DIRAS3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method or location. Please contact your local distributor for specific delivery time information.
Synonyms
DIRA3 antibody; DIRA3_HUMAN antibody; DIRAS family GTP binding RAS like 3 antibody; DIRAS family GTPase 3 antibody; DIRAS family; GTP-binding RAS-like protein 3 antibody; DIRAS3 antibody; Distinct subgroup of the Ras family member 3 antibody; GTP binding protein Di Ras3 antibody; GTP-binding protein Di-Ras3 antibody; NOEY2 antibody; Ras homolog gene family member I antibody; Rho related GTP binding protein RhoI antibody; Rho-related GTP-binding protein RhoI antibody; RHOI antibody
Target Names
DIRAS3
Uniprot No.

Target Background

Gene References Into Functions
  1. Low DIRAS3 expression is associated with metastasis in gastric cancer. PMID: 30043279
  2. lncRNA GAS5 functions as a competing endogenous RNA for miR-221 to suppress cell growth and EMT in osteosarcoma by regulating the miR-221/ARHI pathway. PMID: 28519068
  3. DiRas3 binds to KSR1 independently of its interaction with activated Ras and RAF. PMID: 27368419
  4. DIRAS3 knock-down (KD) in adipose stromal/progenitor cells (ASCs) induces activation of Akt-mTOR signaling and proliferation arrest. DIRAS3 KD ASCs lose their colony-forming potential and are Ki-67 negative. Moreover, DIRAS3 silencing results in a premature senescence phenotype. PMID: 28316325
  5. ARHI-mediated up-regulation of glycolysis and glutaminolysis was found to be autophagy-dependent, and inhibition of these metabolic pathways resulted in decreased cell viability. PMID: 27784287
  6. Overexpression of ARHI promotes colon cancer SW480 cell apoptosis by inhibiting Akt levels. PMID: 27774944
  7. Transcriptional, but not post-transcriptional, silencing of GNG12-AS1 causes concomitant upregulation of DIRAS3, indicating a function in transcriptional interference. PMID: 26832224
  8. ARHI mRNA and protein expression is markedly decreased in osteosarcoma MG-63 cell lines. Overexpression of ARHI inhibits cell viability and proliferation. PMID: 26165148
  9. ARHI-mediated autophagy-associated cell death enhances chemosensitivity to cisplatin in ovarian cancer cell lines and xenografts. PMID: 26247722
  10. EZH2-induced H3K27me3 is associated with epigenetic repression of the ARHI tumor-suppressor gene in epithelial ovarian cancer. PMID: 25077680
  11. ARHI competes with RanGTPase and interacts with importin beta via a basic-acidic patch interaction, leading to inhibition of STAT3 translocation. PMID: 25499977
  12. Letter: ARHI suppresses pancreatic cancer by regulating ERK 1/2 signaling. PMID: 25675421
  13. JMJD2A-dependent silencing of Sp1 in advanced breast cancer promotes metastasis by downregulation of DIRAS3. PMID: 25193278
  14. DIRAS3 not only regulates the autophagosome initiation complex, but also induces autophagy in dormant, nutrient-deprived ovarian cancer cells that remain after conventional chemotherapy, facilitating their survival. PMID: 24879154
  15. ARHI is required for autophagy-mediated cancer cell arrest and inhibits signaling through PI3K/AKT and Ras/MAP by enhancing internalization and degradation of the epidermal growth factor receptor. PMID: 24769729
  16. In vitro, silencing of ARHI expression appears to accelerate the malignant transformation of healthy ovarian cells by restraining apoptosis and autophagy. PMID: 24476894
  17. ARHI acts as a tumor suppressor gene in MDA-MB-231 cells, and while trichostatin A+5-aza-2'-deoxycytidine can block the cells at different cell cycle phases, the antitumor effect is ARHI-dependent. PMID: 24676336
  18. Loss of ARHI expression is associated with glioma. PMID: 24458808
  19. ARHI acts as a tumor suppressor by downregulating the NFkappaB signaling pathway, resulting in the inhibition of cell proliferation, apoptosis, and cell cycle progression in the pancreatic tumor PANC-1 cell line. PMID: 23447002
  20. Overexpression of the ARHI gene may be associated with the inhibition of lung cancer cell growth, proliferation, and invasion, and the promotion of apoptosis. PMID: 23247805
  21. Results indicate that the aplysia ras homolog member I (ARHI) 3'UTR was a direct target of miR-221 in breast cancer MCF-7 cells. PMID: 23801152
  22. Acetylated STAT3 bound to the ARHI promoter and recruited DNA methyltransferase 1 for genetic modification. PMID: 23604529
