MAVS Human

Mitochondrial Antiviral Signaling Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to MAVS Human

MAVS (Mitochondrial Antiviral-Signaling Protein) is a critical protein localized to mitochondrial membranes, peroxisomes, and mitochondrial-associated endoplasmic reticulum membranes (MAMs). It plays dual roles: driving antiviral responses and maintaining mitochondrial homeostasis in human cells. Recent studies reveal its uncanonical function in antagonizing cellular senescence, particularly in human mesenchymal stem cells (hMSCs) .

Gene and Protein Structure

  • Gene: Located on human chromosome 20, with multiple exons and introns.

  • Protein: Full-length MAVS comprises 540 amino acids, including:

    • N-terminal CARD domain: Facilitates interactions with cytosolic sensors (e.g., RIG-I, MDA5).

    • Proline-rich region: Scaffolds signaling complexes (e.g., TRAF proteins).

    • C-terminal transmembrane domain: Anchors MAVS to mitochondrial/peroxisomal membranes .

  • Isoforms: Alternative splicing generates shorter variants (e.g., miniMAVS), lacking the CARD domain .

DomainFunctionKey Interactions
CARDBinds RIG-I/MDA5 to initiate antiviral signalingRIG-I, MDA5, TRAF2/5
Proline-richScaffolds ubiquitination for signal amplificationTRAF6, NEMO, TBK1/IKKε
TransmembraneMembrane localizationMitochondrial OPA1, MFN1/2

Role in Antiviral Immunity

MAVS activates downstream signaling cascades (IRF3, NF-κB) to induce type I interferons (IFN-α/β) and proinflammatory cytokines. This response is critical for clearing RNA viruses (e.g., influenza, SARS-CoV-2) .

Interaction with OPA1

MAVS stabilizes OPA1, a dynamin-like GTPase essential for mitochondrial fusion. Depletion of MAVS or OPA1 disrupts mitochondrial dynamics, leading to:

  • Mitochondrial fragmentation: Increased mitochondrial mass and reduced membrane potential .

  • Functional deficits: Elevated ROS, diminished oxidative respiration, and premature senescence in hMSCs .

ParameterMAVS+/+ hMSCsMAVS−/− hMSCsReplenishment (MAVS/OPA1)
Mitochondrial membrane potentialHighLowRestored
ROS levelsBasalElevatedReduced
Senescence-associated β-gal activityLowHighAlleviated

Cell-Type-Specific Vulnerability

  • hMSCs: Highly sensitive to MAVS deficiency, showing accelerated senescence .

  • hESCs/hNSCs: No significant senescence phenotypes observed in MAVS-knockout models .

Senescence and Aging

MAVS deficiency in hMSCs triggers nuclear lamina disorganization, DNA damage (e.g., γH2AX foci), and upregulation of senescence-associated pathways (e.g., p53) . Replenishing MAVS or OPA1 reverses these defects, highlighting its therapeutic potential for age-related disorders .

Cardiovascular and Metabolic Disorders

Emerging evidence links MAVS to cardiovascular diseases (e.g., myocarditis) and metabolic dysregulation. MAVS-deficient mice exhibit impaired cardiac function, mitochondrial dysfunction, and lipid metabolism abnormalities .

Key Studies

StudyModelKey Findings
Zhang et al. (2023)MAVS-knockout hMSCsMAVS stabilizes OPA1; depletion disrupts mitochondrial fusion and induces senescence .
Wang et al. (2023)CRISPR hESCs/hNSCsMAVS loss does not affect pluripotency or neural stem cell function .
Liu et al. (2025)Cardiovascular modelsMAVS regulates mitochondrial autophagy and lipid metabolism in heart disease .

Molecular Pathways

  • Antiviral: MAVS → TRAF6 → TBK1/IKKε → IRF3/NF-κB → IFN-α/β .

  • Mitochondrial: MAVS → OPA1 stabilization → Mitochondrial fusion → ROS suppression → Senescence delay .

Product Specs

Introduction

The mitochondrial antiviral signaling protein (MAVS) plays a crucial role in the body's initial defense mechanism against viral infections. Serving as a critical link in the signaling pathways activated by viruses to produce interferon-beta (IFN-beta), MAVS is essential for this process. It facilitates the activation of transcription factors responsible for regulating the expression of IFN-beta, thereby contributing to the body's antiviral immune response.

