RAC1 Human, His

Ras-Related C3 Botulinum Toxin substrate 1 Human Recombinant, His Tag
Shipped with Ice Packs
In Stock

Description

Introduction to RAC1 Human, His

RAC1 Human, His is a recombinant human Rac1 protein engineered with a histidine (His) tag at the N-terminus, enabling purification via nickel-affinity chromatography. This protein is widely used in biochemical assays to study Rac1’s role in signaling pathways, cytoskeletal dynamics, and disease mechanisms. Below is a detailed analysis of its technical specifications, biological activity, and research applications.

Biological Activity and Validation

RAC1 Human, His retains functional activity critical for GTPase assays and effector binding studies:

Assay TypeMethodResultSource
GTP BindingGST-PAK-PBD pulldown with GTPγS/GDP-loaded Rac1>80% active GTP-bound form detected
Nucleotide ExchangeAssays with guanine nucleotide exchange factors (GEFs)Supports dynamic GTP/GDP cycling
Effector BindingInteraction with PAK, WAVE, and other Rac1-binding proteinsConfirmed via co-IP or pulldown

Mechanism of Action:

  • GTPase Activity: Hydrolyzes GTP to GDP, regulated by GEFs and GAPs.

  • Effector Recruitment: Binds downstream targets (e.g., PAK, WAVE2) to modulate cytoskeletal reorganization .

Role in Cancer Progression

Study FocusKey FindingsSource
Colorectal AdenocarcinomaRac1 overexpression accelerates tumor growth; inhibition suppresses metastasis
Head and Neck Cancer (HNSCC)RAC1 amplification/A159V mutation drives PI3K pathway activation; sensitive to EHop-016 inhibitor
LeukemiaRac1 inhibition induces apoptosis in leukemic cells (e.g., ZINC69391, 1A-116)

Vascular and Metabolic Studies

ApplicationOutcomeSource
Endothelial DysfunctionRac1 inhibition reduces oxidative stress and improves NO production in veins
Glucose UptakeRac1 regulates GLUT4 translocation in insulin-sensitive tissues (e.g., muscle)

Drug Targeting

  • EHop-016: A Rac inhibitor showing efficacy in RAC1-amplified and A159V-mutated HNSCC xenografts .

  • Constitutively Active Mutant (Q61L): Used to study oncogenic signaling in vitro (e.g., sustained GTP binding) .

Table 1: RAC1 Human, His in Disease Models

DiseaseModelRAC1 ModificationOutcomeSource
HNSCCPDX modelsAmplification/A159VSensitivity to EHop-016
Colorectal CancerSW620 cell line (orthotopic)OverexpressionAccelerated tumor growth
LeukemiaPatient-derived cellsWild-typeApoptosis induced by Rac inhibitors

Table 2: Comparative Analysis of RAC1 Human, His Products

VendorMolecular WeightPurityKey Applications
Cytoskeleton, Inc. (RC01)22 kDa>90%GTPase assays, nucleotide exchange studies
Prospec Bio (PRO-731)23.6 kDa>90%Effector binding, structural studies

Product Specs

Introduction
RAC1, a member of the RAS superfamily of small GTP-binding proteins, functions as a GTPase. This superfamily plays a crucial role in regulating various cellular processes, including cell growth, cytoskeletal reorganization, and protein kinase activation. Specifically, RAC1 regulates the actin cytoskeleton and participates in cell-cell adhesion regulation.
Description
Recombinant Human Ras-Related C3 Botulinum Toxin substrate 1, expressed in E. coli, is a non-glycosylated polypeptide chain. It consists of 212 amino acids (1-192 a.a.) with a 20 amino acid His Tag at the N-terminus, resulting in a molecular weight of 23.6 kDa. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
The RAC1 His Tag protein is supplied in a solution containing 20mM Tris-HCl (pH 7.5), 1mM EDTA, 10% glycerol, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For longer storage, freeze at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms
P21-RAC1, RAC-1, RAC1, RAS-like protein TC25, MIG5, Cell-migration-inducing gene 5 protein,Ras-related C3 botulinum toxin substrate 1, rho family small GTP binding protein Rac1, TC-25, MGC111543.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MQAIKCVVVG DGAVGKTCLL ISYTTNAFPG EYIPTVFDNY SANVMVDGKP VNLGLWDTAG QEDYDRLRPLSYPQTDVFLI CFSLVSPASF ENVRAKWYPE VRHHCPNTPI ILVGTKLDLR DDKDTIEKLK EKKLTPITYP QGLAMAKEIG AVKYLECSALTQRGLKTVFD EAIRAVLCPP PVKKRKRKCL LL.