  23. Imprinted chromatin around DIRAS3 regulates alternative splicing of GNG12-AS1, a long noncoding RNA. PMID: 23871723
  24. Expression of JMJD2A in infiltrating duct carcinoma is higher than in fibroadenoma, and is associated with ARHI, p53, and ER. PMID: 23678541
  25. DiRas3 interacts with C-RAF and downregulates MEK1 activity to restrict cell migration. PMID: 23157514
  26. ARHI expression is present in the endometrium and up-regulated in ectopic endometrium, whereas in the ectopic endometrium of patients with malignant endometriosis its expression is often negative. PMID: 21602127
  27. ARHI expression is downregulated in human gastric cancer and it may be a novel tumor suppressive target for gastric cancer therapy. PMID: 22497484
  28. The consequence of complex formation is DiRas3-mediated recruitment and anchorage of C-RAF to components of the membrane skeleton, suppression of C-RAF/B-RAF heterodimerization, and inhibition of C-RAF kinase activity. PMID: 22605333
  29. Downregulation of ARHI may play a significant role both in the pathogenesis and aggravation of gastric cancer. ARHI gene CpG island methylation is a potential molecular basis of its downregulation. PMID: 22427032
  30. The level of ARHI mRNA was significantly lower in aggressive compared with non-aggressive prostate cancer tissue samples. PMID: 22117988
  31. ARHI has a critical and previously uncharacterized role in the regulation of ovarian cancer cell migration. PMID: 21643014
  32. ARHI has pro-apoptotic effects on HCC cells, which is associated with the inactivation of both Akt and NF-kappaB survival pathways. PMID: 21933150
  33. Data show that ARHI re-expression induces autophagic cell death in breast cancer cells and enhances the inhibitory effects of paclitaxel by promoting autophagy, apoptosis, and G2/M cell cycle arrest. PMID: 21244707
  34. These data highlighted an important role for ARHI in controlling hepatocellular carcinoma growth and angiogenesis. PMID: 21093415
  35. The DIRAS3 gene is imprinted, with monoallelic expression from the paternal allele. PMID: 9874798
  36. Data show that ARHI could compete for Ran-importin binding and induce disruption of importin-binding to cargo proteins, including STAT3. PMID: 19435463
  37. A link between NOEY2 loss expression and the spreading mechanism of breast cancer may possibly exist. PMID: 12485503
  38. ARHI inhibits cell growth, and loss of its expression in cells may contribute to the development of breast and ovarian cancers. PMID: 12771940
  39. Hypermethylation of CpG island II in the promoter region of ARHI is associated with the complete loss of ARHI expression in breast cancer cells. PMID: 12874023
  40. ARHI may have a role in the development and progression of ductal breast carcinoma in situ. PMID: 14506155
  41. The association between STAT3 and ARHI, as well as the functional inhibition of STAT3 transcriptional activity by ARHI, suggests a novel mechanism through which a putative tumor suppressor gene can inhibit STAT3 activity in breast and ovarian cancers. PMID: 16061651
  42. Down-regulation of the ARHI gene is associated with breast cancer. PMID: 16158053
  43. Down-regulation of ARHI due to loss of heterozygosity and promoter methylation is associated with ovarian cancers. PMID: 18286529
  44. DIRAS3 is a novel, prognostically relevant candidate gene that is frequently methylated and silenced in oligodendroglial tumors with 1p deletion. PMID: 18302158
  45. Evidence suggests that ARHI downregulated in HCCs could play a role in liver cancer via acting as a tumor suppressor gene, which is primarily triggered by epigenetic events in HCC specimens. PMID: 18612997
  46. ARHI can induce autophagic cell death, but can also promote tumor dormancy in the presence of factors that promote survival in the cancer microenvironment. PMID: 19033662
  47. The presence of NOEY2 mutations in human breast cancer and early-stage lesions indicates that NOEY2 mutations may be partly associated with breast tumourigenesis. PMID: 19482475
  48. ARHI represents a modulator of cancer cell proliferation and may play a significant role in the development of pancreatic cancer. PMID: 19639215
  49. Loss of heterozygosity at 1p31 (including ARHI) did not correlate with the mitotic activity index nor provide prognostic information. PMID: 19759414

