ARHGEF39 Human

Rho Guanine Nucleotide Exchange Factor 39 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Mechanisms

Rho GTPase Activation

  • Primary Target: Activates RHOA, a GTPase involved in stress fiber formation and cell adhesion .

  • Cellular Effects:

    • Promotes cell de-adhesion and neural progenitor proliferation .

    • Enhances cancer cell viability, migration, and invasion via AKT/ERK signaling .

Key Pathways

PathwayRole of ARHGEF39Associated Cancers
AKT/ERKPhosphorylates AKT (Ser473) and ERK (Thr202/Tyr204), driving cell proliferationccRCC, gastric cancer
RHOAInduces cytoskeletal reorganization, impacting cell motilityHCC, NSCLC

Clinical and Disease Associations

Neurodevelopmental Disorders

  • Developmental Language Disorder (DLD): A functional polymorphism disrupts miRNA regulation of ARHGEF39, implicating it in neurogenesis defects .

Research Tools and Applications

Recombinant Protein Use

  • Applications: SDS-PAGE, Western blot, and imaging assays (denatured form) .

  • Storage: Stable at -80°C; avoid freeze-thaw cycles .

Interactome
ARHGEF39 interacts with:

  • TIFA: Modulates NF-κB signaling .

  • MELK: Regulates cell cycle progression .

  • RHOC/RHOB: Mediates cytoskeletal dynamics .

Therapeutic Potential and Future Directions

ARHGEF39’s dual role in neurodevelopment and oncogenesis positions it as a biomarker and therapeutic target. Inhibitors targeting its RhoGEF domain or downstream AKT/ERK pathways could mitigate cancer progression. Further in vivo studies are needed to validate these mechanisms .

Product Specs

Introduction
Rho Guanine Nucleotide Exchange Factor 39, also known as ARHGEF39, is a protein coding gene. This gene encodes for a protein with one DH (DBL-homology) domain and one PH domain. ARHGEF39 promotes cell proliferation.
Description
Recombinant human ARHGEF39 protein was expressed in E. coli. This protein is a single, non-glycosylated polypeptide chain consisting of 358 amino acids (amino acids 1-335). It has a molecular mass of 40.7 kDa. A 23 amino acid His-tag was fused to the N-terminus of ARHGEF39. This protein was purified using proprietary chromatographic techniques.
Physical Appearance
A clear solution that has been sterilized by filtration.
Formulation
The ARHGEF39 protein solution (1 mg/mL) is supplied in a buffer containing 20 mM Tris-HCl (pH 8.0) and 10% glycerol.
Stability
The product should be stored at 4°C for 2-4 weeks. For long-term storage, freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated cycles of freezing and thawing.
Purity
Purity is greater than 85% as determined by SDS-PAGE analysis.
Synonyms
Rho guanine nucleotide exchange factor 39, ARHGEF39, C9orf100, RP11-331F9.7.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMELSCPG SRCPVQEQRA RWERKRACTA RELLETERRY QEQLGLVATY FLGILKAKGT LRPPERQALF GSWELIYGAS QELLPYLEGG CWGQGLEGFC RHLELYNQFA ANSERSQTTL QEQLKKNKGF RRFVRLQEGR PEFGGLQLQD LLPLPLQRLQ QYENLVVALA ENTGPNSPDH QQLTRAARLI SETAQRVHTI GQKQKNDQHL RRVQALLSGR QAKGLTSGRW FLRQGWLLVV PPHGEPRPRM FFLFTDVLLM AKPRPPLHLL RSGTFACKAL YPMAQCHLSR VFGHSGGPCG GLLSLSFPHE KLLLMSTDQE ELSRWYHSLT WAISSQKN.

Q&A

What is ARHGEF39 and what is its primary function in human cells?

ARHGEF39 belongs to the family of Rho guanine nucleotide exchange factors (RhoGEFs) that activate small Rho GTPases. These factors initiate Rho GTPase activation by stimulating the exchange of GDP for GTP. In experimental settings, ARHGEF39 has been demonstrated to specifically activate RHOA, but not CDC42 or RAC1, as shown through FRET-based Rho GTPase biosensor assays . RHOA is known to be involved in the assembly of cell-matrix interactions via focal adhesions. Functionally, ARHGEF39 appears to play important roles in cell adhesion processes and neural progenitor cell proliferation during brain development .

