RRAS Antibody, HRP conjugated

Shipped with Ice Packs
In Stock

Description

Definition and Mechanism

  • HRP Conjugation: HRP is a 44 kDa glycoprotein tagged to antibodies to amplify signal detection. It reacts with substrates like diaminobenzidine (DAB) or TMB to produce visible precipitates or colorimetric changes .

  • RRAS Protein: RRAS (Ras-related protein) regulates cell adhesion, integrin activation, and signaling pathways involving PI3K and RAF . It is expressed in immune cells, neurons, and cancer tissues .

Detection Techniques

TechniqueDescriptionCitation
ELISAQuantitative detection of RRAS in lysates or serum .
Western BlottingIdentification of RRAS in cell lysates (23 kDa band) .
Immunohistochemistry (IHC)Localization of RRAS in tissue sections (e.g., gliomas, kidney) .
Immunoprecipitation (IP)Purification of RRAS-protein complexes .

Research Focus

  • Cancer Biology: RRAS influences tumor growth and metastasis by modulating integrin signaling .

  • Immunology: Required for dendritic cell maturation and T-cell activation .

  • Neuroscience: Regulates neural activity and cytoskeletal organization .

Cancer Studies

  • Glioblastoma: RRAS overexpression correlates with tumor invasion and poor prognosis .

  • Breast Cancer: RRAS knockdown inhibits cell migration via PI3K/AKT pathway disruption .

Immunological Roles

  • Dendritic Cells: RRAS-deficient cells show reduced surface MHC II and impaired T-cell synapse formation .

  • Neuroinflammation: RRAS regulates microglial activation in Alzheimer’s disease models .

Technical Considerations

  • Conjugation Kits: HRP tagging can be performed using Lightning-Link® kits, avoiding buffer interference .

  • Optimization: Dilutions (1:100–1:200 for IHC; 1:1000–1:4000 for WB) require titration .

Table 2: Research Highlights

StudyFindingsCitation
R-Ras in dendritic cellsImpaired maturation and T-cell activation .
RRAS in glioblastomaPromotes invasion via PI3K/AKT signaling .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please consult your local distributor for specific delivery timeframes.
Synonyms
Oncogene RRAS antibody; p23 antibody; R Ras antibody; Ras related Protein antibody; Ras related protein R Ras antibody; Ras-related protein R-Ras antibody; related RAS viral (r ras) oncogene homolog antibody; RRAS antibody; RRAS_HUMAN antibody
Target Names
Uniprot No.

Target Background

Function
RRAS Antibody, HRP conjugated, regulates the organization of the actin cytoskeleton. In conjunction with OSPBL3, it modulates integrin beta-1 (ITGB1) activity.
Gene References Into Functions

