RHOH Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to RHOH Antibody

RHOH antibodies are specialized immunoglobulins designed to recognize and bind to RhoH protein, a member of the Ras superfamily of GTP-metabolizing enzymes. These antibodies serve as valuable research tools for investigating the expression, localization, and function of RhoH in various biological contexts. Available in multiple formats including polyclonal, monoclonal, and conjugated variants, RHOH antibodies enable researchers to study this protein using techniques such as Western blotting, immunohistochemistry, immunofluorescence, and ELISA .

The target of these antibodies, RhoH (also known as ARHH or TTF), is predominantly expressed in hematopoietic cells where it functions as a negative regulator of cell growth and survival . Unlike typical Rho GTPases, RhoH is constitutively GTP-bound and lacks intrinsic GTPase activity, making it resistant to Rho-specific GTPase-activating proteins . This unique characteristic positions RhoH as an atypical member of the Rho GTPase family with distinct regulatory functions in immune cell signaling.

Cellular Functions

RhoH serves as a critical regulator of multiple immune cell functions, particularly in T cells. Research has demonstrated that RhoH:

  • Acts as a negative regulator of hematopoietic progenitor cell proliferation, survival, and migration

  • Functions as a critical regulator of thymocyte development and T-cell antigen receptor (TCR) signaling by mediating recruitment and activation of ZAP70

  • Is required for phosphorylation of CD3Z, membrane translocation of ZAP70, and subsequent activation of ZAP70-mediated pathways

  • Plays crucial roles in mast cell function by facilitating phosphorylation of SYK in Fc epsilon RI-mediated signal transduction

  • Inhibits the activation of NF-kappa-B by TNF and IKKB and the activation of CRK/p38 by TNF

  • Acts as a molecular brake on actomyosin-mediated neutrophil effector functions, representing an intracellular regulatory axis involved in controlling the strength of antibacterial inflammatory responses

Subcellular Localization

RhoH is primarily localized in the cytoplasm and cell membrane. Notably, it co-localizes with ZAP70 in the immunological synapse . Research has also shown that RhoH and the transcription factor Kaiso co-localize at chemokine-induced actin-containing cell protrusion sites and can translocate to the nucleus in a time-dependent fashion after chemokine stimulation and T cell receptor activation .

Conjugated RHOH antibodies feature chemical modifications that facilitate direct detection without the need for secondary antibodies. The most common conjugation is with fluorescent dyes such as FITC (Fluorescein isothiocyanate), which enables direct visualization in fluorescence microscopy applications .

Key specifications of FITC-conjugated RHOH antibody include:

  • Host: Rabbit

  • Species reactivity: Human

  • Applications: ELISA (other applications may need validation)

  • Immunogen: Recombinant human Rho-related GTP-binding protein RhoH protein (amino acids 1-188)

  • Storage buffer: Contains preservative (0.03% Proclin 300), 50% Glycerol, and 0.01M PBS, pH 7.4

  • Storage conditions: Shipped at 4°C; store at -20°C or -80°C after aliquoting

Recommended Dilutions for Applications

Optimal antibody dilutions vary depending on the specific application and the antibody manufacturer. Typical recommended dilutions for RHOH antibodies include:

ApplicationRecommended DilutionsReference
Western Blot (WB)1:500-1:2000
Immunohistochemistry (IHC)1:40-1:200
Immunofluorescence (IF)1:100-1:500
ELISA1:2000-1:5000

RHOH in T Cell Development and Function

Research using RHOH antibodies has significantly advanced our understanding of T cell biology. Studies in Rhoh-deficient mice have revealed that RhoH plays a crucial role in T cell development and function:

  • Rhoh-deficient T cells exhibit defects in TCR signaling during thymic development

  • In murine allogenic kidney transplantation, Rhoh deficiency caused a significant 75% reduction of acute and chronic transplant rejection accompanied by 75% lower alloantigen-specific antibody levels and significantly better graft function

  • Rhoh deficiency is associated with 85% lower cytotoxicity and 50-80% lower cytokine release in T cells

  • Human RHOH deficiency leads to T cell defects and susceptibility to persistent EV-HPV infections, suggesting that T cells play a role in the pathogenesis of chronic EV-HPV infections

RHOH Protein Interactions

Immunoprecipitation experiments using RHOH antibodies have identified several protein interactions that shed light on RhoH function:

  • RhoH interacts with Kaiso, a 95 kDa dual-specific transcription factor that regulates gene expression and p120 catenin-associated cell-cell adhesions

  • RhoH, Kaiso, and p120 catenin co-localize at chemokine-induced actin-containing cell protrusion sites

  • RhoH knockdown leads to altered Kaiso localization, demonstrating that Kaiso localization depends on RhoH function

  • RhoH and Kaiso co-localize to the nucleus in a time-dependent fashion after chemokine stimulation and with T cell receptor activation

  • RhoH interacts with non-muscle myosin heavy chain IIA (NMHC IIA) in activated neutrophils to inhibit the transport of mitochondria and granules along actin filaments

RHOH in Neutrophil Function

Recent research has uncovered a previously undefined role of RhoH in neutrophil function:

  • RhoH limits neutrophil degranulation and the formation of neutrophil extracellular traps (NETs)

  • RhoH is induced under inflammatory conditions and binds to NMHC IIA in activated neutrophils

  • RhoH inhibits actin polymerization presumably by modulating RAC1 activity

  • Studies using Rhoh-/- mice demonstrate an increased antibacterial defense capability against Escherichia coli, indicating that RhoH acts as a molecular brake for actomyosin-mediated neutrophil effector functions

Future Directions in RHOH Antibody Research

The continued development and application of RHOH antibodies promise to advance our understanding of immune cell regulation and may lead to novel therapeutic strategies:

  1. The role of RhoH in immune-related diseases requires further investigation, particularly given its importance in T cell function and neutrophil activity.

