Recombinant Rat Arrestin domain-containing protein 4 (Arrdc4)

Shipped with Ice Packs
In Stock

Description

General Information

Arrestin domain-containing protein 4 (ARRDC4) is a member of the alpha-arrestin protein family, initially recognized in fungi and yeast before being identified as a novel class of arrestins in mammals . Alpha-arrestins, including ARRDC4, contain arrestin_N and arrestin_C domains, along with PPxY motifs that are unique to this group . In humans, six alpha-arrestins have been identified: ARRDC1, ARRDC2, ARRDC3, ARRDC4, ARRDC5, and TXNIP .

In the context of rat models, recombinant Arrdc4 refers to the protein produced using recombinant DNA technology, allowing for research into its function and interactions . Studies show that ARRDC4 plays a crucial role in glucose metabolism and response to metabolic stress in the heart .

Protein-Protein Interactions

ARRDC4 functions as a scaffold protein, interacting with various proteins involved in cellular processes such as protein degradation, RNA splicing, and protein transport .

  • E3 Ubiquitin Ligases: ARRDC4 interacts with E3 ubiquitin ligases like NEDD4 through its PPxY motifs, facilitating the ubiquitination of target proteins .

  • WW Domain-Containing Proteins: The PPxY motifs in ARRDC4 bind with high affinity to WW domains found in various proteins, including ubiquitin ligases .

  • GLUT1 Interaction: ARRDC4 interacts with GLUT1, modulating its endocytosis and thereby regulating glucose uptake .

Role in Glucose Metabolism

ARRDC4 plays a significant role in regulating glucose metabolism, especially under conditions of metabolic stress such as ischemia and diabetes .

  • Regulation of GLUT1: ARRDC4 interacts with GLUT1 and promotes its endocytosis, affecting glucose uptake in cells .

  • Myocardial Ischemic Injury: Inhibition of ARRDC4 has been shown to reduce myocardial ischemic injury and improve outcomes after acute myocardial infarction .

  • Glucose Deprivation-Induced ER Stress: ARRDC4 contributes to glucose deprivation-induced endoplasmic reticulum (ER) stress, leading to cardiomyocyte death .

Impact on Cardiac Function

Research indicates that ARRDC4 influences cardiac function, particularly in response to diabetes and ischemia .

  • Cardiac Reserve: Studies using Arrdc4-knockout mice demonstrated greater cardiac inotropic/chronotropic reserve and higher exercise endurance compared to wild-type animals under diabetic conditions .

  • Glucose Uptake in Organs: Arrdc4-knockout mice exhibited higher glucose uptake in essential organs such as the heart and brain, particularly under fasting conditions in diabetes .

  • Endoplasmic Reticulum (ER) Stress: Cardiac-specific overexpression of ARRDC4 in diabetic mice increased ER stress within the myocardium, an effect that was abolished by interaction-defective mutants .

ARRDC4 as a Therapeutic Target

Given its role in cardiac metabolism and response to ischemia, ARRDC4 has emerged as a potential therapeutic target for ischemic heart disease and related metabolic disorders .

  • Inhibition of ARRDC4: Studies suggest that inhibiting ARRDC4 could enhance cardiomyocyte survival in metabolically challenging environments .

  • Target for Type 2 Diabetes (T2D): ARRDC4 has been identified as a potential biomarker for T2D diagnosis .

Experimental Evidence

Arrrdc4 knockout mice exhibit improved cardiac function and glucose metabolism, underscoring ARRDC4's regulatory role.

Table 1: Effects of ARRDC4 Deletion on Glucose Metabolism in Diabetic Mice

ParameterWild-Type MiceArrdc4-Knockout Mice
Fasting Blood Glucose LevelsHigherLower
Glucose Uptake in HeartDecreased in diabetic conditionsIncreased in both fasted and fed diabetic conditions
Glucose Uptake in BrainDecreased in diabetic conditionsIncreased in both fasted and fed diabetic conditions
Glucose Uptake in Skeletal MuscleDecreased in diabetic conditionsIncreased in fed state with diabetes
Glucose Uptake in LiverDecreased in diabetic conditionsIncreased in fed state with diabetes

Table 2: ARRDC4 and Interacting Proteins

ProteinInteractionBiological Function
GLUT1Interacts directly, promoting endocytosisFacilitates glucose transport
NEDD4Interacts via PPxY motifsE3 ubiquitin ligase involved in protein degradation
WWP2, WWP1, ITCHInteracts via WW domainsUbiquitin ligases involved in protein degradation
Proteasome DomainsInteracts directlyProtein degradation
RNA-binding domainsInteracts with ARRDC3RNA splicing and processing

