SVIP Human

Small VCP/P97-Interacting Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Functions

SVIP Human regulates cellular processes through its interaction with Valosin-Containing Protein (VCP/p97), an AAA+ ATPase. Key mechanisms include:

  • Lysosomal Stability: SVIP recruits VCP to lysosomes, maintaining tubular lysosomal networks essential for autophagosome-lysosome fusion .

  • ERAD Inhibition: SVIP competes with adaptors like p47 and Ufd1p for VCP binding, modulating ER-associated protein degradation .

  • Autophagy Regulation: Overexpression enhances autophagic flux by sequestering polyubiquitinated proteins and promoting lysosomal biogenesis .

Neurodegenerative Diseases

  • Drosophila models show SVIP loss causes muscle wasting, neuromuscular degeneration, and reduced lifespan .

  • A human SVIP mutation linked to fronto-temporal dementia (FTD) induces pathogenic effects in Drosophila, including motor dysfunction .

Cancer

  • SVIP is downregulated in metastatic breast cancer tissues, correlating with poor prognosis .

  • In vitro studies reveal SVIP suppresses proliferation and migration in HER2-positive breast cancer cells .

Key Research Findings

  1. VCP Recruitment Mechanism: SVIP binds VCP’s ND1 domain via arginine residues (R24, R25), a interaction disrupted by disease-causing VCP mutations (e.g., D32A) .

  2. Lysosomal Dysfunction: SVIP knockout in Drosophila disrupts lysosomal tubules, impairing autophagy and causing age-dependent motor deficits .

  3. Therapeutic Potential: Muscle-specific SVIP overexpression extends lifespan in stress-dependent contexts, suggesting pathways for treating VCP-associated diseases .

Future Directions

Current research focuses on:

  • Developing SVIP-based therapies for ALS, Parkinson’s disease, and muscular dystrophy.

  • Exploring SVIP’s role in cancer metastasis and chemoresistance .

SVIP Human exemplifies a molecular linchpin bridging lysosomal biology, protein quality control, and disease mechanisms. Its study offers translational insights for neurodegeneration and oncology.

Product Specs

Introduction
The small VCP/p97-interacting protein (SVIP) is a regulatory factor in several cellular pathways, such as membrane fusion and ubiquitin-dependent protein degradation. Notably, SVIP acts as an inhibitor of the endoplasmic reticulum-associated degradation (ERAD) pathway. Conversely, SVIP overexpression leads to increased levels of the p62 protein, which enhances starvation-activated autophagy and promotes the sequestration of polyubiquitinated proteins and p62 within autophagosomes.
Description
Recombinant human SVIP, expressed in E. coli, is a non-glycosylated polypeptide chain containing 100 amino acids (residues 1-77) with a molecular weight of 10.8 kDa. This protein is purified using proprietary chromatographic techniques and features a 23 amino acid His-tag fused to the N-terminus.
Physical Appearance
A clear solution, sterilized by filtration.
Formulation
The SVIP solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.15 M NaCl, 20% glycerol, and 2 mM DTT.
Stability
For short-term storage (2-4 weeks), keep at 4°C. For extended storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Repeated freezing and thawing should be avoided.
Purity
Purity is determined to be greater than 85.0% by SDS-PAGE analysis.
Synonyms
Small VCP/P97-Interacting Protein, SVIP.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMGLCFPC PGESAPPTPD LEEKRAKLAE AAERRQKEAA SRGILDVQSV QEKRKKKEKI EKQIATSGPP PEGGLRWTVS.

Q&A

What is SVIP and what are its primary cellular functions?

SVIP (Small VCP Interacting Protein) is a co-factor that specifically recruits VCP (Valosin-Containing Protein) to lysosomes. It plays an essential role in maintaining lysosomal dynamic stability and facilitating autophagosomal-lysosomal fusion processes. SVIP was initially identified as a VCP-interacting protein and a negative regulator of ERAD (Endoplasmic Reticulum-Associated Degradation) .