RRAS Gene Function

  1. R-Ras activation has been linked to melanoma tumorigenesis. PMID: 26993606
  2. Research has identified potential biochemical factors involved in distant recurrence and suggests that R-Ras and Transgelin may serve as post-surgical prognostic biomarkers for Stage III colorectal cancer. PMID: 27270312
  3. RRAS is a key regulator of miR-34/449, which promotes actin network formation during multiciliogenesis. PMID: 26381333
  4. Studies provide evidence of a functional connection between RRAS and MAPK signaling, revealing an unexpected role for enhanced RRAS function in human disease. PMID: 24705357
  5. Girdin regulates VE-cadherin trafficking in synergy with R-Ras. PMID: 25869066
  6. Phosphorylation of ORP3 controls its association with VAPA. Moreover, ORP3-VAPA complexes stimulate R-Ras signaling. PMID: 25447204
  7. Contrary to the positive association reported in prior studies, research indicates that R-Ras activation may negatively regulate the transformation of breast epithelial cells. PMID: 24700169
  8. A polymorphism linked to RRAS, SCAF1, IRF3, and BCL2L12 genes is associated with cirrhosis in hepatitis C virus carriers. PMID: 24131527
  9. Patients with gemistocytic astrocytomas with an RRAS deletion tend to have shorter survival rates than those without deletion. PMID: 25192052
  10. These data suggest that pharmacological inhibition of RRAS signaling might confer therapeutic benefit in Huntington's disease. PMID: 23209424
  11. These findings identify R-Ras as a critical regulator of vessel integrity and function during tumor vascularization. PMID: 22897853
  12. Maintaining endothelial barrier function relies on active R-Ras and association between R-Ras and FLNa. PMID: 21660952
  13. Data suggest that the RRAS gene is a significant regulatory module in the pathogenesis of biliary atresia (BA), which may serve as a novel prognostic marker for BA. PMID: 21390152
  14. Data support a model in which R-Ras functionally associates with FLNa and thereby regulates integrin-dependent migration. PMID: 20585650
  15. Sema3E acts on plexin D1 to initiate a two-pronged mechanism involving R-Ras inactivation and Arf6 stimulation, which affect the status of activation of integrins and their intracellular trafficking, respectively. PMID: 20385769
  16. Studies have investigated the potential contribution of RRAS in generating estrogen-independence in MCF-7 cells. PMID: 12386818
  17. The oncogene R-Ras promotes tumor growth of cervical epithelial cells and increases their migration potential over collagen through a pathway that involves PI 3-K. PMID: 12548599
  18. Activation of R-Ras promotes the formation of focal adhesions and a spread cell shape. PMID: 12890755
  19. Plexin-B1 mediates Sema4D-induced repulsive axon guidance signaling by acting as a GTPase activating protein for R-Ras. PMID: 15297673
  20. R-Ras plays a key role in cell migration by locally regulating the switch from Rac to Rho activity after membrane protrusion and adhesion. PMID: 15525681
  21. GPCRs rapidly activate R-Ras; R-Ras activation by the GPCRs is apparently directly induced by cyclic AMP-regulated Epac proteins; and activated R-Ras specifically controls GPCR-mediated phospholipase D stimulation. PMID: 16754664
  22. These findings show that R-Ras activity mediates inhibition of insulin signaling associated with suppression of estrogen action, implicating this GTPase in a growth-inhibitory mechanism associated with antiestrogen treatment of breast cancer. PMID: 16914723
  23. R-Ras and ILK have roles upstream of GSK-3beta in the regulation of neuronal polarity. PMID: 17107957
  24. Notch1 ligand, Delta-like ligand-4, stimulates R-Ras-dependent alpha 5 beta 1 integrin-mediated adhesion, demonstrating the physiological relevance of this pathway. PMID: 17664272
  25. Results map the functional elements in the R-ras promoter sequence and suggest that the GABP may be critical for transcription of R-ras and for maintenance of normal blood vessel functions through the regulation of this gene. PMID: 19011236
  26. TCF8 activates R-Ras, another class of angiogenic regulator, to suppress angiogenesis by a mechanism other than a transcriptional attenuator. PMID: 19116136

Show More

Hide All

Database Links

HGNC: 10447

OMIM: 165090

KEGG: hsa:6237

STRING: 9606.ENSP00000246792

UniGene: Hs.515536

Protein Families
Small GTPase superfamily, Ras family
Subcellular Location
Cell membrane; Lipid-anchor; Cytoplasmic side. Note=Inner surface of plasma membrane possibly with attachment requiring acylation of the C-terminal cysteine (By similarity with RAS).

Q&A

What is RRAS and what are its primary functions in cellular signaling?

RRAS (Ras-related protein R-Ras) is a small GTPase involved in multiple cellular signaling pathways. It functions primarily in the activation of Ras protein signal transduction cascades and possesses intrinsic GTPase activity . RRAS regulates organization of the actin cytoskeleton and, working in conjunction with OSPBL3, modulates integrin beta-1 (ITGB1) activity . Recent research has revealed RRAS's critical role in vascular development, endothelial cell quiescence, and immune cell function, particularly in dendritic cells . The protein has a molecular weight of approximately 23 kDa and acts as a molecular switch cycling between GDP-bound (inactive) and GTP-bound (active) states, similar to other Ras family proteins.

