SERPINH1 Human

Heat Shock 47kDa Human Recombinant
Shipped with Ice Packs
In Stock

Description

Recombinant Human HSP47 produced in E.Coli is a single, non-glycosylated polypeptide chain containing 439 amino acids (18-418 a.a.) and having a molecular mass of 48.9 kDa. HSP47 human recombinant is fused to a 38 amino acid His Tag at N-terminus and purified by convential chromatogrpahy techniques.

Product Specs

Introduction
SERPINH1, a member of the serpin superfamily, acts as a serine proteinase inhibitor. Its expression is induced by heat shock. This protein resides in the endoplasmic reticulum lumen where it functions as a molecular chaperone for collagen. Its role involves facilitating the folding and assembly of procollagen, retaining unfolded molecules within the ER, and guiding correctly folded molecules from the ER to the Golgi apparatus. Autoantibodies targeting HSP47 have been identified in individuals with rheumatoid arthritis. Notably, SERPINH1 exhibits specific binding to collagen, underscoring its chaperone function in collagen biosynthesis.
Description
Recombinant Human HSP47, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 439 amino acids (residues 18-418). With a molecular weight of 48.9 kDa, this recombinant protein is fused to a 38 amino acid His Tag at its N-terminus and purified using standard chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The SERPINH1 protein solution is provided at a concentration of 1mg/ml in a buffer consisting of 20mM Tris-HCl (pH 8.0) and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the protein should be kept at 4°C. For extended storage, it is recommended to freeze the protein at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. To maintain protein integrity, repeated freeze-thaw cycles should be avoided.
Purity
The purity of the protein is determined to be greater than 90.0% using SDS-PAGE analysis.
Synonyms
HSP47, HSP-47, Colligin-1, CBP1, Collagen Binding Protein-1, Serpin Peptidase Inhibitor Clade-H memebr 1, Serpin H1, Collagen-binding protein, Colligin, 47 kDa heat shock protein, Rheumatoid arthritis-related antigen RA-A47, Arsenic-transactivated protein 3, AsTP3, Cell proliferation-inducing gene 14 protein, SERPINH1, CBP2, gp46, PIG14, PPROM, RA-A47, SERPINH2.
Source
Escherichia Coli.
Amino Acid Sequence

MRGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSHMAA EVKKPAAAAA PGTAEKLSPK AATLAERSAG LAFSLYQAMA KDQAVENILV SPVVVASSLG LVSLGGKATT ASQAKAVLSA EQLRDEEVHA GLGELLRSLS NSTARNVTWK LGSRLYGPSS VSFADDFVRS SKQHYNCEHS KINFRDKRSA LQSINEWAAQ TTDGKLPEVT KDVERTDGAL LVNAMFFKPH WDEKFHHKMV DNRGFMVTRS YTVGVMMMHR TGLYNYYDDE KEKLQIVEMP LAHKLSSLII LMPHHVEPLE RLEKLLTKEQ LKIWMGKMQK KAVAISLPKG VVEVTHDLQK HLAGLGLTEA IDKNKADLSR MSGKKDLYLA SVFHATAFEL DTDGNPFDQD IYGREELRSP KLFYADHPFI FLVRDTQSGS LLFIGRLVRP KGDKMRDEL.

Q&A

What is SERPINH1 and what is its primary function in human cells?

SERPINH1 (Serpin Family H Member 1), also known as HSP47, is a 47 kDa heat shock protein that functions as a collagen-specific molecular chaperone in the endoplasmic reticulum. It binds specifically to collagen and plays a critical role in the biosynthetic pathway of collagen by ensuring proper folding and preventing premature aggregation of procollagen chains .

The protein contains a signal peptide for secretion, indicating it may function in both autocrine and paracrine fashions as an angiocrine factor . SERPINH1 is particularly important during periods of increased collagen synthesis, as it prevents the accumulation of misfolded collagen that could trigger cellular stress responses. Functionally, it stabilizes the triple helical structure of procollagen during its transit through the secretory pathway.

What are the known molecular interactions of SERPINH1 with other proteins?