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order remarks for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please consult your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to settle the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which may serve as a guideline.
Shelf Life
Shelf life depends on various factors including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The specific tag type will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Arrdc4; Ab1-209Arrestin domain-containing protein 4; Liver regeneration-related protein LRRG041/LRRGT00117
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-300
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Rattus norvegicus (Rat)
Target Names
Arrdc4
Target Protein Sequence
MSSQSSILVD FRPLATSFTG KYGSIQYCVR AVLERPQVPD QSVRRELQVV SHVDVNTPPL LTPMLKTQEK MVGCWLFTSG PVSLSVKIER KGYCNGEAIP IYAEIENCSS RLVVPKAAIF QTQTYLASGK TKTVRHMVAN VRGNHIGSGS TDTWNGKMLK IPPVTPSILD CCIIRVYIHI PGAKKLMLEL PLVIGTIPYS GFGRRNSSMA SQFSMDMCWL ALALPEQPEA PPNYADVVSE EEFSRHIPPY PQPSACDGEA CYSMFACIQE FRFQPPPLYS ESHAQLFCLQ PVGPTNRAHF
Uniprot No.

Target Background

Function
Arrdc4 functions as an adapter protein, recruiting ubiquitin-protein ligases to their substrates. It plays a role in the endocytosis of activated G protein-coupled receptors (GPCRs) through a ubiquitination-dependent mechanism and contributes to SLC11A2 incorporation into extracellular vesicles. It may also be involved in glucose uptake.
Database Links
Protein Families
Arrestin family
Subcellular Location
Early endosome. Cell membrane; Peripheral membrane protein; Cytoplasmic side. Cytoplasmic vesicle.

Q&A

How does ARRDC4 compare to other members of the α-arrestin family?

Among the six known α-arrestins in mammals, ARRDC4 shares the highest sequence similarity with thioredoxin-interacting protein (TXNIP), with approximately 60% similar amino acids . Despite this similarity, ARRDC4 and TXNIP operate through distinct molecular pathways. Unlike TXNIP, ARRDC4 does not increase oxidative stress or change the level of the antioxidant glutathione .

The mechanistic differences extend to extracellular vesicle (EV) formation as well. While Arrdc1 relies on ESCRT components Tsg101 and VPS4 for EV formation, ARRDC4 appears to utilize a different mechanism involving the recycling pathway component Rab11a . This functional divergence highlights the specialized roles of different α-arrestins despite their structural similarities.

How can researchers generate and validate ARRDC4 knockout models?

Creating ARRDC4 knockout models has been successfully accomplished using CRISPR/Cas9 genome editing technology. The process typically follows these steps:

  • Design of gRNA pairs: Target the Arrdc4 gene locus with carefully designed guide RNAs.

  • Microinjection: Introduce the gRNA along with Cas9 mRNA into embryos (e.g., C57BL/6 for mouse models).

  • Genomic deletion verification: One published approach resulted in deletion of exons 1-8 in the Arrdc4 gene, confirmed by PCR of genomic DNA using specific primers that amplified different fragment sizes for wild-type (964 bp) versus knockout (768 bp) alleles .

  • Sequence confirmation: DNA sequencing to verify the precise deletion (e.g., loss of 13,630 base pairs of genomic DNA within the Arrdc4 gene locus) .

  • Expression analysis: Quantitative PCR across multiple tissues to confirm the absence of Arrdc4 mRNA expression in the knockout animals .

  • Transcriptome analysis: RNA-seq analysis can be performed to confirm the specificity of the knockout and evaluate compensatory changes in gene expression. In one study, among 14,411 genes identified, Arrdc4 was the only gene whose expression was greatly changed (log2 fold change: −7.0) in knockout hearts compared with wild-type .

What expression systems and purification methods are optimal for producing recombinant ARRDC4?

Successful expression of recombinant ARRDC4 has been reported using several systems:

  • Mammalian expression systems: Used for rat ARRDC4 production with high yield and proper post-translational modifications .

  • Yeast expression systems: Employed for full-length recombinant rat ARRDC4 with >85% purity (SDS-PAGE) .

  • Plasmid vectors: For human ARRDC4, expression-ready ORF plasmids such as pCMV6-AC-GFP with appropriate selection markers (Ampicillin for E. coli, Neomycin for mammalian cells) have been used successfully .