The primary functions of SVIP include:

  • Recruitment of VCP to lysosomes

  • Maintenance of lysosomal dynamic stability

  • Facilitation of autophagosomal-lysosomal fusion

  • Regulation of tubular lysosome formation

  • Enhancement of autophagic activity

Methodologically, researchers have characterized SVIP using genetic knockout models, fluorescent protein tagging approaches, and transgenic expression systems across various model organisms including Drosophila and C. elegans .

How does human SVIP impact autophagy and healthspan in model organisms?

Human SVIP (hSVIP) expression in model organisms has demonstrated significant effects on autophagy and healthspan through the following mechanisms:

Methodologically, researchers assess these impacts through:

  • Fluorescent tagging of lysosomal membrane proteins (e.g., SPIN-1 tagged with mCherry)

  • Quantitative mobility assays in aged animals

  • Comparative lifespan analysis between wild-type and transgenic animals

  • Side-by-side comparison with orthologs from other species

These findings suggest SVIP as a potential therapeutic target for promoting healthy aging through enhanced lysosomal function and autophagy .

What experimental approaches are recommended for generating SVIP transgenic models?

Generating SVIP transgenic models requires specific methodological considerations:

For C. elegans models:

  • Gene optimization: The human SVIP coding sequence should be codon-optimized for expression in C. elegans using tools such as the C. elegans Codon Adaptor .

  • Construct design: A typical construct includes:

    • Tissue-specific promoter (e.g., ges-1 for gut expression)

    • Codon-optimized SVIP coding sequence

    • Appropriate 3' UTR (e.g., unc-54 UTR)

  • Transgene delivery: Microinjection into the gonad of adult hermaphrodites at approximately 25 ng/μl concentration, either individually or in combination with other constructs .

  • Integration and validation: Integration using UV irradiation followed by >5 generations of outcrossing to maintain genetic integrity. Validation through fluorescence microscopy, phenotypic analysis, or Western blotting .

For CRISPR-based mutations:

  • gRNA design: Target specific exons, such as the first exon of SVIP, to generate frame-shift mutations and premature stop codons .

  • Mutation verification: Sequence validation followed by protein expression analysis using specific antibodies to confirm protein disruption .

This methodological framework has been successfully employed to generate transgenic strains expressing human SVIP and to study its functional effects in vivo .

What are the connections between SVIP dysfunction and human degenerative diseases?

SVIP dysfunction has been linked to various degenerative conditions through several mechanistic pathways:

  • Lysosomal network disruption: Loss of SVIP disrupts the muscle lysosomal network, impairs autophagosome-lysosome fusion, and causes degenerative defects including muscle wasting and neuromuscular degeneration .

  • VCP-related disease mechanisms: SVIP is functionally connected to VCP-dependent diseases, as evidenced by:

    • A disease-causing VCP mutation that prevents SVIP binding

    • Deleterious effects of SVIP mutations that prevent VCP binding

    • Identification of a human SVIP mutation in a patient with fronto-temporal dementia (FTD)

  • Neurodegenerative implications: SVIP mutations or dysfunction contribute to:

    • Progressive motor dysfunction

    • Dendrite degeneration in motoneurons

    • Decreased organismal lifespan

  • Model for VCP disease: Research has established a model for VCP disease based on the differential, co-factor-dependent recruitment of VCP to intracellular organelles, highlighting lysosome dysfunction as an underlying cause for VCP-linked diseases of the peripheral and central nervous systems .

These connections suggest that targeting SVIP function could potentially offer therapeutic approaches for treating VCP-related degenerative diseases by enhancing lysosomal stability and autophagic function .

How should researchers design experiments to investigate SVIP's role in lysosomal tubulation?

Investigating SVIP's role in lysosomal tubulation requires specialized experimental approaches:

Imaging methodologies:

  • Live cell imaging: As muscle lysosomes do not retain their dynamic tubular structure following fixation, researchers should employ live imaging techniques using fluorescently tagged proteins .