SERPINH1 demonstrates strong functional partnerships with multiple collagen types and collagen-processing enzymes, forming a critical network in the collagen biosynthesis pathway. According to STRING database analysis, SERPINH1 shows high confidence interactions (scores >0.85) with:

  • COL26A1 (Collagen type XXVI alpha 1 chain) - interaction score: 0.958

  • FKBP10 (Peptidyl-prolyl cis-trans isomerase) - interaction score: 0.947

  • COL1A1 (Collagen alpha-1(I) chain) - interaction score: 0.921

  • P3H1 (Prolyl 3-hydroxylase 1) - interaction score: 0.911

  • CRTAP (Cartilage-associated protein) - interaction score: 0.900

  • COL4A2 (Collagen alpha-2(IV) chain) - interaction score: 0.880

These interactions reflect SERPINH1's central role in a coordinated protein network responsible for proper collagen formation, modification, and assembly. The functional partnerships with enzymes like P3H1 and FKBP10 are particularly important as they facilitate post-translational modifications of collagen that are essential for its proper function.

How is SERPINH1 expression regulated under normal physiological conditions?

SERPINH1 expression is dynamically regulated by multiple physiological factors. Research has demonstrated that its expression is significantly affected by:

  • Age - Aging increases SERPINH1 expression in cardiac endothelial cells

  • Metabolic status - Obesity and high-fat diet upregulate SERPINH1 expression

  • Physical activity - Exercise training downregulates SERPINH1 expression

Gene overlap analysis identified SERPINH1 as one of only four genes significantly affected by all three interventions (aging, high-fat diet, and exercise), making it a critical node in age and metabolism-related cardiovascular transcriptional networks . The protein appears to be regulated as part of stress response pathways, with increased expression during conditions that promote tissue fibrosis and decreased expression during conditions that improve tissue homeostasis.

Experimental models have shown that SERPINH1 regulation occurs primarily at the transcriptional level, with the gene containing response elements for heat shock factors and other stress-responsive transcription factors that control its expression in response to cellular stress conditions.

What are the current methodologies for studying SERPINH1 protein-collagen interactions?

Investigating SERPINH1-collagen interactions requires specialized approaches that preserve the native conformational states of both molecules. Contemporary methodologies include:

  • Surface Plasmon Resonance (SPR): This real-time, label-free technique allows measurement of binding kinetics between purified SERPINH1 and various collagen types. Researchers typically immobilize collagen on a sensor chip and flow SERPINH1 solutions at varying concentrations to determine association/dissociation constants.

  • Fluorescence Resonance Energy Transfer (FRET): By tagging SERPINH1 and collagen with appropriate fluorophore pairs, researchers can monitor their interaction in live cells, revealing spatial and temporal dynamics of chaperone function.

  • Co-immunoprecipitation with domain mapping: Sequential deletion mutants of SERPINH1 can identify specific domains critical for collagen binding. This approach has identified the central region of SERPINH1 as essential for collagen recognition.

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): This technique provides detailed mapping of protein-protein interaction interfaces by measuring the protection of certain regions from deuterium exchange when complexes form.

  • Cryo-electron microscopy: Recent advances enable visualization of SERPINH1-procollagen complexes, revealing structural details of how the chaperone recognizes and stabilizes the triple-helical collagen structure.

For quantitative measurement of SERPINH1, validated ELISA approaches are available with reported coefficient of variation values of 6.1-10.9%, allowing reliable detection in human samples including cerebrospinal fluid, plasma, serum, tissue lysates, and cell extracts .

How can researchers differentiate between the direct effects of SERPINH1 and secondary effects mediated through collagen regulation?

Differentiating primary from secondary effects of SERPINH1 represents a significant challenge in experimental design. Methodological approaches to address this include:

When designing such experiments, researchers should consider that even direct SERPINH1 functions ultimately affect collagen homeostasis, requiring careful interpretation of results within appropriate temporal and mechanistic frameworks.

What are the current approaches for studying SERPINH1 post-translational modifications and their functional significance?

SERPINH1 undergoes several post-translational modifications (PTMs) that regulate its chaperone activity, localization, and stability. Current methodological approaches to study these modifications include:

  • Mass spectrometry-based PTM mapping: Liquid chromatography-tandem mass spectrometry (LC-MS/MS) with enrichment strategies for specific modifications (phosphorylation, glycosylation) allows comprehensive identification of SERPINH1 PTM sites.