Q&A

What is RAC1 and what cellular functions does it regulate?

RAC1 is a member of the Rac family of GTPases, a subfamily of the Rho family of small GTPases, best known for regulating the cytoskeleton and gene expression. Since its discovery, RAC1 has been implicated in numerous cellular functions including cellular plasticity, migration and invasion, cellular adhesions, cell proliferation, apoptosis, reactive oxygen species (ROS) production, and inflammatory responses . These processes are central to normal cell physiology, but when deregulated, RAC1 signaling can contribute to pathological conditions including cancer, cardiovascular diseases, and neurodegenerative disorders .
The protein functions as a molecular switch, cycling between inactive GDP-bound and active GTP-bound states. In its active form, RAC1 interacts with various effector proteins to initiate downstream signaling cascades that ultimately regulate cellular processes. This activation is tightly controlled by guanine nucleotide exchange factors (GEFs), GTPase-activating proteins (GAPs), and guanine nucleotide dissociation inhibitors (GDIs) .

How does His-tagged RAC1 differ from native RAC1 in research applications?

  • The tag may potentially affect certain protein-protein interactions, particularly if these interactions involve the N-terminus

  • For structural studies, the His-tag might introduce conformational changes

  • When using in cellular systems, the tag could potentially influence subcellular localization
    Quality control is essential when working with His-tagged RAC1. Commercial preparations should be verified for purity (typically >90% as determined by SDS-PAGE) and functionality (through effector binding assays) .

What are the key RAC1 mutants used in experimental research and their properties?

Several RAC1 mutants serve as valuable tools in experimental research, each with distinct properties that facilitate the investigation of specific aspects of RAC1 biology:

MutantMutationFunctional EffectResearch Applications
RAC1(Q61L)Glutamine to leucine at position 61Constitutively active; prevents GTPase activityStudying effects of sustained RAC1 activation; effector binding studies; phenotype induction
RAC1(G12V)Glycine to valine at position 12Constitutively active; similar to Q61LAlternative for studying constitutive activation
RAC1(T17N)Threonine to asparagine at position 17Dominant negative; preferentially binds GDPStudying effects of RAC1 inhibition; blocking endogenous RAC1 activation
RAC1(F28L)Phenylalanine to leucine at position 28Fast-cycling mutant; rapid GDP/GTP exchangeStudying effects of accelerated cycling between active/inactive states
The RAC1(Q61L) mutant is particularly significant because the leucine substitution prevents endogenous and GAP-stimulated GTPase activity, keeping RAC1 in the active, GTP-bound state . This mutation parallels oncogenic mutations found in other small GTPases like RAS. RAC1(Q61L) is invaluable for activating RAC1 effectors both in vitro and in vivo and can be microinjected into cells to stimulate RAC1-dependent phenotypes .

What methods are available for detecting active RAC1 in experimental systems?

Researchers can employ several techniques to measure active RAC1 (RAC1-GTP) in experimental systems:

  • Pull-down assays: This gold standard method uses the p21-binding domain (PBD) of p21-activated kinase (PAK), which specifically binds active RAC1-GTP . The bound active RAC1 is then detected by Western blotting. Commercial PAK-PBD beads are available for this purpose.

  • Antibody-based detection: Anti-RAC1-GTP antibodies have been widely used for:

    • Immunofluorescence

    • Proximity ligation assays

    • Immunohistochemistry

    • Flow cytometry

    • Immunoprecipitation
      IMPORTANT CAUTION: Recent research has questioned the specificity of commercially available anti-RAC1-GTP antibodies. Evidence suggests some of these antibodies may actually detect vimentin rather than RAC1-GTP . The antibody staining colocalizes with vimentin, potentially leading to misinterpretation of results, particularly when studying mesenchymal cancer phenotypes .

  • FRET-based biosensors: These allow real-time visualization of RAC1 activation in living cells, though they require overexpression of sensor constructs.