For purification and storage:

  • His-tagging enables purification via ion-exchange column chromatography

  • Recommended storage in PBS buffer

  • For long-term storage, maintain at -20°C to -80°C

  • For lyophilized protein, shelf life is typically 12 months at -20°C/-80°C

Reconstitution protocol:

  • Centrifuge at 5,000×g for 5 min

  • Add 100μl of sterile water to dissolve the DNA

  • Incubate for 10 minutes at room temperature

  • Briefly vortex and quick spin to concentrate at the bottom

  • For long-term experiments requiring sterility, filtration with a 0.22μm filter is recommended

How does ARRDC4 regulate sperm maturation through extracellular vesicle biogenesis?

ARRDC4 plays a critical role in sperm maturation through its control of extracellular vesicle (EV) biogenesis in the reproductive system. Research has revealed several important aspects of this function:

  • Normal testicular development but epididymal maturation defects: Sperm from Arrdc4–/– mice develop normally through the testis but fail to acquire adequate motility and fertilization capabilities as they traverse the epididymis .

  • Specific fertility deficits: These include reduced motility, premature acrosome reaction, reduction in zona pellucida binding, and decreased two-cell embryo production .

  • Reduction in EV production: There is a significant reduction in extracellular vesicle production by Arrdc4–/– epididymal epithelial cells, particularly affecting larger (>200 nm) vesicles .

  • Functional rescue through EV supplementation: Supplementation of Arrdc4–/– sperm with EVs from wild-type epididymal cells dampened the acrosome reaction defect and restored zona pellucida binding, demonstrating that the fertility deficits are directly related to EV deficiency .

  • Mechanistic distinction: Unlike Arrdc1-mediated EV formation which requires ESCRT components, ARRDC4-dependent EV biogenesis appears to rely on a different mechanism involving the recycling pathway component Rab11a and may mediate the transfer of specific cargoes into EVs, such as the divalent metal ion transporter DMT1 .

These findings establish ARRDC4 as an important regulator of sperm maturation by controlling the acquisition of extrinsic signals required for optimal fertilization capacity through epididymal EVs.

What experimental approaches can assess ARRDC4 effects on fertility in animal models?

To investigate ARRDC4's impact on fertility, researchers can employ multiple complementary experimental approaches:

  • Genetic manipulation models:

    • CRISPR/Cas9-generated Arrdc4 knockout mice

    • Conditional knockout models to study tissue-specific effects

    • Transgenic models expressing mutant forms of ARRDC4 (e.g., C-terminal domain mutations)

  • Sperm functional assessments:

    • Computer-assisted sperm analysis (CASA) to evaluate motility parameters

    • Acrosome reaction assays using fluorescent markers

    • Zona pellucida binding assays

    • In vitro fertilization studies with quantification of two-cell embryo formation rates

  • Extracellular vesicle analysis:

    • Isolation of epididymal EVs using differential ultracentrifugation or size exclusion chromatography

    • Nanoparticle tracking analysis to quantify EV size distribution and concentration

    • Proteomic analysis of EV cargo from wild-type and Arrdc4–/– mice

    • Electron microscopy to visualize EV morphology

  • Reconstitution experiments:

    • Collection of EVs from wild-type mice for supplementation studies

    • Co-incubation of Arrdc4–/– sperm with wild-type EVs

    • Assessment of functional recovery using the parameters mentioned above

  • Molecular pathway analysis:

    • Investigation of Rab11a involvement in ARRDC4-mediated EV biogenesis

    • Analysis of DMT1 and other potential EV cargo proteins

    • EV cargo transfer tracking using fluorescent labeling techniques

Through these approaches, researchers can comprehensively evaluate how ARRDC4 influences reproductive function and potentially develop therapeutic strategies for fertility issues related to EV biogenesis defects.

How does ARRDC4 regulate glucose metabolism across different tissues?

ARRDC4 exhibits tissue-specific roles in glucose metabolism, with distinct effects in different organs:

In liver:

  • ARRDC4 knockout (KO) mice show impaired hepatic glucose production during fasting

  • Glucagon-stimulated gluconeogenesis is defective in ARRDC4KO mice

  • Basal hepatic glucose production decreased by approximately 50% in ARRDC4KO mice compared to wild-type

  • ARRDC4 regulates suppression of glucose production in response to insulin

In cardiac tissue:

  • ARRDC4 binds to GLUT1, induces its endocytosis, and blocks cellular glucose uptake in cardiomyocytes

  • Deletion of Arrdc4 increases myocardial glucose uptake and glycogen storage

  • Arrdc4-KO hearts exhibit enhanced glucose transport, protecting against energy crisis during ischemia