Description
Recombinant MAVS Human, produced in E.coli, is a single polypeptide chain that is not glycosylated. It comprises 536 amino acids (specifically, amino acids 1 to 513) and has a molecular weight of 55.9 kDa. The MAVS protein has a 23 amino acid His-tag attached to its N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The MAVS solution is provided at a concentration of 0.25 mg/ml and is prepared in a buffer containing 20mM Tris-HCl (pH 8.0), 0.15M NaCl, 10% glycerol, and 1mM DTT.
Stability
For short-term storage (up to 2-4 weeks), the MAVS solution can be kept at 4°C. For longer storage, it is recommended to store the solution frozen at -20°C. To further enhance stability during long-term storage, adding a carrier protein such as 0.1% HSA or BSA is advised. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of the MAVS protein is determined to be greater than 85% based on SDS-PAGE analysis.
Synonyms

CARDIF, IPS-1, IPS1, VISA, Mitochondrial antiviral-signaling protein, MAVS, Putative NF-kappa-B-activating protein 031N, Virus-induced-signaling adapter, KIAA1271.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMPFAEDK TYKYICRNFS NFCNVDVVEI LPYLPCLTAR DQDRLRATCT LSGNRDTLWH LFNTLQRRPG WVEYFIAALR GCELVDLADE VASVYQSYQP RTSDRPPDPL EPPSLPAERP GPPTPAAAHS IPYNSCREKE PSYPMPVQET QAPESPGENS EQALQTLSPR AIPRNPDGGP LESSSDLAAL SPLTSSGHQE QDTELGSTHT AGATSSLTPS RGPVSPSVSF QPLARSTPRA SRLPGPTGSV VSTGTSFSSS SPGLASAGAA EGKQGAESDQ AEPIICSSGA EAPANSLPSK VPTTLMPVNT VALKVPANPA SVSTVPSKLP TSSKPPGAVP SNALTNPAPS KLPINSTRAG MVPSKVPTSM VLTKVSASTV PTDGSSRNEE TPAAPTPAGA TGGSSAWLDS SSENRGLGSE LSKPGVLASQ VDSPFSGCFE DLAISASTSL GMGPCHGPEE NEYKSEGTFG IHVAENPSIQ LLEGNPGPPA DPDGGPRPQA DRKFQEREVP CHRPSP.

Q&A

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

MAVS (Mitochondrial Antiviral Signaling protein) is a 540 amino acid adapter protein that serves as a critical signaling platform in the innate immune response against RNA viruses. Its primary function is to act as a molecular switch that receives signals from cytosolic RNA sensors (RIG-I and MDA5) and transduces these signals to activate downstream pathways leading to type I interferon production .

When investigating MAVS function, researchers should employ both gain-of-function approaches (overexpression studies) and loss-of-function techniques (siRNA knockdown, CRISPR/Cas9 knockout) to comprehensively assess its role in various cell types. Complementary approaches include co-immunoprecipitation assays to identify protein interaction partners and reporter assays measuring interferon promoter activity.

Where is MAVS localized within human cells?

MAVS primarily localizes to the outer mitochondrial membrane through its C-terminal transmembrane domain (TM) . This localization is essential for its function in antiviral signaling.

For subcellular localization studies, researchers should utilize:

  • Immunofluorescence microscopy with antibodies against MAVS and mitochondrial markers

  • Subcellular fractionation followed by Western blotting

  • Live-cell imaging with fluorescently tagged MAVS constructs

  • Super-resolution microscopy for detailed localization studies

Each approach offers different advantages: fractionation provides biochemical evidence while microscopy offers visual confirmation of MAVS distribution in intact cells.

What are the key structural domains of human MAVS?

Human MAVS contains several functional domains essential for its signaling activities:

DomainPositionFunctionInteracting Partners
CARD (N-terminal)Residues 1-93Mediates homotypic interactions with RIG-I/MDA5 CARD domainsRIG-I, MDA5
Proline-rich regionCentral regionContains binding motifs for downstream signaling moleculesTRAF proteins
Transmembrane (C-terminal)C-terminusAnchors MAVS to mitochondrial membraneMitochondrial membrane

To study domain function, employ domain deletion/mutation approaches coupled with functional readouts of MAVS activity, such as interferon reporter assays, aggregation assessment, and co-immunoprecipitation with binding partners .

How does MAVS form prion-like aggregates during viral infection?

