RRAS Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Biological Role of RRAS

RRAS is a small GTPase regulating:

  • Integrin activation and cell adhesion

  • Actin cytoskeleton organization

  • Immune cell maturation (e.g., dendritic cells)

  • Platelet signaling via GPVI-FcRγ

Mutations in RRAS (e.g., Q87L) lead to constitutive activation, influencing oncogenic pathways and immune responses .

Cellular Localization Studies

  • Actin colocalization: FITC-conjugated RRAS antibodies identified RRAS in membrane ruffles and filopodia, but not lamellipodia, in 32D myeloid cells .

  • Subcellular distribution: Demonstrated plasma membrane targeting via C-terminal lipid modifications .

Immune Regulation

  • Dendritic cell (DC) maturation: Reduced MHC-II and CD86 expression in Rras−/− DCs was quantified using FITC-labeled antibodies in flow cytometry .

  • Treg proliferation: FITC-based assays revealed increased natural Treg populations in Rras−/− mice during experimental autoimmune encephalomyelitis (EAE) .

Signal Transduction Analysis

  • Platelet activation: FITC-conjugated antibodies detected RRAS involvement in GPVI-FcRγ-mediated signaling .

  • Phosphorylation cascades: Used alongside phospho-specific antibodies (e.g., p-AKT, p-ERK) to map RRAS-dependent pathways .

Key Experimental Findings

Table 2: RRAS Functional Insights Using FITC-Conjugated Antibodies

Study FocusKey FindingAssay TypeCitation
DC-T cell interactionsRRAS deficiency reduces immunologic synapse stabilityFlow Cytometry
Integrin activationRRAS Q87L mutant enhances cell adhesionFluorescence Imaging
Actin dynamicsRRAS localizes to peripheral membrane rufflesImmunofluorescence
Autoimmunity modulationRras−/− mice show attenuated EAE severityFlow Cytometry

Optimization Guidelines

  • Photostability: FITC’s susceptibility to photobleaching makes it less suitable for long-term imaging; alternatives like Alexa Fluor® 488 are recommended for extended assays .

  • Multiplexing compatibility: Pair with TRITC (ex: 557 nm) or Cy5 (ex: 649 nm) to avoid spectral overlap .

  • Cross-reactivity: Santa Cruz’s sc-166221 FITC detects RRAS across human, mouse, and rat samples, while Cusabio’s CSB-PA020513PC01HU is human-specific .

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we are able to dispatch the products within 1-3 working days after receiving your order. Delivery time may vary based on the purchasing method or location. Please consult your local distributors 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, FITC conjugated, plays a regulatory role in the organization of the actin cytoskeleton. In conjunction with OSPBL3, it modulates the activity of integrin beta-1 (ITGB1).
Gene References Into Functions

RRAS: A Multifaceted Regulator

The RRAS gene is implicated in a wide range of biological processes, contributing to various cellular functions and pathways. Here's a closer look at its involvement in diverse cellular functions and its potential relevance in disease pathogenesis:

  1. R-Ras activation has been linked to melanoma tumorigenesis. PMID: 26993606
  2. Research has identified R-Ras and Transgelin as potential post-surgical prognostic biomarkers for Stage III colorectal cancer, suggesting their role in predicting the likelihood of recurrence. PMID: 27270312
  3. RRAS is a critical regulator of miR-34/449 in promoting actin network formation during multiciliogenesis, a process essential for the development of cilia, hair-like structures on the surface of cells. PMID: 26381333
  4. Studies have revealed a functional link between RRAS and MAPK signaling, with enhanced RRAS function potentially playing a role in human disease. PMID: 24705357
  5. Girdin, a protein involved in cell signaling, regulates the trafficking of VE-cadherin, a cell-cell adhesion molecule, in synergy with R-Ras. PMID: 25869066
  6. Phosphorylation of ORP3, a protein involved in lipid metabolism, controls its association with VAPA, another protein involved in cell signaling. ORP3-VAPA complexes, in turn, stimulate R-Ras signaling. PMID: 25447204
  7. While previous studies suggested a positive association between R-Ras activation and breast epithelial cell transformation, recent research indicates that R-Ras activation may actually negatively regulate this process. 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, a type of brain tumor, with an RRAS deletion tend to have shorter survival rates compared to those without the deletion. PMID: 25192052
  10. Research suggests that pharmacological inhibition of RRAS signaling may hold therapeutic potential in Huntington's disease. PMID: 23209424
  11. R-Ras has been identified as a crucial regulator of vessel integrity and function during tumor vascularization, highlighting its role in tumor growth and development. PMID: 22897853
  12. Maintaining endothelial barrier function, which is essential for blood vessel integrity, is dependent on active R-Ras and the association between R-Ras and FLNa, a protein involved in cytoskeletal organization. PMID: 21660952
  13. Data suggests that the RRAS gene is a significant regulatory module in the pathogenesis of biliary atresia (BA), a serious liver disease in infants. This gene may serve as a novel prognostic marker for BA. PMID: 21390152
  14. Research supports a model where R-Ras functionally associates with FLNa and thereby regulates integrin-dependent migration, a critical process for cell movement and tissue development. PMID: 20585650
  15. Sema3E, a protein involved in cell signaling, 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 R-Ras in generating estrogen-independence in MCF-7 cells, a breast cancer cell line. PMID: 12386818
  17. The oncogene R-Ras promotes tumor growth of cervical epithelial cells and increases their migration potential over collagen through a pathway involving PI 3-K, a key signaling molecule. PMID: 12548599
  18. Activation of R-Ras promotes the formation of focal adhesions, specialized structures that connect the cell to the extracellular matrix, and a spread cell shape, contributing to cell migration and adhesion. PMID: 12890755
  19. Plexin-B1, a protein involved in cell signaling, mediates Sema4D-induced repulsive axon guidance signaling by acting as a GTPase activating protein for R-Ras, influencing the growth and direction of nerve cells. PMID: 15297673
  20. R-Ras plays a crucial role in cell migration by locally regulating the switch from Rac to Rho activity after membrane protrusion and adhesion, controlling the dynamic changes in cell shape and movement. PMID: 15525681
  21. GPCRs, G protein-coupled receptors, rapidly activate R-Ras. This activation is apparently directly induced by cyclic AMP-regulated Epac proteins, and activated R-Ras specifically controls GPCR-mediated phospholipase D stimulation, a signaling process involved in various cellular functions. PMID: 16754664
  22. Research shows that R-Ras activity mediates inhibition of insulin signaling associated with suppression of estrogen action, suggesting its role in a growth-inhibitory mechanism related to antiestrogen treatment of breast cancer. PMID: 16914723
  23. R-Ras and ILK, a protein involved in cell signaling, have roles upstream of GSK-3beta in the regulation of neuronal polarity, a crucial process for the development and function of nerve cells. 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 in cell-cell adhesion and tissue development. PMID: 17664272
  25. Studies have mapped the functional elements in the R-ras promoter sequence, suggesting that the GABP transcription factor may be critical for transcription of R-ras and for maintaining normal blood vessel functions through the regulation of this gene. PMID: 19011236
  26. TCF8, a transcription factor, activates R-Ras, another class of angiogenic regulator, to suppress angiogenesis, the formation of new blood vessels, 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 why is it an important research target?

RRAS (Ras-related protein R-Ras, p23) is a member of the RAS family of small GTPases with the UniprotID P10301. Unlike classical RAS proteins, RRAS plays distinct roles in regulating integrin activity, cell migration, and various signal transduction pathways. RRAS is particularly important in research because it functions differently from other RAS family members like HRAS - while HRAS can suppress integrin activation, RRAS1 and RRAS2/TC21 can reverse this suppression, indicating its unique signaling properties . R-Ras activity is critical for ECM-mediated β1 integrin activation and subsequent cell migration processes, making it an important target for studying cellular adhesion, migration, and response to extracellular cues .

What are the key characteristics of FITC-conjugated RRAS antibodies?

FITC-conjugated RRAS antibodies are immunological reagents where fluorescein isothiocyanate (FITC) is chemically linked to antibodies targeting RRAS proteins. The commercially available RRAS antibody with FITC conjugation is typically a polyclonal antibody derived from rabbit hosts, designed to recognize human RRAS protein, specifically a peptide sequence from residues 7-23 . The FITC conjugation enables direct visualization of RRAS in fluorescence-based applications without requiring secondary antibodies. These antibodies maintain reactivity to human species and are purified using antigen affinity methods . The green fluorescence of FITC (excitation maximum ~495 nm, emission maximum ~519 nm) makes these antibodies suitable for various imaging techniques including flow cytometry, immunofluorescence microscopy, and fluorescence-based protein detection methods.

How does RRAS relate to other members of the RAS family in terms of function and signaling?

RRAS demonstrates distinct functional characteristics compared to other RAS family members. While sharing structural similarities with classical RAS proteins (HRAS, KRAS, NRAS), RRAS has unique roles in cellular processes. In contrast to HRAS which suppresses integrin activation, RRAS1 and RRAS2/TC21 can reverse this suppression . RRAS is activated in response to specific stimuli such as ECM interaction and agonists for receptors like GPVI/FcRγ in platelets .