In peripheral tissues:

  • Insulin-stimulated glucose uptake rates in skeletal muscle and white adipose tissue are significantly decreased in ARRDC4KO mice

  • ARRDC4KO mice show decreased insulin sensitivity during hyperinsulinemic-euglycemic clamp studies

Whole-body effects:

  • ARRDC4KO mice exhibit mild fasting hypoglycemia at baseline

  • During insulin tolerance tests, ARRDC4KO mice show decreased glucose levels after insulin injection with delayed recovery compared to wild-type mice

This tissue-specific regulation indicates ARRDC4 functions as an important metabolic switch with context-dependent roles in glucose homeostasis.

What molecular mechanisms explain ARRDC4's interaction with glucose transporters?

ARRDC4 regulates glucose uptake through a direct interaction with glucose transporter 1 (GLUT1). The molecular mechanism has been characterized through both functional and structural studies:

  • Direct binding and endocytosis induction:

    • ARRDC4 binds to GLUT1 and induces its endocytosis from the cell surface

    • This results in decreased surface expression of GLUT1 and reduced glucose uptake

    • In cells expressing wild-type ARRDC4, GLUT1 appears in intracellular puncta/vesicles rather than at the plasma membrane

  • Structural interaction interface:

    • Deep-learning methods based on the AlphaFold database identified specific binding residues

    • Key ARRDC4 residues include Lys243, Thr244, Asp290, and Glu308 in the C-terminal domain

    • These residues interact with the intracellular loops of GLUT1

  • Mutational analysis outcomes:

    • When these critical residues are mutated, ARRDC4 completely loses interaction with GLUT1

    • The interaction-defective ARRDC4 fails to inhibit cellular glucose transport

    • Confocal imaging confirms GLUT1 remains at the plasma membrane when the ARRDC4 docking site is mutated

  • Functional consequences:

    • Wild-type ARRDC4 induces ATF4/CHOP mRNA expression (ER stress markers)

    • Interaction-defective ARRDC4 fails to fully induce these stress response genes

    • This demonstrates that GLUT1 interaction is necessary for ARRDC4-mediated metabolic stress

This mechanistic understanding provides a foundation for potential therapeutic targeting of the ARRDC4-GLUT1 interaction in diseases where altered glucose metabolism plays a role.

How does ARRDC4 mediate cardiac responses to ischemic stress?

ARRDC4 plays a critical role in cardiac responses to ischemic stress through several interconnected mechanisms:

  • Glucose transport inhibition:

    • ARRDC4 binds to GLUT1, inducing its endocytosis from the cell surface

    • This reduces glucose uptake in cardiomyocytes, limiting energy substrate availability during ischemia

    • Arrdc4-KO hearts exhibit increased myocardial glucose uptake and glycogen storage, improving energy reserves

  • ER stress induction:

    • ARRDC4 promotes endoplasmic reticulum (ER) stress in cardiomyocytes

    • It increases expression of multiple unfolded protein response (UPR) genes, including ATF6, ATF4, CHOP, and XBP1

    • ARRDC4 enhances XBP1 splicing, a key indicator of ER stress activation

    • Western blot analysis confirms increased protein expression of ATF4 and CHOP

  • Cell death promotion:

    • ARRDC4 expression increases LDH release, a marker of cell damage

    • It activates both apoptotic pathways (caspase-3/7 activation, Annexin V-positive staining) and necrotic pathways (DNA Nuclear Green DCS1-positive staining)

    • These effects are associated with depletion of intracellular ATP

  • Hypoxia sensitivity:

    • Hypoxic stimuli (1% O₂) increase cellular LDH release and expression of Chop in a time-dependent manner

    • ARRDC4 overexpression enhances hypoxia-induced cytotoxicity

    • Cardiomyocytes from Arrdc4-KO mice show greater resistance to hypoxia than those from wild-type animals

  • Protective effects of ARRDC4 deletion:

    • Deletion of Arrdc4 improves energy homeostasis during ischemia

    • It protects cardiomyocytes against myocardial infarction

    • This protection appears mediated by enhanced glucose uptake and reduced ER stress

These findings establish ARRDC4 as a key determinant of cardiomyocyte survival under ischemic conditions, primarily through its regulation of glucose metabolism and ER stress responses.

What therapeutic strategies targeting ARRDC4 might benefit ischemic heart disease?

