ARL15 Human

ADP-Ribosylation Factor-Like 15 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Overview of ARL15 Human

ARL15 Human (ADP-ribosylation factor-like 15) is a small GTP-binding protein encoded by the ARL15 gene (chromosome 5) and belongs to the RAS superfamily. It is structurally related to ARF (ADP-ribosylation factor) proteins, though its precise biochemical functions remain partially characterized. ARL15 is implicated in metabolic regulation, adipocyte differentiation, and signaling pathways, with emerging links to diseases such as diabetes, rheumatoid arthritis, and cardiovascular disorders .

Protein Structure

ARL15 is a 204-amino acid protein with conserved guanine nucleotide-binding motifs (GTPase domains), typical of small GTPases . Structural predictions suggest a globular conformation with dynamic regions for nucleotide binding and effector interactions .

Subcellular Localization

ARL15 localizes primarily to the Golgi apparatus, with secondary presence in plasma membrane and vesicular compartments . This distribution aligns with its proposed roles in intracellular trafficking and secretion .

Key Functions

FunctionMechanism/RoleSource
Adipocyte DifferentiationRegulates preadipocyte maturation via Golgi-associated pathways
Adiponectin SecretionFacilitates adiponectin trafficking from Golgi to cell surface
TGFβ/BMP Signaling ModulationBinds Smad4 to promote Smad-complex assembly and signaling
Lipid MetabolismInfluences lipid trafficking and storage in adipocytes

Tissue-Specific Expression

TissueExpression LevelKey ObservationsSource
Skeletal MuscleHighMost abundant expression
Adipose TissueModerateVisceral and subcutaneous depots
LiverLowDetectable but not prominent
BrainTraceLimited expression in select regions

Cellular Distribution

  • Primary: Golgi apparatus .

  • Secondary: Plasma membrane, endolysosomes .

  • Co-localization: Observed with Smad4 at endolysosomes during TGFβ signaling .

Metabolic Disorders

DiseaseMechanismGenetic EvidenceSource
Type 2 DiabetesImpaired adiponectin secretion and insulin resistanceGWAS-linked SNPs at ARL15 locus
LipodystrophyHaploinsufficiency variants linked to partial lipodystrophyRare nonsense/splice-site mutations
CardiovascularAltered HDL cholesterol and triglyceride metabolismGWAS associations with CAD risk

Autoimmune and Inflammatory Disorders

DiseaseRole of ARL15Source
Rheumatoid ArthritisPotential therapeutic target; modulates TGFβ signaling

Rare Genetic Variants

  • Nonsense Mutation: Identified in a patient with femorogluteal lipodystrophy and congenital adrenal hyperplasia .

  • Splice-Site Mutation: Linked to partial lipodystrophy and childhood yolk sac tumor .

Adipogenesis and Secretion

  • Knockdown in 3T3-L1 Cells:

    • Preadipocytes: Impaired adipogenesis .

    • Adipocytes: Reduced adiponectin secretion (no effect on adipsin or Glut4) .

  • Mechanism: Disrupted Golgi-dependent trafficking of adiponectin .

TGFβ/BMP Signaling

  • Arl15-Smad4 Interaction:

    • Binding: Direct interaction with Smad4’s MH2 domain .

    • Regulation: Activates Smad4 to promote Smad-complex assembly; Smad4 reciprocally acts as a GAP for Arl15 .

  • Functional Impact:

    • TGFβ Activity: Essential for transcriptional output (e.g., PAI-1) .

    • BMP Pathway: Modulates BMP-dependent effects (e.g., Smad1/5/8 activation) .

Targeting ARL15 in Disease

DiseaseProposed StrategyRationaleSource
DiabetesEnhance adiponectin secretionImprove insulin sensitivity
Rheumatoid ArthritisInhibit TGFβ signalingReduce pro-inflammatory responses
LipodystrophyGene therapy to restore ARL15 functionCorrect adipose tissue remodeling