Methodology for studying ARHGEF39's function typically involves overexpression studies in cell models, FRET-based biosensor assays for measuring Rho GTPase activation, and analysis of cell adhesion phenotypes following manipulation of ARHGEF39 expression levels.

How does ARHGEF39 relate to neurodevelopmental processes?

ARHGEF39 has been identified as a marker gene for proliferating neural progenitor cells in the developing brain. Analysis of single-cell RNA sequencing (scRNA-seq) data from both mouse and human neocortex development indicates that ARHGEF39 is co-expressed with genes involved in cell division . This expression pattern suggests a role for ARHGEF39 in neurogenesis, specifically in regulating neural progenitor cell proliferation.

The methodological approach to understanding this relationship involves analyzing existing scRNA-seq datasets to:

  • Identify cell types where ARHGEF39 is expressed

  • Determine co-expressed genes to infer biological pathways

  • Map expression patterns across developmental timepoints

The association between ARHGEF39 and developmental language disorder (DLD) further suggests its importance in neurodevelopment, particularly in language-related circuits .

What techniques are used to measure ARHGEF39 activation of Rho GTPases?

Researchers employ several methodologies to measure ARHGEF39's activation of Rho GTPases, with the most definitive being FRET-based biosensor assays:

  • FRET-Based Rho GTPase Biosensor Assay: This technique uses genetically encoded Förster Resonance Energy Transfer (FRET) biosensors for specific Rho GTPases. The biosensor constructs contain:

    • The Rho GTPase of interest (e.g., RHOA, RAC1, CDC42)

    • A Rho GTPase binding domain (RBD)

    • Two fluorophores (typically mTFP as donor and Venus as acceptor)

When the Rho GTPase is activated and binds to the RBD, the fluorophores are brought into close proximity, allowing energy transfer from the donor to the acceptor fluorophore, which can be detected as a change in the emission spectrum .

The experimental protocol involves:

  • Seeding cells (e.g., HEK293FT) in poly-D-lysine-coated glass-bottom plates

  • Co-transfecting cells with the biosensor plasmid, ARHGDIA plasmid (to increase dynamic range), and varying amounts of ARHGEF39 expression plasmid

  • Measuring fluorescence at specific wavelengths (excitation at 453 nm, emission scan from 487-600 nm)

  • Calculating the ratio between emissions at 528 nm and 492 nm to determine Rho GTPase activation

This approach allows for quantitative measurement of ARHGEF39's specificity for different Rho GTPases and has demonstrated that ARHGEF39 activates RHOA but not CDC42 or RAC1.

What mechanisms underlie ARHGEF39's role in developmental language disorder?

ARHGEF39 was implicated in developmental language disorder (DLD) through a functional polymorphism (rs72727021) located in its 3′UTR region. The mechanism involves disruption of post-transcriptional regulation by microRNAs, specifically miR-215 . The methodological approach to uncovering this mechanism involved:

  • Screening non-coding 3′UTR sequences for variations that could disrupt microRNA binding sites in children with DLD

  • Associating the identified single nucleotide polymorphism with quantitative measures of language impairment (non-word repetition tasks)

  • Conducting functional assays in cell models to demonstrate that the risk allele disrupts regulation of ARHGEF39 by miR-215

  • Analyzing expression quantitative trait loci data, which showed that the DLD-associated allele correlates with higher expression of ARHGEF39 in post-mortem human brain tissue

This suggests that elevated ARHGEF39 expression may contribute to DLD. The connection to language development likely involves ARHGEF39's role in neural progenitor cell proliferation and potentially in cytoskeletal remodeling during neurodevelopment, which are critical for proper neural circuit formation.

How does ARHGEF39 influence cell adhesion in neural contexts?