Based on the molecular mechanisms of ARRDC4 in cardiac ischemia, several therapeutic strategies show promise:

  • Disruption of ARRDC4-GLUT1 interaction:

    • Small molecule inhibitors targeting the identified interaction interface (residues Lys243, Thr244, Asp290, and Glu308 in the C-terminal domain)

    • Peptide-based inhibitors mimicking critical regions of GLUT1 intracellular loops

    • This approach is supported by structural studies showing that mutation of these residues preserves GLUT1 at the plasma membrane and restores glucose transport

  • Modulation of ARRDC4 expression:

    • siRNA or antisense oligonucleotides targeting ARRDC4 mRNA

    • Gene therapy approaches to reduce ARRDC4 expression in cardiac tissue

    • This is supported by studies showing knockdown of ARRDC4 decreases hypoxia-induced cytotoxicity

  • Targeting downstream ER stress pathways:

    • Inhibitors of the ATF4/CHOP pathway to mitigate cell death

    • Compounds that enhance adaptive UPR responses while limiting terminal UPR pathways

    • Rationale: ARRDC4 induces ER stress through glucose deprivation, which contributes to cell death

  • Combinatorial approaches:

    • Combining ARRDC4 inhibition with standard cardioprotective strategies

    • Targeting ARRDC4 together with other key regulators of cardiac metabolism

Validation strategies should include:

  • In vitro screening using cardiomyocyte models under hypoxic conditions

  • Ex vivo isolated heart perfusion studies measuring functional parameters

  • In vivo myocardial infarction models with pre- and post-conditioning treatments

  • Assessment of infarct size, cardiac function, and long-term outcomes

The discovery that ARRDC4 drives glucose deprivation-induced ER stress leading to cardiomyocyte death provides a clear mechanistic basis for therapeutic intervention in ischemic heart disease.

How is ARRDC4 implicated in prostate cancer progression?

ARRDC4 has emerged as a significant factor in prostate cancer (PCa) progression through both genetic association and functional studies:

  • Genetic association with aggressive disease:

    • A germline variant of ARRDC4 (rs200944490 in chromosome 15) is significantly associated with high Gleason scores in prostate cancer

    • The odds ratio (OR) for high Gleason score was 6.459 (95% CI: 3.028–13.78, p = 1.39 × 10⁻⁶)

    • This represents one of the strongest genetic associations with PCa aggressiveness

SNP IDChrAllelesGeneMinor Allele FrequencyOR (95% CI)p-Value
rs20094449015G > CARRDC4GS < 8: 0.0105, GS ≥ 8: 0.060876.459 (3.028–13.78)1.39 × 10⁻⁶
  • Functional effects in prostate cancer cell lines:

    • siRNA-mediated knockdown of ARRDC4 in prostate cancer cell lines (PC3, DU145, LNCaP, and 22Rv1) significantly reduced cell proliferation

    • The effect was particularly pronounced in DU145 and LNCaP cells

    • ARRDC4 knockdown also inhibited cell migration, invasion, and epithelial-mesenchymal transition (EMT)

    • These changes appeared to be mediated through suppression of the PI3K/Akt/NF-κB signaling pathway

  • Potential as biomarker and therapeutic target:

    • The strong association with Gleason score suggests ARRDC4 genetic variants could serve as predictive biomarkers for aggressive prostate cancer

    • The functional role in promoting proliferation and invasion indicates ARRDC4 could be a therapeutic target

    • Targeting ARRDC4 might be particularly valuable in tumors with high Gleason scores

These findings collectively establish ARRDC4 as a potential candidate marker predictive of PCa aggressiveness and suggest its involvement in fundamental processes driving cancer progression.

What methodological approaches can assess ARRDC4's role in tumor development?

To comprehensively investigate ARRDC4's role in tumor development, researchers can employ multiple methodological approaches:

  • Genetic association studies:

    • Exome array analysis of tumor samples to identify ARRDC4 variants associated with disease progression

    • Correlation of variants with clinical parameters such as Gleason score, metastasis, and survival

    • Validation of associations in independent patient cohorts

  • Gene expression analysis:

    • Quantitative PCR and western blotting to assess ARRDC4 expression levels in tumor vs. normal tissues

    • Immunohistochemical staining of tissue microarrays to evaluate protein expression patterns

    • Correlation of expression with clinical outcomes and pathological features

  • Functional assessment in cancer cell lines:

    • siRNA or CRISPR-based knockdown/knockout of ARRDC4 in cancer cell lines

    • Overexpression studies using vectors like pCMV6-AC-GFP containing the ARRDC4 ORF

    • Analysis of effects on:

      • Proliferation (e.g., CCK-8 assay)