  • Dual-labeling approaches: Generate transgenic animals expressing:

    • Fluorescently tagged lysosomal markers (e.g., SPIN-1-mCherry)

    • Tagged SVIP protein or endogenously tagged VCP (e.g., VCP-sfGFP)

  • Exposure optimization: Adjust exposure times for different fluorescent proteins to enable visualization of potentially weak signals at lysosomes .

Experimental manipulations:

  • Genetic perturbations:

    • SVIP overexpression using tissue-specific promoters

    • SVIP knockout through CRISPR-Cas9 genome editing

    • Expression of mutant SVIP variants lacking VCP-binding capacity

  • Nutrient conditions:

    • Compare fed versus food-limited conditions, as tubular lysosomes are normally only stimulated under food limitation in young adult C. elegans

    • Assess whether SVIP expression induces tubular lysosomes constitutively regardless of nutritional status

  • Tissue specificity analysis:

    • Target SVIP expression to specific tissues (e.g., gut, muscle) to determine tissue-specific effects on lysosomal tubulation

These approaches have revealed that SVIP expression is sufficient to induce tubular lysosomes constitutively in C. elegans gut, suggesting it as a key molecular determinant of lysosomal tubulation with implications for cellular adaptation to stress and aging .

What methodological approaches can be used to study the SVIP-VCP interaction in human cells?

Studying the SVIP-VCP interaction requires multiple complementary approaches:

Biochemical methods:

  • Co-immunoprecipitation: To assess physical interactions between SVIP and VCP under various conditions or with different mutations .

  • Domain mapping: Identify critical interaction domains, particularly focusing on the VCP-interacting motif (VIM) in SVIP .

  • Mutation analysis: Test specific mutations, such as those that prevent VCP binding, to establish structure-function relationships .

Imaging approaches:

  • Subcellular localization: Determine whether SVIP alters VCP distribution within cells, particularly to "peri-nuclear granules" and plasma membrane as previously observed in HeLa cells .

  • Co-localization studies: Use fluorescently tagged proteins to visualize:

    • Endogenous VCP protein concentration on nuclear membranes

    • VCP distribution throughout muscle cells

    • VCP concentration at tubular lysosomes

  • Differential exposure imaging: Extend exposure times for VCP-tagged fluorescent proteins to enable visualization of lysosome-associated VCP that might otherwise be obscured by stronger nuclear membrane signals .

Functional assessments:

  • Lysosomal recruitment: Quantify VCP recruitment to lysosomes under normal conditions versus with SVIP overexpression .

  • Pathogenic mutation testing: Assess how disease-associated mutations in either VCP or SVIP affect their interaction and subsequent cellular functions .

These methodological approaches have revealed that endogenous VCP protein concentrates on nuclear membranes in muscle and is weakly distributed throughout muscle cells, with significant concentration at tubular lysosomes when visualized with appropriate imaging parameters .

How can researchers analyze SVIP expression patterns across different cancer types?

Analyzing SVIP expression patterns in cancer contexts requires specific methodological considerations:

Database utilization:

  • TNMplot database: Access the TNMplot database (http://www.tnmplot.com) to analyze SVIP gene expression levels across normal, tumor, and metastatic tissues .

  • All of Us Research Program data: Leverage the extensive genomic and phenotypic data available through the All of Us Researcher Workbench, which includes data from ~250,000 participants with whole genome sequence data .

Analysis approaches:

  • Comparative expression analysis: Compare SVIP expression levels between:

    • Normal versus tumor tissues

    • Primary tumors versus metastases

    • Different molecular subtypes of the same cancer

  • Association studies: Utilize genome-wide association studies (GWAS) and rare variant association studies (RVAS) to identify potential associations between SVIP genetic variants and cancer phenotypes .

  • Cell line studies: Investigate differential expression and function of SVIP across various cancer cell lines, particularly breast cancer cell lines as highlighted in recent research .