ARHGEF39 appears to regulate cell adhesion through its activation of RHOA. Experimental evidence demonstrates that overexpression of ARHGEF39 in cell cultures leads to an increased number of cells in suspension compared to control conditions . The methodological investigation of this phenomenon revealed:

  • Quantitative assessment showed significantly more cells in suspension in ARHGEF39-overexpressing cultures (p=0.0001)

  • Cell viability analysis using trypan blue staining showed that floating cells in the ARHGEF39 overexpression condition remained largely viable (~78%), compared to poor viability (~42%) of the small number of floating cells in control conditions

This suggests that ARHGEF39 overexpression induces cell detachment without causing cell death. The mechanism likely involves ARHGEF39's activation of RHOA, which is known to regulate focal adhesions and cell-matrix interactions. In neural contexts, this function could affect neural progenitor cell migration, neuronal positioning, or axon guidance during development.

Dysregulation of these processes could contribute to neurodevelopmental disorders, including DLD, by affecting the proper formation of neural circuits necessary for language acquisition and processing.

What experimental approaches can help elucidate ARHGEF39's role in neurodevelopmental disorders?

To fully understand ARHGEF39's contribution to neurodevelopmental disorders, researchers should employ multi-level experimental approaches:

  • Single-cell transcriptomics: Analyzing scRNA-seq data from developing brain tissue to:

    • Track ARHGEF39 expression across developmental timepoints

    • Identify cell types with enriched ARHGEF39 expression

    • Map co-expression networks to infer biological pathways

  • Functional studies in neural cell models:

    • CRISPR-Cas9 knockout or knockdown of ARHGEF39 in neural progenitor cells

    • Overexpression studies with wild-type and mutant ARHGEF39

    • Assessment of proliferation, differentiation, migration, and adhesion phenotypes

    • Time-lapse imaging to track cell behavior in real-time

  • Rho GTPase activation assays in neural cells:

    • FRET-based biosensors to measure RHOA activation in neural progenitor cells

    • Pull-down assays to detect GTP-bound (active) RHOA

    • Analysis of downstream effectors of RHOA signaling

  • Animal models with Arhgef39 mutations:

    • Generation of conditional knockout or knockin models

    • Assessment of neurogenesis, neuronal migration, and brain development

    • Behavioral testing related to language and communication (in appropriate models)

  • Patient-derived cellular models:

    • iPSC-derived neural progenitors and neurons from individuals with DLD

    • Comparison of ARHGEF39 expression, RHOA activation, and cellular phenotypes

    • Rescue experiments to restore normal function

What is the relationship between ARHGEF39 expression and cancer progression?

ARHGEF39 has been implicated in multiple cancer types, with evidence suggesting its role as a potential oncogene. In hepatocellular carcinoma (HCC), ARHGEF39 expression is significantly elevated compared to adjacent normal tissues, and high expression correlates with poor clinical outcomes .

Key findings from HCC studies include:

Similarly, in clear cell renal cell carcinoma (ccRCC), ARHGEF39 promotes cell viability, migration, and invasion by regulating the AKT/ERK signaling pathway .

Methodologically, these associations have been investigated through:

  • Analysis of TCGA database expression data

  • RT-qPCR and immunohistochemistry of patient samples

  • Functional studies in cancer cell lines

  • Survival analyses using Kaplan-Meier and Cox regression models

The oncogenic function of ARHGEF39 may relate to its role in cell proliferation, as observed in neural progenitor cells, suggesting common cellular mechanisms across different contexts.

How do cellular mechanisms of ARHGEF39 in neural development potentially relate to its role in language disorders?

The cellular mechanisms by which ARHGEF39 functions in neural development provide insight into potential pathways affecting language acquisition and processing. Based on current evidence, several potential connections can be proposed:

  • Neural progenitor cell proliferation: ARHGEF39 is a marker gene for proliferating neural progenitor cells and is co-expressed with genes involved in cell division . Proper neurogenesis is essential for establishing the correct number and types of neurons in language-related circuits. Dysregulation of this process could lead to structural abnormalities in language processing networks.

  • Cell adhesion regulation: ARHGEF39 activation of RHOA affects cell adhesion properties , which is critical for:

    • Neural migration during development

    • Axon guidance and targeting

    • Synapse formation and maintenance

Each of these processes is essential for establishing functional neural circuits, including those involved in language processing.

  • Cytoskeletal reorganization: Through RHOA activation, ARHGEF39 influences cytoskeletal dynamics , which affects:

    • Dendritic spine development

    • Axonal growth and branching

    • Structural plasticity of synapses

These processes contribute to the fine-tuning of neural connections during language acquisition and learning.

