ARF6 Human

ADP-Ribosylation Factor 6 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles in Cellular Processes

ARF6 regulates membrane trafficking, cytoskeletal dynamics, and signaling pathways.

Membrane Trafficking

  • Endocytosis: Mediates clathrin-independent internalization of plasma membrane proteins (e.g., integrins, receptors) .

  • Recycling: Promotes recycling of internalized cargo (e.g., transferrin receptors) via endosomal compartments .

  • Vesicular Transport: Coordinates exocytosis of vesicles (e.g., GLUT1, synaptic vesicles) .

Cytoskeletal Remodeling

  • Actin Polymerization: Activates PIP5-kinase to generate PIP2, which recruits actin-binding proteins (e.g., N-WASP) .

  • Cytokinesis: Regulates vesicle transport to the midbody during cell division .

Signaling Pathways

PathwayRole of ARF6Downstream Effects
MAPK/ERK1/2Activated by PDGF-BB and Ang IIMMP14 expression, invasion
PI3K/AKTRegulated by PDGF-BBCell survival, proliferation
PAKStimulated by both growth factorsCytoskeletal reorganization

Regulation by GEFs and GAPs

ARF6 activity is controlled by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs).

Key Regulators

GEFsGAPs
ARNOACAP1
BRAG2ARAP2
CYTH3CENTRINβ1

GEFs (e.g., ARNO) localize ARF6 to membranes via lipid-binding domains, enabling GDP/GTP exchange . GAPs (e.g., ACAP1) accelerate GTP hydrolysis, returning ARF6 to its inactive state .

Expression and Tissue Distribution

ARF6 is expressed in diverse tissues, with elevated levels in immune and endothelial cells.

Tissue Expression Profile

TissueExpression LevelKey Functions
Immune CellsHighPhagocytosis, TLR signaling
Vascular SMCModerateMigration, ECM degradation
MelanomaHighMetastasis, immune evasion
PlacentaHighTrophoblast invasion

Data from the Human Protein Atlas confirm ubiquitous expression, with enriched activity in tissues requiring dynamic membrane remodeling.

Pathological Roles in Disease

ARF6 dysregulation is implicated in cancer and cardiovascular diseases.

Cancer Progression

  • Metastasis: ARF6-GTP promotes melanoma cell invasion by upregulating MMP14 and suppressing immune surveillance .

  • Immune Evasion: Recycling of IFNγ receptors in tumor cells limits antitumor T-cell responses .

Cardiovascular Disease

  • Vascular SMC Invasion: PDGF-BB and Ang II activate ARF6, driving MMP2/14-dependent ECM degradation .

  • Atherosclerosis: ARF6 depletion reduces MMP14 expression, attenuating plaque formation .

PDGF-BB and Ang II Signaling

In human aortic SMCs, PDGF-BB and Ang II transiently activate ARF6-GTP, peaking at 2 minutes . This activation:

  • PDGF-BB: Triggers MAPK/ERK, PI3K/AKT, and PAK pathways.

  • Ang II: Selectively activates MAPK/ERK and PAK pathways.

Melanoma Microenvironment

ARF6 promotes immunosuppression by:

  1. Recycling IFNγ Receptors: Limits cytokine signaling in tumor cells.

  2. Suppressing CTL Activity: Enhances Treg infiltration and PD-L1 expression .

Therapeutic Implications

  • Cancer: Inhibiting ARF6-GTP (e.g., using small molecules) may improve responses to immune checkpoint therapies .

  • Atherosclerosis: Targeting ARF6-MMP14 axis could reduce plaque instability .