The signaling pathways downstream of RRAS also differ from classical RAS proteins. RRAS prominently signals through PI3-K pathways to regulate integrin activation, whereas classical RAS proteins preferentially activate the RAF-MEK-ERK pathway. Notably, the R-Ras-mediated increase in affinity of β1 integrins is dependent on PI3-K activity, and this pathway is critical for processes like cell migration and adhesion . R-Ras activity can be measured using pull-down assays with GST-RBD (RAS binding domain of c-Raf-1), similar to other RAS proteins, indicating some conservation in the effector binding regions .

What are the recommended protocols for detecting RRAS activation using FITC-conjugated antibodies?

While direct detection of activated RRAS is typically performed using pull-down assays rather than antibodies, FITC-conjugated RRAS antibodies can be valuable in complementary analyses. For comprehensive RRAS activation studies, researchers should employ a combined approach:

  • Pull-down assay for activated RRAS:

    • Lyse cells in appropriate buffer (25 mM Hepes-NaOH, pH 7.5, 150 mM NaCl, 1% NP-40, 0.25% sodium deoxycholate, 0.1% SDS, 10% glycerol, 10 mM MgCl₂, 1 mM EDTA, 1 mM DTT, and protease inhibitors)

    • Incubate lysates with GST-fused Ras binding domain of c-Raf-1 (GST-RBD) (~75 μg)

    • Isolate complexes using glutathione-sepharose beads

    • Analyze by Western blotting using RRAS-specific antibodies

  • Complementary visualization with FITC-conjugated RRAS antibodies:

    • Fix cells with 4% paraformaldehyde (10 minutes, room temperature)

    • Permeabilize with 0.1% Triton X-100 (5 minutes)

    • Block with 3% BSA in PBS (1 hour)

    • Incubate with FITC-conjugated RRAS antibody (1:100-1:500 dilution, optimal concentration determined empirically)

    • Counterstain nuclei with DAPI

    • Mount and analyze by fluorescence microscopy

For flow cytometry applications:

  • Harvest cells and fix with 2% paraformaldehyde

  • Permeabilize with 0.1% saponin in PBS with 0.5% BSA

  • Incubate with FITC-conjugated RRAS antibody (typically 1-5 μg per 10⁶ cells)

  • Wash and analyze using appropriate flow cytometer settings for FITC detection

Remember that storage conditions (-20°C or -80°C, avoiding repeated freeze-thaw cycles) are critical for maintaining antibody performance .

How can I design experiments to study RRAS-mediated regulation of integrins?

To study RRAS-mediated regulation of integrins, consider the following experimental design approach:

  • Measurement of integrin activation states:

    • Use conformation-specific antibodies like HUTS-4 that detect active β1 integrins

    • Employ both immunoprecipitation and flow cytometry methods for quantification

    • For flow cytometry: prepare cell suspensions, stain with HUTS-4 followed by PE-conjugated secondary antibodies

    • For ELISA-based detection: immobilize cells on fibronectin-coated surfaces, stain with HUTS-4, and quantify binding

  • Manipulation of RRAS activity:

    • Express constitutively active (R-Ras-QL) or dominant negative RRAS mutants

    • Use R-Ras-specific GAP (like the myristoylated GAP domain of p98-R-RasGAP) to downregulate endogenous R-Ras activity

    • Implement RNA interference using R-Ras siRNA to knockdown expression

    • Apply pathway modulators like the PI3-K inhibitor LY294002 to block downstream signaling

  • Readouts for functional consequences:

    • Measure FAK tyrosine phosphorylation as a marker of integrin-mediated signaling

    • Assess PI3-K activity through Akt phosphorylation status

    • Quantify cell migration using transwell migration assays

    • Monitor adhesion strength and spreading dynamics on ECM proteins

  • Visualization of molecular interactions:

    • Use FITC-conjugated RRAS antibodies to track RRAS localization

    • Implement co-immunoprecipitation to detect RRAS interactions with effector proteins

    • Apply proximity ligation assays to visualize RRAS-integrin proximity

This experimental approach allows comprehensive analysis of how RRAS regulates integrin activity, from molecular interactions to functional cellular outcomes.

What are the optimal conditions for storing and handling FITC-conjugated RRAS antibodies?