  • Site-directed mutagenesis: Systematic mutation of identified or predicted modification sites to non-modifiable residues (e.g., serine to alanine for phosphorylation sites) helps establish functional relevance in cellular models.

  • Modification-specific antibodies: Phospho-specific or other modification-specific antibodies enable tracking of SERPINH1 modification status under different physiological conditions or disease states.

  • Pharmacological modulation: Using kinase inhibitors, phosphatase inhibitors, or other enzymes that regulate specific PTMs helps determine the upstream regulatory pathways controlling SERPINH1 function.

  • Proximity labeling proteomics: BioID or APEX2 fusions with SERPINH1 identify proteins that interact with the chaperone in a modification-dependent manner, revealing how PTMs reshape the SERPINH1 interactome.

Interpretation of PTM studies should consider that SERPINH1 function occurs within the context of the endoplasmic reticulum environment, where redox conditions and calcium levels may influence modification patterns. Additionally, researchers should be aware that some PTMs may be species-specific, necessitating validation across multiple model systems before extrapolating to human biology.

What is the evidence for SERPINH1's role in cardiovascular disease progression?

SERPINH1 has emerged as a critical mediator in cardiovascular disease (CVD) pathogenesis through several mechanistic pathways. Extensive research has established that:

  • Endothelial cell transcriptomic remodeling: Cardiovascular risk factors including aging and obesity significantly increase SERPINH1 expression in cardiac endothelial cells, while exercise training represses its expression. This positions SERPINH1 as a molecular integrator of CVD risk factors at the transcriptomic level .

  • Induction of mesenchymal properties: Mechanistic studies demonstrate that increased SERPINH1 in human endothelial cells induces mesenchymal properties, promoting a phenotypic switch associated with endothelial dysfunction. Conversely, silencing SERPINH1 inhibits collagen deposition, suggesting its direct role in vascular fibrosis .

  • Stress, senescence and fibrosis pathway activation: All cardiovascular risk factors studied (age, weight, inactivity, heart defects) caused endothelial cells to activate mechanisms leading to stress, senescence, and fibrosis, with SERPINH1 serving as a key mediator in these pathways .

  • Conservation across species: The regulatory patterns of SERPINH1 in response to cardiovascular risk factors are conserved between mouse models and human cells, strengthening the translational relevance of these findings .

According to the World Health Organization, cardiovascular disease accounts for 10% of the global disease burden, and SERPINH1 represents a promising therapeutic target at the intersection of multiple risk factor pathways. The molecular evidence positions SERPINH1 inhibition as a potential strategy for mitigating the adverse effects of cardiovascular risk factors on vasculature.

How does SERPINH1 contribute to cancer progression and immune evasion?

SERPINH1 demonstrates significant oncogenic potential across multiple tumor types, with comprehensive pan-cancer analysis revealing several key mechanisms through which it promotes cancer progression:

  • Prognostic significance: SERPINH1 overexpression is associated with worse survival status across numerous cancer types, functioning as an independent risk factor and predictor of poor prognosis .

  • Correlation with disease progression: High SERPINH1 expression positively correlates with advanced tumor stage, suggesting its involvement in cancer progression rather than just initiation .

  • Tumor microenvironment modulation: SERPINH1 plays a critical role in shaping the tumor microenvironment and regulating immune responses. Research has established strong correlations between SERPINH1 expression and immune cell infiltration profiles .

  • Immune checkpoint pathway interaction: Analysis of TCGA samples demonstrates that SERPINH1 expression correlates significantly with immune checkpoints, chemokines, and immune regulatory molecules, suggesting it may contribute to immune evasion mechanisms .

  • Immunosuppressive potential: High SERPINH1 expression appears to contribute to tumor immune-suppressive status, potentially limiting anti-tumor immune responses and creating a permissive environment for cancer growth .

These findings position SERPINH1 as not merely a prognostic biomarker but as a functional contributor to cancer biology with particular relevance to cancer immunology. The dual role of SERPINH1 in both structural aspects of the tumor microenvironment (via collagen regulation) and immunomodulation makes it a particularly interesting target for combination therapies that might address both tumor architecture and immune activation.

What methodologies are most effective for studying SERPINH1 in rare genetic disorders like Osteogenesis Imperfecta?