  • Analysis of downstream effector phosphorylation: Examining the phosphorylation state of RAC1 effectors such as PAK can provide indirect evidence of RAC1 activation.
    When selecting a method, researchers should implement appropriate controls, such as using RAC1 knockout cells or constitutively active/dominant negative RAC1 mutants to validate their approach.

How is RAC1 implicated in neurodegenerative disorders like Alzheimer's disease?

RAC1 dysregulation plays a significant role in neurodegenerative disorders, particularly Alzheimer's disease (AD), as evidenced by multiple lines of research:

  • Altered expression in AD brain tissue: Analysis of human brain samples reveals that RAC1 mRNA expression is significantly downregulated in the entorhinal cortex (EC) of AD patients (FDR-adjusted P-value = 0.046) . This reduction shows a regional specificity, being statistically significant across Braak NFT stages specifically in the EC, but not in the temporal cortex (TC) or frontal cortex (FC) .

  • Relationship to disease progression: The reduction in RAC1 expression correlates with disease progression, with greater reductions seen in more advanced Braak NFT stages . Importantly, this reduction is not simply correlated with aging but specifically linked to AD pathology.

  • Functional consequences of RAC1 reduction: Using Drosophila as an in vivo model, researchers demonstrated that neuronal knockdown of Rac1 (which shares 83% identity and 87% similarity with human RAC1) causes:

    • Age-dependent locomotor deficits (manifesting after 28 days)

    • Neurodegeneration

    • These effects occur without affecting lifespan, indicating specific neuronal dysfunction rather than general toxicity

  • Regulatory mechanism - miRNA involvement: The miRNA hsa-miR-101-3p shows the strongest negative correlation with RAC1 expression in AD brains (Spearman correlation coefficient = -0.501, P-value = 6.66 × 10^-5) . This miRNA:

    • Increases with progressive Braak NFT stage specifically in the EC

    • Experimentally suppresses RAC1 expression when overexpressed in human neuronal cells

    • Has been previously linked to AD through effects on amyloid-β precursor protein
      These findings suggest a pathway where increased expression of hsa-miR-101-3p leads to RAC1 downregulation, contributing to neurodegeneration in AD. This makes RAC1 signaling a potential therapeutic target for preventing or treating neurodegeneration, possibly through approaches aimed at restoring normal RAC1 levels or activity in affected brain regions.

What role does RAC1 play in cancer progression and metastasis?

RAC1 has emerged as a critical regulator of cancer progression and metastasis, with particularly compelling evidence in tongue squamous cell carcinoma (TSCC):

How do microRNAs regulate RAC1 expression and what are the implications for disease mechanisms?

MicroRNAs (miRNAs) represent a sophisticated regulatory mechanism controlling RAC1 expression, with significant implications for disease pathogenesis:

  • Identification and validation of RAC1-targeting miRNAs:

    • Comprehensive analysis using the starBase database identified 62 miRNAs that potentially bind to RAC1 mRNA

    • Nine miRNAs showed negative correlation with RAC1 gene expression in brain samples

    • hsa-miR-101-3p demonstrated the strongest negative correlation (Spearman correlation coefficient = -0.501, P-value = 6.66 × 10^-5)

    • Experimental validation in human neuronal SH-SY5Y cells confirmed that overexpression of hsa-miR-101-3p significantly decreases RAC1 gene expression (P-value = 3.32 × 10^-3)

  • Disease-specific miRNA-RAC1 interactions:

    • In Alzheimer's disease:

      • hsa-miR-101-3p expression increases with progressive Braak NFT stage specifically in the entorhinal cortex

      • This correlates inversely with decreased RAC1 expression in the same region

      • The SCC between RAC1 and hsa-miR-101-3p showed negative correlation in all brain regions examined: entorhinal cortex, temporal cortex, and frontal cortex

      • hsa-miR-101-3p is differentially expressed in AD serum, suggesting potential biomarker value

      • Both precursor and mature forms of miR-101 regulate amyloid-β precursor protein expression

  • Broader implications in other diseases:

    • miR-101 has been implicated in:

      • Innate immune responses in macrophages

      • Hepatocellular carcinoma pathogenesis

      • Other cancer types where RAC1 signaling is dysregulated

  • Therapeutic potential based on miRNA regulation:

    • Anti-miR strategies could potentially restore RAC1 levels in conditions like AD where RAC1 is downregulated

    • miRNA mimics might reduce RAC1 expression in cancers where RAC1 is overexpressed

    • miRNA-based biomarkers could help identify patients with RAC1 dysregulation
      The identification of miRNAs as regulators of RAC1 expression provides new insights into disease mechanisms and suggests novel therapeutic approaches. For neurodegenerative disorders like AD, strategies to inhibit specific miRNAs (such as hsa-miR-101-3p) could potentially restore RAC1 levels and function. Conversely, for cancers where RAC1 is overexpressed, miRNA mimics could help reduce RAC1 expression and limit disease progression.