Product Specs

Introduction
ARF6, a member of the ADP ribosylation factor family of GTP-binding proteins, is localized to the plasma membrane. It regulates vesicle trafficking, membrane lipid remodeling, and signaling pathways associated with actin remodeling. Additionally, ARF6 plays a crucial role in maintaining organelle integrity, assembling coat proteins, and activating phospholipase D.
Description
Recombinant ARF6, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 195 amino acids (1-175a.a.) with a molecular weight of 22.2 kDa. It includes a 20 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic methods.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The ARF6 protein solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 2 mM DTT, 0.2 mM PMSF, and 20% glycerol.
Purity
Purity exceeds 95% as determined by SDS-PAGE analysis.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For long-term storage, freeze at -20°C. Avoid repeated freeze-thaw cycles.
Synonyms

ADP-ribosylation factor 6, ARF6, DKFZp564M0264.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGKVLSKIFG NKEMRILMLG LDAAGKTTIL YKLKLGQSVT TIPTVGFNVE TVTYKNVKFN VWDVGGQDKI RPLWRHYYTG TQGLIFVVDC ADRDRIDEAR QELHRIINDR EMRDAIILIF ANKQDLPDAM KPHEIQEKLG LTRIRDRNWY VQPSCATSGD GLYEGLTWLT SNYKS

Q&A

What is ARF6 and what are its primary functions in human cells?

ARF6 is a member of the ADP-ribosylation factor (ARF) family of small GTPases and belongs to the larger RAS GTPase superfamily. Unlike other ARF family members (ARF1-5), ARF6 constitutes the sole member of class III ARFs and has distinct functions .

ARF6 primarily regulates:

  • Endocytosis and recycling of membrane proteins

  • Plasma membrane reorganization

  • Actin cytoskeleton remodeling

  • Vesicular transport between cell compartments

  • Exocytosis of secretory granules

These functions are accomplished through ARF6's ability to cycle between inactive GDP-bound and active GTP-bound states, which is regulated by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs) . In human cells, ARF6 has become increasingly recognized for its role in cancer cell invasion and migration, vascular stability, and inflammatory processes .

How is ARF6 activation regulated in human cells?

ARF6 activation occurs through a precisely controlled GDP/GTP exchange cycle mediated by specific regulatory proteins. The activation process follows these general steps:

  • In the inactive state, ARF6 is bound to GDP

  • GEF proteins (including ARNO, EFA6, BRAG, and GEP100) catalyze the exchange of GDP for GTP

  • This exchange triggers conformational changes, particularly in the switch regions

  • Activated ARF6-GTP can then interact with effector proteins

  • GAPs later promote GTP hydrolysis, returning ARF6 to its inactive state

In human cells, specific stimuli such as PDGF-BB and Angiotensin II (Ang II) can rapidly activate ARF6, as demonstrated in human aortic smooth muscle cells (HASMCs). The activation is typically transient, with peak activation occurring around 2 minutes post-stimulation before returning to basal levels .

Different GEFs use distinct mechanisms to activate ARF6. For example, EFA6 families are not autoinhibited by the PH domain, while ARNO has less regulation of ARF6 activation, with the PH domain (not Sec7 domain) mediating binding to ARF6 .

What structural changes occur during ARF6 activation?

The GDP/GTP cycle of ARF6 involves significant structural rearrangements:

  • In GDP-bound form, ARF6 adopts a specific conformation with its N-terminal helix tucked into a hydrophobic pocket

  • Upon GTP binding, a rigid body translation of approximately 6.5 Å occurs in the interswitch region

  • This destroys the binding site for the N-terminal helix, displacing it from the protein core

  • The N-terminus likely becomes less organized relative to the protein core

  • Switch I and Switch II regions undergo substantial reorganization

Interestingly, while ARF6 and ARF1 have different conformations in their GDP-bound forms, their GTP-bound structures are remarkably similar, particularly in the switch regions that interact with regulators and effectors . This suggests that discrimination between ARF isoforms occurs primarily in their inactive states rather than active states .

What are the key differences between ARF6 and other ARF family members in experimental systems?