Product Specs

Introduction
As a member of the ARF family, ARL15 plays a crucial role in eukaryotic vesicular trafficking pathways. It is essential for activating phospholipase D. Variations in ARL15 can affect the levels of Acrp30, an adipocyte-derived protein strongly linked to genetics and inversely associated with the risk of developing type 2 diabetes mellitus and coronary heart disease.
Description
Recombinant human ARL15, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 224 amino acids (1-204). It has a molecular weight of 25.0 kDa. The protein is fused to a 20 amino acid His-tag at the N-terminus and purified using proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, colorless solution.
Formulation
The ARL15 solution is provided at a concentration of 1mg/ml and contains 20mM Tris-HCl buffer (pH 8.0), 1mM DTT, 0.1M NaCl, and 10% glycerol.
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. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the product is greater than 85% as determined by SDS-PAGE analysis.
Synonyms
ADP-ribosylation factor-like 15, ADP-ribosylation factor related protein 2, ARFRP2, ARF-related protein 2, FLJ20051.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MSDLRITEAF LYMDYLCFRA LCCKGPPPAR PEYDLVCIGL TGSGKTSLLS KLCSESPDNV VSTTGFSIKA VPFQNAILNV KELGGADNIR KYWSRYYQGS QGVIFVLDSA SSEDDLEAAR NELHSALQHP QLCTLPFLIL ANHQDKPAAR SVQEIKKYFE LEPLARGKRW ILQPCSLDDM DALKDSFSQL INLLEEKDHE AVRM 

Q&A

What is ARL15 and what protein family does it belong to?

ARL15 is a member of the ADP-ribosylation factor (ARF) family within the RAS superfamily of small GTPases. The human ARL15 protein consists of 204 amino acids and functions as a GTP-binding protein . Like other members of the ARF family, ARL15 cycles between GTP-bound (active) and GDP-bound (inactive) states, which enables it to function as a molecular switch in various cellular processes . ARL15 shares structural features with other small GTPases, including conserved GTP-binding motifs, but has unique characteristics that distinguish it within the ARF family.

What is the subcellular localization of ARL15?

ARL15 exhibits distinct subcellular localization patterns that are critical to its function. Studies have demonstrated that endogenous ARL15 is palmitoylated and primarily localizes to the Golgi apparatus in mouse liver cells . Interestingly, during adipocyte differentiation, ARL15 undergoes translocation within the Golgi complex—it predominantly co-localizes with markers of the cis-Golgi face in preadipocytes and then redistributes to other Golgi compartments following differentiation induction . Additionally, active ARL15 has been observed to colocalize with Smad4 at the endolysosome . The palmitoylation status of ARL15 appears critical for its correct localization, as palmitoylation-deficient ARL15 redistributes to the cytoplasm in adipocytes .

How does ARL15 function in TGFβ signaling pathways?

ARL15 serves as a positive regulator of transforming growth factor β (TGFβ) family signaling through a novel mechanism involving the Smad protein complex . The process occurs as follows:

  • Active ARL15 (GTP-bound) specifically binds to the MH2 domain of Smad4 and colocalizes with it at the endolysosome.

  • This binding disrupts the autoinhibitory conformation of Smad4 by relieving the intramolecular interaction between its MH1 and MH2 domains.

  • The activated Smad4 becomes capable of interacting with phosphorylated receptor-regulated Smads (R-Smads), facilitating the assembly of the Smad complex.

  • Notably, Smad4 appears to function not only as an effector but also as a GTPase activating protein (GAP) for ARL15.

  • Once the Smad complex is assembled, the GAP activity of Smad4 toward ARL15 increases, causing dissociation of ARL15 before the nuclear translocation of the Smad complex .

This regulatory mechanism demonstrates ARL15's significance in modulating a fundamental signaling pathway involved in numerous developmental and disease processes.

What role does ARL15 play in adipocyte biology and metabolism?

ARL15 appears to be a critical regulator of adipocyte differentiation and metabolic function through several mechanisms:

  • Adipocyte Differentiation: ARL15 undergoes translocation within Golgi compartments during adipocyte differentiation, suggesting a dynamic role in this process .

  • Gene Expression Regulation: Palmitoylation-deficient ARL15 expression in adipocytes results in reduced expression of adipogenesis-related genes, indicating that properly localized ARL15 is required for normal adipogenic gene expression .

  • Insulin Signaling: Loss of ARL15 reduces insulin secretion in human β-cell lines, suggesting a role in insulin production or secretion pathways .

  • Adiponectin Regulation: Genetic variants in ARL15 are associated with altered adiponectin levels, indicating a potential role in adipokine regulation .