What are the challenges and opportunities in targeting RAC1 therapeutically?

Targeting RAC1 therapeutically presents both significant challenges and promising opportunities:

  • Key challenges in RAC1 inhibition:

    • RAC1 is involved in numerous essential physiological processes, raising concerns about on-target toxicity

    • As a GTPase, RAC1 lacks deep binding pockets that typically facilitate small molecule drug development

    • High homology between RAC1 and other Rho GTPases makes achieving selectivity difficult

    • RAC1 has complex regulatory mechanisms involving multiple proteins (GEFs, GAPs, and GDIs) that complicate intervention strategies

  • Current therapeutic approaches:
    a. Direct RAC1 inhibitors:

    • NSC23766: Prevents RAC1-GEF interaction

    • EHop-016: Blocks RAC1 activation with higher potency than NSC23766

    • AZA1: Derivative of NSC23766 with improved potency
      b. Targeting post-translational modifications:

    • Statins: Inhibit prenylation required for RAC1 membrane localization

    • Cerivastatin: Shows anti-metastatic effects through RAC1 inhibition
      c. Targeting RAC1 regulators:

    • P-REX1 inhibitors: Block this GEF that activates RAC1 in cancer

    • Tiam1 inhibitors: Target another important RAC1 GEF
      d. Targeting downstream effectors:

    • PAK inhibitors: Block p21-activated kinases, key RAC1 effectors

    • WAVE/Arp2/3 inhibitors: Disrupt cytoskeletal remodeling downstream of RAC1

  • Disease-specific therapeutic considerations:

    • For cancer: Inhibition approaches may be appropriate, as RAC1 is often overexpressed or hyperactivated

    • For neurodegenerative disorders: Activation or restoration of RAC1 signaling might be beneficial, as RAC1 is often downregulated

    • Tissue-specific delivery systems could help mitigate systemic toxicity concerns

  • Emerging and future directions:

    • Structure-based drug design to identify novel druggable pockets in RAC1

    • Development of proteolysis-targeting chimeras (PROTACs) for RAC1

    • RNA therapeutics targeting RAC1 or its regulators (e.g., miRNA-based approaches)

    • Combination approaches targeting multiple nodes in RAC1 signaling networks
      Given the adverse effects associated with RAC1 signaling perturbation, a deeper understanding of the cellular mechanisms regulating RAC1 activity is essential for developing effective therapeutic strategies . The ideal approach would restore normal RAC1 signaling rather than simply blocking its activity, potentially requiring disease-specific and even patient-specific interventions.

What are the optimal conditions for using His-tagged RAC1 proteins in binding studies?

For optimal results with His-tagged RAC1 proteins in binding studies, researchers should adhere to specific handling and experimental conditions:

  • Protein preparation and storage:

    • Reconstitute lyophilized His-RAC1 in distilled water to a concentration of 1 mg/ml

    • After reconstitution, the protein should be in buffer containing 2 mM Tris pH 7.6, 0.5 mM MgCl₂, 0.5% sucrose, and 0.1% dextran

    • Store aliquots at -70°C to avoid freeze-thaw cycles

    • For short-term storage (1-2 weeks), 4°C is acceptable

    • Protein concentration can be determined using the Precision Red Advanced Protein Assay Reagent

  • Critical assay considerations:

    • For studies with effector proteins like PAK, use PAK-PBD beads for pull-down assays

    • Maintain Mg²⁺ in all buffers (typically 0.5-5 mM) as it is essential for nucleotide binding

    • Include appropriate controls:

      • Wild-type RAC1 loaded with GDP (inactive control)

      • Wild-type RAC1 loaded with non-hydrolyzable GTP analogs (active control)

      • For constitutively active RAC1(Q61L), compare with wild-type protein

  • Protein quality assessment:

    • Verify protein purity using SDS-PAGE (commercial preparations should be >90% pure)

    • His-RAC1(Q61L) has a molecular weight of approximately 25 kDa on SDS-PAGE

    • For activity verification, use a functional assay such as PAK-PBD binding

  • Applications and specifications:

    • RAC1(Q61L) is particularly useful for:

      • RAC1 effector identification and binding studies

      • GAP binding studies

      • Activation of RAC1 effectors in vitro

      • Microinjection into cells to stimulate RAC1 phenotypes

    • The constitutively active form contains a glutamine to leucine substitution at residue 61, preventing endogenous and GAP-stimulated GTPase activity
      By maintaining these conditions, researchers can ensure optimal protein functionality and minimize experimental artifacts when working with His-tagged RAC1 proteins in binding and functional studies.

How should researchers validate the specificity of anti-RAC1-GTP antibodies?

Given recent concerns about the specificity of anti-RAC1-GTP antibodies, rigorous validation is essential before using these reagents in research:

  • Essential genetic controls:

    • Test antibody staining in RAC1 knockout cells generated using CRISPR/Cas9

    • Compare with isogenic wild-type cells

    • Any persistent signal in knockout cells indicates non-specific binding

    • This approach revealed that some anti-RAC1-GTP antibodies may actually detect vimentin rather than active RAC1

  • Biochemical validation:

    • Serum-starve cells to reduce basal RAC1 activation

    • Stimulate cells with known RAC1 activators (e.g., EGF, PDGF)

    • Treat cells with RAC1 inhibitors (e.g., NSC23766)

    • Signal changes should correlate with activation status as confirmed by PAK-PBD pulldown assays

  • Vimentin cross-reactivity assessment:

    • Perform dual immunofluorescence with anti-RAC1-GTP and anti-vimentin antibodies

    • Assess colocalization using confocal microscopy

    • Studies have shown that anti-RAC1-GTP antibody staining colocalizes with vimentin

    • Compare staining in cell lines characterized with low and high RAC1-GTP levels, as determined by PAK-PBD pulldown assays

  • Cell phenotype correlation analysis:

    • Be aware that some antibodies may show correlation with mesenchymal cancer cell phenotype rather than actual RAC1 activation

    • Compare staining in epithelial versus mesenchymal cell types

    • Assess whether signal correlates with epithelial-mesenchymal transition (EMT) markers

  • Alternative validation approaches:

    • Peptide competition assays to test specificity

    • Expression of constitutively active RAC1(Q61L) versus dominant negative RAC1(T17N)

    • Use of multiple antibodies from different sources and clones
      The widespread use of anti-RAC1-GTP antibodies in various applications including immunofluorescence, proximity ligation, immunohistochemistry, flow cytometry, and immunoprecipitation underscores the importance of proper validation. Researchers should be cautious when interpreting results obtained with these antibodies, particularly in contexts involving mesenchymal cell types or EMT processes.

What methodologies are recommended for analyzing RAC1 activity in patient-derived samples?

Analyzing RAC1 activity in patient-derived samples requires specialized approaches to overcome challenges related to sample preservation and heterogeneity:

  • Critical sample processing considerations:

    • Process fresh samples rapidly (within 30 minutes of collection) to preserve RAC1 activation status

    • Use buffers containing protease inhibitors, phosphatase inhibitors, and GTPase inhibitors

    • Maintain consistent temperature (4°C) during processing to minimize artificial GTPase activity changes

    • For tissue banking, flash freezing in liquid nitrogen is preferred over chemical fixation

  • Recommended analytical methods:
    a. Gold standard: PAK-PBD pulldown assays:

    • Most reliable for quantitative assessment of RAC1-GTP levels

    • Requires sufficient fresh/frozen tissue (typically >50mg)

    • Western blot detection of pulled-down RAC1

    • Include positive controls (samples treated with GTPγS) and negative controls (GDP-loaded)
      b. Antibody-based approaches (with important caveats):

    • CRITICAL CAUTION: Research has demonstrated that some commercial anti-RAC1-GTP antibodies may cross-react with vimentin rather than detecting active RAC1

    • If using immunohistochemistry, include parallel vimentin staining

    • Compare results with PAK-PBD pulldowns whenever possible

    • Consider using phospho-specific antibodies against RAC1 effectors (e.g., phospho-PAK) as indirect markers

