ARF4 Human

ADP-Ribosylation Factor 4 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Organization

The ARF4 gene spans approximately 12 kb on chromosome 3 (reverse strand: GRCh38:CM000665.2) and contains six exons and five introns . It encodes nine transcript variants, with the canonical isoform (ENST00000303436.11) producing a 180-amino acid (aa) protein (UniProt: P18085) . Recombinant ARF4 is typically expressed in E. coli with a His-tag, yielding a 23-kDa protein .

Membrane Trafficking and Organelle Dynamics

ARF4 regulates vesicular transport across compartments:

  • Golgi Apparatus: Localizes to the cis-Golgi and ER-Golgi intermediate compartment (ERGIC), working redundantly with ARF1 in early secretory pathways .

  • Ciliary Trafficking: Part of a complex with Rab11 and ASAP1, directing post-Golgi vesicles to the mother centriole during ciliogenesis .

  • Retrograde Transport: Mediates endosome-to-trans-Golgi network (TGN) trafficking, influencing receptor recycling and chemoresistance .

Specialized Roles

  • N-Cadherin Trafficking: Regulates radial migration in cerebral cortex development by controlling N-cadherin surface expression .

  • Exocytosis: Binds CAPS (calcium-dependent activator protein for secretion) to facilitate dense-core vesicle release .

Cerebral Cortex Development

Knockdown of ARF4 in mice disrupted radial migration, causing Golgi disorientation and cytoplasmic N-cadherin accumulation. Supplementation of N-cadherin partially restored migration defects, highlighting ARF4’s role in neuronal adhesion and guidance .

Key Observations

ObservationEffect
ARF4 KnockdownStalled neurons in intermediate zone (IZ), reduced migration speed
N-Cadherin RescuePartial recovery of migration, confirming trafficking dependency

Glioblastoma (GBM) Chemoresistance

ARF4 drives resistance to temozolomide (TMZ) by enhancing retrograde trafficking of EGFR to the nucleus, upregulating DNA repair proteins. CRISPR screens identified ARF4 as a therapeutic target for GBM .

Recombinant ARF4 Proteins

Recombinant ARF4 is widely used in biochemical assays:

SupplierProduct Details
Abcam>95% purity, 1–180 aa, E. coli-derived, suitable for SDS-PAGE/MS
ProSpec Bio23-kDa, His-tagged, 0.5 mg/mL in Tris-HCl buffer
Molecular Depot100 µg vials, used in studies on pancreatic degeneration and yeast rescue

Product Specs

Introduction
ARF4, a member of the ARF gene family, is a small guanine nucleotide-binding protein. It plays a crucial role in vesicular trafficking and acts as an activator of phospholipase D. ARF proteins, encompassing five ARF proteins and eleven ARF-like proteins, belong to the RAS superfamily and are categorized into class I, II, and III. ARF4 falls under class II. Each class exhibits a shared gene organization. The ARF4 gene, spanning approximately 12kb, comprises 6 exons and 5 introns. Notably, ARF4 is recognized as the most divergent member among human ARFs.
Description
Recombinant human ARF4, expressed in E. coli, is a single, non-glycosylated polypeptide chain with a molecular weight of 23kDa. It comprises 204 amino acids, including a 24 amino acid His tag at the N-terminus (1-180 a.a.). Purification of ARF4 is achieved using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
ARF4 is supplied as a 0.5 mg/ml solution in 20mM Tris-HCl buffer (pH 8.0), containing 0.2M NaCl, 5mM DTT, and 20% glycerol.
Stability
For short-term storage (up to 2-4 weeks), keep at 4°C. For longer storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95.0% as determined by SDS-PAGE analysis.
Synonyms
ADP-ribosylation factor 4, ARF4, ARF2.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMGLTIS SLFSRLFGKK QMRILMVGLD AAGKTTILYK LKLGEIVTTI PTIGFNVETV EYKNICFTVW DVGGQDRIRP LWKHYFQNTQ GLIFVVDSND RERIQEVADE LQKMLLVDEL RDAVLLLFAN KQDLPNAMAI SEMTDKLGLQ SLRNRTWYVQ ATCATQGTGL YEGLDWLSNE LSKR.