Research methodologies to investigate these connections should include:

  • Analysis of ARHGEF39 expression in language-related brain regions during development

  • Correlation of ARHGEF39 polymorphisms with neuroimaging data in DLD patients

  • Investigation of synaptic plasticity in neural models with altered ARHGEF39 expression

  • Assessment of dendritic spine morphology and density in neurons with ARHGEF39 mutations

What are the optimal experimental systems for studying ARHGEF39 function?

When investigating ARHGEF39 function, researchers should consider multiple experimental systems, each with specific advantages:

  • Cell line models:

    • HEK293 cells provide a tractable system for basic biochemical studies, such as FRET-based Rho GTPase activation assays

    • Neuroblastoma cell lines (SH-SY5Y, Neuro2A) offer a neuronal-like background for studying ARHGEF39 in a more relevant context

    • Primary neural progenitor cells represent a more physiologically relevant system for studying ARHGEF39's role in neurogenesis

  • Organoid models:

    • Brain organoids derived from human iPSCs can recapitulate aspects of human brain development

    • Allow for the study of ARHGEF39 in a three-dimensional context that includes multiple cell types and developmental processes

    • Can be generated from patient-derived cells carrying ARHGEF39 variants

  • Animal models:

    • Mouse models with Arhgef39 knockout or knockin mutations can reveal developmental functions

    • Conditional and inducible systems allow for temporal and spatial control of Arhgef39 expression

    • In utero electroporation can be used for acute manipulation of Arhgef39 in specific brain regions during development

  • Patient-derived samples:

    • Post-mortem brain tissue from individuals with language disorders

    • iPSC-derived neural cells from patients with DLD or ARHGEF39 variants

    • Genetic association studies linking ARHGEF39 variants to language phenotypes

The choice of experimental system should be guided by the specific research question, with consideration of the trade-offs between physiological relevance and experimental tractability.

How can single-cell RNA sequencing data be leveraged to understand ARHGEF39's cell-type specific functions?

Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful tool for understanding gene expression at cellular resolution. For ARHGEF39 research, scRNA-seq data can be leveraged in several ways:

  • Cell type identification and marker analysis:

    • Determine which specific cell types express ARHGEF39

    • Establish if ARHGEF39 serves as a marker gene for particular cell states

    • Previous analyses have identified ARHGEF39 as a marker for proliferating neural progenitor cells

  • Developmental trajectory analysis:

    • Track ARHGEF39 expression across developmental pseudotime

    • Identify when ARHGEF39 expression is upregulated or downregulated during neurogenesis

    • Associate expression changes with specific developmental transitions

  • Co-expression network analysis:

    • Identify genes that are co-expressed with ARHGEF39

    • Infer biological pathways and processes associated with ARHGEF39 function

    • Previous analyses have shown co-expression with genes involved in cell division

  • Spatial context integration:

    • Combine scRNA-seq with spatial transcriptomics data

    • Map ARHGEF39-expressing cells to specific brain regions and developmental structures

    • Correlate expression patterns with known language-related circuits

  • Cell-cell interaction analysis:

    • Predict ligand-receptor interactions between ARHGEF39-expressing cells and other cell types

    • Understand how ARHGEF39-mediated signals might influence neighboring cells

Methodological approaches should include:

  • Reanalysis of existing datasets (e.g., developing human and mouse neocortex )

  • Generation of new scRNA-seq data from relevant experimental models

  • Integration of multiple datasets for cross-species and cross-developmental stage comparisons

  • Validation of key findings using orthogonal methods such as in situ hybridization, immunohistochemistry, or FISH

What are the critical unresolved questions about ARHGEF39 in human neurodevelopment?

Several critical questions remain unanswered regarding ARHGEF39's role in human neurodevelopment:

  • Temporal and spatial specificity:

    • When and where is ARHGEF39 expression most critical during brain development?

    • Are there specific developmental windows during which ARHGEF39 dysfunction most severely impacts language circuit formation?

  • Regulatory mechanisms:

    • What factors regulate ARHGEF39 expression during neurodevelopment?

    • How do microRNAs beyond miR-215 contribute to ARHGEF39 regulation?

    • What epigenetic mechanisms control ARHGEF39 expression in neural progenitor cells?