Upon viral infection, RIG-I or MDA5 recognize viral RNA and undergo conformational changes that expose their CARD domains. These activated CARD domains interact with the CARD domain of MAVS, triggering a remarkable conformational change that promotes MAVS to form functional, prion-like aggregates . These aggregates serve as signaling platforms that recruit and activate downstream effectors.

Methodological approaches to study MAVS aggregation include:

  • Semi-denaturing detergent agarose gel electrophoresis (SDD-AGE)

  • Sucrose gradient centrifugation followed by Western blotting

  • Fluorescence microscopy using split-fluorescent protein constructs

  • Electron microscopy of purified MAVS complexes

Researchers should note that MAVS aggregates are highly stable and resistant to detergent solubilization, requiring specialized techniques for proper analysis.

What factors regulate MAVS activation and signaling?

MAVS regulation occurs through multiple mechanisms that precisely control its activation:

  • Protein-protein interactions: LGP2 interacts with the TM domain of MAVS to prevent recruitment of TRAF3, thereby regulating signal transduction .

  • Mitochondrial dynamics: Factors affecting mitochondrial physical state influence MAVS aggregation:

    • Mitochondrial fusion promotes MAVS signaling

    • Changes in membrane potential alter MAVS aggregation capacity

    • Reactive oxygen species (ROS) levels modulate MAVS activity

  • Post-translational modifications:

    • Phosphorylation: c-Abl positively regulates RLR signaling by phosphorylating MAVS at Y9 and Y3 residues

    • Ubiquitination: Various E3 ligases target MAVS for different ubiquitin modifications

To effectively study these regulatory mechanisms, researchers should employ a combination of biochemical, genetic, and imaging approaches in both resting and virus-stimulated conditions.

What are the optimal methods for detecting MAVS aggregation in experimental systems?

MAVS aggregation represents a critical step in antiviral signaling. Researchers can employ several complementary techniques to detect and quantify this phenomenon:

MethodApplicationAdvantagesLimitations
SDD-AGEBiochemical detection of high-molecular-weight MAVS aggregatesDirectly visualizes aggregate formationCell disruption required
Confocal microscopyVisualization of MAVS redistributionPreserves cellular contextLower resolution
Proximity ligation assayDetection of MAVS self-associationHigh sensitivityIndirect measure
FRET analysisReal-time monitoring of MAVS clusteringDynamic informationTechnical complexity

When interpreting aggregation data, researchers should consider that different cell types may display varying aggregation kinetics and that fixation methods can influence visualization results.

How can researchers distinguish between MAVS-dependent and MAVS-independent antiviral responses?

Differentiating MAVS-dependent from MAVS-independent responses requires careful experimental design:

  • Generate MAVS knockout cell lines using CRISPR/Cas9 technology

  • Compare interferon responses to stimuli known to activate:

    • RIG-I/MDA5 pathway (MAVS-dependent)

    • cGAS-STING pathway (MAVS-independent)

    • TLR pathways (MAVS-independent)

  • Use pathway-specific inhibitors alongside genetic approaches

  • Employ time-course experiments to capture differential kinetics

  • Analyze downstream signaling components specific to each pathway (IRF3 vs. IRF7 activation patterns)

Data interpretation should account for potential compensatory mechanisms that may emerge in MAVS-deficient systems and consider cell-type specific variations in pathway utilization.

What post-translational modifications regulate human MAVS activity?

MAVS undergoes multiple post-translational modifications that fine-tune its signaling capacity:

ModificationEnzymes InvolvedEffect on MAVSDetection Methods
Phosphorylationc-Abl (Y9, Y3)ActivationPhospho-specific antibodies, mass spectrometry
UbiquitinationTRAF family E3 ligasesContext-dependentUbiquitin pulldown, mass spectrometry
SUMOylationSUMO ligasesTypically inhibitorySUMO-IP, SUMO-specific antibodies
AcetylationVarious acetyltransferasesModulates activityAcetylation-specific antibodies

When studying MAVS modifications, researchers should consider that:

  • Modifications often occur sequentially or interdependently

  • Different cell types may exhibit varying modification patterns

  • Virus-specific effects may target particular modification sites

How do mitochondrial dynamics influence MAVS signaling capacity?