For maintaining optimal activity of FITC-conjugated RRAS antibodies, follow these storage and handling guidelines:

  • Storage temperature:

    • Upon receipt, store at -20°C or -80°C for long-term preservation

    • Avoid repeated freeze-thaw cycles that can degrade both antibody function and FITC fluorescence

    • For working aliquots, store at 4°C for up to one month

  • Buffer conditions:

    • The antibody is typically supplied in a buffer containing 50% glycerol, 0.01M PBS (pH 7.4), and 0.03% Proclin 300 as preservative

    • Maintain these conditions during storage and avoid diluting the stock solution unnecessarily

  • Light protection:

    • FITC is sensitive to photobleaching, so protect from light during storage and handling

    • Use amber tubes for aliquots and cover with aluminum foil

    • Minimize exposure to bright laboratory lighting during experiments

  • Working with the antibody:

    • Centrifuge briefly before opening the vial to collect liquid at the bottom

    • Handle with clean, DNase/RNase-free pipette tips

    • For dilutions, use fresh, high-quality buffers with appropriate pH (usually PBS pH 7.4)

    • For flow cytometry, optimal working concentrations are typically 1-5 μg per 10⁶ cells

    • For immunofluorescence, starting dilutions of 1:100-1:500 are recommended

  • Quality control:

    • Before critical experiments, verify antibody performance using positive control samples

    • Consider including an isotype control to account for non-specific binding

Following these guidelines will help maintain antibody performance and FITC fluorescence intensity for reliable experimental results.

How can FITC-conjugated RRAS antibodies be used to study RRAS in platelet activation pathways?

FITC-conjugated RRAS antibodies can be strategically employed to investigate RRAS in platelet activation through multiple sophisticated approaches:

  • Visualization of RRAS distribution during platelet activation:

    • Isolate washed platelets using established protocols

    • Fix activated vs. resting platelets at various time points after stimulation with GPVI agonists

    • Permeabilize and stain with FITC-conjugated RRAS antibody

    • Co-stain with markers of platelet activation (e.g., P-selectin) using compatible fluorophores

    • Image using high-resolution confocal microscopy to track RRAS relocalization

  • Flow cytometric analysis of platelets:

    • Prepare platelets as described in published protocols

    • Dual-label with FITC-conjugated RRAS antibody and phycoerythrin-conjugated activation markers (like JON/A for activated αIIbβ3 or anti-P-selectin)

    • Analyze using flow cytometry to correlate RRAS expression/localization with activation status

    • Gate specifically on platelets using forward/side scatter characteristics

  • Correlation with RRAS activation status:

    • Perform GST-RBD pull-down assays in parallel samples to measure RRAS-GTP levels

    • Correlate active RRAS with integrin activation states and aggregate formation

    • Use phospho-specific antibodies against RRAS effectors to map signaling pathways

  • Inhibitor studies:

    • Pretreat platelets with Src inhibitors before agonist stimulation

    • Monitor changes in RRAS localization using FITC-conjugated antibodies

    • Correlate with functional readouts like aggregation and adhesion

  • Genetic approaches:

    • Isolate platelets from TC21/RRAS2 knockout mice or platelets treated with siRNA

    • Compare RRAS staining patterns and activation profiles with wild-type controls

This multifaceted approach allows researchers to understand how RRAS contributes to the complex signaling networks in platelet activation, particularly in response to GPVI/FcRγ immunoreceptor tyrosine-based activation motif (ITAM)-containing collagen receptor stimulation .

What methods can be used to study the interaction between Plexin-B1 and RRAS in cell migration?

The interaction between Plexin-B1 and RRAS in cell migration can be investigated using several sophisticated methodological approaches:

  • GTPase activation assays:

    • Use GST-RBD pull-down assays to quantify active RRAS levels after Sema4D stimulation

    • Compare RRAS activity in cells expressing wild-type Plexin-B1 versus R-Ras GAP-deficient mutants (Plexin-B1-GGA or Plexin-B1-RA)

    • Monitor changes in RRAS activation during ECM-stimulated cell migration

  • Functional migration studies:

    • Perform transwell migration assays with cells expressing various Plexin-B1 constructs

    • Treat cells with soluble Sema4D-Fc to activate Plexin-B1 signaling

    • Quantify migration in response to ECM proteins like fibronectin or collagen

    • Use function-blocking antibodies against β1 integrins (e.g., P5D2) or activating antibodies (e.g., 8A2) to manipulate integrin activity

  • Visualization of protein interactions:

    • Implement fluorescence resonance energy transfer (FRET) between labeled Plexin-B1 and RRAS

    • Use FITC-conjugated RRAS antibodies in combination with differentially labeled Plexin-B1

    • Perform proximity ligation assays to detect close association between proteins

    • Track co-localization during Sema4D-induced cell repulsion events

  • Analysis of downstream signaling:

    • Monitor PI3-K activity through Akt phosphorylation as a readout of RRAS signaling

    • Compare cells expressing wild-type and mutant forms of Plexin-B1

    • Examine β1 integrin activation using HUTS-4 antibody binding under various conditions

    • Quantify FAK phosphorylation as a measure of integrin-mediated adhesion signaling

  • Structure-function analyses:

    • Generate specific mutations in the Plexin-B1 R-Ras GAP domain

    • Assess effects on RRAS binding, GAP activity, and cellular migration

    • Combine with live cell imaging to visualize migration dynamics

This multifaceted approach allows comprehensive analysis of how Plexin-B1-mediated regulation of RRAS controls cell migration through modulation of integrin activity and downstream signaling pathways.

