RASSF1A Human

Ras association domain-containing protein 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to RASSF1A Human

RASSF1A (Ras Association Domain Family 1 Isoform A) is a tumor suppressor protein encoded by the RASSF1 gene located at chromosome 3p21.3. It is one of eight isoforms produced by alternative splicing, with RASSF1A and RASSF1C being the most biologically relevant. RASSF1A functions as a molecular scaffold, regulating critical cellular processes such as apoptosis, cell cycle arrest, microtubule stability, and genomic integrity . Its inactivation, primarily through promoter hypermethylation, is observed in over 70% of human cancers, including lung, breast, renal, and colorectal carcinomas, underscoring its role as a key guardian against tumorigenesis .

Protein Structure

RASSF1A comprises three distinct domains:

  1. N-terminal Cysteine-Rich Domain (CRD): Mediates interactions with death receptors (e.g., TNF-R1, TRAIL-R1) and modulates apoptosis .

  2. Ras Association (RA) Domain: Weakly binds activated Ras GTPases, though its primary role is scaffolding tumor suppressor complexes .

  3. C-terminal SARAH Domain: Facilitates interactions with Hippo pathway kinases (e.g., MST1/2) and Salvador (WW45), linking RASSF1A to cell growth regulation .

Key Biological Functions

FunctionMechanismOutcome
Cell Cycle ArrestInhibits cyclin D1/A2 via p120E4F and JNK pathways; binds SalvadorG1/S checkpoint control
Apoptosis PromotionActivates Bax via MOAP1; enhances TNF-α and K-Ras-induced apoptosisTumor cell elimination
Microtubule StabilizationInhibits HDAC6, increasing acetylated α-tubulin; binds MAPsPrevents chromosomal instability
Hypoxia ResponseForms a feedforward loop with HIF-1α; stabilizes HIF-1α proteinModulates glycolysis in tumors

Epigenetic Inactivation

RASSF1A is silenced in cancers primarily via CpG island promoter hypermethylation, a reversible epigenetic modification. This mechanism is observed in:

Cancer TypeMethylation FrequencyClinical ImpactSource
Lung (NSCLC)~76%Poor prognosis, metastasis
Renal (Clear RCC)91%Early carcinogenesis
Breast~50%Tumor progression
Colorectal~60%Genetic instability

Functional Consequences of Loss

  • Cell Cycle Dysregulation: Accumulation of cyclin D1 drives unchecked proliferation .

  • Metastasis Promotion: Loss of RhoB activation via GEF-H1 deregulation enhances EMT and invasion .

  • HIF-1α Amplification: Hypoxia-induced RASSF1A stabilizes HIF-1α, promoting glycolysis and angiogenesis .

Mouse Models

ModelPhenotypeSignificanceSource
Rassf1a KOSpontaneous lung/lymphoma tumorsConfirms tumor suppressor role
Rassf1a +/−Enhanced benzo(a)pyrene-induced tumorsSuggests haploinsufficiency
Rassf1a −/−Resistance to hypoxia-induced PHLinks RASSF1A to vascular remodeling

Interactions with Oncogenic Pathways

  • RAS Signaling: Binds DAB2IP (RasGAP), reducing Ras-GTP levels. Loss of RASSF1A elevates Ras activity in wild-type tumors .

  • Hippo Pathway: Interacts with Salvador and MST kinases, enabling YAP nuclear exclusion and growth inhibition .

Reactivation Strategies

ApproachMechanismPreclinical EfficacySource
Demethylating AgentsReverses promoter methylationRestores RASSF1A in lung cancer cells
RAS InhibitorsTargets hyperactive Ras in RASSF1A−/− tumorsSynergizes with DAB2IP upregulation
HIF-1α InhibitorsDisrupts RASSF1A-HIF-1α loopReduces Warburg effect in hypoxic tumors

Biomarker Potential

RASSF1A promoter methylation is a validated biomarker for early detection in lung and renal cancers. Its expression levels correlate with prognosis, guiding personalized therapy .