  • Controls and validation strategies:

    • Use paired normal adjacent tissue as internal control when possible

    • Include cell line standards with known RAC1 activation levels

    • Consider employing RAC1 knockout or knockdown controls in parallel experiments

    • Validate findings with multiple methodological approaches

  • Molecular correlates and alternative approaches:

    • Analyze RAC1 mRNA expression levels

    • Examine expression of known RAC1-targeting miRNAs (e.g., hsa-miR-101-3p)

    • Assess RAC1 regulators (GEFs, GAPs) and effectors

    • Consider single-cell approaches to address tumor heterogeneity

  • Clinical-pathological correlation:

    • Integrate RAC1 activation data with clinical parameters

    • In cancer samples, correlate with invasion, metastasis, and patient outcomes

    • In neurodegenerative disorders, correlate with disease progression markers (e.g., Braak staging)
      These approaches enable more reliable assessment of RAC1 activation status in patient samples, potentially identifying clinically relevant patterns that could inform diagnosis, prognosis, or treatment selection.

What are the most effective strategies for manipulating RAC1 activity in cellular models?

Researchers can employ multiple strategies to manipulate RAC1 activity in cellular models, each offering distinct advantages for specific experimental applications:

  • Genetic manipulation approaches:
    a. Expression of RAC1 variants:

    • Constitutively active RAC1(Q61L): Prevents GTP hydrolysis, maintaining RAC1 in active state

    • Dominant negative RAC1(T17N): Sequesters GEFs, blocking endogenous RAC1 activation

    • Fast-cycling RAC1(F28L): Exhibits rapid GDP/GTP exchange

    • Consider using inducible expression systems (Tet-on/off) for temporal control
      b. Gene silencing methods:

    • siRNA/shRNA: For temporary reduction of RAC1 expression

    • CRISPR/Cas9: For complete knockout

    • Verify knockdown/knockout efficiency via Western blot

    • Analyze potential compensatory upregulation of related GTPases

  • Pharmacological modulators:
    a. Direct RAC1 inhibitors:

    • NSC23766: Blocks RAC1-GEF interaction (50-100 μM)

    • EHop-016: More potent derivative (1-5 μM)

    • Consider time-course studies to determine optimal treatment duration
      b. Pathway modulators:

    • PI3K inhibitors: Reduce activity of RAC1 GEFs

    • Statins: Impair prenylation and membrane localization

    • Growth factors (EGF, PDGF): Stimulate RAC1 activation

  • Advanced manipulation strategies:
    a. Optogenetic approaches:

    • Photoactivatable RAC1 (PA-RAC1): Enables spatiotemporal control of activation

    • Light-inducible GEF recruitment systems

    • Allows subcellular manipulation of RAC1 activity
      b. miRNA-based approaches:

    • Transfection with miRNA mimics (e.g., hsa-miR-101-3p) to reduce RAC1 expression

    • Anti-miR strategies to increase RAC1 expression

    • Stable expression of miRNA sponges for long-term effects

  • Validation and monitoring strategies:
    a. Activity assessment:

    • PAK-PBD pulldown assays to confirm manipulation of RAC1-GTP levels

    • Phenotypic validation (lamellipodia formation, migration assays)

    • Phosphorylation of downstream effectors (PAK, LIMK)
      b. Live-cell monitoring:

    • FRET-based biosensors for real-time visualization

    • Combined manipulation and activity sensing

    • Analysis of subcellular activation patterns
      These diverse approaches provide researchers with a comprehensive toolkit for manipulating RAC1 activity in cellular models. The choice of method should be guided by the specific research question, required temporal and spatial resolution, and available resources. Combining multiple approaches can provide more robust evidence for RAC1's role in the biological process under investigation.

What emerging technologies are advancing RAC1 research?