Integration of multi-omic data:

  • Correlation with clinical outcomes: Associate SVIP expression levels with patient survival, treatment response, or other clinical parameters.

  • Pathway analysis: Place SVIP in the context of known cancer-related pathways, particularly those involving autophagy and lysosomal function.

  • Protein-level validation: Confirm RNA-level findings with protein expression analysis using immunohistochemistry or Western blotting.

These approaches can reveal cancer-specific patterns of SVIP expression and potential functional implications, as demonstrated by recent studies showing differential expression in breast cancer cell lines .

What factors should researchers consider when studying SVIP mutations in human patients?

When investigating SVIP mutations in human patients, researchers should consider several critical factors:

Genetic analysis considerations:

  • Mutation identification: Sequence the SVIP gene (located on Chromosome 11) in patient cohorts with relevant phenotypes, particularly neurodegenerative conditions like frontotemporal dementia which has been associated with SVIP mutations .

  • Variant classification: Distinguish between:

    • Pathogenic mutations

    • Likely pathogenic variants

    • Variants of uncertain significance

    • Benign polymorphisms

  • Domain-specific impacts: Prioritize variants affecting functional domains, particularly those impacting the VCP-interacting motif (VIM) .

Functional validation approaches:

  • Model system validation: Confirm pathogenicity of identified human mutations using model organisms like Drosophila, as has been successfully demonstrated for an SVIP mutation found in a frontotemporal dementia patient .

  • Binding assays: Determine whether the mutation affects SVIP's ability to bind VCP using biochemical approaches .

  • Cellular phenotypes: Assess effects on:

    • Lysosomal morphology and distribution

    • Autophagosome-lysosome fusion

    • VCP recruitment to subcellular compartments

Clinical correlation methods:

  • Phenotype documentation: Carefully document clinical presentations, particularly:

    • Muscle wasting

    • Neuromuscular degeneration

    • Motor function abnormalities

    • Cognitive changes

  • Family studies: When possible, conduct segregation analysis in families to strengthen evidence for pathogenicity.

  • Population frequency: Consider the frequency of identified variants in population databases to distinguish rare pathogenic mutations from common polymorphisms.

This multifaceted approach has successfully identified SVIP mutations in human patients and validated their pathogenicity through model organism studies, establishing SVIP's relevance to human degenerative conditions .

How can researchers quantitatively assess the impact of SVIP on autophagic activity?

Quantitative assessment of SVIP's impact on autophagic activity requires rigorous methodological approaches:

Direct measurement techniques:

  • Autophagy reporter systems: Utilize fluorescent reporters such as GFP-tagged LC3 to visualize and quantify autophagosome formation .

  • Autophagy flux assays: Measure the rate of autophagosome formation and degradation, which provides more functional information than static autophagosome counts.

  • Western blot analysis: Quantify changes in autophagy markers such as LC3-II/LC3-I ratio and p62/SQSTM1 levels in response to SVIP manipulation .

Comparative analysis frameworks:

  • Control-normalized quantification: Compare autophagic activity between:

    • Wild-type animals

    • SVIP knockout models

    • Human SVIP transgenic animals

    • Drosophila SVIP transgenic animals

  • Time-course studies: Assess changes in autophagic activity across different age points to determine whether SVIP-mediated effects are sustained throughout lifespan.

  • Tissue-specific analysis: Quantify autophagic activity in specific tissues where SVIP is expressed, such as gut tissue in C. elegans models .

Research findings:

Recent studies have demonstrated that while human SVIP overexpression heightens autophagic activity in C. elegans, it does not increase autophagy to the same extent as Drosophila SVIP. Interestingly, despite this difference in autophagic stimulation, both orthologs produced similar physiological benefits, suggesting that maximum autophagic activity may not be required to achieve optimal physiological effects .

These quantitative approaches provide crucial insights into the mechanistic basis of SVIP's impact on cellular homeostasis and healthy aging through modulation of autophagic processes.