How can I measure the dynamics of RRAS activation in response to ECM stimulation?

Measuring the dynamics of RRAS activation in response to ECM stimulation requires a combination of biochemical, imaging, and functional approaches:

  • Time-course pull-down assays:

    • Plate cells on ECM proteins (e.g., fibronectin or collagen) for varying durations (0-60 minutes)

    • Lyse cells directly on dishes with ice-cold cell lysis buffer containing GST-RBD

    • Perform GST-RBD pull-down assays to isolate active, GTP-bound RRAS

    • Analyze by Western blotting using RRAS-specific antibodies

    • Quantify the ratio of active RRAS to total RRAS at each timepoint

  • Live-cell FRET-based biosensors:

    • Design or obtain FRET-based RRAS activation reporters

    • Transfect cells and plate on ECM-coated surfaces

    • Perform live-cell imaging to monitor RRAS activation in real-time

    • Correlate activation patterns with cell morphology changes and migration events

  • Spatial analysis of activation:

    • Fix cells at different timepoints after ECM stimulation

    • Perform immunofluorescence using conformation-specific antibodies or proximity ligation assays

    • Use FITC-conjugated RRAS antibodies for co-localization studies with activated integrins

    • Analyze subcellular distribution of active RRAS, particularly at adhesion sites

  • Correlation with integrin activation:

    • In parallel samples, measure β1 integrin activation using HUTS-4 antibody

    • Correlate the timing of RRAS activation with integrin activation

    • Use flow cytometry for population-level analysis and immunofluorescence for spatial information

  • Manipulation studies:

    • Compare dynamics in cells expressing different levels of RRAS (wild-type, knockdown, overexpression)

    • Assess the effects of pathway inhibitors (e.g., PI3-K inhibitor LY294002)

    • Test the influence of integrin-modulating antibodies (blocking vs. activating)

  • Experimental conditions to consider:

    • For suspension cells: Maintain cells in serum-free medium with 1% BSA before plating

    • Compare different ECM protein concentrations (e.g., 1 μg/ml vs. 10 μg/ml collagen)

    • Include non-adherent (suspension) controls for baseline RRAS activity

    • Consider cell density effects on activation dynamics

This comprehensive approach allows precise measurement of RRAS activation kinetics in response to ECM stimulation, providing insights into how this signaling node regulates cell adhesion and migration.

What are the common pitfalls when using FITC-conjugated antibodies in flow cytometry, and how can they be addressed?

When using FITC-conjugated RRAS antibodies in flow cytometry, researchers may encounter several challenges:

  • Autofluorescence interference:

    • Problem: Cellular autofluorescence in the FITC channel can reduce signal-to-noise ratio

    • Solution: Include unstained and isotype controls; use compensation settings to subtract autofluorescence; consider alternative fluorophores with emission spectra outside the autofluorescence range for highly autofluorescent samples

  • Photobleaching:

    • Problem: FITC is relatively susceptible to photobleaching, leading to signal loss

    • Solution: Minimize light exposure during sample preparation; analyze samples promptly after staining; consider more photostable fluorophores for lengthy procedures

  • pH sensitivity:

    • Problem: FITC fluorescence is optimal at pH 8.0 and decreases at lower pH values

    • Solution: Ensure buffers are maintained at appropriate pH (7.4-8.0); avoid acidic conditions during fixation and staining

  • Insufficient permeabilization for intracellular targets:

    • Problem: RRAS is often intracellular, requiring effective permeabilization

    • Solution: Optimize permeabilization protocols (test different detergents and concentrations); ensure adequate fixation before permeabilization; validate with known intracellular controls

  • Non-specific binding:

    • Problem: High background due to Fc receptor binding or hydrophobic interactions

    • Solution: Include blocking step with serum or commercial blocking buffers; use Fc receptor blocking reagents; perform titration experiments to determine optimal antibody concentration

  • Signal spillover:

    • Problem: FITC emission spectrum overlaps with other fluorophores

    • Solution: Apply proper compensation if using multiple fluorophores; design panels with minimal spectral overlap; use single-stained controls for compensation setup

  • Inadequate cell fixation:

    • Problem: Poor fixation can alter epitope accessibility or cell morphology

    • Solution: Optimize fixation conditions (time, temperature, fixative concentration); consider mild fixatives for sensitive epitopes

  • Sample preparation variability:

    • Problem: Inconsistent results between experiments

    • Solution: Standardize protocols; prepare master mixes of antibodies; include internal controls; establish consistent gating strategies

By anticipating these challenges and implementing appropriate solutions, researchers can obtain reliable and reproducible flow cytometry data using FITC-conjugated RRAS antibodies.