Recent technological innovations are significantly enhancing our ability to study RAC1 function and regulation with unprecedented precision:

  • Advanced imaging technologies:

    • Super-resolution microscopy techniques (PALM, STORM, SIM) enable visualization of RAC1 localization and activation at nanoscale resolution

    • Light-sheet microscopy allows long-term 3D imaging of RAC1 dynamics in developing tissues and organoids

    • Correlative light and electron microscopy (CLEM) connects RAC1 activation patterns with ultrastructural features

  • Spatiotemporal control systems:

    • Optogenetic RAC1 tools enable precise activation or inhibition with subcellular specificity

    • Improved photoactivatable RAC1 variants with reduced dark-state activity

    • Dual-wavelength systems allowing simultaneous manipulation of multiple pathways

  • Single-cell analysis approaches:

    • Single-cell RNA sequencing reveals cell-specific RAC1 regulatory networks

    • Mass cytometry (CyTOF) for high-dimensional analysis of RAC1 pathway components

    • Spatial transcriptomics and proteomics for mapping RAC1 expression and activity in tissue context

  • Structural and interaction analysis:

    • Cryo-EM structures of RAC1 in complex with regulators and effectors

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) for analyzing RAC1 conformational dynamics

    • Proximity labeling approaches (BioID, APEX) to map the RAC1 interactome in living cells

  • Genome editing innovations:

    • Base editing and prime editing for introducing precise RAC1 mutations without double-strand breaks

    • CRISPR activation/inhibition systems for modulating endogenous RAC1 expression

    • CRISPR screens targeting RAC1 pathway components
      These emerging technologies are driving rapid advances in our understanding of RAC1 biology and providing new opportunities for developing more effective therapeutic strategies targeting RAC1 and its regulatory pathways.

How is our understanding of RAC1 regulation evolving?

Our understanding of RAC1 regulation is expanding beyond the classical GTPase cycle to encompass multiple layers of control:

  • Post-translational modifications:

    • Phosphorylation at specific residues can alter RAC1 activity and subcellular localization

    • SUMOylation influences RAC1 stability and nuclear translocation

    • Ubiquitination regulates RAC1 degradation and recycling

    • Prenylation is essential for membrane association and function

  • Spatiotemporal regulation:

    • Subcellular compartmentalization restricts RAC1 activity to specific locations

    • Membrane microdomains serve as signaling platforms for RAC1 activation

    • Vesicular trafficking modulates RAC1 availability at different cellular locations

    • Nuclear RAC1 functions are increasingly recognized as important in gene regulation

  • Metabolic control:

    • Cellular energy status affects RAC1 activation through AMPK signaling

    • Redox regulation influences RAC1 function through oxidation of critical cysteine residues

    • Lipid metabolism impacts RAC1 membrane association and signaling capabilities

  • Non-coding RNA regulation:

    • miRNAs like hsa-miR-101-3p directly regulate RAC1 expression levels

    • Long non-coding RNAs can act as miRNA sponges, indirectly affecting RAC1 expression

    • Circular RNAs emerging as additional regulators of RAC1 signaling networks

  • Crosstalk with other signaling pathways:

    • Reciprocal regulation between RAC1 and Rho/Cdc42 coordinates cytoskeletal dynamics

    • Integration with growth factor signaling determines cell fate decisions

    • Inflammatory pathways interact with RAC1 to regulate immune cell function
      This evolving understanding highlights the complexity of RAC1 regulation and explains why dysregulation can contribute to diverse pathologies from cancer to neurodegeneration. It also suggests multiple intervention points for therapeutic targeting beyond direct RAC1 inhibition.

What significant gaps remain in our knowledge of RAC1 biology?

Despite significant advances, several critical knowledge gaps persist in RAC1 biology that limit our ability to fully exploit it as a therapeutic target:

  • Isoform-specific functions:

    • The unique roles of RAC1, RAC2, and RAC3 remain incompletely defined

    • Splicing variants (particularly RAC1b) have distinct functions that need further characterization

    • Isoform-specific interactomes are poorly mapped

  • Tissue-specific regulatory mechanisms:

    • How RAC1 regulation differs across tissues is not well understood

    • The basis for tissue-specific vulnerability to RAC1 dysregulation requires investigation

    • Developmental stage-specific functions of RAC1 need further clarification

  • Dynamic RAC1 signaling:

    • The temporal dynamics of RAC1 activation in physiological contexts remain difficult to measure

    • How cells interpret different patterns of RAC1 activation (pulsatile vs. sustained) is unclear

    • The mechanisms restoring homeostasis after RAC1 perturbation are poorly defined

  • RAC1 in disease pathogenesis:

    • Whether RAC1 alterations are drivers or consequences in many diseases remains debated

    • The genetic and environmental factors predisposing to RAC1 dysregulation are not fully characterized

    • Patient-specific differences in RAC1 signaling that affect disease progression or treatment response need exploration

  • Therapeutic targeting challenges:

    • Strategies for achieving tissue-specific RAC1 modulation remain limited

    • Methods to restore normal RAC1 function (rather than simple inhibition) are underdeveloped

    • Biomarkers to identify patients likely to benefit from RAC1-targeted therapies are lacking
      Addressing these knowledge gaps will require interdisciplinary approaches combining structural biology, systems biology, genetic models, and clinical studies. Progress in these areas promises to unlock new therapeutic opportunities for the many diseases associated with RAC1 dysregulation.