What criteria should researchers consider when selecting an RRAS antibody for their experiments?

When selecting an RRAS antibody, researchers should evaluate multiple critical parameters: (1) Specificity: confirm the antibody recognizes your target RRAS isoform without cross-reactivity to other Ras family members; (2) Application compatibility: verify the antibody has been validated for your intended application (WB, IHC-P, etc.) ; (3) Species reactivity: ensure compatibility with your experimental model system (human, mouse, etc.) ; (4) Clonality: determine whether monoclonal (greater specificity) or polyclonal (broader epitope recognition) better suits your needs; (5) Immunogen sequence: check if the antibody recognizes specific regions of interest in RRAS protein; and (6) Published validation: review literature citations where the antibody has been successfully utilized . For HRP-conjugated antibodies specifically, also consider enzyme stability, optimal storage conditions, and recommended working dilutions for your application.

What are the optimal dilution ratios for HRP-conjugated RRAS antibodies in western blotting applications?

Optimal dilution ratios for HRP-conjugated RRAS antibodies vary by product and application but generally range from 1:1000 to 1:5000 for western blotting applications. For example, the HRP Anti-Ras antibody [EPR3255] (ab199557) performs optimally at a 1:5000 dilution when detecting Ras proteins in HEK293 whole cell lysates . In contrast, unconjugated RRAS antibodies like ab154962 are typically used at higher concentrations (1:500) and require secondary antibody incubation . These dilution differences reflect the direct enzymatic activity of HRP-conjugated antibodies. To determine the optimal dilution for your specific experimental conditions, perform a dilution series (1:1000, 1:2000, 1:5000, 1:10000) using positive control lysates containing known RRAS expression levels. Select the dilution that provides the clearest specific band at the expected molecular weight (23 kDa for RRAS) with minimal background.

How should researchers design western blotting protocols when using HRP-conjugated RRAS antibodies?

An optimized western blotting protocol for HRP-conjugated RRAS antibodies should include the following methodological steps: (1) Sample preparation: extract proteins using lysis buffers containing 25mM HEPES-KOH (pH 7.4), 200mM NaCl, 1% NP-40, 2mM EDTA, 5mM MgCl₂, and 0.25% NaDOC ; (2) Gel electrophoresis: load 10-30μg protein per lane on a 4-12% Bis-tris gel; run at 200V for 35 minutes using MES buffer system ; (3) Transfer: transfer proteins to nitrocellulose membrane at 30V for 70 minutes; (4) Blocking: block membrane with 3% milk for one hour at room temperature ; (5) Primary antibody: incubate with diluted HRP-conjugated RRAS antibody (typically 1:5000) overnight at 4°C; (6) Washing: wash thoroughly with TBST buffer; (7) Detection: visualize using ECL substrate kit (high sensitivity recommended) . When optimizing, pay particular attention to blocking conditions, as inadequate blocking can result in high background with direct HRP-conjugated antibodies.

What controls should be included when using HRP-conjugated RRAS antibodies in research applications?

Rigorous experimental design with HRP-conjugated RRAS antibodies requires several critical controls: (1) Positive control: include lysates from cell lines known to express RRAS (e.g., Molt4, Raji, or HEK293 cells) ; (2) Negative control: incorporate lysates from cells with RRAS knockdown/knockout or tissues from Rras−/− mouse models ; (3) Loading control: use antibodies against housekeeping proteins such as actin to normalize protein loading; (4) Antibody specificity control: perform peptide competition assays using the immunizing peptide; (5) Signal specificity control: include a membrane incubated with secondary HRP-conjugated antibody alone (for non-specific binding assessment) and a membrane with ECL reagent only (for endogenous peroxidase activity); and (6) Molecular weight marker: always include to confirm the detected band matches the expected 23 kDa size of RRAS . These controls enable proper interpretation of results and troubleshooting of technical issues.

What are common causes of false positives or background issues when using HRP-conjugated RRAS antibodies?

