RRAGC Human

Ras-Related GTP Binding C Human Recombinant
Shipped with Ice Packs
In Stock

Description

RRAGC Human Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 423 amino acids (1-399 a.a) and having a molecular mass of 46.7kDa.
RRAGC is fused to a 24 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Ras-related GTP binding C (RRAGC) is a monomeric guanine nucleotide-binding protein, also known as a G protein. Small G proteins like RRAGC bind to GTP or GDP, acting as molecular regulators in various cellular processes and signaling pathways. Specifically, RRAGC regulates the organization of the actin cytoskeleton and possesses intrinsic GTPase activity. It is believed to be involved in the amino acid-induced relocalization of mTORC1 to lysosomes and its subsequent activation by the RHEB GTPase, a crucial step in activating the TOR signaling cascade by amino acids.
Description
Recombinant human RRAGC, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 423 amino acids (with amino acids 1-399 present) and possessing a molecular weight of 46.7 kDa. The RRAGC protein is fused to a 24 amino acid His-tag at its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized through filtration.
Formulation
The RRAGC protein solution is provided at a concentration of 0.5 mg/ml and contains 20 mM Tris-HCl buffer (pH 8.0), 1 mM DTT, 10% glycerol, and 0.1 M NaCl.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To ensure optimal stability during long-term storage, consider adding a carrier protein (0.1% HSA or BSA). It is important to avoid repeated freeze-thaw cycles to maintain product integrity.
Purity
The purity of the RRAGC protein is greater than 85.0% as determined by SDS-PAGE analysis.
Synonyms
Ras-related GTP-binding protein C, Rag C, RagC, GTPase-interacting protein 2, TIB929, RRAGC, GTR2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMSLQYG AEETPLAGSY GAADSFPKDF GYGVEEEEEE AAAAGGGVGA GAGGGCGPGG ADSSKPRILL MGLRRSGKSS IQKVVFHKMS PNETLFLEST NKIYKDDISN SSFVNFQIWD FPGQMDFFDP TFDYEMIFRG TGALIYVIDA QDDYMEALTR
LHITVSKAYK VNPDMNFEVF IHKVDGLSDD HKIETQRDIH QRANDDLADA GLEKLHLSFY LTSIYDHSIF EAFSKVVQKL IPQLPTLENL LNIFISNSGI EKAFLFDVVS KIYIATDSSP VDMQSYELCC DMIDVVIDVS CIYGLKEDGS GSAYDKESMA IIKLNNTTVL YLKEVTKFLA
LVCILREESF ERKGLIDYNF HCFRKAIHEV FEVGVTSHRS CGHQTSASSL KALTHNGTPR NAI.

Q&A

What is RRAGC and what is its function in human cells?

RRAGC is a monomeric guanine nucleotide-binding protein (G protein) encoded by the RRAGC gene located on human chromosome 1. It functions as a molecular switch by binding either GTP or GDP, allowing it to participate in numerous cellular processes and signaling pathways . RRAGC plays a critical role in amino acid sensing and mechanistic target of rapamycin complex 1 (mTORC1) regulation. As part of the RagGTPase family, RRAGC forms heterodimers with either RagA or RagB to signal intracellular amino acid concentrations to mTORC1, thereby controlling cellular metabolism, growth, and protein synthesis .

How does RRAGC interact with other proteins in the mTORC1 pathway?

RRAGC primarily interacts with RRAGA (RagA) to form functional heterodimers . These heterodimers play an essential role in the amino acid-dependent activation of mTORC1. When amino acids are abundant, RagC (in its GDP-bound state) and RagA (in its GTP-bound state) form an active heterodimer that binds to the Raptor component of mTORC1, facilitating its localization to the lysosomal surface where it can be activated by Rheb .

Additionally, RRAGC interacts with the protein product of the tumor suppressor gene FLCN (folliculin). Research has shown that mutations in RRAGC can decrease these interactions with folliculin while increasing binding to Raptor, resulting in aberrant mTORC1 activation .

What experimental models are commonly used to study RRAGC function?

Several experimental models have been developed to study RRAGC function:

  • Cell Culture Systems: HEK293T cells and lymphoma cell lines are commonly used for stable retroviral or lentiviral transduction of wild-type or mutant RRAGC .

  • Genetically Engineered Mouse Models: Mice engineered to endogenously express RagC mutants have been developed to study the physiological impact of nutrient signaling on normal and pathological B cell responses .