Despite high sequence homology among ARF proteins, ARF6 exhibits several unique characteristics that distinguish it from other family members:

FeatureARF6Other ARFs (primarily ARF1)
Subcellular localizationPrimarily plasma membrane and endosomal compartmentsPrimarily Golgi apparatus
GDP-bound conformationUnique structureDifferent conformation
GTP-bound conformationSimilar to ARF1Similar to ARF6
Sequence in switch regionsNearly identical to ARF1Nearly identical to ARF6
Key sequence differencesGln37Ser38 in switch IGlu41Ile42 in ARF1 switch I
Developmental roleEssential for embryogenesisLess critical for development
Adult physiological roleDispensable at therapeutic inhibition levelsEssential for Golgi function

The striking observation that active ARF6 and ARF1 adopt similar conformations despite having different functions suggests that specificity is achieved through: (1) different inactive conformations, (2) recognition of regions outside the switch regions, or (3) cellular context and protein complexes rather than isolated proteins .

Experimentally, these differences mean that researchers must carefully consider the cellular context and binding partners when studying ARF6 function, as the isolated protein structure may not fully explain its specificity in vivo .

How does ARF6 contribute to pathological processes and what are the implications for experimental design?

ARF6 has been implicated in multiple pathological processes, with particular relevance to cardiovascular disease and cancer:

Cancer progression mechanisms:

  • Promotes cell motility and invasion

  • Regulates membrane trafficking and matrix metalloproteinase secretion

  • Contributes to drug resistance mechanisms

  • Facilitates cancer cell survival

Vascular pathology:

  • Regulates vascular smooth muscle cell invasion (VSMC)

  • Controls barrier function

  • Mediates inflammatory responses

  • Activated by growth factors (PDGF-BB) and hormones (Ang II)

When designing experiments to study ARF6 in pathological contexts, researchers should consider:

  • Cell-type specificity: ARF6 functions differently in various cell types; thus, experiments should be performed in disease-relevant cells

  • Activation dynamics: ARF6 activation is typically rapid and transient, peaking at ~2 minutes in HASMCs

  • Downstream pathway analysis: Different stimuli can activate different downstream pathways (e.g., PDGF-BB activates both MAPK and PI3K, while Ang II primarily activates MAPK in HASMCs)

  • Compensatory mechanisms: Long-term ARF6 inhibition may trigger compensatory pathways

Importantly, while animal models have provided valuable insights, human cells can exhibit distinct signaling patterns. For example, while rat VSMC studies have been informative, validation in human VSMCs is crucial, as demonstrated by recent work showing ARF6 regulation of invasion in human aortic smooth muscle cells .

What are the critical considerations when interpreting contradictory data in ARF6 research?

Researchers frequently encounter contradictory findings when studying ARF6. Several factors may contribute to these discrepancies:

  • Isoform specificity issues: The high sequence similarity between ARF proteins (particularly in switch regions) can lead to misinterpretation of results from non-specific antibodies or inhibitors

  • Context-dependent functions: ARF6 may play different roles depending on:

    • Cell type (epithelial vs. endothelial vs. smooth muscle)

    • Activation stimulus (growth factor vs. hormone)

    • Cellular microenvironment

    • Available GEFs/GAPs in the system

  • Experimental approach differences:

    • Acute vs. chronic manipulation

    • Overexpression vs. knockdown methodologies

    • In vitro vs. in vivo settings

  • Activation measurement challenges:

    • The transient nature of ARF6 activation (peak at ~2 min) means timing is critical

    • Different pull-down assays may yield varying results

    • Post-lysis activation changes can confound results

When facing contradictory data, researchers should carefully consider experimental conditions, validate findings using multiple approaches, and explicitly acknowledge the specific cellular and molecular context in which the experiments were performed .

What are the optimal methods for measuring ARF6 activation in human samples?

Measuring ARF6 activation requires detecting the GTP-bound form of the protein. Several methodological approaches are available:

1. GGA3-PBD pull-down assay:

  • Uses the protein-binding domain (PBD) of GGA3, which specifically binds ARF6-GTP

  • Allows quantification of active ARF6 by immunoblotting

  • Timing is critical; samples must be processed rapidly due to transient activation

2. Immunofluorescence with conformation-specific antibodies:

  • Antibodies that preferentially recognize the GTP-bound conformation

  • Allows visualization of active ARF6 localization within cells

  • Requires validation of antibody specificity

3. FRET-based biosensors:

  • Allows real-time monitoring of ARF6 activation in living cells

  • Enables spatial and temporal resolution of activation dynamics

  • Requires specialized equipment and careful controls

Key methodological considerations:

  • Sample timing: As seen in HASMCs, ARF6 activation peaks around 2 minutes after stimulation

  • Lysis conditions: Must prevent post-lysis nucleotide exchange

  • Controls: Include positive controls (GTPγS-loaded samples) and negative controls (GDP-loaded samples)

  • Normalization: Always normalize active ARF6 to total ARF6 levels

For analyzing ARF6 activation in patient-derived samples, the GGA3-PBD pull-down approach is most commonly used due to its reliability and compatibility with frozen samples .

How can researchers effectively manipulate ARF6 activity for functional studies?

Multiple approaches exist for manipulating ARF6 activity in experimental systems:

Genetic approaches:

  • siRNA/shRNA knockdown: Reduces total ARF6 protein levels

  • CRISPR/Cas9 knockout: Eliminates ARF6 expression

  • Overexpression systems:

    • Wild-type ARF6: Increases total ARF6 levels

    • Constitutively active mutants (Q67L): Locked in GTP-bound state

    • Dominant negative mutants (T27N): Locked in GDP-bound state

Pharmacological approaches:

  • Direct ARF6 inhibitors:

    • NAV-2729: Directly inhibits ARF6 activation

    • Shows efficacy in animal models without observable toxicity

  • GEF inhibitors:

    • SecinH3: Inhibits cytohesins (including ARNO)

    • Has demonstrated therapeutic potential in disease models

Experimental design considerations:

  • Timing: Acute vs. chronic manipulation may yield different results

  • Specificity: Validate effects are due to ARF6 and not other ARFs

  • Compensation: Check for compensatory upregulation of other ARFs

  • Cell viability: Monitor for non-specific cytotoxicity

  • Pathway analysis: Determine which downstream pathways are affected

The evidence suggests that reducing ARF6 activity through pharmacological inhibition does not produce detrimental effects in adult organisms and may have therapeutic potential for diseases characterized by aberrant ARF6 activation .

What experimental approaches can delineate ARF6-specific signaling pathways in human cells?

Distinguishing ARF6-specific pathways from those mediated by other ARF family members requires sophisticated experimental approaches:

1. Chimeric protein analysis:

  • Creation of ARF1-ARF6 chimeras to identify functional domains

  • Studies have shown regions beyond switch regions contribute to specificity

  • Allows mapping of domains responsible for specific effector interactions

2. Proteomic approaches:

  • Proximity labeling (BioID, APEX) to identify ARF6 interactome

  • Comparative proteomics of ARF6-GTP vs. ARF6-GDP associated proteins

  • Phosphoproteomic analysis to identify downstream signaling events

3. Pathway delineation experiments:

  • Specific inhibitors of downstream pathways (e.g., MAPK, PI3K)

  • Sequential activation analysis (e.g., ARF6 → PAK → MMP14)

  • Epistasis experiments with multiple knockdowns/inhibitors

4. Cell type-specific analysis:

  • Comparison of ARF6 signaling across cell types

  • Identification of cell-specific GEFs/GAPs and effectors

  • Context-dependent signaling networks

In human aortic smooth muscle cells, researchers have successfully delineated that PDGF-BB stimulation of ARF6 regulates both MAPK and PI3K pathways, while Ang II stimulation activates only MAPK pathways . Both stimuli promote activation of PAK, leading to MMP14 membrane expression and activation, which regulates extracellular matrix degradation .

What are the emerging targets in ARF6 research that might lead to novel therapeutic approaches?