  • Vesicular and Lipid Trafficking: ARL15 functions in these cellular processes, which are essential for adipocyte function and lipid metabolism .

These diverse roles position ARL15 as a multifaceted regulator of metabolic processes, particularly in adipose tissue biology.

Which protein interactions are critical for ARL15 function?

Several protein interactions have been identified that mediate ARL15's biological functions:

  • ARL6IP5 Interaction: Co-immunoprecipitation and mass spectrometry studies have identified the ER-localized protein ARL6IP5 as a potential interacting partner of ARL15, suggesting ARL15 may regulate adipocyte differentiation through this interaction .

  • Smad4 Binding: Active ARL15 specifically binds to the MH2 domain of Smad4, a crucial interaction for TGFβ family signaling regulation .

  • R-Smad Complex Formation: While not directly interacting with R-Smads, ARL15 facilitates the formation of the Smad complex by activating Smad4, which then interacts with phosphorylated R-Smads .

  • Trafficking Machinery: Though specific interactions are still being characterized, ARL15 likely interacts with components of the vesicular trafficking machinery given its role in vesicular and lipid trafficking .

Understanding these protein-protein interactions provides insight into the molecular mechanisms through which ARL15 influences various cellular processes and disease states.

What are the recommended methods for studying ARL15 subcellular localization?

To effectively study ARL15 subcellular localization, researchers should consider the following methodological approaches:

  • Immunofluorescence Microscopy with Golgi Markers:

    • Co-stain with markers of different Golgi compartments (e.g., GM130 for cis-Golgi, TGN46 for trans-Golgi)

    • Use confocal microscopy to precisely determine co-localization patterns

    • Implement super-resolution techniques for detailed analysis of Golgi subcompartments

  • Live Cell Imaging:

    • Express fluorescently-tagged ARL15 (e.g., GFP-ARL15) to monitor dynamic changes in localization

    • Use photoactivatable or photoconvertible fluorescent proteins to track ARL15 movement between compartments

  • Subcellular Fractionation:

    • Isolate Golgi, ER, endolysosomal, and cytosolic fractions

    • Perform Western blotting with anti-ARL15 antibodies and organelle markers

    • Quantify the distribution of ARL15 across fractions

  • Palmitoylation-Site Mutants:

    • Generate palmitoylation-site mutants (e.g., cysteine to alanine substitutions)

    • Compare localization patterns between wild-type and mutant proteins

  • Electron Microscopy:

    • Use immunogold labeling to precisely localize ARL15 at the ultrastructural level

    • Quantify gold particle distribution across cellular compartments

These approaches, used in combination, provide comprehensive insight into the dynamic localization patterns of ARL15 under various cellular conditions and experimental perturbations.

How can researchers effectively manipulate ARL15 expression and activity in cellular models?

Several approaches can be employed to modulate ARL15 expression and activity for functional studies:

  • Genetic Knockdown/Knockout Approaches:

    • siRNA or shRNA for transient or stable knockdown

    • CRISPR-Cas9 genome editing for complete knockout

    • Conditional knockout systems (e.g., Cre-lox) for tissue-specific or inducible deletion

  • Overexpression Systems:

    • Transient transfection with expression vectors

    • Stable cell lines using lentiviral/retroviral systems

    • Inducible expression systems (e.g., Tet-On/Off)

  • Activity Modulation:

    • Express constitutively active mutants (GTP-locked)

    • Express dominant negative mutants (GDP-locked)

    • Use palmitoylation inhibitors to disrupt localization

  • Rescue Experiments:

    • Knockdown endogenous ARL15 and reintroduce wild-type or mutant proteins

    • Useful for structure-function analyses and determining critical domains

  • Pharmacological Approaches:

    • While specific ARL15 inhibitors are not yet available, TGFβ pathway modulators can be used to study ARL15's role in this signaling context

When designing these experiments, researchers should consider cell type-specificity, as ARL15 functions may vary between tissues. Adipocyte cell lines, pancreatic β-cells, and cells responsive to TGFβ signaling are particularly relevant models based on ARL15's known functions.

What are the recommended approaches for investigating ARL15's role in TGFβ signaling?