Several factors can contribute to false positives or high background when using HRP-conjugated RRAS antibodies: (1) Insufficient blocking: inadequate blocking allows non-specific antibody binding—extend blocking time to 2 hours or increase blocking agent concentration to 5%; (2) Overconcentrated antibody: excessive HRP-conjugated antibody increases background—dilute further (1:10000) if high background persists; (3) Degraded ECL substrate: old or improperly stored ECL reagents can cause non-specific chemiluminescence—use fresh substrate and optimize exposure times; (4) Membrane drying: dried membranes increase non-specific binding—keep membranes consistently wet throughout the protocol; (5) Endogenous peroxidase activity: sample tissues may contain naturally occurring peroxidases—pretreat lysates with hydrogen peroxide; and (6) Cross-reactivity with other Ras family members: Ras proteins share structural homology—perform validation with Rras knockout samples . Addressing these issues through methodical optimization will significantly improve signal-to-noise ratio and data reliability.

How can researchers distinguish between specific RRAS signal and other Ras family proteins in western blotting?

Distinguishing RRAS from other Ras family proteins requires deliberate experimental approaches: (1) Antibody selection: choose antibodies raised against unique N-terminal regions of RRAS that differ from classical Ras proteins (H-Ras, K-Ras, N-Ras) ; (2) Molecular weight comparison: RRAS appears at approximately 23 kDa, whereas other Ras proteins may vary slightly—carefully analyze band positions relative to molecular weight markers ; (3) Knockout/knockdown validation: include samples from RRAS-deficient systems as negative controls—Rras−/− mouse-derived cells or RRAS siRNA-treated cells provide definitive negative controls ; (4) Pull-down assays: perform R-Ras-GTP-specific pull-down assays using GST-RalGDS-RBD fusion proteins that preferentially bind active R-Ras over other Ras proteins ; and (5) Competition assays: pre-incubate antibodies with purified RRAS protein to demonstrate binding specificity. This multi-faceted approach ensures accurate identification of RRAS-specific signals.

What approaches can address inconsistent detection when using HRP-conjugated RRAS antibodies?

Inconsistent detection with HRP-conjugated RRAS antibodies can be resolved through systematic troubleshooting: (1) Antibody storage: HRP conjugates are sensitive to repeated freeze-thaw cycles—aliquot antibodies upon receipt and store at -20°C; (2) Sample preparation: standardize lysis conditions using buffers containing phosphatase and protease inhibitors to prevent protein degradation; (3) Loading consistency: normalize protein loading (10-30μg) across all samples and verify with loading controls ; (4) Transfer efficiency: ensure complete protein transfer by staining membranes with Ponceau S before blocking; (5) Incubation conditions: maintain consistent temperature (4°C overnight) and time for antibody incubation ; (6) ECL substrate sensitivity: match substrate sensitivity to expression level—high sensitivity substrates for low expression samples; and (7) Exposure optimization: capture multiple exposure times to identify the linear detection range. Methodically addressing these variables will significantly improve reproducibility.

How can HRP-conjugated RRAS antibodies be used to investigate RRAS activation states in disease models?

Investigating RRAS activation states in disease models requires specialized methodological approaches: (1) Activity-specific detection: combine HRP-conjugated RRAS antibodies with GST-RalGDS-RBD pull-down assays to selectively capture and detect GTP-bound (active) RRAS—30μg of fusion protein coupled to 25μL glutathione-Sepharose beads is optimal for 1mg protein lysate ; (2) Comparative analysis: analyze both total RRAS (direct lysate blotting) and active RRAS (pull-down) to determine activation ratios in disease versus normal samples; (3) Time-course experiments: perform kinetic analyses of RRAS activation following stimulation (e.g., lipopolysaccharide treatment shows increased R-Ras-GTP levels within 10 minutes) ; (4) Pathway inhibitor studies: combine RRAS activation detection with inhibitors targeting downstream effectors (e.g., p38, Akt) to dissect signaling networks ; and (5) Disease model validation: apply these techniques in relevant models such as Huntington's disease models where abnormal RRAS activation and downstream RAF1 activity have been observed . These approaches provide mechanistic insights into disease-specific RRAS dysregulation.

