knockdown studies in cell lines, validated siRNA approaches targeting RND3 have been documented in the literature .

  • Culture conditions:

    • Both 2D and 3D culture conditions have been employed to study RND3 functions in cancer cells, with the 3D culture system providing a more physiologically relevant environment .

    • For in vivo validation, tumor xenograft models have successfully demonstrated the effects of RND3 silencing on tumor growth .

  • Experimental validation approaches:

    • Combining molecular techniques (western blot, PCR) with functional assays (proliferation, apoptosis, migration) provides comprehensive assessment of RND3 function.

    • Cell cycle analysis using flow cytometry has been effective in quantifying RND3's effects on cell cycle progression .

How can researchers effectively manipulate RND3 expression for functional studies?

Researchers have successfully employed several approaches to modulate RND3 expression:

  • RNA interference (RNAi):

    • siRNA targeting RND3 has been effectively used to knockdown expression in various cell types. This approach has revealed RND3's roles in regulating proliferation, with knockdown promoting cell proliferation in trophoblast cells .

    • Specific siRNA sequences validated for RND3 knockdown can be found in the literature, though they aren't explicitly provided in the search results.

  • Overexpression systems:

    • GFP-RND3 overexpression plasmids have been successfully employed with approximately 50% transfection efficiency when using GFP as a reporter gene .

    • Overexpression approaches have demonstrated that increased RND3 levels suppress cell proliferation by preventing S-phase entry and blocking G1 phase cell cycle progression .

  • Genetic knockout models:

    • For in vivo studies, Rnd3-knockout mice have been generated by inserting a targeting vector into RND3 intron 2 .

    • These knockout models allow for the assessment of RND3's physiological functions in intact organisms and have revealed its roles in brain apoptosis regulation .

  • Re-expression studies:

    • After RND3 knockdown, re-expression experiments have demonstrated that cells can bypass senescence and regain proliferative capacity, providing insights into the reversibility of RND3-mediated phenotypes .

  • Experimental validation:

    • qRT-PCR and western blot analysis should be performed to confirm successful manipulation of RND3 expression at both mRNA and protein levels.

    • Functional assays (proliferation, apoptosis, migration) should be conducted to assess the phenotypic consequences of altered RND3 expression.

What are the key considerations when studying RND3 in cancer contexts?

Researchers investigating RND3 in cancer should consider several important factors:

How might RND3 expression serve as a biomarker in human diseases?

RND3 shows promising potential as a biomarker in several human diseases:

  • Recurrent miscarriage (RM):

    • RND3 expression is significantly elevated in the cytotrophoblasts of patients with RM compared to normal villi .

    • This increased expression correlates with decreased proliferation and increased apoptosis of trophoblast cells, suggesting RND3 could serve as a diagnostic marker for RM risk.

    • Combined assessment of RND3 with its transcriptional regulator FOXD3 might provide enhanced predictive value for RM .

  • Hepatocellular carcinoma (HCC):

    • Downregulation of RND3 in HCC tissues compared to normal liver suggests potential utility as a diagnostic marker .

    • Low RND3 expression is associated with the presence of satellite nodules in HCC, indicating its potential as a prognostic marker for disease progression and metastatic potential .

  • Neurological disorders:

    • Given RND3's role in regulating brain apoptosis and its association with hydrocephalus in knockout models, altered RND3 expression could potentially serve as a biomarker for certain neurological conditions .

  • Biomarker development considerations:

    • Standardized detection methods (IHC, qPCR, western blot) with validated antibodies and primers are essential for reliable biomarker development.

    • Correlation with clinical outcomes in larger patient cohorts is necessary to establish the clinical utility of RND3 as a biomarker.

    • Tissue-specific expression patterns must be considered when developing RND3-based biomarkers.

What therapeutic opportunities exist for targeting RND3 or its pathways?

Several potential therapeutic strategies targeting RND3 or its pathways emerge from the research:

  • Pregnancy complications:

    • Modulating RND3 expression in trophoblasts could potentially address recurrent miscarriage. Since elevated RND3 is associated with reduced proliferation and increased apoptosis in trophoblasts from RM patients, strategies to normalize RND3 levels might improve pregnancy outcomes .

    • The identified FOXD3-RND3 regulatory axis offers a potential upstream target for therapeutic intervention .

  • Cancer therapeutics:

    • The complex role of RND3 in cancer necessitates context-specific approaches:

      • In cancers where RND3 functions as a tumor suppressor, strategies to restore or enhance its expression might be beneficial.

      • In contexts where RND3 promotes cancer progression, targeted inhibition could be therapeutic.

    • The transient senescence induced by RND3 knockdown in HCC cells suggests potential for senescence-inducing therapies, though careful consideration of potential senescence escape mechanisms is needed .

  • Pathway-based approaches:

    • Targeting downstream effectors of RND3 might provide therapeutic opportunities:

      • RhoA-ROCK1 pathway inhibitors could mimic RND3's effects on cell migration and proliferation.

      • Modulators of ERK1/2 signaling could influence RND3-mediated apoptosis regulation.

      • NF-κB pathway intervention might alter RND3's impact on inflammatory and apoptotic processes.

  • Development considerations:

    • Tissue-specific delivery systems would be crucial given RND3's varied roles across different tissues.