How can I distinguish between specific and non-specific binding when using RRAS antibodies in immunofluorescence?

Distinguishing between specific and non-specific binding is crucial for accurate interpretation of immunofluorescence results with RRAS antibodies:

  • Essential controls:

    • Negative controls: Include samples with isotype-matched control antibodies at the same concentration as the RRAS antibody

    • Blocking peptide controls: Pre-incubate FITC-conjugated RRAS antibody with excess immunizing peptide (RRAS protein 7-23AA) before staining to block specific binding sites

    • Knockdown controls: Compare staining in cells with RRAS knockdown via siRNA or CRISPR-Cas9

    • Secondary-only controls: For indirect immunofluorescence, include samples with secondary antibody but no primary

  • Validation through multiple detection methods:

    • Confirm staining patterns using alternative RRAS antibodies targeting different epitopes

    • Correlate immunofluorescence results with Western blot or flow cytometry data

    • Compare distribution pattern with GFP-tagged RRAS in transfected cells

  • Optimization strategies:

    • Titration experiments: Test a range of antibody concentrations to identify optimal signal-to-noise ratio

    • Fixation comparison: Try different fixation methods (paraformaldehyde, methanol, or combination) as epitope accessibility can vary

    • Blocking optimization: Test different blocking agents (BSA, normal serum, commercial blockers) and durations

    • Permeabilization adjustment: Compare different permeabilization reagents and concentrations

  • Signal pattern analysis:

    • Specific binding typically shows:

      • Consistent subcellular distribution matching known RRAS localization (membrane association with potential cytoplasmic component)

      • Reduced signal in knockdown cells

      • Correlation with stimulation conditions (e.g., increased membrane localization upon ECM stimulation)

    • Non-specific binding often presents as:

      • Uniform background staining

      • Nuclear staining (RRAS is not typically nuclear)

      • Persistent staining in knockdown controls

      • Variable patterns between identical samples

  • Advanced validation approaches:

    • Co-localization with known RRAS interacting partners

    • Differential staining patterns in cells expressing constitutively active versus dominant negative RRAS mutants

    • Correlation with functional readouts like integrin activation

Implementation of these strategies will help ensure that observed signals represent authentic RRAS localization rather than artifactual staining.

How do I design appropriate experimental controls when studying RRAS-mediated integrin activation?

Designing robust experimental controls is essential for rigorous studies of RRAS-mediated integrin activation:

  • Controls for RRAS manipulation:

    • Expression controls: Empty vector controls for overexpression studies; non-targeting siRNA for knockdown experiments

    • Activity controls: Compare constitutively active (R-Ras-QL) with dominant-negative mutants

    • Specificity controls: Include effector domain mutants like R-Ras-QL-64A that selectively disrupt PI3-K binding

    • Pharmacological controls: Include appropriate vehicle controls for inhibitors like LY294002

  • Controls for integrin activation measurement:

    • Antibody controls: Use isotype-matched control antibodies alongside HUTS-4 for β1 integrin activation detection

    • Positive controls: Include manganese treatment (Mn²⁺), which directly activates integrins independently of inside-out signaling

    • Negative controls: Use integrin-blocking antibodies like P5D2 to establish baseline levels

    • Technical controls: Include detergent conditions to expose all epitopes as a maximum binding control for HUTS-4

  • ECM stimulation controls:

    • Substrate controls: Include poly-L-lysine surfaces that support adhesion without integrin engagement

    • Concentration controls: Test multiple ECM protein concentrations (e.g., 1 μg/ml vs. 10 μg/ml)

    • Specificity controls: Use ECM proteins that engage different integrin heterodimers

    • Timing controls: Include multiple timepoints to capture activation dynamics

  • Signaling pathway controls:

    • Pathway inhibitor controls: Include specific inhibitors of PI3-K (LY294002), as well as inhibitors of other pathways

    • Downstream readout controls: Monitor multiple outputs (Akt phosphorylation, FAK phosphorylation)

    • Cross-pathway controls: Assess potential contributions from related pathways (e.g., Rap1)

  • Cell type considerations:

    • Use multiple cell lines to ensure findings are not cell-type specific

    • Include primary cells where feasible to confirm relevance in non-transformed systems

    • For specialized contexts (e.g., platelets), include appropriate positive controls (e.g., GPVI agonists)

  • Functional validation:

    • Correlate molecular findings with functional outcomes like cell migration

    • Use rescue experiments (e.g., expressing siRNA-resistant RRAS constructs in knockdown cells)

    • Implement dose-response relationships to establish causality

What are the recommended protocols for quantifying RRAS expression levels using FITC-conjugated antibodies?