  2. The potential of targeting RhoH in transplantation medicine deserves exploration, considering the significant reduction in transplant rejection observed in Rhoh-deficient models.

  3. The development of more specific antibodies targeting different epitopes or post-translational modifications of RhoH could provide new insights into its regulation and function.

  4. Further studies of human RHOH deficiency may reveal new therapeutic targets for treating persistent viral infections and other immune-related disorders.

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide.
Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method or location. Please consult your local distributors for specific delivery times.
Synonyms
ARHH antibody; GTP binding protein TTF antibody; GTP-binding protein TTF antibody; OTTHUMP00000125213 antibody; Ras homolog gene family member H antibody; Rho related GTP binding protein antibody; Rho related GTP binding protein RhoH antibody; Rho-related GTP-binding protein RhoH antibody; RHOH antibody; RHOH_HUMAN antibody; Translocation three four protein antibody; TTF antibody; TTF translocation three four antibody
Target Names
Uniprot No.

Target Background

Function
RHOH Antibody is a negative regulator of hematopoietic progenitor cell proliferation, survival, and migration. It plays a critical role in thymocyte development and T-cell antigen receptor (TCR) signaling by mediating the recruitment and activation of ZAP70. RHOH Antibody is essential for the phosphorylation of CD3Z, the membrane translocation of ZAP70, and subsequent activation of ZAP70-mediated pathways. It is crucial for efficient beta-selection and positive selection by promoting the ZAP70-dependent phosphorylation of the LAT signalosome during pre-TCR and TCR signaling. RHOH Antibody is vital for thymocyte maturation during the DN3 to DN4 transition and during positive selection. It also plays critical roles in mast cell function by facilitating the phosphorylation of SYK in Fc epsilon RI-mediated signal transduction. RHOH Antibody is essential for the phosphorylation of LAT, LCP2, PLCG1, and PLCG2 and for Ca(2+) mobilization in mast cells. It binds GTP but lacks intrinsic GTPase activity and is resistant to Rho-specific GTPase-activating proteins. RHOH Antibody inhibits the activation of NF-kappa-B by TNF and IKKB and the activation of CRK/p38 by TNF. It also inhibits the activities of RAC1, RHOA, and CDC42. Finally, it negatively regulates leukotriene production in neutrophils.
Gene References Into Functions
  1. Extracellular microenvironment signals regulate RhoH and Kaiso to modulate actin-cytoskeleton structure and transcriptional activity during T cell migration. PMID: 27574848
  2. These results highlight a novel negative regulatory role for RhoH in eosinophil differentiation, most likely due to altered GATA-2 levels. PMID: 27740624
  3. Mutations in hematopoiesis-specific Rho GTPases Rac2 and RhoH lead to a wide range of human blood disorders. (Review) PMID: 23850828
  4. Our current results indicate differential requirements for RhoH in the development of TCRalphabeta CD8alphaalpha IELs compared to other subsets of T cells, including agonist selected T cells. PMID: 23499578
  5. RHOH deficiency leads to T cell defects and persistent epidermodysplasia verruciformis-HPV infections. PMID: 22850876
  6. We conclude that RhoH expression provides a key molecular determinant that allows T cells to switch between sensing chemokine-mediated go signals and TCR-dependent stop signals. PMID: 22689994
  7. RhoH plays a critical role in prosurvival chronic lymphocytic leukemia cell-cell and cell-microenvironment interactions with this agent. PMID: 22474251
  8. Repression of the RHOH gene by JunD. PMID: 21473742
  9. RhoH is a key adapter protein that maintains LCK in the inactive state, contributing to the regulation of both pre-TCR and TCR signaling during T-cell development. PMID: 20851766
  10. We propose that RhoH functions as a negative regulator for IL3-induced signals through modulation of the JAK-STAT pathway. PMID: 20738848
  11. Data suggest that RhoH function might be regulated by lysosomal degradation of RhoH protein following TCR complex but not BCR activation. PMID: 19950172
  12. RhoH has a function in the progression of B-cell chronic lymphocytic leukemia (CLL) in a murine model and show RhoH expression is altered in human primary lymphocytic (CLL) samples. PMID: 19847197
  13. 5' end heterogeneity of RhoH mRNAs was observed, due to alternative splicing of some 5' exons and to the use of these different transcription start sites. PMID: 15563831
  14. In both Richter's transformation and prolymphocytic transformation, high-levels of AID mRNA expression and high-frequency mutations of RhoH protein genes were detected. PMID: 16541139
  15. Aberrant somatic hypermutation of RhoH gene is associated with diffuse large B cell lymphoma. PMID: 17443219
  16. RHOH mutation is associated with splenic and nodal marginal zone B-cell lymphomas. PMID: 17476282
  17. Underexpression of RhoH is associated with Hairy Cell Leukemia. PMID: 18559497
  18. Low expression of the RhoH transcript was a predictor of worse prognosis in both overall and disease-free survival. PMID: 18691253
  19. RhoH/TTF represents an inducible feedback inhibitor in neutrophils that is involved in the limitation of innate immune responses. PMID: 19414807