Q&A

What is ARF4 and how does it function in human cells?

ARF4 is a member of the ADP-ribosylation factor (Arf) family of small GTPases, which belongs to the larger Ras superfamily of small guanine nucleotide-binding proteins. ARF4 primarily functions in regulating membrane trafficking pathways within the cell. Like other GTPases, ARF4 cycles between active (GTP-bound) and inactive (GDP-bound) states, which are regulated by guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs) . In its active state, ARF4 can interact with various effector proteins to mediate vesicle formation, sorting of cargo proteins, and vesicle trafficking. ARF4 is primarily associated with the Golgi apparatus and the trans-Golgi network (TGN), where it plays crucial roles in organizing membrane trafficking in both the endocytic and secretory pathways . The functional specificity of ARF4 is determined by its interactions with specific GEFs, GAPs, and effector proteins in different cellular contexts.

How is ARF4 expression regulated in human cells?

ARF4 expression is regulated at both transcriptional and translational levels. One important regulatory mechanism involves the transcription factor small leucine zipper protein (sLZIP) . Research has demonstrated that sLZIP binds directly to the CRE motif located in the -43 to -35 region of the ARF4 promoter . This binding interaction is crucial for controlling ARF4 expression, particularly in response to certain stimuli. For example, treatment with phorbol 12-myristate 13-acetate (PMA) has been shown to increase ARF4 expression through this regulatory pathway . The PMA-induced expression occurs at both transcriptional and translational levels, suggesting multiple layers of regulation. Understanding these regulatory mechanisms is particularly important in cancer research, as altered ARF4 expression has been implicated in cancer cell migration and potentially in metastatic processes. These regulatory pathways represent potential targets for therapeutic intervention in diseases where ARF4 dysregulation plays a role.

What are the structural differences between human ARF4 and its homologs in other species?

Interestingly, in mice and rats, two acidic amino acid residues in this region are replaced by hydrophobic amino acids, creating a hydrophobic stretch instead of a negatively charged one . This type of substitution typically disrupts protein-protein interactions and likely affects the ability of rodent ARF4 to interact with rhodopsin or does so with significantly reduced affinity. This structural difference may explain some of the discrepancies observed in ARF4 function between different model organisms and highlights the importance of considering species-specific variations when extrapolating findings from animal models to human biology.

How does ARF4 contribute to ciliary membrane targeting in human cells?

ARF4 plays a specialized role in the ciliary membrane-targeting complex that directs sensory receptors to primary cilia. In its active GTP-bound state, ARF4 directly binds to the VxPx ciliary-targeting signal (CTS) found in ciliary-destined cargo proteins such as rhodopsin . This interaction occurs at the trans-Golgi network and represents the initial step in the assembly of the ciliary-targeting complex. The process begins with the activation of ARF4 by the guanine nucleotide exchange factor GBF1, creating a functional ternary complex between GBF1, activated ARF4, and the ciliary cargo protein .

This complex formation triggers a positive feedback loop that drives further ARF4 activation through an autocatalytic amplification mechanism. Once activated, ARF4 regulates the assembly of a targeting nexus containing the Arf GAP ASAP1 and the Rab11a–FIP3–Rabin8 dual effector complex . This complex subsequently controls the assembly of the highly conserved Rab11a–Rabin8–Rab8 ciliary-targeting module, which is essential for the final stages of ciliary membrane protein trafficking. Disruption of this pathway, particularly through mutations affecting ARF4's ability to bind cargo or undergo proper GTP hydrolysis, can lead to ciliopathies and related disorders. The process demonstrates how ARF4 functions as a critical regulatory node in ensuring the proper spatiotemporal delivery of proteins to the primary cilium.

What methodologies are most effective for studying ARF4 activation dynamics in live cells?