  • Yeast Models: Saccharomyces cerevisiae expressing mutations in the RRAGC homolog Gtr2 provides a simpler eukaryotic system to study conserved functions .

  • In vitro Kinase Assays: These are used to study phosphorylation of RRAGC by kinases such as mTOR and Akt2 .

What methods are used to measure RRAGC activity in cellular contexts?

Researchers typically assess RRAGC activity through several methodologies:

  • Phosphorylation Assays: Monitoring downstream substrate phosphorylation (e.g., RPS6KB/S6-kinase phosphorylation) as a readout of mTORC1 activation .

  • Amino Acid Starvation and Refeeding Experiments: Analyzing mTORC1 activity in cells expressing wild-type or mutant RRAGC under conditions of nutrient withdrawal and reintroduction, particularly leucine and arginine .

  • Protein-Protein Interaction Assays: Co-immunoprecipitation or proximity-ligation assays to assess binding of RRAGC to partners like Raptor and folliculin .

  • Cell Size Determination: Using particle size counters like Coulter Z2 to measure changes in cell size as a phenotypic readout of altered mTORC1 signaling .

What are the mechanisms behind RRAGC mutations in follicular lymphoma?

RRAGC mutations occur in approximately 9.4% of follicular lymphoma cases, with mutations distinctly clustering on one protein surface area surrounding the GTP/GDP-binding sites . The mechanistic consequences include:

  • Enhanced Raptor Binding: Mutated RRAGC proteins demonstrate increased binding to RPTOR (raptor), a component of mTORC1 .

  • Reduced Folliculin Interaction: RRAGC mutations substantially decrease interactions with the product of the tumor suppressor gene FLCN (folliculin) .

  • Constitutive mTORC1 Activation: Multiple RRAGC mutations demonstrate elevated MTOR activation even under nutrient-deprived conditions, as evidenced by increased RPS6KB/S6-kinase phosphorylation .

  • Nutrient-Independent Signaling: RRAGC mutations confer partial resistance to amino acid withdrawal, specifically leucine and arginine, allowing continued mTORC1 signaling when these nutrients are scarce .

These mechanisms suggest RRAGC mutations provide follicular lymphoma cells with a growth advantage by maintaining anabolic processes even under nutrient-limited conditions.

How do RRAGC mutations specifically impact B cell function and lymphomagenesis?

RRAGC mutations exert several specific effects on B cell function that may contribute to lymphomagenesis:

  • Enhanced Germinal Center Formation: RagC mutant mice show approximately 3-fold increase in germinal center B cells following immunization with sheep red blood cells (SRBC) .

  • Increased Plasma Cell Production: Mice with RagC mutations demonstrate elevated plasma cell formation and higher titers of class-switched, IgG1 antibodies in serum .

  • Altered B Cell Transcriptional Programs: Transcriptional profiling of B cells from RagC mutant follicular lymphomas reveals enrichment of mTORC1 signaling signatures, translation, c-myc targets, and suppression of lysosomal biogenesis .

  • Accelerated Lymphomagenesis: In mouse models, RagC mutations accelerate experimental lymphomagenesis, while creating a vulnerability to mTORC1 inhibitors .

  • Selective Activation of B Cells: Mutations in the nutrient signaling pathway enhance B cell autonomous activation, potentially corrupting nutrient-dependent control of paracrine positive signals from the T cell microenvironment .

These findings provide evidence for a critical oncogenic role of RRAGC mutations in human follicular lymphoma and suggest potential therapeutic vulnerabilities.

What is the three-dimensional significance of RRAGC mutation hotspots?

Analysis of RRAGC mutations in follicular lymphoma reveals that they cluster distinctly on one protein surface area surrounding the GTP/GDP-binding sites . This non-random distribution suggests functional significance:

  • Structural Impact: The clustering of mutations around nucleotide-binding sites likely affects the GTP/GDP binding or hydrolysis properties of the protein.

  • Conformational Changes: These mutations may lock the protein in a specific conformation that favors interaction with certain binding partners (like Raptor) while disfavoring others (like folliculin).

  • Protein Complex Assembly: The affected surface area might be critical for the proper assembly of the heterodimeric Rag complex or its interaction with regulatory proteins.

  • Nucleotide Exchange Rates: Mutations may alter the intrinsic rate of GDP-GTP exchange or GTP hydrolysis, affecting the molecular switch function of RRAGC.