Show More

Hide All

Database Links

HGNC: 687

OMIM: 605193

KEGG: hsa:9077

STRING: 9606.ENSP00000360020

UniGene: Hs.194695

Protein Families
Small GTPase superfamily, Di-Ras family
Subcellular Location
Cell membrane; Lipid-anchor; Cytoplasmic side.
Tissue Specificity
Expressed in normal ovarian and breast epithelial cells but not in ovarian and breast cancers.

Q&A

What is DIRAS3 and why is it important in cancer research?

DIRAS3 (DIRAS family GTPase 3, also known as ARHI or NOEY2) is a maternally imprinted tumor suppressor gene that encodes a 26 kDa GTPase with approximately 60% amino acid homology to RAS proteins. Its distinctive feature is a 34-amino-acid N-terminal extension, which is essential for its tumor suppressive functions .

DIRAS3 is significant in cancer research for several reasons:

  • It's the first endogenous non-RAS protein discovered that heterodimerizes with RAS, disrupts RAS clustering, blocks RAS signaling, and inhibits cancer cell growth

  • It's frequently downregulated in multiple cancer types including ovary, breast, lung, prostate, colon, brain, and thyroid cancers

  • It functions as a tumor suppressor by inhibiting signaling through PI3K/AKT, JAK/STAT, and RAS/MAPK pathways

  • DIRAS3 plays critical roles in autophagy, cancer cell dormancy, and oxidative stress, making it a potential target for novel cancer therapies

Re-expression of DIRAS3 inhibits cancer cell growth, prevents angiogenesis, induces autophagy, and creates tumor dormancy in xenograft models, highlighting its potential as a therapeutic target .

What experimental applications are DIRAS3 antibodies suitable for?

DIRAS3 antibodies have been validated for multiple experimental applications in molecular and cellular biology research:

ApplicationDescriptionNotes
Western Blot (WB)Detection of DIRAS3 protein in cell/tissue lysatesMost commonly used; dilutions typically 1:500-1:2000
Enzyme-Linked Immunosorbent Assay (ELISA)Quantitative detection of DIRAS3Useful for high-throughput screening
Immunohistochemistry (IHC)Detection of DIRAS3 in tissue sectionsUsed for both paraffin-embedded and frozen sections
Immunofluorescence (IF)Cellular localization studiesBoth cellular (IF-cc) and paraffin (IF-p) applications
Immunoprecipitation (IP)Isolation of DIRAS3 and associated proteinsCritical for protein-protein interaction studies
Immunogold electron microscopyUltrastructural localizationUsed for precise subcellular localization studies

When selecting a DIRAS3 antibody, researchers should consider the specific experimental application, target species, and binding specificity (e.g., N-terminal vs. C-terminal epitopes) most relevant to their research question .

What positive controls should I use when working with DIRAS3 antibodies?