To effectively study ARF4 activation dynamics in live cells, researchers should employ a multi-modal approach combining advanced imaging techniques with molecular tools that can detect the GTP-bound state of ARF4. A particularly effective methodology involves the use of fluorescence resonance energy transfer (FRET)-based biosensors that can detect the conformational changes associated with ARF4 activation. These biosensors typically consist of ARF4 sandwiched between appropriate fluorophores that undergo changes in FRET efficiency upon GTP binding and activation.

For studying the spatiotemporal dynamics of ARF4 activation, spinning disk confocal microscopy or total internal reflection fluorescence (TIRF) microscopy provides the necessary temporal and spatial resolution. These approaches can be complemented with the use of photoactivatable or photoconvertible fluorescent protein tags that allow for pulse-chase experiments to track the movement and activation state of ARF4 pools within specific cellular compartments.

To manipulate ARF4 activity, researchers can use optogenetic tools that allow for light-induced activation or inactivation of ARF4 in specific subcellular regions. Additionally, small molecule inhibitors of GBF1, such as Golgicide A, which has been shown to disrupt the functional complex between GBF1, activated ARF4, and rhodopsin, provide pharmacological means to probe the consequences of disrupting ARF4 activation .

For biochemical validation of activation states, pull-down assays using the binding domains of ARF4 effectors that specifically recognize the GTP-bound form can be employed. Together, these methodologies provide a comprehensive toolkit for dissecting the complex dynamics of ARF4 activation in various cellular contexts.

What role does ARF4 play in neuronal migration during cerebral cortical development?

ARF4 plays a crucial role in regulating radial migration during cerebral cortical development through its involvement in N-cadherin trafficking. During the formation of cortical layers, N-cadherin facilitates radial migration by enabling cell-to-cell adhesion between migrating neurons and radial glial fibers or Cajal-Retzius cells . Research using knock-down experiments in mouse embryos has revealed that reducing ARF4 expression results in the stalling of transfected neurons in the upper intermediate zone (IZ) with disorientation of the Golgi apparatus . Additionally, neurons with reduced ARF4 expression showed decreased migration speed in both the intermediate zone and cortical plate (CP).

Mechanistically, ARF4 knock-down leads to cytoplasmic accumulation of N-cadherin within migrating neurons, along with disturbed organelle morphology and distribution . This suggests that ARF4 is essential for the proper trafficking of N-cadherin to cell surfaces where it can mediate adhesive interactions necessary for migration. Support for this mechanistic model comes from experiments showing that supplementation of exogenous N-cadherin partially rescues the migration defect caused by ARF4 knock-down . These findings establish ARF4 as a key regulator of neuronal positioning during brain development through its control of adhesion molecule trafficking, and suggest that disruptions in ARF4 function could potentially contribute to neurodevelopmental disorders characterized by abnormal cortical layering or neuronal positioning.

How is ARF4 implicated in cancer progression, particularly in breast cancer?

ARF4 has been implicated in cancer progression through its roles in cellular signaling and migration. In breast cancer specifically, ARF4 contributes to cell migration processes that are crucial for invasion and metastasis . Research has demonstrated that ARF4 interacts with the epidermal growth factor receptor (EGFR) and mediates EGF-dependent signaling pathways, which are frequently dysregulated in cancer . Additionally, ARF4 has demonstrated anti-apoptotic functions in human glioblastoma-derived U373MG cells, suggesting a potential role in promoting cancer cell survival .

The molecular mechanisms linking ARF4 to breast cancer migration involve transcriptional regulation through the AP-1 (Activator Protein-1) promoter. Studies have shown that phorbol 12-myristate 13-acetate (PMA) stimulates ARF4 expression, which subsequently increases AP-1 promoter activity . This cascade leads to the induction of breast cancer cell migration. The regulatory pathway involves the small leucine zipper protein (sLZIP), which binds directly to the CRE motif in the ARF4 promoter and regulates PMA-induced ARF4 expression .