Show More

Hide All

Database Links

HGNC: 686

OMIM: 602037

KEGG: hsa:399

STRING: 9606.ENSP00000371219

UniGene: Hs.654594

Involvement In Disease
A chromosomal aberration involving RHOH is found in a non-Hodgkin lymphoma cell line. Translocation t(3;4)(q27;p11) with BCL6.
Protein Families
Small GTPase superfamily, Rho family
Subcellular Location
Cytoplasm. Cell membrane; Lipid-anchor; Cytoplasmic side.
Tissue Specificity
Expressed only in hematopoietic cells. Present at very high levels in the thymus, less abundant in the spleen, and least abundant in the bone marrow. Expressed at a higher level in the TH1 subtype of T-helper cells than in the TH2 subpopulation. Expressed

Q&A

What is RHOH and why are antibodies against it important in research?

RHOH (also known as ARHH or TTF) is a hematopoietic-specific member of the Rho family of small GTPases that functions as a negative regulator of hematopoietic progenitor cell proliferation, survival, and migration. Unlike typical Rho GTPases, RHOH is GTPase deficient, remaining constitutively in a GTP-bound activated state without cycling . This unique characteristic makes it an important subject for immunological research.

RHOH antibodies are critical research tools because they enable the detection, quantification, and localization of RHOH protein in experimental systems. These antibodies help researchers investigate RHOH's crucial roles in T-cell development and T-cell antigen receptor (TCR) signaling, particularly through its mediation of ZAP70 recruitment and activation . Additionally, RHOH antibodies facilitate the study of how this protein regulates immune cell functions in both normal physiological conditions and disease states.

What are the main applications of RHOH antibodies in laboratory research?

RHOH antibodies can be utilized in multiple laboratory techniques depending on the specific research question:

ApplicationDescriptionCommon FormatsRecommended Dilutions
Western Blot (WB)Detection of RHOH protein in cell/tissue lysatesPolyclonal rabbit antibodies1:1,000 - 1:2,000
Immunohistochemistry (IHC-P)Visualization of RHOH in fixed tissue sectionsPolyclonal rabbit antibodies1:100 - 1:500
Immunocytochemistry/Immunofluorescence (ICC/IF)Cellular localization of RHOH proteinPolyclonal rabbit antibodies1:100 - 1:400
ELISAQuantification of RHOH in solutionPolyclonal rabbit antibodiesAs recommended by manufacturer
Immunoprecipitation (IP)Isolation of RHOH and associated proteinsHA-tagged antibodies1:50 - 1:100

These applications allow researchers to study RHOH expression levels, localization patterns, protein-protein interactions, and signaling pathway involvement in various experimental settings .

What are the key structural and functional characteristics of RHOH protein that antibodies typically target?

RHOH possesses several structural and functional domains that antibodies may recognize. The protein contains regions that are critical for its unique signaling properties:

RHOH is characterized by key functional elements:

  • GTP-binding domain (but with mutations in critical GTPase activity residues)

  • A unique carboxyl-terminal insert domain that distinguishes it from other Rho GTPases

  • Interaction interfaces for binding partners like ZAP70

Functionally, RHOH serves as:

  • A negative regulator of hematopoietic progenitor cell functions

  • A critical regulator of thymocyte development and TCR signaling

  • A mediator for ZAP70 recruitment and activation

  • A required component for CD3Z phosphorylation and ZAP70 membrane translocation

  • An essential factor for the phosphorylation of LAT, LCP2, PLCG1, and PLCG2 in mast cells

  • An inhibitor of RAC1, RHOA, and CDC42 activities

Most commercial antibodies are generated against synthetic peptides or recombinant proteins corresponding to regions within human RHOH, often conjugated to carriers like Keyhole Limpet Hemocyanin for enhanced immunogenicity .

How do RHOH expression levels change during T-cell development and activation?

RHOH expression exhibits dynamic regulation during T-cell development and activation, which is critical for researchers to understand when designing experiments:

During T-cell development:

  • RHOH is crucial for thymocyte maturation during the double-negative (DN) 3 to DN4 transition

  • It is essential for efficient beta-selection and positive selection processes

  • RHOH promotes ZAP70-dependent phosphorylation of the LAT signalosome during pre-TCR and TCR signaling

During T-cell activation:

  • RHOH levels can be dramatically downregulated after phorbol myristate acetate (PMA) treatment

  • In Th1 cells, RHOH is downregulated after anti-CD3-mediated activation

  • RHOH is expressed at higher levels in Th1 cells compared to Th2 cells under basal conditions

The transcriptional regulation of RHOH provides a mechanism for controlling its function, as it lacks the typical GTP/GDP cycling seen in other Rho GTPases. This regulation at the mRNA level allows for modulation of RHOH's inhibitory activities on other signaling pathways . Researchers should account for these dynamic changes when designing experiments to study RHOH's role in T-cell biology.

What are the technical considerations when using RHOH antibodies for co-immunoprecipitation experiments?