  • Downstream effectors:

    • What are the key downstream targets of ARHGEF39-activated RHOA in neural progenitor cells?

    • How does ARHGEF39 signaling intersect with other neurodevelopmental pathways?

  • Functional specificity in language circuits:

    • Does ARHGEF39 have specialized functions in brain regions associated with language processing?

    • How do ARHGEF39 variants specifically affect language-related neural circuits rather than causing broader neurodevelopmental impairments?

  • Therapeutic potential:

    • Can modulation of ARHGEF39 or its downstream pathways ameliorate language impairments in DLD?

    • Are there critical periods during which intervention would be most effective?

Addressing these questions will require integrated approaches combining:

  • Advanced genetic models with cell-type and temporal specificity

  • Human neuroimaging studies correlating ARHGEF39 variants with structural and functional connectivity

  • Longitudinal studies of language development in individuals with ARHGEF39 variants

  • Pharmacological and genetic rescue experiments in model systems

How might findings from ARHGEF39 cancer research inform neurodevelopmental studies?

The parallels between ARHGEF39's roles in cancer progression and neural development offer opportunities for cross-disciplinary insights:

  • Cellular proliferation mechanisms:

    • ARHGEF39's association with increased proliferation in cancer cells mirrors its expression in proliferating neural progenitor cells

    • Understanding the molecular mechanisms by which ARHGEF39 enhances proliferation in cancer could inform how it regulates neurogenesis

  • Signaling pathway integration:

    • In ccRCC, ARHGEF39 regulates the AKT/ERK signaling pathway

    • Investigating whether similar pathway interactions occur in neural progenitor cells could reveal how ARHGEF39 influences neuronal differentiation and survival

  • Cell adhesion and migration:

    • ARHGEF39 promotes migration and invasion in cancer cells

    • These functions likely involve the same RHOA-mediated cytoskeletal regulation that affects cell adhesion in other contexts

    • Understanding these mechanisms could inform how ARHGEF39 influences neuronal migration during development

  • Biomarker and therapeutic target insights:

    • Methods developed to detect and target ARHGEF39 in cancer research

    • Adaptation of these approaches for neurodevelopmental research, particularly for early detection of language disorders

Methodological approaches should include:

  • Comparative analysis of ARHGEF39-associated gene expression profiles across cancer and neural datasets

  • Testing of cancer-derived ARHGEF39 inhibitors in neural models

  • Investigation of whether ARHGEF39 polymorphisms associated with DLD also influence cancer susceptibility or progression

Product Science Overview

Introduction

Rho Guanine Nucleotide Exchange Factor 39 (ARHGEF39), also known as C9orf100, is a protein-coding gene that plays a crucial role in cellular processes. It is a member of the Dbl-family of guanine nucleotide exchange factors (GEFs), which are responsible for activating Rho GTPases by facilitating the exchange of GDP for GTP .

Gene and Protein Structure

The ARHGEF39 gene is located on chromosome 9 and encodes a protein that is involved in the regulation of cell migration and proliferation . The protein structure includes domains that are essential for its function as a guanine nucleotide exchange factor, enabling it to interact with Rho GTPases and promote their activation .

Function and Mechanism

ARHGEF39 is predicted to enable guanyl-nucleotide exchange factor activity, which is crucial for the activation of Rho GTPases . Rho GTPases act as molecular switches that regulate various cellular functions, including actin cytoskeleton organization, cell motility, polarity, growth, survival, and gene transcription . By activating Rho GTPases, ARHGEF39 plays a significant role in these cellular processes.

Pathways and Interactions

ARHGEF39 is involved in several signaling pathways, including the p75 NTR receptor-mediated signaling and signaling by Rho GTPases . It promotes cell proliferation and migration by activating the Rac1/P38 MAPK/ATF2 signaling pathway . This activation leads to the increased expression of Cyclin A2, Cyclin D1, and MMP2, which are essential for cell cycle progression and extracellular matrix degradation .

Clinical Significance

ARHGEF39 has been associated with various diseases, including glutaric acidemia I and non-small cell lung cancer (NSCLC) . In NSCLC, ARHGEF39 promotes tumor progression and invasion, and its expression correlates with poor prognosis . The protein’s role in cell migration and proliferation makes it a potential target for therapeutic interventions in cancer treatment.

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.