What methodological approaches enable investigation of RRAS protein interactions in different cellular contexts?

Investigating RRAS protein interactions across cellular contexts requires specialized techniques: (1) Co-immunoprecipitation with HRP-conjugated antibodies: precipitate RRAS protein complexes followed by direct detection of interacting proteins; (2) Proximity ligation assays: visualize and quantify protein-protein interactions in situ by combining RRAS antibodies with antibodies against potential binding partners; (3) Co-localization studies: perform immunofluorescence microscopy using fluorescently-labeled RRAS antibodies with markers for cellular compartments or interacting proteins (particularly relevant for studies of RRAS and mutant huntingtin co-localization in mouse striatum) ; (4) Cell-type specific interaction analysis: compare RRAS interactome between different cell types (e.g., endothelial cells versus immune cells) using tissue-specific antibody applications ; and (5) Active-state specific interactions: combine GST-RalGDS-RBD pull-downs with interaction studies to identify proteins specifically binding to active RRAS . These methodological approaches reveal context-dependent interaction networks and functional relationships.

How can researchers utilize HRP-conjugated RRAS antibodies to study post-translational modifications of RRAS?

Studying RRAS post-translational modifications (PTMs) requires specific methodological adaptations: (1) Two-dimensional gel electrophoresis: separate RRAS protein variants by both molecular weight and isoelectric point before detection with HRP-conjugated antibodies to resolve modified forms; (2) Phosphorylation studies: combine phosphatase treatments of samples with subsequent western blotting to identify phosphorylated RRAS states; (3) Farnesylation analysis: use farnesyltransferase inhibitors in conjunction with RRAS detection to study this critical lipid modification—particularly relevant given that FNTA/B farnesyltransferase inhibition suppresses mutant huntingtin toxicity ; (4) Ubiquitination detection: perform immunoprecipitation under denaturing conditions followed by ubiquitin blotting to identify degradation-targeted RRAS; and (5) Mass spectrometry validation: combine immunoprecipitation with mass spectrometry analysis to comprehensively map all PTMs on RRAS. These approaches provide critical insights into regulatory mechanisms controlling RRAS activity, localization, and stability across different physiological and pathological contexts.

How does RRAS signaling contribute to neurodegenerative disease pathology?

RRAS signaling demonstrates significant involvement in neurodegenerative conditions, particularly Huntington's disease (HD). Genome-scale RNAi screening identified multiple components of the RRAS signaling pathway as loss-of-function suppressors of mutant huntingtin toxicity in both human and mouse cell models . Mechanistically, abnormal activation of RRAS and its downstream effector RAF1 has been observed in cellular and mouse models of HD . Co-localization studies have revealed physical association between RRAS and mutant huntingtin in cells and mouse striatum, suggesting pathogenic RRAS activation may occur through direct protein interaction . This dysregulation creates a promising intervention opportunity, as targeting multiple points in the pathway—including RRAS itself, FNTA/B (farnesyltransferase), PIN1, and PLK1—can suppress mutant huntingtin toxicity . These findings position RRAS signaling as both a critical pathogenic mechanism and a potential therapeutic target in neurodegenerative disorders.

What methodological approaches best demonstrate RRAS function in vascular and immune systems?

Investigating RRAS function in vascular and immune contexts requires specialized methodological approaches: (1) Endothelial-specific genetic models: utilize cdh5-Cre;Rras mouse models for endothelial-specific gene ablation to study vascular development and stabilization ; (2) Immunologic synapse formation analysis: quantify dendritic cell-T cell interactions using Rras−/− mouse-derived dendritic cells to assess immunological synapse stability ; (3) Activation markers: measure MHC class II and CD86 surface expression in lipopolysaccharide-stimulated dendritic cells from wild-type versus Rras−/− mice using flow cytometry ; (4) Downstream signaling analysis: assess phosphorylation of p38 and Akt in response to stimulation using phospho-specific antibodies ; (5) Barrier function assessment: measure extravascular fibrinogen accumulation in brain tissue to evaluate blood-brain barrier integrity in Rras−/− models ; and (6) Cell-specific pathway analysis: compare R-Ras-induced upregulation of Hey1, Hes1, p21, and p53 across different tissue contexts . These methodological approaches reveal tissue-specific roles of RRAS in maintaining vascular stability and promoting proper immune cell function.