Three-dimensional modeling and structural biology approaches are essential for fully understanding how these hotspot mutations influence RRAGC function in disease states.

What experimental approaches can be used to screen for pharmacological inhibitors of mutant RRAGC?

Several experimental approaches can be employed to identify and validate potential inhibitors of mutant RRAGC:

  • High-Throughput Screening: Using cell lines expressing mutant RRAGC to screen compound libraries for those that selectively inhibit mutant RRAGC-driven mTORC1 signaling.

  • Structure-Based Drug Design: Leveraging knowledge of the three-dimensional location of RRAGC hotspot mutations to design small molecules that specifically target these altered regions .

  • Phenotypic Screening: Assessing the ability of compounds to reverse cellular phenotypes associated with RRAGC mutations, such as increased cell size or enhanced survival under nutrient-limited conditions .

  • In Vivo Validation: Testing promising compounds in mouse models of RagC mutant follicular lymphoma to assess their efficacy and toxicity profiles .

  • Combination Therapy Assessment: Evaluating potential synergies between direct RRAGC inhibitors and other targeted therapies, such as mTOR inhibitors like rapamycin .

What is the concept of "ragopathies" and how does RRAGC contribute to these diseases?

"Ragopathies" is a term used to describe diseases associated with RagGTPase dysfunction . These conditions may result from mutations in genes encoding the Rags themselves or in their upstream regulators, leading to various clinical manifestations:

  • Cancer Development: Mutations in RRAGC have been identified in approximately 9.4% of follicular lymphoma cases, where they contribute to oncogenesis through constitutive activation of mTORC1 signaling .

  • Metabolic Disorders: Dysfunction in Rag-mediated nutrient sensing may contribute to metabolic abnormalities due to aberrant mTORC1 signaling.

  • Developmental Abnormalities: Given the role of mTORC1 in growth and development, ragopathies may manifest as developmental disorders.

  • Tissue-Specific Pathologies: Clinical features of ragopathies may include cataract, kidney tubulopathy, dilated cardiomyopathy, and various cancer types .

Understanding RRAGC's contribution to these pathologies provides potential avenues for therapeutic intervention targeting the Rag-mTORC1 axis.

How can RRAGC mutations be targeted therapeutically in follicular lymphoma?

RRAGC mutations in follicular lymphoma create both oncogenic drivers and potential therapeutic vulnerabilities:

  • mTORC1 Inhibitors: Follicular lymphomas with RRAGC mutations show selective sensitivity to pharmacological inhibition of mTORC1 in vivo, suggesting mutations in the nutrient signaling pathway could serve as potential markers for patients who would benefit from treatment with mTOR inhibitors .

  • Rapamycin and Analogs: Studies show that the exacerbated humoral response in RagC mutant mice is sensitive to rapamycin treatment, indicating potential efficacy of rapalogs in treating RRAGC-mutant follicular lymphoma .

  • Targeting Nutrient Sensing: Approaches that disrupt the aberrant nutrient sensing caused by RRAGC mutations could potentially restore normal regulation of mTORC1.

  • Combinatorial Approaches: Combining mTORC1 inhibitors with other targeted therapies could enhance efficacy against RRAGC-mutant follicular lymphoma.

  • Direct RRAGC Targeting: Development of small molecules that specifically bind mutant RRAGC and interfere with its interaction with Raptor could provide a more selective therapeutic approach.

What is the differential impact of mutations in RagA versus RagC in human pathology?

An intriguing aspect of Rag mutations in human disease is the apparent differential impact of mutations in RagA versus RagC:

  • Mutation Frequency: Mutations in RRAGC are found in approximately 9.4% of follicular lymphoma cases, while mutations in RRAGA are rare in lymphoid and solid malignancies .

  • Functional Asymmetry: Experimental evidence demonstrates a biochemical asymmetry between the Rag heterodimeric components. Heterozygous RagA GTP/+ cells show no detectable signaling perturbation, while RagC S74C/+ and RagC T89N/+ cells demonstrate partial resistance to amino acid withdrawal .

  • B Cell Specificity: The selective occurrence of RRAGC mutations in follicular lymphoma suggests a particular effect of such genetic alterations on the physiology and pathology of B lymphocytes .