      • Apoptosis

      • Migration (wound healing assays)

      • Invasion (transwell assays)

      • EMT markers

  • Signaling pathway analysis:

    • Western blotting to assess effects on PI3K/Akt/NF-κB pathway components

    • Phosphoproteomic analysis to identify broader signaling changes

    • Rescue experiments using pathway activators or inhibitors

  • In vivo models:

    • Xenograft models using ARRDC4-modulated cancer cell lines

    • Genetically engineered mouse models with ARRDC4 alterations

    • Patient-derived xenografts with varying ARRDC4 expression levels

    • Assessment of tumor growth, metastasis, and response to therapy

  • Translational implications:

    • Development of ARRDC4-targeting strategies (small molecules, peptides, etc.)

    • Testing combination approaches with standard therapies

    • Biomarker development for patient stratification

These multifaceted approaches would provide comprehensive insights into ARRDC4's role in cancer biology and its potential as a therapeutic target.

How does ARRDC4 regulate innate immune signaling during viral infections?

ARRDC4 serves as an important regulator of innate immune signaling during viral infections, particularly in the context of Enterovirus 71 (EV71) infection:

  • Upregulation during viral infection:

    • ARRDC4 expression increases after EV71 infection in THP-1-derived macrophages

    • This upregulation has been verified in EV71-infected hand, foot, and mouth disease (HFMD) patients

  • Correlation with inflammatory markers:

    • ARRDC4 expression levels positively correlate with serum concentrations of pro-inflammatory cytokines

    • These include IL-6, TNF-α, and CCL3 in clinical specimens from infected patients

  • Molecular mechanism:

    • ARRDC4 interacts with MDA5 (melanoma differentiation-associated protein 5), a key pattern recognition receptor for viral RNA

    • This interaction occurs via ARRDC4's arrestin-like N domain

    • ARRDC4 recruits TRIM65 (tripartite motif-containing protein 65) to enhance K63 ubiquitination of MDA5

    • This ubiquitination activates downstream innate signaling pathways

    • The result is increased transcription of proinflammatory cytokines during viral infection

  • Pathological implications:

    • While innate immune activation is critical for viral clearance, excessive inflammatory responses can lead to tissue damage

    • In EV71 infection, high levels of cytokines and chemokines, with impaired production of type I interferon, contribute to severe complications

    • ARRDC4 appears to be a key mediator of this inflammatory response

This research highlights a previously unknown function of ARRDC4 in innate immunity, contributing to the understanding of how MDA5 activation is regulated during viral infection and suggesting ARRDC4 as a potential target for intervention in virus-induced inflammatory responses.

What experimental systems can be used to study ARRDC4's role in pathogen response?

To investigate ARRDC4's role in pathogen response, researchers can utilize several experimental systems and approaches:

  • Cell culture models:

    • Immune cell lines (THP-1, RAW264.7) with ARRDC4 modulation

    • Primary immune cells (macrophages, dendritic cells) from wild-type and Arrdc4-KO mice

    • Viral infection models using EV71 and other viruses

    • Assessment of cytokine production, signaling pathway activation, and cell survival

  • Protein interaction studies:

    • Co-immunoprecipitation assays to confirm interactions between ARRDC4 and immune signaling components (MDA5, TRIM65)

    • Domain mapping experiments to identify critical interaction regions

    • Ubiquitination assays to assess K63 ubiquitination of MDA5

    • Proximity ligation assays to visualize protein interactions in situ

  • Signaling pathway analysis:

    • Western blotting to assess activation of innate immune signaling components

    • Reporter assays (e.g., luciferase-based) to measure pathway activation

    • RNA-seq to assess global transcriptional responses

    • CRISPR screens to identify additional components of ARRDC4-mediated pathways

  • In vivo infection models:

    • EV71 infection in wild-type versus Arrdc4-KO mice

    • Assessment of viral loads, inflammatory markers, and disease severity

    • Tissue-specific deletion of ARRDC4 to determine cell type-specific contributions

    • Therapeutic modulation of ARRDC4 during infection

  • Clinical correlation studies:

    • Analysis of ARRDC4 expression in patient samples from various infectious diseases

    • Correlation with disease severity, inflammatory markers, and outcomes

    • Genetic association studies to identify ARRDC4 variants linked to infection susceptibility or severity

  • High-throughput approaches:

    • Proteomics to identify additional ARRDC4-interacting proteins during infection

    • Single-cell RNA-seq to assess cell type-specific responses

    • CRISPR activation/inhibition screens to identify genes that modify ARRDC4 function

These experimental systems would provide comprehensive insights into ARRDC4's role in pathogen response and potentially identify novel therapeutic targets for controlling excessive inflammation during viral infections.