When conducting co-immunoprecipitation (co-IP) experiments to study RHOH interactions, researchers should consider several technical aspects:

Buffer Composition:

  • Use lysis buffers containing 8% glycerol to stabilize protein complexes

  • Include phosphatase inhibitors (10 mM sodium fluoride, 1 mM sodium orthovanadate)

  • Add complete protease inhibitor mixture to prevent protein degradation

  • Consider using specialized buffers like Ca2+ lysis/wash buffer for certain applications

Controls and Validation:

  • Include negative controls using non-specific antibodies of the same isotype

  • Perform reverse IP experiments to confirm interactions

  • Validate RHOH antibody specificity using RHOH-deficient cell lines or knockdown samples

  • Consider using epitope-tagged RHOH (e.g., HA-tagged) for more specific pull-downs

Detection Methods:

  • For immunoblotting of precipitated complexes, use 12% SDS-polyacrylamide gels

  • Transfer to PVDF membranes for optimal protein retention

  • Use anti-RHOH antibodies at 1:1,000 dilution for detection

  • Include loading controls such as β-actin (1:10,000 dilution)

Known Interacting Partners:
RHOH co-IP experiments have successfully identified several interacting partners:

Interacting PartnerFunctionDetection MethodReference
ZAP70T-cell signaling kinaseCo-IP with anti-RHOH or anti-ZAP70
RACK1Scaffold proteinCo-IP with anti-RHOH
NMHC IIAMyosin heavy chainCo-IP with anti-RHOH or anti-NMHC IIA
p120 catenin/KaisoTranscriptional regulationCo-IP with anti-RHOH

These considerations will help ensure successful co-IP experiments when studying RHOH protein interactions.

How can researchers investigate the role of RHOH in neutrophil function using antibodies?

Recent research has revealed RHOH's involvement in neutrophil function, particularly in the context of inflammation and NET (Neutrophil Extracellular Trap) formation. Researchers can investigate this role using several antibody-based approaches:

Experimental Design for Neutrophil Studies:

  • RHOH Expression Analysis:

    • Immunoblotting with anti-RHOH antibodies to assess expression levels in resting versus activated neutrophils

    • Flow cytometry with intracellular staining to quantify RHOH at the single-cell level

    • Compare RHOH expression in neutrophils under various inflammatory stimuli

  • RHOH-NMHC IIA Interaction Studies:

    • Co-immunoprecipitation to detect physical interaction between RHOH and NMHC IIA

    • Confocal microscopy using fluorescently labeled antibodies to visualize co-localization

    • Proximity ligation assays for more sensitive detection of protein-protein interactions

  • Functional Assays with RHOH Modulation:

    • NET formation assessment: Confocal microscopy using DNA stains combined with neutrophil elastase (NE) antibodies

    • Quantification of released double-stranded DNA in culture supernatants

    • Neutrophil degranulation analysis by flow cytometry

    • Comparison between wild-type, RHOH-knockdown, or RHOH-overexpressing neutrophils

Research findings indicate that RHOH is induced under inflammatory conditions and can bind to NMHC IIA in activated neutrophils, acting as a molecular brake on actomyosin-mediated neutrophil functions. Experiments have shown that reduction of RHOH levels leads to enhanced neutrophil degranulation and NET formation in response to stimuli such as GM-CSF in combination with C5a . These findings suggest that RHOH plays an important regulatory role in restraining excessive neutrophil activation during inflammatory responses.

What approaches can be used to study RHOH protein stability and degradation?

RHOH protein regulation can occur post-translationally through degradation pathways. Researchers can investigate these mechanisms using antibody-based techniques combined with inhibitor studies:

Experimental Approaches:

  • Protein Stability Assays:

    • Treat cells with cycloheximide (50 μM CHX) to inhibit protein synthesis

    • Collect samples at various time points (0, 1, 3, and 6 hours)

    • Perform immunoblot analysis using anti-RHOH or anti-HA antibodies (for tagged RHOH)

    • Include β-actin as loading control and reference

    • Calculate protein half-life based on degradation kinetics

  • Degradation Pathway Identification:

    • Treat cells with various inhibitors:

      • Proteasome inhibitors (30 μM MG132, 30 μM MG115, 50 μM ALLN)

      • Caspase inhibitors (30 μM)

      • AICAR (1 mM) to inhibit autophagy

    • Compare RHOH protein levels by immunoblotting

    • Determine which pathway predominantly contributes to RHOH turnover

  • Lysosomal Degradation Assessment:

    • Co-localization studies with lysosomal markers using immunofluorescence

    • Density gradient fractionation followed by immunoblot analysis

    • Treatment with lysosomal inhibitors (e.g., chloroquine, bafilomycin A1)

Research Findings on RHOH Stability:

Studies have demonstrated that RHOH can be regulated through protein degradation mechanisms, which may be particularly important given its constitutively active nature. Evidence suggests that RHOH levels can be modulated through:

  • Proteasomal degradation pathways

  • Lysosomal degradation

  • Regulation through somatic mutations and transcriptional repressors

Understanding these degradation mechanisms provides insight into how this GTPase-deficient protein can be regulated at the post-translational level, complementing its known transcriptional regulation.

How can researchers validate the specificity of RHOH antibodies in their experimental systems?