To thoroughly characterize ARL15's function in TGFβ signaling, researchers should consider these methodological approaches:

  • Protein-Protein Interaction Studies:

    • Co-immunoprecipitation to confirm ARL15-Smad4 interactions

    • Proximity ligation assays to visualize interactions in situ

    • FRET/BRET assays to monitor dynamic interactions

    • Yeast two-hybrid or mammalian two-hybrid screens to identify additional interaction partners

  • Smad Complex Assembly Assays:

    • Analyze R-Smad phosphorylation via Western blotting

    • Assess Smad complex formation using size exclusion chromatography

    • Monitor Smad nuclear translocation through subcellular fractionation or live imaging

  • GTPase Activity Measurements:

    • GTP-binding assays using radiolabeled GTP

    • GAP activity assays to measure Smad4's effect on ARL15 GTPase activity

    • Use of GTP/GDP-locked ARL15 mutants to assess pathway dependence on nucleotide binding state

  • Transcriptional Readouts:

    • Luciferase reporter assays using TGFβ-responsive elements

    • qRT-PCR analysis of TGFβ target genes

    • ChIP assays to assess Smad binding to target promoters following ARL15 manipulation

  • Functional Consequences:

    • Assess cellular responses to TGFβ (e.g., growth inhibition, EMT) following ARL15 perturbation

    • Analyze pathway activation kinetics with and without ARL15

These approaches together provide a comprehensive framework for understanding ARL15's specific contributions to TGFβ signaling regulation .

How do ARL15 variants contribute to metabolic disease pathogenesis?

ARL15 variants likely contribute to metabolic disease pathogenesis through multiple mechanisms, though the complete picture is still emerging:

  • Altered Insulin Signaling:

    • ARL15 loss-of-function reduces insulin secretion in β-cell lines, potentially contributing to hyperglycemia in diabetes

    • GWAS studies associate ARL15 variants with elevated fasting insulin levels, suggesting insulin resistance

  • Adiponectin Dysregulation:

    • ARL15 variants are associated with lower adiponectin levels

    • Reduced adiponectin contributes to insulin resistance, inflammation, and vascular dysfunction

  • Impaired Adipocyte Differentiation:

    • ARL15 regulates adipogenesis-related gene expression

    • Disruption may lead to dysfunctional adipose tissue and altered lipid storage

  • Vesicular and Lipid Trafficking Defects:

    • ARL15 functions in these processes are crucial for normal cellular metabolism

    • Disruption could alter cellular lipid handling and contribute to lipotoxicity

  • TGFβ Signaling Dysregulation:

    • ARL15 positively regulates TGFβ signaling

    • Alterations could affect various TGFβ-dependent processes implicated in metabolic and inflammatory diseases

  • Lipodystrophy Connection:

    • Loss-of-function mutations in ARL15 are found in some lipodystrophy patients

    • Lipodystrophy is associated with severe metabolic dysfunction and insulin resistance

These diverse mechanisms suggest that different ARL15 variants may contribute to distinct aspects of metabolic syndrome, potentially explaining its associations with multiple metabolic traits.

What are the current challenges in developing ARL15-targeted therapeutics?

Despite its promising therapeutic potential, several challenges must be addressed in developing ARL15-targeted therapeutics:

  • Target Specificity:

    • ARL15 belongs to the ARF family with many structurally similar members

    • Developing compounds that specifically target ARL15 without affecting related GTPases is challenging

  • Activation State Selectivity:

    • Distinguishing between GTP-bound and GDP-bound forms requires sophisticated screening approaches

    • Different disease contexts may require stabilizing different activation states

  • Protein-Protein Interaction Complexity:

    • ARL15 has multiple interaction partners including Smad4 and ARL6IP5

    • Determining which interactions to target for specific diseases remains unclear

  • Tissue-Specific Delivery:

    • ARL15 functions in multiple tissues including pancreatic β-cells and adipocytes

    • Tissue-targeted delivery systems would be necessary to avoid unwanted effects

  • Incomplete Understanding of Biology:

    • The precise molecular mechanisms of ARL15 in disease contexts are still being characterized

    • Additional research is needed to identify the most effective intervention points

  • Potential Compensatory Mechanisms:

    • Other ARF family members might compensate for ARL15 inhibition

    • Long-term efficacy could be limited by pathway adaptation

  • Validation in Disease Models:

    • More extensive testing in relevant disease models is needed before clinical development

Despite these challenges, ARL15 remains a promising target, particularly for metabolic disorders and rheumatoid arthritis, as highlighted in recent review literature .