  • Evolutionary Conservation: The differential impact of RagA versus RagC mutations appears to be conserved, as similar effects are observed in yeast models expressing mutations in their respective homologs, Gtr1 and Gtr2 .

This asymmetry provides a potential explanation for the exclusive occurrence of RRAGC heterozygous mutations, and the absence of mutations in RRAGA in human follicular lymphoma.

What techniques are used to assess the impact of RRAGC phosphorylation on mTORC1 regulation?

Several specialized techniques are employed to study RRAGC phosphorylation and its effects on mTORC1 regulation:

  • In Vitro Kinase Analysis: This approach involves preparing equal aliquots of purified FLAG-RRAGC and incubating them with active mTOR fragment or other kinases (like Akt2) in the presence or absence of specific inhibitors (such as Torin or GDC-0068) .

  • Phospho-specific Antibodies: Development and use of antibodies that specifically recognize phosphorylated residues on RRAGC (such as S2, S21, and T394) to monitor phosphorylation status in various conditions.

  • Phosphorylation Site Mutants: Generation of RRAGC constructs with phospho-mimetic (e.g., S2E/S21E/T394E) or phospho-deficient (e.g., S2A/S21A/T394A) mutations to study the functional consequences of phosphorylation .

  • Mass Spectrometry: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis of purified RRAGC to identify and quantify phosphorylation sites under different cellular conditions.

  • Functional Readouts: Assessment of downstream mTORC1 activation through monitoring phosphorylation of substrates like S6K and 4E-BP1 in cells expressing wild-type versus phosphorylation site mutants of RRAGC .

What experimental systems are most effective for studying the role of RRAGC in humoral immune responses?

Research on RRAGC's role in humoral immunity employs several complementary experimental systems:

  • Immunization Models: Intraperitoneal immunization with sheep red blood cells (SRBC) or hapten-carrier complexes like NP-KLH in mice expressing wild-type or mutant RRAGC, followed by analysis of germinal center formation and antibody production .

  • Flow Cytometry: Quantification of different B cell populations (including germinal center B cells and plasma cells) in immunized mice to assess the impact of RRAGC mutations on B cell differentiation and function .

  • Affinity-Dependent ELISA: Measurement of high-affinity antibodies in serum over time to evaluate the effect of RRAGC mutations on affinity maturation .

  • Rapamycin Treatment Protocols: Administration of rapamycin for varying durations during the humoral response to determine the temporal requirements for mTORC1 signaling in different phases of the response .

  • Transcriptional Profiling: Analysis of gene expression in sorted B cells from wild-type versus RRAGC mutant mice to identify altered gene expression patterns associated with enhanced B cell activation .

These systems collectively provide insights into how RRAGC mutations affect B cell activation, differentiation, and function during humoral immune responses.

What are the advantages and limitations of different model systems for studying RRAGC mutations?

Various model systems offer distinct advantages and limitations for investigating RRAGC mutations:

Model SystemAdvantagesLimitations
Cell Lines (HEK293T, lymphoma)- Easy genetic manipulation
- Rapid experimental turnover
- Well-suited for biochemical studies
- Good for high-throughput screening
- Lack physiological context
- May not recapitulate tissue-specific effects
- Often contain other genetic alterations
- Limited cell-cell interactions
Mouse Models- Physiologically relevant
- Allow study of immune responses in vivo
- Enable assessment of lymphomagenesis
- Permit testing of therapeutic interventions
- Expensive and time-consuming
- Species differences from humans
- Genetic background effects
- May require specialized facilities
Yeast (S. cerevisiae)- Simple eukaryotic system
- Rapid generation time
- Highly amenable to genetic manipulation
- Conserved Rag functions
- Lacks mammalian complexity
- No adaptive immune system
- Differences in nutrient sensing
- Limited relevance to cancer
In vitro Biochemical Assays- Precise control of experimental conditions
- Direct assessment of protein activities
- Quantitative measurements
- Mechanistic insights
- Artificial conditions
- Lack cellular context
- May not reflect in vivo regulation
- Limited to purifiable components

Selecting the appropriate model system depends on the specific research question, with complementary approaches often providing the most comprehensive understanding of RRAGC function and mutation effects.

How might single-cell technologies advance our understanding of RRAGC's role in follicular lymphoma?