Validating antibody specificity is crucial for ensuring reliable experimental results. For RHOH antibodies, researchers should implement several validation strategies:

Recommended Validation Methods:

  • Genetic Approaches:

    • Compare antibody reactivity in wild-type versus RHOH knockout or knockdown models

    • Use shRNA-mediated depletion (e.g., Myh9 shRNA has been used in related studies)

    • Overexpress RHOH in a non-expressing cell line and confirm detection

  • Peptide Competition Assays:

    • Pre-incubate the antibody with the immunizing peptide

    • Run parallel experiments with blocked and unblocked antibody

    • Loss of signal indicates specificity for the target epitope

  • Multiple Antibody Validation:

    • Use different antibodies targeting distinct epitopes of RHOH

    • Compare results using monoclonal and polyclonal antibodies

    • Consistent results across antibodies suggest specific detection

  • Cross-Reactivity Testing:

    • Test antibody against related Rho family members (RhoA, Rac1, Cdc42)

    • Ensure antibody doesn't detect these structurally similar proteins

  • Application-Specific Validation:

    • For Western blotting: Confirm single band at expected molecular weight (~21 kDa)

    • For IHC/ICC: Include positive controls (lymphoid tissues) and negative controls (non-hematopoietic tissues)

    • For flow cytometry: Compare with isotype controls and known expression patterns

Validation Data Example:

Validation MethodExpected Result for Specific AntibodyPotential Issues
RHOH knockdownReduced/absent signalIncomplete knockdown may give partial signal
Peptide competitionSignal eliminationNon-specific binding may persist
Western blot sizingSingle band at ~21 kDaMultiple bands suggest cross-reactivity
Tissue specificitySignal in hematopoietic tissues onlySignal in non-hematopoietic tissues indicates non-specificity
Tagged proteinDetection matches tag detectionDiscrepancy suggests antibody problems

Following these validation procedures will help ensure that experimental results accurately reflect RHOH biology rather than antibody artifacts.

What are the best experimental designs to study RHOH's role in hematopoietic malignancies?

RHOH has been implicated in various B-cell malignancies, with non-coding mutations found in B-cell lymphomas . Researchers investigating RHOH's role in these diseases can employ several antibody-based experimental approaches:

Experimental Designs for Malignancy Studies:

  • Expression Analysis in Clinical Samples:

    • Immunohistochemistry on lymphoma tissue microarrays using RHOH antibodies

    • Western blot analysis of patient-derived samples versus normal controls

    • Flow cytometry to assess RHOH levels in primary malignant B cells

    • Correlation of RHOH expression with clinical outcomes and disease subtypes

  • Functional Studies in Cell Models:

    • Modulation of RHOH levels (overexpression or knockdown) in lymphoma cell lines

    • Assessment of proliferation, survival, and migration phenotypes

    • Evaluation of signaling pathway alterations using phospho-specific antibodies

    • Analysis of RHOH's impact on transcriptional programs (e.g., BCL6 gene regulation)

  • Interaction Studies in Malignant Contexts:

    • Co-immunoprecipitation to identify altered binding partners in malignant versus normal B cells

    • Assessment of RHOH-Kaiso-p120 catenin complex formation and its impact on Rac1 signaling

    • Investigation of RHOH's effects on p53 tumor suppressor levels through BCL6 repression

Research Findings on RHOH in Malignancies:

Studies have shown that RHOH can function as a tumor suppressor in hematopoietic cells by:

  • Inhibiting cell survival, migration, and invasion

  • Repressing the BCL6 gene through interaction with Kaiso, leading to increased p53 expression

  • Attenuating Rac1 signaling, which is often hyperactivated in malignancies

Interestingly, in non-hematopoietic contexts (e.g., prostate cancer), RHOH has been reported to promote cell migratory polarity by directing Rac1 and PAK2 to membrane protrusions, highlighting the context-dependent nature of its functions .

How can RHOH antibodies be used to study its role in immune-related diseases?

Beyond cancer, RHOH has been implicated in various immune-related diseases, including primary immunodeficiencies, systemic lupus erythematosus, and psoriasis . Researchers can utilize RHOH antibodies to investigate these connections:

Research Approaches for Immune Disease Studies:

  • Comparative Expression Analysis:

    • Quantify RHOH protein levels in patient samples versus healthy controls

    • Compare expression across different immune cell subsets in disease states

    • Correlate RHOH levels with disease activity markers

  • Functional Studies in Patient-Derived Cells:

    • Isolate primary T cells or other immune cells from patients

    • Assess TCR signaling strength using phospho-flow cytometry

    • Measure ZAP70 recruitment and activation in response to stimulation

    • Evaluate the impact of disease-associated RHOH variants on protein function

  • Signaling Pathway Analysis:

    • Investigate RHOH interactions with known partners (ZAP70, Vav1, RhoGDI)

    • Examine cross-talk with other small GTPases (RhoA, Rac1, Cdc42)

    • Assess NFκB and p38 pathway activation states in relation to RHOH levels

  • Therapeutic Target Exploration:

    • Screen for compounds that modulate RHOH expression or function

    • Evaluate the impact of current immunomodulatory drugs on RHOH levels

    • Develop tools to normalize RHOH expression in disease states

Research Findings in Immune-Related Diseases:

Studies have revealed that RHOH dysfunction can contribute to immune dysregulation through:

  • Altered T-cell development and reduced T-cell responsiveness

  • Impaired ZAP70-dependent signaling and LAT signalosome formation

  • Dysregulated neutrophil function and NET formation

  • Abnormal inhibition of other Rho GTPases, affecting immune cell migration and function

These findings suggest that RHOH serves as a critical regulator of immune homeostasis, and its dysregulation may contribute to both immunodeficiency and autoimmune pathologies.