How might researchers reconcile contradictory findings about ARL15 function across different studies?

When faced with contradictory findings about ARL15 function, researchers should consider the following approaches:

  • Evaluate Experimental Systems:

    • Different cell types may exhibit distinct ARL15 functions

    • Compare primary cells vs. cell lines vs. in vivo models

    • Assess species differences (human vs. mouse) that might explain discrepancies

  • Consider ARL15 Activation State:

    • Results may differ based on whether studies examined GTP-bound or GDP-bound ARL15

    • Some phenotypes may be specific to constitutively active or dominant negative forms

  • Analyze Tissue Specificity:

    • ARL15 may have tissue-specific roles in adipose tissue, pancreatic islets, etc.

    • Contradictory findings might reflect legitimate biological differences between tissues

  • Assess Technical Approaches:

    • Different knockdown/knockout strategies may yield varying results

    • Acute vs. chronic ARL15 depletion could produce different phenotypes

    • Overexpression artifacts might explain some discrepancies

  • Examine Environmental Conditions:

    • ARL15 function may be context-dependent (e.g., high glucose, insulin stimulation)

    • Experimental conditions should be carefully compared across studies

  • Conduct Direct Replication Studies:

    • Design experiments that directly test contradictory findings under identical conditions

    • Collaborate with groups reporting different results for joint analyses

  • Consider Post-Translational Modifications:

    • Palmitoylation status significantly affects ARL15 localization and function

    • Other modifications might also influence activity in a context-dependent manner

By systematically addressing these factors, researchers can develop more nuanced models of ARL15 function that reconcile apparently contradictory findings and advance understanding of this protein's complex biology.

What are the emerging technologies that could advance ARL15 research?

Several cutting-edge technologies show promise for advancing ARL15 research:

  • Proximity-Based Proteomics:

    • BioID, APEX, or TurboID approaches to identify the complete ARL15 interactome in different cellular compartments

    • Identification of activation state-specific interaction partners

  • Single-Cell Transcriptomics/Proteomics:

    • Characterize cell-type specific expression patterns of ARL15

    • Identify heterogeneous responses to ARL15 perturbation within tissues

  • CRISPR Screens:

    • Genome-wide or targeted CRISPR screens to identify genetic modifiers of ARL15 function

    • Synthetic lethal approaches to discover context-dependent vulnerabilities

  • Cryo-EM and Structural Biology:

    • Determine high-resolution structures of ARL15 in different activation states

    • Characterize ARL15-Smad4 complex structure for rational drug design

  • Organoid Models:

    • Study ARL15 function in more physiologically relevant 3D culture systems

    • Examine tissue-specific roles in adipose, pancreatic, or other relevant organoids

  • In Vivo CRISPR Editing:

    • Generate tissue-specific or inducible ARL15 modifications in animal models

    • Create human disease-associated variants for mechanistic studies

  • Novel Chemical Biology Approaches:

    • Development of small molecule modulators of ARL15 activity

    • Targeted protein degradation approaches (PROTACs) specific for ARL15

  • Advanced Imaging Techniques:

    • Live super-resolution imaging to track ARL15 dynamics

    • Correlative light and electron microscopy to characterize subcellular localization at nanoscale resolution

These technologies could rapidly accelerate understanding of ARL15 biology and facilitate translation to therapeutic applications.

How might epigenetic regulation influence ARL15 expression in different disease contexts?

Epigenetic regulation likely plays an important role in modulating ARL15 expression across different tissues and disease states:

  • DNA Methylation:

    • The ARL15 promoter region may be subject to differential methylation in metabolic diseases

    • GWAS-identified ARL15 variants might create or destroy CpG sites affecting methylation patterns

    • Researchers should consider performing methylation analysis of the ARL15 locus in relevant tissues from disease vs. healthy samples

  • Histone Modifications:

    • Activating (H3K4me3, H3K27ac) or repressive (H3K27me3, H3K9me3) marks may dynamically regulate ARL15 expression

    • ChIP-seq analysis of histone modifications at the ARL15 locus could reveal tissue-specific regulatory mechanisms

    • TGFβ signaling itself influences epigenetic modifications, potentially creating feedback loops affecting ARL15 expression