For accurate quantification of RRAS expression using FITC-conjugated antibodies, researchers should implement these standardized protocols:

  • Flow cytometry-based quantification:

    • Sample preparation:

      • Harvest cells in log-phase growth

      • Fix with 2-4% paraformaldehyde (10 minutes, room temperature)

      • Permeabilize with 0.1% saponin or 0.1% Triton X-100

      • Block with 2-3% BSA in PBS (30 minutes)

      • Stain with titrated FITC-conjugated RRAS antibody (typically 1-5 μg per 10⁶ cells)

    • Data acquisition:

      • Include unstained, isotype, and single-stained controls

      • Acquire data using consistent PMT voltages across experiments

      • Collect sufficient events (minimum 10,000 per sample)

    • Analysis metrics:

      • Mean fluorescence intensity (MFI)

      • Median fluorescence intensity (more robust to outliers)

      • Coefficient of variation (CV) to assess population homogeneity

      • Calculate fold change relative to appropriate control samples

  • Fluorescence microscopy quantification:

    • Sample preparation:

      • Culture cells on appropriate substrates (coverslips, chamber slides)

      • Fix, permeabilize, block, and stain as above

      • Include DAPI counterstain for nuclear identification

    • Image acquisition:

      • Capture multiple fields (minimum 10) per condition

      • Use consistent exposure settings across all samples

      • Include calibration standards in each imaging session

    • Analysis approach:

      • Measure integrated density or mean fluorescence intensity

      • Define regions of interest (whole cell, specific compartments)

      • Apply background subtraction using cell-free areas

      • Normalize to cell number or area if comparing different samples

  • Quantitative considerations:

    • Dynamic range assessment:

      • Include samples with known varying levels of RRAS expression

      • Verify linearity of detection within the expected range

    • Standardization approaches:

      • Use quantitative fluorescent beads to calibrate instruments

      • Include internal control cells in each experiment

      • Express results as molecules of equivalent soluble fluorochrome (MESF)

    • Statistical analysis:

      • Apply appropriate statistical tests based on data distribution

      • Consider using non-parametric tests for flow cytometry data

      • Report both statistical and biological significance

  • Validation strategies:

    • Confirm expression levels using alternative methods (Western blot, qPCR)

    • Correlate fluorescence intensity with functional readouts

    • Verify specificity using RRAS knockdown samples

Following these protocols ensures reliable and reproducible quantification of RRAS expression levels across experimental conditions and between different studies.

How can I integrate RRAS signaling data with functional outcomes in cell migration studies?

Integrating RRAS signaling data with functional cell migration outcomes requires a multidimensional approach that connects molecular events to cellular behaviors:

  • Temporal correlation analysis:

    • Experimental design:

      • Collect RRAS activation data (GST-RBD pull-downs) at multiple timepoints after ECM stimulation

      • In parallel samples, measure migration parameters at corresponding timepoints

      • Include conditions that modulate RRAS activity (e.g., R-Ras siRNA, R-RasGAP expression)

    • Analysis approach:

      • Plot RRAS activation kinetics alongside migration metrics

      • Calculate correlation coefficients between RRAS-GTP levels and migration parameters

      • Identify temporal relationships (immediate, delayed, sustained responses)

  • Pathway perturbation analysis:

    • Intervention strategies:

      • Manipulate RRAS activity using genetic approaches (siRNA, dominant mutants)

      • Target specific downstream effectors (PI3-K inhibition with LY294002)

      • Modulate upstream regulators (Plexin-B1, integrin-modulating antibodies)

    • Integrated readouts:

      • Measure both signaling outputs (Akt phosphorylation, integrin activation) and migration behaviors

      • Construct dose-response relationships between pathway activity and functional outcomes

      • Perform rescue experiments to establish causality

  • Single-cell correlation approaches:

    • Methodology:

      • Implement live-cell imaging of RRAS activity using biosensors

      • Simultaneously track individual cell migration parameters

      • Apply FITC-conjugated RRAS antibodies in fixed timepoint analyses

    • Advanced analytics:

      • Calculate correlation at single-cell level between RRAS parameters and migration metrics

      • Identify cell subpopulations with distinct signaling-migration relationships

      • Apply machine learning algorithms to discover complex relationships

  • Spatial signal integration:

    • Experimental approach:

      • Use high-resolution imaging to map RRAS activity in subcellular regions

      • Correlate RRAS localization with migration structures (leading edge, focal adhesions)