Based on current knowledge of ARF6 biology, several promising research directions are emerging:

1. ARF6 in immune regulation:

  • Role in immune cell migration and adhesion

  • Potential target for inflammatory disorders

  • Involvement in antigen presentation and immune synapse formation

2. Tissue-specific ARF6 functions:

  • Differential roles in various tissues and cell types

  • Potential for tissue-targeted therapies

  • Specialized functions in polarized cells

3. ARF6 in metabolic signaling:

  • Connections to insulin signaling and glucose metabolism

  • Role in vesicular transport of metabolic regulators

  • Potential implications for metabolic disorders

4. Novel ARF6 inhibition strategies:

  • Development of isoform-specific inhibitors

  • Targeted disruption of specific ARF6-effector interactions

  • Combination approaches targeting multiple points in ARF6 signaling

5. ARF6 in disease-specific contexts:

  • Cancer subtype-specific functions

  • Vascular disease-specific mechanisms

  • Neurological disorder implications

The therapeutic potential of ARF6 inhibition is supported by animal studies showing that systemic administration of small molecules targeting ARF6 or its GEFs effectively ameliorated disease phenotypes without toxicity or adverse effects . This suggests ARF6 inhibition may be a viable therapeutic strategy for multiple pathological conditions.

How can researchers address the challenges of studying ARF6 in primary human tissues?

Studying ARF6 in primary human tissues presents several challenges but is essential for translational research. Recommended approaches include:

1. Tissue preservation methods:

  • Rapid fixation to preserve ARF6 activation state

  • Optimized protocols for ARF6 detection in tissue sections

  • Validation using multiple antibodies and controls

2. Ex vivo tissue models:

  • Short-term culture of fresh human tissue explants

  • Treatment with ARF6 activators/inhibitors

  • Analysis of downstream effects on tissue architecture

3. Patient-derived primary cell cultures:

  • Establishment of primary cells from patient samples

  • Comparison of ARF6 signaling in disease vs. healthy cells

  • Correlation with clinical parameters

4. Organoid and 3D culture systems:

  • Development of human organoids to study ARF6 in tissue context

  • Analysis of ARF6's role in 3D epithelial structures

  • Recapitulation of tissue-specific ARF6 functions

5. Single-cell approaches:

  • Single-cell RNA-seq to examine cell-specific ARF6 pathway expression

  • Single-cell proteomics for ARF6 activation analysis

  • Spatial transcriptomics to map ARF6 signaling in tissue architecture

The importance of validating findings in human cells is highlighted by studies showing that while rat VSMC models have been helpful, specific pathways activated by ARF6 in human vascular smooth muscle cells needed direct examination to confirm relevance to human pathology .

Product Science Overview

Structure and Localization

ARF6 is localized to the plasma membrane and is involved in the regulation of vesicular trafficking and membrane lipid remodeling . The protein’s structure allows it to interact with various other proteins and cellular components, facilitating its role in these processes.

Functions

ARF6 has several key functions within the cell:

  1. Vesicular Trafficking: ARF6 regulates the trafficking of biological membranes and transmembrane protein cargo . This includes the endocytosis of plasma membrane proteins and, to a lesser extent, their recycling back to the plasma membrane .
  2. Actin Cytoskeleton Remodeling: ARF6 is involved in signaling pathways that lead to the remodeling of the actin cytoskeleton . This is crucial for various cellular activities, including cell shape changes, motility, and division.
  3. Immune Signaling: ARF6 plays a pivotal role in Toll-like receptor 9 (TLR9)-mediated immune signaling . It regulates the cellular uptake of CpG oligodeoxynucleotides (CpG ODN) into endosomes, which is a critical step in eliciting innate immune responses .
Mechanisms of Action

ARF6 functions by cycling between an active GTP-bound state and an inactive GDP-bound state. This cycling is regulated by guanine nucleotide exchange factors (GEFs) and GTPase-activating proteins (GAPs). In its active state, ARF6 can interact with various effector proteins to carry out its functions .

Research and Applications

Research on ARF6 has revealed its importance in various physiological and pathological processes. For instance, inhibition of ARF6 by dominant mutants and siRNA has been shown to impair CpG ODN/TLR9-mediated responses, highlighting its role in immune signaling . Additionally, ARF6 has been implicated in cancer progression, as it can influence cell migration and invasion through its effects on the actin cytoskeleton .

Recombinant human ARF6 is used in research to study these processes and to develop potential therapeutic strategies targeting ARF6-related pathways.

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.