Product Specs

Introduction
RASSF1A protein, a potential tumor suppressor similar to RAS effector proteins, might be involved in cancer development. Its presence or absence is linked to various cancers, suggesting its role as a tumor suppressor. RASSF1A inactivation is frequently observed alongside hypermethylation of its CpG-island promoter region. Additionally, RASSF1A is crucial for death receptor-mediated apoptosis.
Description
Recombinant human RASSF1A, produced in E. coli, is a non-glycosylated polypeptide chain with 376 amino acids (1-340 a.a.) and a molecular weight of 42.9 kDa. This protein comprises amino acids 1-340 of the RASSF1A protein and a 36 amino acid His-tag at the N-terminus. It undergoes purification through proprietary chromatographic methods.
Physical Appearance
A clear solution, sterilized by filtration.
Formulation
The RASSF1A protein solution is provided at a concentration of 1mg/ml and contains the following components: 20mM Tris-HCl buffer (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), keep the solution refrigerated at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. To maintain stability, avoid repeated freezing and thawing cycles.
Purity
The purity of the protein is greater than 85%, as determined by SDS-PAGE analysis.
Synonyms
123F2, NORE2A, RASSF1, RDA32, REH3P21, Ras association domain-containing protein 1.
Source
Escherichia Coli.
Amino Acid Sequence
MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSMSGE PELIELRELA PAGRAGKGRT RLERANALRI ARGTACNPTR QLVPGRGHRF QPAGPATHTW CDLCGDFIWG VVRKGLQCAH CKFTCHYRCR ALVCLDCCGP RDLGWEPAVE RDTNVDEPVE WETPDLSQAE IEQKIKEYNA QINSNLFMSL NKDGSYTGFI KVQLKLVRPV SVPSSKKPPS LQDARRGPGR GTSVRRRTSF YLPKDAVKHL HVLSRTRARE VIEALLRKFL VVDDPRKFAL FERAERHGQV YLRKLLDDEQ PLRLRLLAGP SDKALSFVLK ENDSGEVNWD AFSMPELHNF LRILQREEEE HLRQILQKYS YCRQKIQEAL HACPLG.

Q&A

What is RASSF1A and what is its role in human cells?

RASSF1A (Ras Association Domain Family Protein 1 isoform A) is a tumor suppressor protein that functions as a molecular scaffold, coordinating numerous signaling pathways that control cellular homeostasis. It's located at chromosome 3p21.3, a region frequently showing loss of heterozygosity (LOH) in various cancers . RASSF1A prevents tumorigenesis through multiple mechanisms including cell cycle arrest, inhibition of migration/metastasis, microtubular stabilization, and promotion of apoptosis . It serves as a critical nexus for the coordination of signaling pathways controlling cell fate, metabolism, communication, motility, growth, division, and death .

How is RASSF1A commonly inactivated in human cancers?

RASSF1A is predominantly inactivated through promoter hypermethylation, which leads to transcriptional silencing. This epigenetic mechanism is the most widespread form of RASSF1A inactivation in human cancers, rather than gene deletion or germline mutations which do occur but less frequently . Studies have shown hypermethylation of RASSF1A's GC-rich putative promoter region in approximately 91% of primary renal cell carcinoma tumors and all 18 RCC cell lines analyzed in one study . This methylation pattern causally relates to loss of transcription of the corresponding mRNA, and expression can be reactivated through treatment with demethylating agents such as 5-aza-2′-deoxycytidine .

What are the major signaling pathways involving RASSF1A?

RASSF1A primarily functions at the crossroads of three intertwined molecular signaling mechanisms:

  • Ras/Rho GTPases pathway: RASSF1A binds to K-RAS, H-RAS, RAP1/2, and RAN GTPases through its RA (Ras Association) domain with varying affinity .

  • Hippo pathway: RASSF1A directly binds to Hippo kinases MST1 and MST2 through its SARAH domain, promoting downstream Hippo pathway signaling to YAP1 .

  • Microtubule regulation: RASSF1A interacts with tubulin through its RA domain, contributing to hyperstabilization of microtubules .
    Additionally, RASSF1A plays a role in nuclear actin transport regulation through interaction with exportin-6 (XPO6) and RAN-GTPase, affecting the MRTF/SRF signaling axis .

How does RASSF1A regulate nuclear actin levels and what are the functional consequences?

RASSF1A regulates nuclear actin levels by facilitating the association of exportin-6 (XPO6) with RAN-GTPase, which is essential for the nuclear export of XPO6 cargoes, including actin and profilin . When RASSF1A is absent, as commonly observed in human tumors, this leads to:

  • Accumulation of actin within the nucleus

  • Reduced Myocardin-related transcription factor A (MRTF-A) nuclear localization

  • Decreased serum response factor (SRF) activity and expression

  • Downregulation of SRF target genes, many involved in cytoskeletal regulation

  • Significant decrease in cell adhesion properties
    Research has shown that restoring normal nuclear actin levels by silencing IPO9 (importin-9) can rescue these effects in RASSF1A-depleted cells . This mechanism may explain the widespread prognostic associations of RASSF1A loss across different cancer types, as nuclear actin levels influence multiple cellular processes including hippo pathway regulation, apoptosis, differentiation, and DNA damage repair .