Single-cell technologies offer promising avenues for deeper insights into RRAGC's role in follicular lymphoma:

  • Single-Cell RNA Sequencing: This can reveal heterogeneity within RRAGC-mutant follicular lymphoma populations, potentially identifying distinct transcriptional programs in subclones with different functional properties or therapeutic vulnerabilities.

  • Single-Cell Proteomics: Mass cytometry (CyTOF) or other single-cell protein analysis methods can map the activation state of mTORC1 signaling networks at the individual cell level, revealing how RRAGC mutations affect signaling heterogeneity within tumors.

  • Spatial Transcriptomics/Proteomics: These approaches can map the distribution of RRAGC-mutant cells within lymphoma tissue microenvironments, potentially uncovering spatial relationships between mutant cells and supporting stromal or immune cells.

  • Single-Cell Metabolomics: Emerging technologies for single-cell metabolic profiling could reveal how RRAGC mutations affect nutrient utilization and metabolic reprogramming in individual lymphoma cells.

  • Lineage Tracing: Combining genetic barcoding with single-cell sequencing in mouse models could track the clonal evolution of RRAGC-mutant B cells during lymphomagenesis, revealing selection pressures and cooperative genetic events.

What approaches could identify synthetic lethal interactions with RRAGC mutations?

Identifying genetic interactions that create vulnerabilities specifically in RRAGC-mutant cells could lead to novel therapeutic strategies:

  • CRISPR-Cas9 Screens: Genome-wide or focused CRISPR knockout or activation screens in RRAGC-mutant versus wild-type cells can identify genes whose disruption or activation is selectively lethal to mutant cells.

  • Drug Combination Screens: Testing libraries of approved drugs or investigational compounds in combination with mTOR inhibitors could identify synergistic combinations specifically effective against RRAGC-mutant lymphomas.

  • Metabolic Dependency Profiling: Systematic testing of nutrient dependencies or metabolic pathway inhibitors may uncover metabolic vulnerabilities specific to cells with dysregulated nutrient sensing due to RRAGC mutations.

  • Computational Approaches: Network analysis and computational modeling of signaling pathways affected by RRAGC mutations could predict potential synthetic lethal interactions for experimental validation.

  • Patient-Derived Models: Testing candidate synthetic lethal interactions in patient-derived xenografts or organoids with natural RRAGC mutations could validate clinically relevant vulnerabilities.

These approaches could identify targetable dependencies that could translate into more effective and selective therapies for RRAGC-mutant follicular lymphoma.

Product Science Overview

Introduction

Ras-Related GTP Binding C (RRAGC) is a protein encoded by the RRAGC gene in humans. It belongs to the Ras superfamily of small GTPases, which are involved in various cellular processes, including cell growth, differentiation, and intracellular trafficking . RRAGC is particularly notable for its role in the regulation of the mechanistic target of rapamycin (mTOR) signaling pathway, which is crucial for cell growth and metabolism .

Structure and Function

RRAGC is a GTP-binding protein that cycles between an active GTP-bound state and an inactive GDP-bound state. This cycling is essential for its function as a molecular switch in signaling pathways. The protein is composed of several conserved domains that facilitate its interaction with other proteins and its ability to bind and hydrolyze GTP .

Role in mTOR Signaling

The mTOR signaling pathway is a central regulator of cell growth and metabolism, responding to various environmental cues such as nutrient availability, energy status, and growth factors. RRAGC, along with its binding partners RRAGA, RRAGB, and RRAGD, forms a heterodimeric complex that is crucial for the activation of mTORC1 (mechanistic target of rapamycin complex 1) in response to amino acids . This activation is essential for the proper regulation of protein synthesis, autophagy, and other metabolic processes.

Clinical Significance

Mutations or dysregulation of RRAGC have been associated with several diseases, including cancer and metabolic disorders . For instance, alterations in the mTOR signaling pathway, where RRAGC plays a pivotal role, have been implicated in the development and progression of various cancers. Additionally, RRAGC has been linked to rare genetic disorders such as Long-Olsen-Distelmaier Syndrome and Birt-Hogg-Dube Syndrome .

Therapeutic Potential

Given its central role in mTOR signaling, RRAGC is a potential target for therapeutic interventions. Inhibitors or modulators of RRAGC function could be developed to treat diseases associated with mTOR dysregulation, such as cancer and metabolic disorders . Research is ongoing to better understand the precise mechanisms by which RRAGC regulates mTOR signaling and to develop targeted therapies that can modulate its activity.

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.