Implementing appropriate positive controls is crucial for ensuring the validity and reproducibility of experiments using DIRAS3 antibodies:

Cell lines with known DIRAS3 expression:

  • NIH/3T3 cells

  • Normal ovarian epithelial cells

  • CAOv3 ovarian cancer cells (high endogenous DIRAS3 expression and basal autophagy)

Tissue samples:

  • Mouse liver, mouse kidney, rat liver (as indicated in product information)

Expression systems:

  • DIRAS3-inducible cell lines (typically Tet-on/DOX-inducible systems)

  • Transiently transfected cells overexpressing DIRAS3

Recombinant proteins:

  • Purified recombinant DIRAS3 protein is useful as a standard in quantitative assays and specificity testing

For inducible systems (such as DOX-inducible DIRAS3 expression), comparing induced versus non-induced samples provides an excellent internal control for antibody validation .

How can I optimize DIRAS3 antibody performance for detecting low-level expression in cancer samples?

Detecting low levels of DIRAS3 expression in cancer samples requires optimized sensitivity approaches:

Enhanced immunohistochemistry protocols:

  • Implement heat-induced epitope retrieval (HIER) with optimized buffer conditions

  • Utilize signal amplification systems such as tyramide signal amplification (TSA)

  • Extend primary antibody incubation time (overnight at 4°C)

  • Use high-sensitivity detection systems (polymer-based or biotin-free)

Enhanced Western blot detection:

  • Increase protein loading (50-100 μg total protein)

  • Use high-sensitivity chemiluminescent substrates

  • Employ concentration steps like immunoprecipitation before Western blotting

  • Consider using gradient gels for better resolution near 26 kDa

  • Extend exposure time with cooled CCD camera systems

Alternative detection methods:

  • Proximity ligation assay (PLA): Can detect protein-protein interactions with single-molecule sensitivity

  • RNAscope in situ hybridization: To detect DIRAS3 mRNA as complementary approach

  • Mass spectrometry: For targeted detection of DIRAS3 peptides

DIRAS3 expression is often downregulated in cancer tissues through epigenetic mechanisms, so combining protein detection with epigenetic analysis (e.g., promoter methylation status) can provide a more complete picture of DIRAS3 status .

How can I design experiments to study DIRAS3-RAS interactions using antibody-based approaches?

Studying DIRAS3-RAS interactions requires carefully designed antibody-based approaches to detect these protein-protein interactions:

Co-immunoprecipitation (Co-IP) strategies:

  • Forward and reverse Co-IP: Immunoprecipitate with anti-DIRAS3 and probe for RAS, then reverse the approach

  • Use mild detergents (e.g., 1% NP-40 or 0.5% Triton X-100) to preserve membrane associations

  • Include appropriate controls: IgG control, lysates from cells not expressing one protein

Proximity-based detection methods:

  • Proximity Ligation Assay (PLA): Detect DIRAS3-RAS interactions in situ with single-molecule resolution

  • Immunofluorescence co-localization using specific antibodies for DIRAS3 and RAS

Advanced electron microscopy:

  • Immunogold electron microscopy with differentially sized gold particles (e.g., DIRAS3: 2nm, K-RAS: 4.5nm)

  • Bi-variate K-function analysis to quantify co-localization at ultrastructural level

Functional validation approaches:

  • Assess effects of DIRAS3 and mutant constructs (ΔNT DIRAS3, C226S) on RAS clustering

  • Use membrane sheet preparations to focus on plasma membrane interactions

  • Study interactions under different cellular conditions (starvation, growth factor stimulation)

Research has demonstrated that DIRAS3 directly binds to RAS, forming heteromers that disrupt RAS clustering and inhibit downstream signaling. The N-terminal extension of DIRAS3 and its membrane association through the CAAX-box domain are both critical for this interaction .

What strategies can I use to study DIRAS3-mediated autophagy using antibody-based approaches?