Given these findings, both sLZIP and ARF4 have been identified as potential therapeutic target molecules for treating breast cancer invasion and metastasis . Developing inhibitors that target the sLZIP-ARF4-AP-1 axis could potentially reduce the migratory capacity of breast cancer cells and limit metastatic spread. Further research is needed to determine whether ARF4 expression levels correlate with clinical outcomes in breast cancer patients and to develop effective therapeutic approaches targeting this pathway.

What are the optimal experimental models for studying ARF4 function in human contexts?

When selecting experimental models for studying ARF4 function in human contexts, researchers should consider several factors including evolutionary conservation, tissue-specific expression patterns, and the specific biological process under investigation. For studying basic ARF4 functions, human cell lines such as HEK293, HeLa, or tissue-specific cell lines (e.g., neuronal or breast cancer lines) provide accessible systems that faithfully represent human ARF4 biology. These cell lines can be manipulated using CRISPR/Cas9 genome editing to generate ARF4 knockouts or to introduce specific mutations that affect ARF4 function.

For more complex developmental processes, researchers should be cautious about using rodent models due to the significant differences in the ARF4 α3 helix region between humans and rodents . The substitution of acidic amino acids with hydrophobic ones in mouse and rat ARF4 likely affects protein-protein interactions, particularly with rhodopsin . In these cases, the frog model system may be more appropriate as frog ARF4 shares 95% sequence identity with human ARF4 . The frog model has been particularly valuable for studying high-volume membrane trafficking processes, such as those occurring in photoreceptors.

For studying ciliary trafficking, specialized cell culture models that readily form primary cilia, such as hTERT-RPE1 cells or IMCD3 cells, are recommended. These can be combined with fluorescently tagged ciliary cargo proteins to visualize trafficking processes. Organoid systems, particularly cerebral organoids for studying neuronal migration or mammary organoids for breast cancer research, offer more physiologically relevant three-dimensional contexts. Finally, patient-derived cells with specific ciliopathies or neurodevelopmental disorders may provide valuable insights into pathological ARF4 function. The choice of model should ultimately be guided by the specific research question and the need to accurately represent human ARF4 biology.

What techniques are available for measuring ARF4-mediated protein trafficking in neuronal cells?

Studying ARF4-mediated protein trafficking in neuronal cells requires specialized techniques that can capture the spatial and temporal dynamics of cargo movement within these complex, polarized cells. Live-cell imaging approaches using spinning disk confocal microscopy are particularly valuable, as they allow for high temporal resolution imaging while minimizing phototoxicity. To visualize ARF4-dependent trafficking, researchers can use fluorescently tagged cargo proteins such as N-cadherin, which has been shown to rely on ARF4 for proper trafficking during neuronal migration .

Pulse-chase experiments using photoconvertible fluorescent proteins fused to ARF4 or its cargo can reveal the dynamics of protein movement from the trans-Golgi network to the cell surface or other destinations. For instance, a pulse of photoconversion at the Golgi followed by time-lapse imaging can track the movement of newly synthesized proteins through ARF4-dependent trafficking routes. Super-resolution microscopy techniques such as STORM or PALM can provide nanoscale resolution of trafficking intermediates and colocalization with specific cellular compartments.

For biochemical approaches, subcellular fractionation combined with western blotting can assess the distribution of ARF4 and its cargo proteins across different cellular compartments. Cell surface biotinylation assays can specifically measure the efficiency of cargo delivery to the plasma membrane. Additionally, proximity ligation assays (PLA) can detect interactions between ARF4 and its partner proteins such as GBF1 or ASAP1 within intact neurons.

To specifically study the role of ARF4 in N-cadherin trafficking during neuronal migration, in utero electroporation techniques can be used to knock down ARF4 in developing neurons, followed by time-lapse imaging of migration dynamics and N-cadherin localization . These combined approaches provide a comprehensive toolkit for dissecting the complex trafficking pathways mediated by ARF4 in neuronal contexts.