Investigating SERPINH1 in rare genetic disorders like Osteogenesis Imperfecta Type X requires specialized approaches that overcome the challenges of limited patient samples and complex genotype-phenotype relationships:

  • Patient-derived iPSC models: Generating induced pluripotent stem cells from patient samples with SERPINH1 mutations allows creation of disease-relevant cell types (osteoblasts, fibroblasts) that preserve the genetic background. This approach enables detailed study of collagen processing defects in a patient-specific context.

  • CRISPR-engineered isogenic cell lines: Creating isogenic cell lines differing only in SERPINH1 mutation status eliminates confounding genetic background effects, allowing precise attribution of phenotypic changes to specific mutations.

  • Mouse models with humanized SERPINH1: Knock-in mouse models containing human SERPINH1 variants provide in vivo systems to study how mutations affect development, bone mineralization, and response to potential therapeutics.

  • Secretome analysis: Quantitative proteomics of the cellular secretome from patient-derived cells reveals how SERPINH1 mutations affect not only collagen processing but also the broader extracellular matrix composition.

  • High-content imaging of collagen assembly: Advanced microscopy with quantitative image analysis allows visualization of defects in collagen fibril formation and organization in 3D cellular models.

  • Thermal stability assays: Differential scanning fluorimetry and related techniques can assess how SERPINH1 mutations affect the thermal stability of both the chaperone itself and its collagen substrates, providing mechanistic insights into disease pathogenesis.

When designing such studies, researchers should consider the highly tissue-specific manifestations of SERPINH1 deficiency and incorporate analysis of tissue mechanical properties alongside molecular and cellular characterization. Collaboration with clinical specialists is essential for correlating in vitro findings with patient phenotypes to establish clinical relevance.

How do environmental and physiological stressors modulate SERPINH1 expression and function?

SERPINH1 responds dynamically to various environmental and physiological stressors through complex regulatory mechanisms that affect both its expression and molecular function:

  • Metabolic regulation: Obesity and high-fat diet significantly upregulate SERPINH1 expression in cardiac endothelial cells, suggesting sensitivity to metabolic stress and potential involvement in obesity-related cardiovascular complications. Gene overlap analysis identified SERPINH1 as one of only four genes significantly affected by multiple metabolic interventions .

  • Age-related modulation: Aging consistently increases SERPINH1 expression in cardiac tissues, positioning it as a potential mediator of age-related cardiovascular decline. This upregulation appears to be part of a broader reprogramming of endothelial cells toward stress, senescence, and fibrotic phenotypes with advancing age .

  • Exercise-mediated suppression: Physical exercise training downregulates SERPINH1 expression, representing a molecular mechanism through which exercise may counteract both age and obesity-related cardiovascular risk. This finding suggests SERPINH1 as a potential target for exercise-mimetic interventions .

  • Heat shock response: As indicated by its alternative name HSP47, SERPINH1 belongs to the heat shock protein family and responds to thermal stress through heat shock factor (HSF)-mediated transcriptional activation. This response is particularly important during conditions that might disrupt collagen folding.

  • Redox sensitivity: The function of SERPINH1 is modulated by the redox environment of the endoplasmic reticulum, with oxidative stress potentially affecting its chaperone activity through modification of critical cysteine residues.

Understanding these regulatory mechanisms provides insights into how SERPINH1 integrates diverse physiological signals into coordinated responses affecting collagen biosynthesis. The sensitivity of SERPINH1 to modifiable factors like diet and exercise makes it a particularly interesting target for lifestyle-based interventions aimed at improving collagen homeostasis in aging and metabolic disorders.

What are the key transcriptional regulators of SERPINH1 and how can they be experimentally manipulated?