DIRAS3-mediated autophagy can be comprehensively studied using various antibody-based strategies:

Monitoring autophagic flux:

  • Track LC3-I to LC3-II conversion by Western blot using anti-LC3B antibodies

  • Measure SQSTM1/p62 degradation as marker of autophagic flux

  • Use chloroquine to block autophagosome-lysosome fusion and assess accumulation

  • Use tandem mCherry-GFP-LC3 reporter systems to differentiate autophagosomes (yellow) from autolysosomes (red)

Detecting DIRAS3-autophagy protein interactions:

  • Co-immunoprecipitation of DIRAS3 with key autophagy proteins (BECN1, ATG12, LC3)

  • Immunofluorescence co-localization studies of DIRAS3 with autophagy markers

  • Immunogold electron microscopy to visualize DIRAS3 localization to autophagosomal membranes

Monitoring autophagy pathway activation:

  • Assess phosphorylation status of key autophagy regulators:

    • Decreased p-MTOR (Ser2448)

    • Decreased p-STK11/LKB1 (Ser428)

    • Increased p-AMPK (Thr172)

  • Track nuclear translocation of autophagy-related transcription factors:

    • TFEB

    • FOXO3

Visualizing autophagosome formation:

  • Immunofluorescence staining for ATG proteins (ULK1, ATG13, ATG14, BECN1)

  • Quantify puncta formation as measure of autophagosome initiation

Research has shown that DIRAS3 is essential for autophagy and triggers this process through multiple mechanisms, including downregulation of the PtdIns3K-AKT-MTOR pathway, interaction with BECN1, and disruption of the BECN1-BCL2 complex .

How can I use DIRAS3 antibodies to investigate tumor dormancy mechanisms?

DIRAS3 antibodies can be valuable tools for investigating tumor dormancy mechanisms, particularly given DIRAS3's role in inducing and maintaining dormancy:

Monitoring DIRAS3 expression in dormant models:

  • Use immunohistochemistry to detect DIRAS3 in dormant tumor regions

  • Compare DIRAS3 expression in proliferating vs. dormant regions using dual staining with proliferation markers

  • Quantify DIRAS3 levels during dormancy induction, maintenance, and exit phases

In vivo dormancy models:

  • Create inducible DIRAS3 expression systems (e.g., DOX-inducible) in xenograft models

  • Monitor tumor growth and angiogenesis before/during/after DIRAS3 induction

  • Sample tumors at various timepoints to track molecular changes via IHC and Western blotting

  • Assess effects of autophagy inhibitors (chloroquine, DC661) on dormant tumor recurrence

Pathway analysis in dormant cells:

  • Analyze key signaling pathways modulated by DIRAS3 during dormancy:

    • PI3K/AKT/MTOR pathway inhibition

    • RAS/MAPK signaling suppression

    • Autophagy pathway activation

    • Angiogenesis inhibition

  • Use phospho-specific antibodies to track signaling dynamics

Studies have shown that re-expression of DIRAS3 induces dormancy in xenograft models, inhibiting cancer cell growth and angiogenesis. DIRAS3-mediated induction of autophagy facilitates the survival of dormant cancer cells in a nutrient-poor environment, suggesting that targeting DIRAS3-positive dormant cells could eliminate residual disease after conventional therapy .

What are the optimal conditions for using DIRAS3 antibodies in co-immunoprecipitation experiments?