What structure-function relationships in ARRDC4 could be exploited for targeted therapeutics?

Understanding structure-function relationships in ARRDC4 reveals several potential targets for therapeutic development:

  • C-terminal arrestin-fold domain:

    • Specific residues in this domain (Lys243, Thr244, Asp290, and Glu308) form the interaction interface with GLUT1

    • When these residues are mutated, ARRDC4 completely loses its ability to interact with GLUT1

    • This preserves GLUT1 at the plasma membrane and restores glucose transport

    • Small molecules or peptides targeting this interface could selectively disrupt GLUT1 interaction while preserving other ARRDC4 functions

  • Arrestin-like N domain:

    • This domain mediates interaction with MDA5 in the context of immune signaling

    • Targeting this interaction could modulate inflammatory responses during viral infections

    • Structure-based drug design could develop selective inhibitors of this domain

  • PY motifs:

    • ARRDC4 contains C-terminal PY motifs that bind WW domains of Nedd4 family ubiquitin ligases

    • These interactions are important for ARRDC4's adaptor functions

    • Compounds that interfere with these motifs could modulate ARRDC4's ability to recruit ubiquitin ligases

  • Tissue-specific targeting:

    • ARRDC4 exhibits tissue-specific effects (e.g., different roles in liver versus heart)

    • Tissue-targeted delivery of inhibitors could exploit these differences

    • For example, liver-targeted inhibition might improve glucose production during fasting, while cardiac-targeted inhibition could protect against ischemic injury

  • Pathway-specific modulation:

    • Different domains of ARRDC4 mediate distinct cellular functions

    • Selective modulation of specific interactions could achieve pathway-specific effects

    • This could minimize off-target effects while maximizing therapeutic benefit

  • Integration with computational approaches:

    • Deep-learning methods based on AlphaFold have already successfully predicted the ARRDC4-GLUT1 interaction interface

    • Similar approaches could identify other druggable pockets or interaction surfaces

    • Virtual screening against these targets could identify lead compounds for experimental validation

These structure-function insights provide a foundation for developing targeted therapeutics against ARRDC4 for conditions including ischemic heart disease, metabolic disorders, and inflammatory conditions.

How might ARRDC4 interact with other cellular pathways in stress response and disease progression?

ARRDC4 appears to be a critical integrator of multiple cellular pathways, particularly under stress conditions:

  • Integration of metabolic and stress signaling:

    • ARRDC4 links glucose metabolism (via GLUT1 regulation) to ER stress responses

    • ARRDC4 overexpression induces ER stress markers (ATF4, CHOP, XBP1)

    • This connection may represent a metabolic checkpoint where nutrient limitation triggers stress responses

  • Cross-talk between metabolism and inflammation:

    • ARRDC4 regulates both glucose metabolism and inflammatory signaling

    • In viral infections, ARRDC4 enhances inflammatory cytokine production

    • This suggests ARRDC4 may coordinate metabolic adaptation with immune responses

  • Cell death pathway integration:

    • ARRDC4 activates both apoptotic and necrotic death pathways

    • This occurs through ATP depletion, a central mediator of cell death

    • ARRDC4 may serve as a master regulator determining cell fate under stress

  • Possible interactions with autophagy:

    • Given ARRDC4's role in vesicle formation and metabolic stress response

    • Autophagy is a major adaptive response to nutrient limitation

    • ARRDC4 might regulate autophagic processes through its effects on vesicle trafficking

  • Potential circadian regulation:

    • Many metabolic processes show circadian rhythmicity

    • ARRDC4's role in glucose metabolism suggests potential temporal regulation

    • This could link circadian rhythms to metabolic adaptation in different tissues

  • Extracellular vesicle-mediated intercellular communication:

    • ARRDC4 controls EV biogenesis in multiple cell types

    • EVs mediate intercellular communication by transferring proteins, lipids, and nucleic acids

    • ARRDC4 may therefore coordinate tissue-level responses to stress through EV-mediated signaling

  • Cancer progression pathways:

    • ARRDC4 has been implicated in cancer progression through the PI3K/Akt/NF-κB pathway

    • It appears to regulate proliferation, migration, and EMT

    • These effects may involve both metabolic reprogramming and direct signaling effects

Future research should aim to elucidate these diverse interactions, potentially revealing ARRDC4 as a central node in cellular stress responses with significant implications for disease pathogenesis and therapeutic intervention.