What is the relationship between RASSF1A and the Hippo pathway in tumor suppression?

RASSF1A connects to the Hippo pathway through direct binding to MST1 and MST2 kinases via its SARAH domain . In response to DNA damage, nuclear RASSF1A undergoes phosphorylation on Ser131 by ATM or ATR kinases, which promotes:

  • Dimerization and trans-autophosphorylation of MST2 required for its activation

  • Downstream Hippo pathway signaling to YAP1

  • Regulation of cell growth and apoptosis
    Additionally, nuclear actin levels (which are regulated by RASSF1A) also contribute to Hippo pathway regulation, specifically SRF regulation of YAP . The dual role of RASSF1A in both directly activating MST kinases and regulating nuclear actin levels that affect YAP activity highlights its central position in coordinating tumor suppression through Hippo signaling.

How does forced expression of RASSF1A affect cancer cell phenotypes?

Forced expression of RASSF1A in cancer cell lines has demonstrated significant tumor suppressive effects. In renal carcinoma cell line KRC/Y (which contains a normal and expressed VHL gene), introduction of RASSF1A transcripts:

  • Suppressed growth on plastic dishes

  • Inhibited anchorage-independent colony formation in soft agar

  • Showed reduced growth suppression activity when mutant RASSF1A was expressed instead
    These findings suggest that RASSF1A restoration could be a potential therapeutic approach for cancers with RASSF1A silencing. Interestingly, the tumor suppressive effects appear to be independent of VHL status, as RASSF1A hypermethylation was observed in both VHL-caused clear RCC tumors and clear RCC without VHL inactivation .

What techniques are used to assess RASSF1A promoter methylation status?

Several methodologies are employed to evaluate RASSF1A promoter methylation:

  • Methylation-Specific PCR (MSP): This technique uses primers designed to amplify either methylated or unmethylated versions of the RASSF1A promoter region.

  • Bisulfite Sequencing: This provides a comprehensive analysis of individual CpG sites within the promoter region, allowing for quantification of methylation at specific positions.

  • Pyrosequencing: A quantitative method that measures the degree of methylation at each CpG site.

  • Methylation-Sensitive High-Resolution Melting (MS-HRM): A sensitive screening method that can detect low levels of methylation.

  • Methylation Arrays: Genome-wide approaches to analyze methylation patterns of multiple genes including RASSF1A.
    When conducting methylation analysis, researchers should focus on the GC-rich putative promoter region of RASSF1A, which has been shown to be causally related to transcriptional silencing when methylated .

How can RASSF1A expression be restored in experimental settings?

RASSF1A expression can be restored through several experimental approaches:

  • Demethylating Agents: Treatment with 5-aza-2′-deoxycytidine (5-Aza-dC) has been shown to reactivate RASSF1A expression by reversing promoter hypermethylation .

  • Forced Expression: Transfection with RASSF1A expression vectors allows for exogenous expression in cell lines lacking endogenous RASSF1A.

  • CRISPR-dCas9 Systems: Epigenome editing using CRISPR-dCas9 fused to demethylases can be used to specifically demethylate the RASSF1A promoter.

  • Histone Deacetylase (HDAC) Inhibitors: These compounds can sometimes restore expression of genes silenced by promoter hypermethylation through chromatin remodeling.
    When designing experiments to restore RASSF1A expression, researchers should consider potential off-target effects of demethylating agents and validate restoration of expression through both mRNA and protein analysis.

What experimental models are most appropriate for studying RASSF1A functions?

The following experimental models provide valuable insights into RASSF1A functions:

  • Cell Line Models:

    • RCC cell lines (with partial or complete RASSF1A promoter methylation)

    • HeLa cells (for studying nuclear actin regulation)

    • Cell lines with engineered RASSF1A knockout or knockdown

  • Primary Tumor Samples:

    • Clear cell renal cell carcinomas show high frequency of RASSF1A inactivation (91% in one study)

    • Breast, lung, and gastrointestinal cancer tissues with variable RASSF1A status

  • Animal Models:

    • RASSF1A knockout mice

    • Xenograft models with RASSF1A-expressing or RASSF1A-deficient cells

  • 3D Culture Systems:

    • Organoids derived from primary tumors

    • Spheroid cultures for studying invasion and metastasis processes
      When selecting experimental models, researchers should consider the specific RASSF1A function they aim to study (e.g., cell cycle regulation, microtubule stability, nuclear actin transport) and choose models that best represent the biological context of interest.