  • Chromatin Accessibility:

    • ATAC-seq or DNase-seq approaches could identify differential chromatin accessibility at the ARL15 locus

    • Metabolic stress might alter chromatin structure to influence ARL15 expression

  • Non-coding RNAs:

    • miRNAs targeting ARL15 mRNA might be dysregulated in disease states

    • Long non-coding RNAs could regulate ARL15 expression through various mechanisms

    • Comprehensive RNA-seq analysis could identify regulatory non-coding RNAs

  • Environmental Influences:

    • Metabolic stressors (high glucose, free fatty acids) may induce epigenetic changes affecting ARL15

    • Aging-associated epigenetic drift might contribute to altered ARL15 expression in age-related metabolic decline

Understanding these epigenetic regulatory mechanisms could reveal new therapeutic opportunities through targeting the upstream regulation of ARL15 rather than the protein itself.

What are the most promising translational research directions for ARL15?

Several translational research directions show particular promise for ARL15:

  • Biomarker Development:

    • Evaluate circulating ARL15 protein levels as potential biomarkers for metabolic disease risk

    • Develop assays for ARL15 activation states in accessible tissues

    • Create genetic risk scores incorporating ARL15 variants for personalized medicine approaches

  • Therapeutic Target Validation:

    • Conditional knockout models in metabolic disease-relevant tissues

    • Humanized mouse models carrying disease-associated ARL15 variants

    • Validation in patient-derived cells and organoids

  • Drug Discovery Approaches:

    • Structure-based design of small molecules targeting ARL15-Smad4 interaction

    • Development of allosteric modulators of ARL15 GTPase activity

    • Screening for compounds that influence ARL15 palmitoylation or localization

  • Rheumatoid Arthritis Applications:

    • Investigate ARL15 function in immune cells relevant to RA pathogenesis

    • Explore connections between ARL15, TGFβ signaling, and inflammatory processes

    • Develop tissue-specific delivery approaches for synovial targeting

  • Diabetes and Obesity Interventions:

    • Target ARL15 to improve β-cell function and insulin secretion

    • Modulate ARL15 in adipose tissue to enhance adiponectin production

    • Explore approaches to normalize vesicular trafficking in insulin-responsive tissues

  • Combinatorial Approaches:

    • Identify synergistic targets that enhance beneficial effects of ARL15 modulation

    • Develop combined biomarkers incorporating ARL15 status with other disease indicators

  • Precision Medicine Strategies:

    • Stratify patients based on ARL15 genetic variants for targeted interventions

    • Develop companion diagnostics for future ARL15-targeted therapies

These translational directions could accelerate the development of ARL15-based diagnostics and therapeutics for metabolic and inflammatory diseases, positioned as "emerging therapeutic targets" as highlighted in recent literature .

Product Science Overview

Structure and Function

ARL15 is a small GTPase that binds to guanosine triphosphate (GTP) and guanosine diphosphate (GDP), cycling between an active GTP-bound state and an inactive GDP-bound state . This cycling is essential for its function as a molecular switch in various cellular processes. ARL15 has been identified as a binding partner of CNNM (Cyclin M) proteins and an inhibitor of divalent cation influx by TRPM7 (Transient Receptor Potential Melastatin 7) .

Role in Ion Transport

ARL15 plays a significant role in regulating ion transport, particularly magnesium (Mg²⁺) and zinc (Zn²⁺) ions. It binds to CNNM proteins and inhibits their Mg²⁺ efflux activity . This regulation is crucial for maintaining cellular ion homeostasis, which is vital for various cellular functions, including energy production and protein synthesis .

Structural Insights

Recent structural studies have provided insights into the interaction between ARL15 and CNNM proteins. The crystal structure of the ARL15-CNNM2 complex reveals the molecular basis for their binding and identifies specific mutations that can block this interaction . For example, the R95A mutant of ARL15 fails to inhibit CNNM and TRPM7 transport of Mg²⁺ and Zn²⁺ ions .

Biological Significance

The regulation of ion transport by ARL15 is essential for various physiological processes. Disruptions in ARL15 function can lead to imbalances in ion homeostasis, which may contribute to various diseases. For instance, ARL15 has been associated with conditions such as cholestasis and spastic paraplegia .

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.