SERPINH1 transcription is controlled by multiple regulatory elements and transcription factors that respond to different cellular states and stressors. Key transcriptional regulators and methodologies for their experimental manipulation include:

  • Heat Shock Factors (HSFs): As a heat shock protein, SERPINH1 contains heat shock elements (HSEs) in its promoter region that bind HSF family transcription factors. These can be experimentally manipulated through:

    • Small molecule HSF1 activators (e.g., celastrol)

    • Heat shock preconditioning protocols

    • CRISPR-mediated mutation of specific HSEs in the promoter

    • HSF1/2 siRNA knockdown or overexpression systems

  • TGF-β/SMAD pathway: SERPINH1 is responsive to TGF-β signaling, a major pro-fibrotic pathway. Experimental approaches include:

    • TGF-β receptor kinase inhibitors

    • SMAD3/4 knockdown or overexpression

    • CRISPR-mediated editing of SMAD binding elements in the SERPINH1 promoter

    • Constitutively active or dominant-negative TGF-β receptor constructs

  • Hypoxia-inducible factors (HIFs): Evidence suggests SERPINH1 responds to hypoxic conditions, which can be manipulated through:

    • Hypoxia chambers with controlled oxygen levels

    • Chemical HIF stabilizers (e.g., cobalt chloride, dimethyloxalylglycine)

    • HIF-1α knockdown or overexpression systems

    • HIF binding site mutations in the SERPINH1 promoter

  • Metabolic sensors: Given SERPINH1's response to metabolic status, manipulation approaches include:

    • AMPK activators (e.g., metformin, AICAR)

    • mTOR inhibitors (e.g., rapamycin)

    • PPARγ agonists/antagonists

    • Fasting/feeding experimental paradigms

  • Epigenetic regulators: SERPINH1 expression is influenced by DNA methylation and histone modifications, which can be manipulated using:

    • HDAC inhibitors (e.g., trichostatin A)

    • DNA methyltransferase inhibitors (e.g., 5-azacytidine)

    • CRISPR-dCas9 systems with epigenetic effector domains

    • ChIP-seq analysis to map regulatory element activity

When designing experiments to manipulate these pathways, researchers should consider potential cross-talk between regulatory systems and validate that observed effects on SERPINH1 are direct rather than secondary to broader cellular responses. Complementary approaches combining pharmacological, genetic, and epigenetic manipulations provide the most robust characterization of regulatory mechanisms.

What are the most reliable methods for quantifying SERPINH1 expression in human tissue and fluid samples?

Accurate quantification of SERPINH1 in human samples requires consideration of several methodological factors to ensure reliability and reproducibility. Current best practices include:

  • Enzyme-Linked Immunosorbent Assay (ELISA): Commercially validated sandwich ELISA kits demonstrate good precision with coefficient of variation values ranging from 6.1-10.9%. These assays are suitable for quantifying SERPINH1 in multiple human sample types including cerebral spinal fluid, plasma, serum, tissue lysates, and cell extracts .

  • Immunohistochemistry (IHC): For tissue localization studies, established protocols using validated antibodies allow assessment of cell-type specific expression patterns. Digital image analysis with algorithms for positivity scoring provides semi-quantitative data on expression levels and distribution.

  • Western Blotting: Quantitative western blotting with fluorescent secondary antibodies provides reliable protein quantification when normalized to appropriate housekeeping proteins. This approach is particularly valuable for distinguishing between different molecular weight forms of SERPINH1.

  • Quantitative PCR: For mRNA quantification, well-designed qPCR primers targeting conserved regions of SERPINH1 transcript show high specificity and efficiency. Reference genes should be carefully selected based on the specific tissue being studied, with validation of their stability under experimental conditions.

  • Droplet Digital PCR (ddPCR): For absolute quantification without standard curves, ddPCR provides high precision for SERPINH1 transcript quantification, particularly valuable in samples with low abundance or high inhibitor content.

  • Mass Spectrometry: For highest specificity, targeted proteomics approaches using selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) with stable isotope-labeled internal standards provide absolute quantification of SERPINH1 protein.

When selecting quantification methods, researchers should consider the specific research question, required sensitivity, sample availability, and potential confounding factors. For clinical studies, standardized sampling procedures and processing times are critical as SERPINH1 levels may be affected by sample handling conditions.

How can researchers effectively design gene knockout or knockdown experiments to study SERPINH1 function?

Designing effective genetic manipulation studies for SERPINH1 requires careful consideration of experimental approaches that account for its essential role in collagen biosynthesis. Recommended methodological strategies include:

  • Inducible knockdown systems: Doxycycline-regulated shRNA or CRISPR interference (CRISPRi) systems allow titratable and temporal control of SERPINH1 suppression, critical for studying a protein where complete knockout may cause severe phenotypes that complicate interpretation.