Mitochondrial dynamics significantly impact MAVS signaling through multiple mechanisms:

  • Mitochondrial fusion: Promotes MAVS aggregation by increasing proximity of MAVS molecules

  • Mitochondrial fission: Generally inhibits MAVS signaling by dispersing signaling platforms

  • Membrane potential: Affects the conformation and accessibility of MAVS

  • ROS production: Modulates the redox environment, influencing MAVS activation threshold

Methodological approaches to study these relationships include:

  • Pharmacological manipulation of mitochondrial dynamics (e.g., mdivi-1 for fission inhibition)

  • Genetic manipulation of fusion/fission machinery (Mfn1/2, Drp1)

  • Live-cell imaging with mitochondrial and MAVS fluorescent reporters

  • Simultaneous monitoring of membrane potential, ROS, and MAVS activation

How do viruses evade or antagonize MAVS-dependent immunity?

RNA viruses have evolved diverse strategies to counteract MAVS-mediated antiviral signaling:

Virus FamilyEvasion StrategyViral Proteins InvolvedResearch Methods
FlaviviridaeProteolytic cleavage of MAVSNS3/4A proteaseCleavage-resistant MAVS mutants
CoronaviridaeRedistribution of MAVSVarious proteinsSubcellular fractionation, microscopy
ParamyxoviridaeInhibition of MAVS aggregationV proteinsAggregation assays, binding studies
PicornaviridaeDegradation of MAVS3C proteaseProtein stability assays

To study viral evasion mechanisms:

  • Express individual viral proteins to identify the specific antagonist

  • Create viral mutants lacking the antagonistic function

  • Design MAVS variants resistant to viral interference

  • Perform structure-function analyses of MAVS-viral protein interactions

Understanding viral evasion strategies provides insights into both viral pathogenesis and the critical nodes of the MAVS pathway.

What is the experimental evidence linking MAVS dysfunction to human diseases?

MAVS dysfunction has been implicated in several human pathologies beyond viral susceptibility:

  • Autoimmune disorders: Excessive MAVS activation contributes to type I interferonopathies

  • Inflammatory diseases: Aberrant MAVS signaling amplifies inflammatory responses

  • Metabolic disorders: MAVS influences mitochondrial metabolism and insulin signaling

  • Neurodegenerative conditions: MAVS-mediated inflammation may contribute to neurodegeneration

Research methodologies to establish these connections include:

  • Patient-derived samples with MAVS sequencing

  • Functional testing of patient MAVS variants

  • Mouse models with tissue-specific MAVS manipulation

  • Integration of genome-wide association studies with functional validation

When investigating MAVS in disease contexts, researchers should distinguish between primary causative roles and secondary effects due to altered immune responses or mitochondrial function.

Product Science Overview

Introduction

Mitochondrial Antiviral Signaling Protein (MAVS), also known as CARDIF, IPS-1, and VISA, is a crucial component of the innate immune response against viral infections. MAVS is located on the outer membrane of mitochondria, peroxisomes, and the mitochondrial-associated endoplasmic reticulum membrane (MAM). It plays a pivotal role in the detection and response to viral pathogens, particularly RNA viruses .

Structure and Function

MAVS is a protein encoded by the MAVS gene, which is located on chromosome 20 in humans. The protein contains a caspase activation and recruitment domain (CARD) that is essential for its function in antiviral signaling. Upon viral infection, MAVS acts as a central hub for signal transduction initiated by RIG-I-like receptors (RLRs), which predominantly recognize viral RNA .

Mechanism of Action

When a virus infects a cell, viral RNA is detected by RLRs such as RIG-I and MDA5. These receptors then interact with MAVS through their CARD domains, leading to the formation of prion-like aggregates of MAVS. This aggregation is crucial for the activation of downstream signaling pathways that result in the production of type I interferons and other antiviral cytokines .

Regulation

The expression and function of MAVS are tightly regulated by various post-transcriptional and post-translational mechanisms, including ubiquitination and phosphorylation. These modifications can either enhance or inhibit MAVS activity, thereby modulating the antiviral response. Viruses have evolved strategies to interfere with MAVS signaling, allowing them to evade the host immune response .

Clinical Significance

MAVS is essential for the host’s defense against a wide range of RNA viruses, including influenza, hepatitis C, and SARS-CoV-2. Dysregulation of MAVS signaling can lead to impaired immune responses and increased susceptibility to viral infections. Understanding the mechanisms of MAVS regulation and function can provide insights into the development of novel antiviral therapies .

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.