What is the prognostic significance of RASSF1A methylation in human cancers?

RASSF1A methylation has been widely associated with poor patient outcomes across multiple cancer types . The prognostic significance includes:

  • Correlation with tumor aggressiveness and metastatic potential

  • Association with decreased disease-free survival

  • Potential prediction of response to certain therapeutic approaches
    Research has shown that in breast and liver cancers specifically, low SRF mRNA (which is linked to RASSF1A loss) serves as a poor prognostic factor . The widespread prognostic associations suggest that RASSF1A loss affects fundamental biological processes contributing to cancer progression, potentially through its role in regulating nuclear actin levels and subsequent effects on multiple cellular functions .

How might targeting RASSF1A pathways contribute to cancer therapy?

The RASSF1A pathway represents a promising therapeutic target through several potential approaches:

  • Epigenetic Therapies: Demethylating agents like 5-aza-2′-deoxycytidine could restore RASSF1A expression .

  • Nuclear Actin Regulation: Targeting nuclear actin transport components might normalize cellular functions disrupted by RASSF1A loss .

  • SRF Pathway Modulation: Strategies to restore SRF activity could counteract the downstream effects of RASSF1A silencing .

  • Combination Approaches: Targeting RASSF1A-related pathways in combination with conventional therapies might enhance treatment efficacy.
    The tumor-suppressive role of RASSF1A across multiple cellular processes suggests that restoring its function or compensating for its loss could have far-reaching effects on tumor behavior. Understanding how RASSF1A functions as a nexus in the coordination of signaling pathways provides multiple potential intervention points for therapeutic development .

Can RASSF1A methylation serve as a biomarker for early detection or treatment response?

RASSF1A methylation has potential as both a diagnostic and predictive biomarker:

  • Early Detection: The high frequency of RASSF1A methylation in various cancers (91% in RCC) makes it a promising candidate for early detection strategies, particularly in liquid biopsy approaches analyzing circulating tumor DNA.

  • Treatment Response Prediction: RASSF1A has been identified as a prognostic biomarker that predicts chemosensitivity in cancer , potentially allowing for more personalized treatment approaches.

  • Monitoring Disease Progression: Changes in RASSF1A methylation levels may reflect disease progression or response to therapy.

  • Risk Stratification: RASSF1A methylation patterns could help categorize patients into different risk groups, informing treatment intensity decisions. For clinical implementation, standardized methodologies for assessing RASSF1A methylation and established thresholds for positivity would need to be developed and validated in large cohort studies.

Product Science Overview

Overview

Ras association domain-containing protein 1 (RASSF1) is a member of the Ras association domain family, which includes proteins that share structural similarities with Ras effector proteins . The RASSF1 gene is located on chromosome 3p21.3 and encodes multiple isoforms through alternative splicing . Among these isoforms, RASSF1A and RASSF1C are the most abundantly expressed .

Structure and Isoforms

RASSF1 proteins contain a Ras association (RA) domain, which is crucial for their interaction with Ras and Ras-like small GTPases . The RA domain is located at the C-terminus in most RASSF proteins, but in some isoforms, it is found at the N-terminus . Additionally, RASSF1A, the longest isoform, harbors a cysteine-rich diacylglycerol (DAG) or C1 lipid-binding domain . The RA domain shares structural similarities with the Ras-binding protein Raf, although RASSF1 proteins lack catalytic domains and function primarily as adaptors to recruit other proteins .

Function and Mechanisms

RASSF1A is a well-known tumor suppressor gene that plays a significant role in cell cycle control, apoptosis, cell migration, and mitosis regulation . It is involved in the repression of cyclin A2 and cyclin D1, leading to cell cycle arrest . RASSF1A also contributes to microtubule stability by inhibiting histone deacetylase 6 (HDAC6), resulting in increased acetylation of microtubules . Furthermore, RASSF1A interacts with microtubule-associated proteins (MAPs) to regulate microtubule stability and modulates apoptosis through the activation of the MST2-LATS1 pathway .

Role in Cancer

The inactivation of RASSF1A is frequently observed in various cancers, including lung, breast, kidney, and nasopharyngeal cancers . The primary mechanism of inactivation is the hypermethylation of its promoter region, leading to the loss of expression . RASSF1A is one of the most frequently silenced genes in over 40 types of cancer . The loss of RASSF1A expression disrupts its tumor suppressor functions, contributing to uncontrolled cell proliferation and tumor development .

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.