How do the functions of ARRDC4 differ between cardiovascular, hepatic, and reproductive tissues?

ARRDC4 demonstrates remarkable tissue-specific functions that may reflect adaptation to the unique metabolic and physiological demands of different organs:

Cardiovascular system:

  • Binds GLUT1 and induces its endocytosis, reducing glucose uptake in cardiomyocytes

  • Promotes ER stress through glucose deprivation, activating the ATF4/CHOP pathway

  • Enhances cardiomyocyte death under ischemic conditions

  • Deletion of Arrdc4 protects against myocardial infarction by improving energy homeostasis

  • ARRDC4 is upregulated by ischemic stimuli, suggesting a role in maladaptive response to cardiac stress

Hepatic tissue:

  • Regulates hepatic glucose production during fasting

  • Controls gluconeogenesis in response to pyruvate

  • Modulates insulin's ability to suppress glucose production

  • ARRDC4 knockout mice show impaired hepatic glucose production with basal rates decreased by ~50%

  • May play a role in maintaining blood glucose levels during fasting

Reproductive system:

  • Critical for sperm maturation through the epididymis

  • Controls extracellular vesicle biogenesis in epididymal epithelial cells

  • Regulates the transfer of extrinsic signals required for optimal fertilization

  • Arrdc4-/- mice show reduced sperm motility, premature acrosome reaction, and reduced zona pellucida binding

  • Uses a mechanism involving the recycling pathway component Rab11a for EV formation

Metabolic integration:

  • While ARRDC4 promotes glucose uptake in peripheral tissues (muscle, fat), it inhibits uptake in the heart

  • This differential regulation suggests tissue-specific adaptations to varying metabolic demands

  • These opposing effects highlight the context-dependent nature of ARRDC4 function

These tissue-specific functions suggest that ARRDC4 has evolved specialized roles that may be regulated by tissue-specific interacting partners, post-translational modifications, or signaling environments. Understanding these differences is crucial for developing tissue-targeted therapeutic approaches.

What methodological approaches can distinguish between direct and indirect effects of ARRDC4 in different cell types?

Distinguishing between direct and indirect effects of ARRDC4 across different cell types requires sophisticated methodological approaches:

  • Tissue-specific genetic manipulation:

    • Conditional knockout mouse models using tissue-specific Cre recombinase systems

    • Inducible expression systems to control timing of ARRDC4 modulation

    • Cell type-specific CRISPR editing in vivo using AAV delivery systems

    • These approaches help isolate primary effects in specific tissues from secondary systemic responses

  • Acute versus chronic manipulation:

    • Acute knockdown using siRNA or inducible shRNA systems

    • Comparison with constitutive knockout models

    • Time-course studies to track the progression of molecular and cellular changes

    • This temporal analysis can separate immediate direct effects from adaptive responses

  • Structure-function analysis with mutant proteins:

    • Expression of domain-specific mutants (e.g., GLUT1-binding mutants)

    • Targeted disruption of specific protein-protein interactions

    • Comparison of phenotypes between different functional mutants

    • This approach can isolate which molecular interactions mediate specific cellular effects

  • Proximity labeling approaches:

    • BioID or APEX2 fusion proteins to identify proteins in close proximity to ARRDC4

    • Cell type-specific application to map tissue-specific interaction networks

    • Temporal analysis following stimulus application (e.g., hypoxia, nutrient deprivation)

    • This reveals direct binding partners that may mediate ARRDC4's effects

  • Multi-omics integration:

    • Comparison of transcriptomics, proteomics, and metabolomics data

    • Pathway analysis to identify direct versus secondary effects

    • Network modeling to predict causal relationships

    • Time-resolved analysis to establish sequence of events

  • Ex vivo and organoid systems:

    • Isolated organ perfusion models (e.g., Langendorff heart preparation)

    • Organoid cultures from different tissues with ARRDC4 manipulation

    • Co-culture systems to assess cell-cell communication effects

    • These approaches bridge the gap between in vitro and in vivo studies

  • Rescue experiments:

    • Selective restoration of ARRDC4 in specific cell types of global knockout models

    • Expression of tissue-specific binding partners in ARRDC4-deficient systems

    • Targeted intervention in downstream pathways

    • These approaches help establish necessity and sufficiency of direct mechanisms

These methodological approaches, applied systematically across different tissues and cell types, would provide a comprehensive understanding of ARRDC4's direct molecular actions versus its indirect effects on cellular physiology.

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.