What are the technical considerations for using HRP-conjugated RRAS antibodies in therapeutic target validation studies?

When employing HRP-conjugated RRAS antibodies for therapeutic target validation, researchers should implement these technical considerations: (1) Target engagement assessment: develop assays using HRP-conjugated RRAS antibodies to verify binding of candidate therapeutics to RRAS protein; (2) Activation state monitoring: combine with GST-RalGDS-RBD pull-down assays to measure changes in RRAS-GTP levels following therapeutic intervention ; (3) Downstream effector analysis: simultaneously monitor p38, Akt, and RAF1 phosphorylation states to assess pathway inhibition completeness ; (4) Cross-target specificity: evaluate effects on related Ras family members (H-Ras, K-Ras, N-Ras) to determine therapeutic selectivity; (5) Tissue penetration verification: perform immunohistochemistry with HRP-conjugated antibodies to confirm target engagement in relevant tissues; and (6) Dose-response characterization: establish quantitative relationships between therapeutic dose, RRAS inhibition, and functional outcomes. For farnesyltransferase inhibitors specifically, measure RRAS membrane localization to verify disruption of this critical post-translational modification . These technical approaches provide robust validation of RRAS-targeting therapeutic strategies.

How might multiplexed detection systems incorporating HRP-conjugated RRAS antibodies advance complex pathway analysis?

Emerging multiplexed detection systems with HRP-conjugated RRAS antibodies offer transformative potential for complex pathway analysis: (1) Sequential multiplexing: implement sequential stripping and reprobing protocols optimized for HRP-conjugated antibodies to analyze multiple pathway components on a single membrane; (2) Spectrally distinct substrates: utilize HRP substrates with different emission spectra to simultaneously detect RRAS alongside other signaling proteins; (3) Microfluidic western blotting: adapt HRP-conjugated RRAS antibodies to microfluidic platforms allowing parallel analysis of dozens of proteins from limited samples; (4) Mass cytometry adaptation: develop metal-conjugated RRAS antibodies for high-dimensional single-cell analysis of RRAS in heterogeneous populations; and (5) Spatial analysis: implement advanced tissue imaging systems combining HRP-conjugated RRAS antibodies with other key pathway markers. These methodological advances will enable integrated analysis of RRAS within its complete signaling network context, particularly important for understanding its role in complex disease states like Huntington's disease where multiple pathway interactions determine pathological outcomes .

What methodological innovations might improve detection sensitivity and specificity for low-abundance RRAS in tissue samples?

Advanced methodological innovations to enhance RRAS detection include: (1) Tyramide signal amplification: combine HRP-conjugated RRAS antibodies with tyramide-based signal amplification to dramatically increase sensitivity while maintaining spatial resolution in tissue sections; (2) Automated western blotting platforms: utilize capillary-based automated western systems with optimized detection parameters for RRAS; (3) Single-molecule detection: adapt HRP-conjugated antibodies for single-molecule pull-down assays to detect extremely low RRAS concentrations; (4) Nanobody-based detection: develop HRP-conjugated anti-RRAS nanobodies with improved tissue penetration and epitope accessibility; and (5) Digital protein quantification: implement digital ELISA technologies for absolute quantification of RRAS at femtomolar concentrations. These methodological advances will enable detection of physiologically relevant RRAS in challenging samples like cerebrospinal fluid or specific brain regions, critical for monitoring RRAS in neurodegenerative disease progression and therapeutic response .

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.