What are the best methods for quantifying RHOH protein levels using antibody-based techniques?

Accurate quantification of RHOH protein levels is essential for many research applications. Several antibody-based methods can be employed, each with specific advantages:

Quantitative Methods Comparison:

MethodAdvantagesLimitationsSample Requirements
Western Blot- Semi-quantitative
- Allows size verification
- Compatible with lysates
- Lower throughput
- Requires normalization
10-30 μg total protein
ELISA- Highly quantitative
- High throughput
- Good for multiple samples
- No size verification
- Requires validated kit
Purified protein or lysate
Flow Cytometry- Single-cell resolution
- Allows subset analysis
- Can combine with other markers
- Requires cell fixation/permeabilization
- Less absolute quantification
1x10^6 cells per sample
Mass Spectrometry- Absolute quantification
- No antibody dependence
- Complex workflow
- Expensive equipment
- Requires sample enrichment
Highly purified samples

Recommended Protocols for RHOH Quantification:

  • Western Blot Quantification:

    • Use RIPA buffer with protease inhibitors for cell lysis

    • Separate proteins on 12% SDS-PAGE gels

    • Transfer to PVDF membranes

    • Block with 5% non-fat milk

    • Incubate with RHOH antibody (1:1,000)

    • Use β-actin as loading control (1:10,000)

    • Employ HRP-coupled secondary antibodies (1:2,000)

    • Detect using chemiluminescence

    • Quantify band intensity using image analysis software

  • Flow Cytometry Protocol:

    • Fix cells with 4% paraformaldehyde

    • Permeabilize with 0.1% Triton X-100

    • Block with 2% BSA

    • Stain with anti-RHOH antibody at manufacturer-recommended dilution

    • Use fluorophore-conjugated secondary antibody

    • Include appropriate isotype controls

    • Analyze by standard flow cytometry, quantifying mean fluorescence intensity

When comparing RHOH levels across different conditions or samples, researchers should implement appropriate normalization strategies and statistical analyses to ensure meaningful quantitative comparisons.

How can researchers optimize immunofluorescence techniques for studying RHOH localization?

Immunofluorescence and confocal microscopy are valuable techniques for studying RHOH subcellular localization and co-localization with interacting partners. Optimizing these approaches requires attention to several key parameters:

Optimization Strategies:

  • Fixation Methods:

    • 4% paraformaldehyde (10-15 minutes) preserves most epitopes and cellular structures

    • Methanol fixation (-20°C, 10 minutes) may better expose some intracellular epitopes

    • Test both methods to determine optimal epitope accessibility for your RHOH antibody

  • Permeabilization Conditions:

    • 0.1-0.5% Triton X-100 (5-10 minutes) for cytoplasmic proteins

    • 0.05-0.1% saponin for membrane-associated proteins

    • Optimize concentration and time for best signal-to-noise ratio

  • Blocking Parameters:

    • 5-10% normal serum (species of secondary antibody)

    • 1-3% BSA in PBS with 0.1% Tween-20

    • Include 0.1-0.3 M glycine to reduce background from aldehyde fixation

  • Antibody Incubation:

    • Primary RHOH antibody: Test dilutions from 1:50 to 1:500

    • Incubation time: 1 hour at room temperature or overnight at 4°C

    • Secondary antibody: Typically 1:200 to 1:1,000 for 1 hour at room temperature

    • Include appropriate controls (no primary, isotype control, known positive/negative samples)

  • Co-localization Studies:

    • For T-cell studies: Co-stain for ZAP70, CD3, or LAT

    • For neutrophil studies: Co-stain for NMHC IIA, neutrophil elastase, or CD63

    • For nuclear translocation: Co-stain with nuclear markers (DAPI, Hoechst)

    • Use secondary antibodies with spectrally distinct fluorophores

Analysis Recommendations:

  • Quantify co-localization using Pearson's coefficient in colocalized volume

  • Use software like Imaris for numerical analysis of co-localization

  • Perform z-stack imaging to capture the full 3D distribution of RHOH

  • Consider super-resolution microscopy (STED, STORM) for detailed localization studies

Research findings using these techniques have revealed important insights about RHOH localization, including its co-localization with NMHC IIA in activated neutrophils and its involvement in directing ZAP70 to the membrane in T cells .

What are the latest findings regarding RHOH's role in neutrophil function and inflammatory responses?