How can researchers effectively distinguish between ARF4's direct functions and indirect effects mediated through interaction partners?

Distinguishing between direct ARF4 functions and indirect effects mediated through interaction partners requires a strategic combination of biochemical, genetic, and imaging approaches. One fundamental approach is to perform structure-function analyses using ARF4 mutants that specifically disrupt interactions with individual partners. For example, researchers can generate mutations in the α3 helix region known to be crucial for binding to rhodopsin and other cargo proteins . By comparing the phenotypes of these specific interaction-deficient mutants with complete ARF4 knockouts, researchers can differentiate direct from indirect effects.

Proximity-dependent labeling techniques such as BioID or APEX2 provide powerful tools for mapping the immediate protein interaction network of ARF4 in living cells. These approaches involve fusing ARF4 to an enzyme that biotinylates nearby proteins, allowing for the identification of proximal interactors in specific subcellular locations or under different conditions. Combined with quantitative proteomics, these methods can reveal the dynamic interactome of ARF4.

Temporal control of ARF4 activity using rapid inducible systems (such as auxin-inducible degrons or chemical dimerization approaches) allows researchers to distinguish immediate effects of ARF4 loss (likely direct) from those that develop over longer timeframes (potentially indirect). Similarly, acute pharmacological inhibition of specific ARF4 regulators, such as using Golgicide A to inhibit GBF1-mediated ARF4 activation , can help separate direct from indirect pathways.

For specific trafficking processes, researchers can employ in vitro reconstitution assays using purified components to test whether ARF4 alone is sufficient to drive particular membrane remodeling or vesicle formation events, or whether additional factors are required. Finally, computational approaches such as network analysis can help predict whether observed phenotypes are likely to be direct consequences of ARF4 perturbation or secondary effects propagated through interaction networks. By integrating these diverse approaches, researchers can build a comprehensive understanding of ARF4's direct functional roles versus those mediated by its extensive interaction network.

What is the potential for targeting ARF4 pathways in cancer therapies?

ARF4 represents a promising therapeutic target for cancer intervention, particularly in breast cancer where its role in promoting cell migration has been established . The potential for targeting ARF4 pathways stems from several key findings. First, ARF4 has demonstrated anti-apoptotic functions in human glioblastoma-derived cells, suggesting that its inhibition could potentially sensitize cancer cells to apoptotic stimuli . Second, ARF4 mediates EGF-dependent signaling pathways through its interaction with EGFR, a well-established oncogenic driver in multiple cancer types . Third, the regulatory pathway involving sLZIP and ARF4 has been directly linked to breast cancer cell migration, a critical process in metastasis .

For therapeutic development, several approaches could be pursued. Small molecule inhibitors targeting the GTPase activity of ARF4 could prevent its activation and subsequent downstream signaling. Alternatively, disrupting the interaction between ARF4 and specific effector proteins could selectively inhibit pathways relevant to cancer progression while preserving other functions. Another promising approach involves targeting the transcriptional regulation of ARF4 by developing compounds that inhibit the binding of sLZIP to the CRE motif in the ARF4 promoter .

RNA interference or antisense oligonucleotides specifically targeting ARF4 could also be developed as therapeutic modalities. Additionally, since ARF4 functions within a complex network of GTPases and trafficking proteins, combination therapies targeting multiple nodes in this network might provide synergistic anti-cancer effects while reducing the likelihood of resistance development. As research advances, biomarkers based on ARF4 expression or activation status could potentially be developed to identify patients most likely to benefit from ARF4-targeted therapies, enabling a precision medicine approach to cancer treatment.

What emerging technologies might advance our understanding of ARF4 function in the next decade?

The next decade will likely see significant advances in our understanding of ARF4 function through the application of emerging technologies across multiple disciplines. In structural biology, cryo-electron microscopy (cryo-EM) advancements will likely enable visualization of ARF4 in complex with its interaction partners at near-atomic resolution, providing unprecedented insights into the conformational dynamics of these complexes during various stages of activation and effector binding. AlphaFold and similar AI-powered structure prediction tools will further complement these experimental approaches by generating testable hypotheses about ARF4 interactions.