What are the optimal experimental designs for studying SVIP's role in healthy aging?

Designing experiments to investigate SVIP's role in healthy aging requires careful consideration of multiple factors:

Model selection and preparation:

  • Transgenic models: Generate transgenic organisms expressing:

    • Wild-type SVIP

    • Mutant SVIP variants

    • Tissue-specific SVIP expression

  • Model organism selection: C. elegans offers particular advantages for aging studies due to its short lifespan and well-characterized aging phenotypes .

  • Genetic background control: Perform multiple generations of outcrossing (>5 generations recommended) after transgene integration to maintain genetic background integrity .

Experimental parameters:

  • Lifespan assessment: Track survival of multiple cohorts of transgenic versus control animals under standardized conditions .

  • Healthspan metrics: Measure physiological parameters including:

    • Late-age mobility (particularly strong proxy for healthspan in C. elegans)

    • Stress resistance

    • Reproductive capacity

    • Tissue integrity maintenance

  • Intervention timing: Express SVIP during specific life stages to determine critical periods for intervention.

  • Measurement timepoints: Perform measurements at multiple age points to capture the full trajectory of age-related changes.

Data analysis frameworks:

  • Healthspan/lifespan gap analysis: Quantify the relationship between healthspan extension and lifespan effects to determine whether SVIP promotes compression of morbidity .

  • Comparative ortholog assessment: Compare effects of human SVIP with orthologs from other species to identify conserved mechanisms .

  • Dose-response relationships: Evaluate whether different expression levels of SVIP produce proportional or threshold effects on aging parameters.

Research has demonstrated that expression of human SVIP in C. elegans improves healthspan with minimal effects on lifespan, similar to the effects observed with Drosophila SVIP expression. This suggests that SVIP primarily promotes healthy aging rather than simply extending lifespan, potentially through its effects on lysosomal function and autophagy .

Product Science Overview

Introduction

Small VCP/P97-Interacting Protein (SVIP) is a crucial adaptor protein localized on the cytosolic surface of the endoplasmic reticulum (ER) membrane. It plays a significant role in the endoplasmic reticulum-associated degradation (ERAD) pathway by inhibiting the activity of p97/VCP, a protein involved in various cellular processes such as cell cycle regulation, post-mitotic membrane fusion, and ubiquitination of misfolded proteins .

Structure and Localization

SVIP is characterized by its ability to directly interact with p97/VCP. It is predominantly found on the cytosolic surface of the ER membrane, where it exerts its inhibitory effects on p97/VCP. This interaction is crucial for maintaining the balance of protein degradation within the ER, ensuring that misfolded proteins are properly managed .

Biological Functions

SVIP has been identified as an androgen-responsive gene, indicating its regulation by androgens. It plays a pivotal role in the ERAD pathway, which is responsible for the degradation of misfolded proteins within the ER. By inhibiting p97/VCP, SVIP helps regulate the degradation process, preventing the accumulation of misfolded proteins that could otherwise lead to cellular stress and dysfunction .

Role in Steroidogenesis

Recent studies have highlighted the involvement of SVIP in steroidogenesis, particularly in Leydig cells. Leydig cells are responsible for the production of testosterone, a process that involves several key proteins such as StAR, CYP17A1, and 3β-HSD. Research has shown that SVIP, along with p97/VCP, colocalizes with these proteins in Leydig cells, suggesting a regulatory role in steroidogenesis .

Experimental Findings

Experimental studies using Leydig cell lines (HLC, TM3, and MA-10) have demonstrated that the suppression of SVIP and p97/VCP using siRNAs leads to a decrease in the expression of StAR, CYP17A1, and 3β-HSD. This suppression also results in reduced testosterone levels, indicating that SVIP and p97/VCP are essential for the proper synthesis of testosterone. Additionally, it was observed that SVIP expression increases in response to hCG stimulation, further supporting its role in steroidogenesis .

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.