    • Therapeutic window identification is essential, as complete ablation of RND3 in animal models leads to developmental abnormalities including hydrocephalus and heart failure .

What are the critical knowledge gaps in RND3 biology that require further investigation?

Several significant knowledge gaps exist in our understanding of RND3 biology:

  • Tissue-specific functions:

    • While RND3's roles have been characterized in brain, placenta, and liver contexts, its functions in many other tissues remain poorly understood.

    • The apparently contradictory effects of RND3 on apoptosis in different cell types (promoting apoptosis in brain, inhibiting it in trophoblasts) require further investigation to understand the molecular basis for these context-dependent functions .

  • Post-translational regulation:

    • As an atypical GTPase that cannot cycle between active and inactive states, RND3 must be regulated through alternative mechanisms including post-translational modifications and protein-protein interactions that remain incompletely characterized.

    • The specific modifications that control RND3 stability, localization, and interactions in different cellular contexts require further study.

  • Transcriptional and epigenetic control:

    • While FOXD3 has been identified as a regulator of RND3 expression in trophoblasts , the broader transcriptional and epigenetic regulatory network controlling RND3 expression in different tissues remains to be fully elucidated.

  • Physiological roles in development:

    • The consequences of RND3 knockout in mice (hydrocephalus, heart failure) suggest important developmental functions , but the precise molecular mechanisms underlying these phenotypes require further investigation.

  • Interactions with other Rho family members:

    • How RND3 coordinates with or antagonizes other Rho GTPases in regulating cellular processes is not fully understood.

How might emerging technologies advance RND3 research?

Emerging technologies offer exciting opportunities to address knowledge gaps in RND3 research:

  • CRISPR/Cas9 genome editing:

    • Generation of conditional, tissue-specific knockout models would allow more precise dissection of RND3's functions while avoiding the developmental abnormalities observed in global knockout mice.

    • Precise genomic editing to introduce specific mutations or tags at the endogenous RND3 locus would facilitate the study of RND3 regulation and interactions under physiological conditions.

  • Single-cell technologies:

    • Single-cell RNA sequencing could reveal cell type-specific expression patterns and responses to RND3 modulation, particularly important given its context-dependent functions.

    • Single-cell proteomics approaches might identify cell-specific interaction partners and post-translational modifications.

  • Advanced imaging techniques:

    • Super-resolution microscopy combined with fluorescently tagged RND3 could provide insights into its dynamic subcellular localization and interactions.

    • Live-cell imaging of RND3 activity using biosensors would advance our understanding of its spatial and temporal regulation.

  • Proteomics approaches:

    • Comprehensive interactome analysis using proximity labeling techniques (BioID, APEX) could identify novel RND3 interaction partners in different cellular contexts.

    • Phosphoproteomics and other post-translational modification analyses would elucidate the regulatory modifications of RND3.

  • Systems biology integration:

    • Integration of transcriptomics, proteomics, and functional data through computational modeling could provide a more holistic understanding of RND3's role in cellular networks and signaling pathways.

Product Science Overview

Introduction

The Rho family of GTPases is a subfamily of the Ras superfamily of small (~21 kDa) signaling G proteins. These proteins play a crucial role in regulating various aspects of intracellular actin dynamics, which are essential for numerous cellular functions such as morphogenesis, polarity, movement, cell division, gene expression, and cytoskeleton reorganization .

Discovery and Classification

The identification of the Rho family of GTPases began in the mid-1980s. The first member, RhoA, was isolated in 1985, followed by Rac1 and Rac2 in 1989, and Cdc42 in 1990 . Over time, additional members were identified, leading to the classification of the Rho family into 20 members distributed across 8 subfamilies: Rho, Rnd, RhoD/F, RhoH, Rac, Cdc42, RhoU/V, and RhoBTB .

Structure and Function

Rho GTPases act as molecular switches by cycling between an inactive GDP-bound state and an active GTP-bound state. This switching mechanism allows them to interact with various downstream effectors, thereby regulating a multitude of biochemical processes . The Rho family members are involved in the formation of cellular projections such as filopodia, lamellipodia, and stress fibers, which are critical for cell movement and morphology .

Rho Family GTPase 3 (RhoG)

Rho Family GTPase 3, also known as RhoG, is one of the members of the Rho family. It shares structural and functional similarities with other Rho GTPases and is involved in various cellular processes. RhoG has been shown to regulate actin cytoskeleton dynamics, cell migration, and cell signaling pathways .

Human Recombinant RhoG

Human recombinant RhoG is a laboratory-produced version of the naturally occurring RhoG protein. Recombinant proteins are typically produced using genetic engineering techniques, where the gene encoding the protein is inserted into an expression system, such as bacteria or yeast, to produce large quantities of the protein. This recombinant form is used in various research applications to study the protein’s function, interactions, and potential therapeutic uses.

Significance in Research

RhoG and other Rho GTPases are of significant interest in biomedical research due to their roles in various cellular processes and their association with human diseases. Altered signaling through Rho GTPases has been implicated in the progression of several malignancies, making them potential targets for therapeutic intervention . Understanding the regulation and function of RhoG can provide insights into the mechanisms underlying these diseases and aid in the development of targeted therapies.

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.