How might RAC1-targeted therapies evolve in the coming years?

The therapeutic landscape for RAC1-targeted interventions is poised for significant evolution:

  • Precision targeting approaches:

    • Structure-guided design of inhibitors targeting specific conformational states of RAC1

    • Allosteric modulators that fine-tune rather than completely block RAC1 function

    • Context-dependent degraders (PROTACs) that selectively remove RAC1 in disease contexts

    • Disruption of specific RAC1-effector interactions while preserving others

  • Disease-tailored strategies:

    • For neurodegenerative disorders: RAC1 activators or miRNA inhibitors to restore downregulated RAC1

    • For cancers: Inhibitors targeting specific oncogenic RAC1 signaling nodes

    • For inflammatory diseases: Modulators targeting RAC1's role in immune cell function

    • Combination approaches targeting multiple points in the RAC1 pathway

  • Advanced delivery systems:

    • Tissue-specific delivery to minimize off-target effects

    • Cell-penetrating peptides that disrupt specific RAC1 interactions

    • Nanoparticle formulations for enhanced bioavailability

    • Blood-brain barrier penetrating formulations for neurodegenerative applications

  • Nucleic acid therapeutics:

    • Antisense oligonucleotides targeting RAC1 or its regulators

    • miRNA mimics or antagonists to modulate RAC1 expression

    • mRNA therapeutics for temporary RAC1 pathway modulation

    • CRISPR-based in vivo editing of RAC1 pathway components

  • Personalized medicine approaches:

    • Patient-specific analysis of RAC1 pathway alterations to guide treatment selection

    • Biomarker development to predict response to RAC1-targeted therapies

    • Companion diagnostics measuring RAC1 activation status

    • Real-time monitoring of treatment response using circulating markers These emerging approaches reflect a shift from blunt inhibition to sophisticated modulation of RAC1 signaling, potentially overcoming the limitations of current strategies. The ideal RAC1-targeted therapeutics will restore normal signaling balance rather than simply blocking activity, minimizing side effects while maximizing disease-specific benefits.

Product Science Overview

Introduction

Ras-related C3 botulinum toxin substrate 1, commonly known as Rac1, is a small (~21 kDa) signaling G protein and a member of the Rac subfamily of the Rho family of GTPases. It is encoded by the RAC1 gene in humans . Rac1 plays a crucial role in various cellular processes, including cell growth, cytoskeletal reorganization, and the activation of protein kinases .

Structure and Function

Rac1 is a pleiotropic regulator involved in many cellular processes such as the cell cycle, cell-cell adhesion, motility, and epithelial differentiation . It is particularly important in the formation of lamellipodia, which are essential for cell motility . Rac1 is ubiquitously expressed in tissues and is vital for maintaining epidermal stem cells .

Role in Disease

Rac1 has been implicated in several diseases, particularly cancer and diabetic nephropathy. In cancer, Rac1 is known to drive cell motility, which is a hallmark of cancer cell invasion and metastasis . Overexpression of Rac1 has been linked to tumor formation and metastasis . In diabetic nephropathy, Rac1 activation accelerates renal damage through the activation of the MLK3-MKK7-JNK signaling module .

Recombinant Rac1 (Human, His Tag)

Recombinant Rac1 proteins are often tagged with a His (histidine) tag to facilitate purification and detection. The His tag is a string of histidine residues that binds to nickel ions, allowing the protein to be easily isolated using nickel-affinity chromatography. This recombinant form is used in various research applications to study the protein’s function and its role in disease.

Research Applications

Recombinant Rac1 is widely used in biochemical and cellular studies to understand its role in signaling pathways. It is also used in drug discovery research to identify potential inhibitors that could modulate Rac1 activity and provide therapeutic benefits in diseases like cancer and diabetic nephropathy .

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.