Recent research has uncovered new roles for RHOH in neutrophil biology and inflammatory responses that were previously unrecognized. These discoveries provide new avenues for investigation using RHOH antibodies:

Recent Research Findings:

Recent studies have revealed that RHOH acts as a molecular brake on actomyosin-mediated activation of neutrophils . Specifically:

  • RHOH is induced under inflammatory conditions in neutrophils

  • It physically binds to non-muscle myosin heavy chain IIA (NMHC IIA)

  • This interaction inhibits excessive neutrophil activation

  • RHOH deficiency leads to enhanced neutrophil degranulation and NET formation in response to stimuli like GM-CSF combined with C5a

Experimental Data:

Studies using various inhibitors and genetic approaches have shown that:

Experimental ConditionEffect on Neutrophil FunctionMethod of Assessment
RHOH knockdownIncreased degranulationFlow cytometry
RHOH knockdownEnhanced NET formationConfocal microscopy, DNA quantification
Myosin IIA inhibitionReduced neutrophil activationConfocal microscopy, flow cytometry
NMHC IIA knockdownAltered neutrophil responsesImmunoblot analysis

These findings suggest that RHOH plays an important regulatory role in restraining excessive neutrophil activation during inflammatory responses, representing a novel mechanism by which immune responses are fine-tuned.

How can researchers use RHOH antibodies to study its transcriptional regulation?

Since RHOH lacks the typical GTP/GDP cycling mechanism for regulation, transcriptional control is a primary means of modulating its activity . Researchers can investigate this unique regulatory mechanism using several approaches:

Research Strategies:

  • Expression Analysis Across Cell States:

    • Compare RHOH protein levels using antibodies in:

      • Resting versus activated T cells

      • Th1 versus Th2 T-cell subtypes

      • Different stages of thymocyte development

      • Various stimulation conditions (PMA, anti-CD3, cytokines)

  • Correlation of Protein and mRNA Levels:

    • Perform parallel analysis of RHOH protein (by western blot) and mRNA (by qRT-PCR)

    • Determine if changes in protein levels correspond to transcriptional regulation

    • Assess the kinetics of protein versus mRNA changes during cell activation

  • Transcription Factor Studies:

    • Identify potential transcription factor binding sites in the RHOH promoter

    • Use chromatin immunoprecipitation (ChIP) to detect transcription factor binding

    • Correlate transcription factor activity with RHOH protein levels detected by antibodies

  • Epigenetic Regulation:

    • Investigate DNA methylation and histone modifications at the RHOH locus

    • Determine how these epigenetic marks correlate with RHOH protein expression

    • Study the effects of epigenetic modifiers on RHOH protein levels

Research Findings:

Studies have shown that:

  • RHOH is dramatically downregulated after phorbol myristate acetate (PMA) treatment

  • In Th1 cells, RHOH is downregulated after anti-CD3-mediated activation

  • RHOH is expressed at higher levels in Th1 cells compared to Th2 cells under basal conditions

  • Modulation of RHOH mRNA levels can alter the effective activities of other Rho GTPases

These findings highlight the importance of transcriptional regulation as a mechanism for controlling RHOH function in hematopoietic cells.

What are common problems encountered when using RHOH antibodies and how can they be resolved?

Researchers working with RHOH antibodies may encounter several technical challenges. Here are common issues and their solutions:

Common Problems and Solutions:

  • Weak or No Signal:

    • Potential Causes:

      • Insufficient antigen

      • Epitope masking during fixation

      • Low antibody concentration

      • Degraded antibody

    • Solutions:

      • Increase protein loading (for WB) or cell concentration

      • Try alternative fixation methods

      • Optimize antibody concentration

      • Use fresh antibody aliquots

      • Include protease inhibitors during sample preparation

      • For immunoprecipitation, include 8% glycerol in lysis buffer

  • High Background:

    • Potential Causes:

      • Insufficient blocking

      • Too high antibody concentration

      • Non-specific binding

      • Cross-reactivity

    • Solutions:

      • Increase blocking time or concentration

      • Optimize antibody dilution

      • Add 0.1-0.3% Tween-20 to wash buffers

      • Pre-absorb antibody with cell/tissue lysate from non-expressing samples

  • Multiple Bands in Western Blot:

    • Potential Causes:

      • Cross-reactivity with related proteins

      • Protein degradation

      • Post-translational modifications

    • Solutions:

      • Validate with RHOH-deficient controls

      • Add protease inhibitors to prevent degradation

      • Consider if bands represent physiologically relevant modifications

      • Use gradient gels for better separation

  • Inconsistent Results Between Experiments:

    • Potential Causes:

      • Variable expression levels

      • Technical variations

      • Lot-to-lot antibody variations

    • Solutions:

      • Standardize protocols carefully

      • Include positive and negative controls in each experiment

      • Use the same antibody lot for related experiments

      • Document complete experimental conditions

Quality Control Measures:

Implement these quality control steps in your RHOH antibody experiments:

  • Include appropriate positive controls (lymphoid tissues, T-cell lines)

  • Use negative controls (non-hematopoietic cells, RHOH knockdown samples)

  • Perform parallel experiments with multiple antibodies when possible

  • Validate new antibody lots before use in critical experiments

  • Document lot numbers, dilutions, and exact protocols for reproducibility

These troubleshooting strategies and quality control measures will help ensure reliable and consistent results when working with RHOH antibodies.

How can researchers distinguish between RHOH and other Rho family GTPases in their experiments?