In cellular imaging, developments in lattice light-sheet microscopy combined with adaptive optics will allow for long-term, high-resolution imaging of ARF4-mediated trafficking events in living cells with minimal phototoxicity. Super-resolution microscopy techniques will continue to evolve, potentially allowing for nanometer-scale visualization of ARF4 and its cargo molecules in native cellular environments. These approaches could be combined with emerging methods for detecting transient protein-protein interactions in living cells, such as split fluorescent proteins or advanced FRET sensors.

The application of CRISPR-based technologies will extend beyond simple gene knockout to include base editing, prime editing, and epigenetic modulation of ARF4 and its regulatory elements. These precise genome editing tools will enable the creation of sophisticated cellular and animal models carrying specific ARF4 variants identified in human populations or diseases. Single-cell multi-omics approaches will provide comprehensive views of how ARF4 expression and function vary across different cell types and states, potentially revealing cell type-specific roles that have been overlooked in bulk studies.

Finally, advances in organoid technology and microphysiological systems ("organs-on-chips") will create more physiologically relevant contexts for studying ARF4 function in complex tissue environments, including the nervous system and cancer microenvironments. These technologies, combined with patient-derived cells, will bridge the gap between basic research and clinical applications, accelerating the translation of ARF4 research into therapeutic strategies.

Product Science Overview

Introduction

ADP-Ribosylation Factor 4 (ARF4) is a member of the ADP-ribosylation factor family of GTP-binding proteins. These proteins are involved in various cellular processes, including vesicle trafficking, cytoskeletal organization, and lipid metabolism. ARF4, in particular, plays a crucial role in the regulation of intracellular trafficking and membrane dynamics. The human recombinant form of ARF4 is produced using recombinant DNA technology, which allows for the expression of the protein in a host organism, typically E. coli, to obtain large quantities for research and therapeutic purposes.

Preparation Methods

The preparation of human recombinant ARF4 involves several key steps:

  1. Gene Cloning: The gene encoding ARF4 is first isolated and cloned into an expression vector. This vector contains the necessary regulatory elements for the transcription and translation of the ARF4 gene in the host organism.

  2. Transformation: The expression vector is introduced into a suitable host organism, such as E. coli, through a process called transformation. The host cells take up the vector and incorporate it into their own genetic material.

  3. Expression: The host cells are cultured under conditions that promote the expression of the ARF4 gene. This typically involves the use of specific growth media and induction agents that activate the transcription of the gene.

  4. Purification: Once the ARF4 protein is expressed, it is extracted from the host cells and purified using various chromatographic techniques. These may include affinity chromatography, ion exchange chromatography, and size exclusion chromatography, among others. The goal is to obtain a highly pure and biologically active form of the protein.

Analysis of Chemical Reactions

ARF4 is involved in several biochemical reactions and pathways within the cell:

  1. GTP Binding and Hydrolysis: Like other GTP-binding proteins, ARF4 cycles between an active GTP-bound state and an inactive GDP-bound state. The binding and hydrolysis of GTP are critical for its function in regulating vesicle trafficking and membrane dynamics.

  2. Vesicle Trafficking: ARF4 plays a key role in the formation and transport of vesicles within the cell. It is involved in the recruitment of coat proteins to the membrane, which are necessary for the budding and scission of vesicles. This process is essential for the transport of proteins and lipids between different cellular compartments.

  3. Cytoskeletal Organization: ARF4 also interacts with components of the cytoskeleton, such as actin filaments, to regulate cell shape and motility. This interaction is important for processes like cell migration and division.

  4. Lipid Metabolism: ARF4 is implicated in the regulation of lipid metabolism, particularly in the transport and distribution of cholesterol within the cell. It helps maintain the balance of lipids in various cellular membranes, which is crucial for proper cell function.

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.