      • Implement fluorescence ratio imaging to detect active vs. total RRAS

    • Analytical methods:

      • Perform spatial correlation analyses between RRAS activity and membrane protrusions

      • Quantify temporal relationships between local RRAS activation and directional changes

      • Develop computational models that predict migration based on RRAS signaling patterns

  • Comprehensive data integration framework:

    • Data structure:

      ParameterBaselineRRAS ActivationRRAS InhibitionPI3-K Inhibition
      RRAS-GTP levels1.03.2 ± 0.40.3 ± 0.10.9 ± 0.2
      β1 integrin activation1.02.8 ± 0.30.4 ± 0.10.5 ± 0.1
      FAK phosphorylation1.02.5 ± 0.40.3 ± 0.10.4 ± 0.1
      Migration velocity (μm/hr)12 ± 328 ± 55 ± 27 ± 2
      Directional persistence0.4 ± 0.10.7 ± 0.10.2 ± 0.10.3 ± 0.1
    • Visualization approaches:

      • Generate correlation matrices between all measured parameters

      • Develop pathway maps with overlay of functional consequences

      • Create predictive models that translate signaling states to migration outcomes

This integrated approach enables researchers to establish mechanistic connections between RRAS signaling events and their functional consequences in cell migration, providing deeper insights into how this GTPase regulates cellular behavior.

What are emerging techniques for studying RRAS dynamics in live cells?

The field of RRAS research is advancing rapidly with several innovative techniques for studying dynamics in live cells:

  • FRET-based RRAS activity biosensors:

    • Design principles: Intramolecular sensors containing the RRAS protein flanked by fluorescent proteins (e.g., CFP/YFP pair) with an RBD domain that binds GTP-RRAS

    • Applications: Real-time visualization of RRAS activation with subcellular resolution

    • Advantages: Provides spatiotemporal information unobtainable with biochemical assays

    • Considerations: Requires optimization of linker sequences; potential interference with normal RRAS function

  • Optogenetic control of RRAS activity:

    • Implementation: Light-sensitive domains (CRY2/CIB, LOV) fused to RRAS regulators (GEFs, GAPs)

    • Applications: Precise temporal and spatial control of RRAS activation/inactivation

    • Advantages: Allows direct testing of cause-effect relationships between RRAS activity and cellular responses

    • Integration with imaging: Combine with FITC-conjugated antibodies for fixed timepoint validation

  • Advanced microscopy techniques:

    • Super-resolution microscopy: Techniques like STORM, PALM, or STED to visualize RRAS nanoclusters

    • Lattice light-sheet microscopy: Reduced phototoxicity for long-term imaging of RRAS dynamics

    • Fluorescence correlation spectroscopy (FCS): Measurement of RRAS diffusion and interaction kinetics

    • Considerations: May require specialized RRAS labeling strategies beyond traditional antibodies

  • Genome editing for endogenous tagging:

    • CRISPR-Cas9 approaches: Knock-in of fluorescent tags or split reporters at the endogenous RRAS locus

    • Applications: Study RRAS dynamics without overexpression artifacts

    • Validation: Use FITC-conjugated RRAS antibodies to verify tagged protein localization

    • Advanced systems: Conditional degron tags for acute RRAS depletion

  • Single-molecule tracking:

    • Implementation: Sparse labeling of RRAS with photoconvertible fluorophores or quantum dots

    • Applications: Track individual RRAS molecules to determine diffusion characteristics, confinement zones

    • Integration: Correlate with functional markers of integrin activation or migration structures

  • Emerging protein-protein interaction techniques:

    • Split luciferase complementation: Real-time detection of RRAS interactions with effectors or regulators

    • Proximity biotinylation: BioID or TurboID fused to RRAS to identify proximal proteins in living cells

    • Advanced FRET approaches: Three-color FRET to study RRAS within multiprotein complexes

  • Next-generation antibody technologies:

    • Nanobodies: Small single-domain antibodies conjugated to FITC for reduced perturbation of living systems

    • Intrabodies: Antibody fragments expressed intracellularly to track RRAS in specific compartments

    • Considerations: Validation against traditional FITC-conjugated antibodies for consistency

These emerging techniques promise to provide unprecedented insights into RRAS dynamics and functional relationships in living cells, potentially revealing new mechanisms in integrin regulation and cell migration.

How might artificial intelligence and machine learning enhance the analysis of RRAS-integrin signaling data?

Artificial intelligence and machine learning approaches are revolutionizing the analysis of complex signaling networks like the RRAS-integrin axis:

These AI/ML approaches promise to accelerate discovery by extracting maximal information from complex datasets, generating testable hypotheses, and guiding experimental design in RRAS-integrin research.

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.