Distinguishing RHOH from other Rho family members is crucial given their structural similarities. Researchers can employ several strategies to ensure specificity:

Differentiation Strategies:

  • Antibody Selection:

    • Choose antibodies targeting unique regions of RHOH

    • The carboxyl-terminal insert domain is particularly distinctive

    • Validate antibodies against recombinant RHOH and related Rho proteins

  • Expression Pattern Analysis:

    • RHOH is predominantly expressed in hematopoietic cells

    • Non-hematopoietic cells generally lack RHOH expression

    • Use tissue/cell type specificity as an additional confirmation

  • Functional Assays:

    • RHOH lacks GTPase activity, unlike typical Rho GTPases

    • RHOH does not regulate actin reorganization in non-hematopoietic cells (unlike RhoA, Rac1, and Cdc42)

    • RHOH inhibits NFκB and p38, while most Rho proteins activate these pathways

  • Molecular Weight Discrimination:

    • RHOH has a distinct molecular weight (~21 kDa)

    • Use high-resolution gels (12-15%) for clear separation from other Rho GTPases

Distinguishing Features of RHOH:

FeatureRHOHTypical Rho GTPases (RhoA, Rac1, Cdc42)
GTPase activityAbsent (constitutively GTP-bound) Present (cycle between GDP/GTP states)
Expression patternHematopoietic-specific Broadly expressed
Effect on actinNo direct effect in non-hematopoietic cells Strong promoters of actin reorganization
Effect on NFκB/p38Inhibitory Usually activating
Regulation mechanismPrimarily transcriptional GTP/GDP cycling and various modifications
Role in T cellsCritical for TCR signaling Various roles in cytoskeletal regulation

By combining these approaches, researchers can confidently distinguish RHOH from other Rho family members in their experimental systems.

What are emerging technologies that could enhance RHOH research beyond traditional antibody approaches?

While antibodies remain essential tools for RHOH research, several emerging technologies offer complementary approaches that may address current limitations and open new avenues of investigation:

Emerging Technologies:

  • CRISPR-Based Technologies:

    • CRISPR/Cas9-mediated genome editing to generate RHOH knockout or knock-in models

    • CRISPRa/CRISPRi for conditional modulation of RHOH expression

    • CRISPR-based genetic screens to identify novel RHOH interactors or regulatory pathways

  • Proximity Labeling Methods:

    • BioID or TurboID fusion with RHOH to identify proximal proteins in living cells

    • APEX2 fusion for temporally controlled proximity labeling

    • These methods may reveal transient or weak interactions missed by traditional co-IP

  • Advanced Microscopy Techniques:

    • Super-resolution microscopy (STORM, STED) for detailed localization studies

    • FRET/FLIM to detect direct protein-protein interactions in living cells

    • Lattice light-sheet microscopy for dynamic tracking of RHOH in live immune cells

  • Single-Cell Technologies:

    • Single-cell RNA-seq to correlate RHOH transcription with cell states

    • CyTOF/mass cytometry for high-dimensional analysis of RHOH in immune cell subsets

    • Spatial transcriptomics to analyze RHOH expression in tissue context

  • Structural Biology Approaches:

    • Cryo-EM structure determination of RHOH complexes

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map interaction interfaces

    • Molecular dynamics simulations to understand RHOH's unique structural properties

Potential Applications:

These technologies could enable researchers to:

  • Map the complete RHOH interactome across different immune cell types and activation states

  • Visualize dynamic changes in RHOH localization during immune cell activation

  • Determine how RHOH's structure contributes to its unique GTPase-deficient state

  • Identify novel regulatory mechanisms beyond transcriptional control

  • Develop targeted approaches to modulate RHOH function in disease states

How might future research on RHOH antibodies contribute to therapeutic developments?

Future research using RHOH antibodies could potentially contribute to therapeutic developments for various diseases, particularly immune-related disorders and hematological malignancies:

Potential Therapeutic Applications:

  • Diagnostic Tools:

    • RHOH antibody-based assays could help stratify patients with immune disorders

    • Detection of altered RHOH levels might serve as biomarkers for disease activity

    • Monitoring RHOH in response to therapy could provide predictive information

  • Target Validation:

    • RHOH antibodies can help validate this protein as a therapeutic target

    • Understanding RHOH's role in disease pathogenesis using antibody-based methods

    • Identifying patient populations most likely to benefit from RHOH-targeted therapies

  • Drug Discovery:

    • Screening for compounds that modulate RHOH expression or function

    • Developing antibody-based assays to monitor drug effects on RHOH pathways

    • Using RHOH antibodies to identify and validate downstream effectors as alternative targets

  • Therapeutic Approaches:

    • Modulating RHOH levels could potentially help normalize T-cell function in immunodeficiencies

    • Targeting RHOH interactions might help treat certain B-cell malignancies

    • Regulating RHOH in neutrophils could potentially address inflammatory conditions

Disease-Specific Opportunities:

Disease CategoryRHOH's RolePotential Therapeutic Approach
B-cell malignanciesMutations found in lymphomas Restore normal RHOH function or target compensatory pathways
Primary immunodeficienciesCritical for T-cell development Enhance RHOH signaling to improve T-cell function
Autoimmune diseasesImplicated in SLE and psoriasis Modulate RHOH to normalize immune cell activation
Inflammatory conditionsRegulates neutrophil function Target RHOH-NMHC IIA interaction to control neutrophil activation

While direct therapeutic targeting of RHOH remains challenging due to its intracellular location and constitutively active nature, understanding its biology through antibody-based research will continue to reveal downstream pathways and interaction partners that may serve as more accessible therapeutic targets.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.