Co-immunoprecipitation (Co-IP) experiments with DIRAS3 antibodies require careful optimization to detect authentic protein interactions while minimizing artifacts:

Buffer optimization:

  • Use buffers containing 0.5-1% non-ionic detergents (NP-40, Triton X-100, Digitonin) to preserve membrane protein interactions

  • Include protease and phosphatase inhibitors to prevent degradation and preserve post-translational modifications

  • Consider adding GTP/GDP for small GTPase interactions

Antibody selection and validation:

  • Validate antibody specificity before Co-IP experiments

  • Consider epitope location: Ensure the epitope is not involved in protein-protein interactions

  • Test multiple antibodies targeting different regions of DIRAS3

Cell lysis considerations:

  • For inducible systems, optimize induction time for maximal expression

  • Consider membrane fractionation to enrich for DIRAS3-RAS interactions occurring at the plasma membrane

Controls and validation:

  • Input control: Check expression levels of target proteins in lysate before IP

  • IgG control: Parallel IP with isotype-matched control IgG

  • Reverse Co-IP: Confirm interaction by immunoprecipitating binding partner

For studying DIRAS3-BECN1 interactions, research has demonstrated direct protein-protein binding that can be detected by co-immunoprecipitation. This interaction is enhanced during nutrient deprivation and plays a crucial role in autophagy induction .

How do I design experiments to differentiate between DIRAS3 and other RAS family proteins?

Differentiating DIRAS3 from other RAS family proteins requires careful experimental design that leverages their structural and functional differences:

Antibody-based differentiation:

  • Use antibodies targeting the unique 34-amino-acid N-terminal extension of DIRAS3

  • Validate antibody specificity against recombinant DIRAS3 and RAS proteins

Size-based differentiation:

  • DIRAS3 (26 kDa) vs. standard RAS proteins (~21 kDa) can be resolved on higher-percentage SDS-PAGE gels

  • Use gradient gels (10-20%) for optimal resolution in the 20-30 kDa range

Expression pattern analysis:

  • Exploit tissue-specific expression differences (DIRAS3 is highly expressed in normal ovarian and breast epithelial cells but downregulated in corresponding cancers)

  • Analyze imprinting status (DIRAS3 is maternally imprinted, unlike RAS genes)

Functional differentiation:

  • Subcellular localization studies: Both localize to membranes, but distribution patterns may differ

  • Downstream signaling effects: DIRAS3 inhibits pathways that RAS activates

  • Autophagy induction: DIRAS3 strongly induces autophagy, unlike classical RAS proteins

Genetic approaches:

  • Use siRNA sequences targeting unique regions of DIRAS3 mRNA

  • Design PCR primers that distinguish DIRAS3 from RAS family members

The distinctive N-terminal extension of DIRAS3 is critical for its tumor suppressive functions and provides a key distinguishing feature from other RAS family proteins .

What are the challenges in detecting DIRAS3 in cancer tissues with imprinting abnormalities?

Detecting DIRAS3 in cancer tissues with imprinting abnormalities presents several challenges that require specialized approaches:

Challenges related to imprinting status:

  • DIRAS3 is normally expressed only from the paternal allele due to maternal imprinting

  • Cancer cells may exhibit loss of imprinting (LOI) or allele-specific silencing

  • Heterogeneous expression patterns within the same tumor sample

  • Loss of expression can occur in a single "hit" through multiple mechanisms

Methodological challenges:

  • Standard antibody-based methods cannot distinguish allele-specific expression

  • Expression levels may be below detection threshold of conventional techniques

  • Presence of multiple mechanisms of downregulation (genetic, epigenetic, post-transcriptional)

  • Distinguishing DIRAS3 protein from closely related RAS family members

Advanced approaches to address these challenges:

  • Combined genetic-protein analysis:

    • Allele-specific PCR followed by protein analysis from the same sample

    • Laser capture microdissection to isolate specific cell populations

  • Epigenetic assessment:

    • Methylation-specific PCR to analyze DIRAS3 promoter status

    • Correlate epigenetic status with protein expression levels

  • Enhanced detection methods:

    • Signal amplification systems for immunohistochemistry

    • Mass spectrometry-based proteomics for DIRAS3 detection

DIRAS3's unique status as a maternally imprinted tumor suppressor gene makes it particularly vulnerable to expression loss in cancer, and comprehensive analysis requires integration of genetic, epigenetic, and protein-level approaches .

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.