  • Cell type-specific manipulation: Using tissue-specific promoters to drive Cre recombinase in floxed SERPINH1 mouse models or cell type-specific CRISPR delivery systems enables investigation of SERPINH1 function in specific contexts while avoiding systemic effects.

  • Acute vs. chronic depletion strategies: Comparing rapid depletion (e.g., auxin-inducible degron systems) with gradual knockdown approaches helps distinguish direct consequences of SERPINH1 loss from adaptive responses that may mask phenotypes.

  • Rescue experiments: Complementing knockdown/knockout with expression of:

    • Wild-type SERPINH1 to confirm specificity

    • Structure-guided mutants affecting specific functions

    • SERPINH1 orthologs from other species to assess functional conservation

    • Tagged versions for localization and interaction studies

  • Domain-specific CRISPR editing: Rather than complete gene knockout, precise editing of functional domains (collagen-binding region, ER retention signal) allows dissection of specific protein functions.

  • Combinatorial manipulation: Simultaneous modulation of SERPINH1 and interacting partners or downstream effectors helps establish epistatic relationships and pathway organization.

Critical controls include:

When interpreting results, researchers should consider that complete loss of SERPINH1 may trigger unfolded protein responses or other stress pathways that could confound phenotypic analysis, necessitating careful distinction between direct and indirect effects.

What are the most informative in vivo models for studying SERPINH1 function in development and disease?

Selecting appropriate in vivo models for SERPINH1 research requires balancing physiological relevance with experimental tractability. The most informative models and their specific applications include:

  • Conditional knockout mouse models: Tissue-specific and temporally controlled Cre-loxP systems targeting SERPINH1 provide versatile platforms for studying its role in:

    • Cardiovascular development and disease (e.g., endothelial-specific deletion using Tie2-Cre)

    • Bone formation and osteogenesis imperfecta models (e.g., osteoblast-specific deletion with Osx-Cre)

    • Fibrosis models in multiple organs (e.g., inducible fibroblast deletion with Col1a2-CreERT)

    These models circumvent the embryonic lethality observed in global knockouts while enabling tissue-specific phenotypic analysis.

  • Hypomorphic/point mutation models: Knock-in mice harboring specific SERPINH1 mutations identified in human disease provide clinically relevant models that maintain some protein function while disrupting specific aspects, avoiding the confounding effects of complete protein loss.

  • Humanized mouse models: Replacement of murine SERPINH1 with human variants allows testing of human-specific functions and evaluation of therapeutics targeting human SERPINH1 in an in vivo context.

  • Reporter mouse lines: Knock-in fluorescent protein fusions or transcriptional reporters for SERPINH1 enable real-time visualization of expression dynamics during development, injury response, and disease progression.

  • Zebrafish models: Morpholino knockdown or CRISPR knockout of zebrafish SERPINH1 orthologs provides accessible systems for high-throughput phenotypic screening and live imaging of collagen dynamics during early development.

  • Transgenic overexpression models: Tissue-specific SERPINH1 overexpression models help investigate gain-of-function effects in contexts like fibrosis or tumor progression, complementing loss-of-function approaches.

When designing in vivo studies, researchers should consider:

  • The evolutionary conservation of SERPINH1 structure and regulation across species

  • Potential differences in collagen types and processing between model organisms and humans

  • The need for physiologically relevant stressors (e.g., exercise, high-fat diet) to reveal conditional phenotypes

  • Integration of molecular, cellular, and physiological endpoints to connect biochemical functions to organism-level outcomes

Combining multiple model systems provides complementary insights, with simpler organisms facilitating mechanistic discovery and mammalian models validating translational relevance.

How might SERPINH1 function beyond its role as a collagen chaperone?

While SERPINH1's primary function as a collagen chaperone is well-established, emerging evidence suggests additional roles that expand its biological significance beyond collagen biosynthesis:

  • Potential secreted signaling functions: Analysis using the MetaSecKB database reveals that SERPINH1 contains a signal peptide for secretion, suggesting it may function as an angiocrine factor in both autocrine and paracrine manners . This positions SERPINH1 as a potential extracellular signaling molecule rather than solely an intracellular chaperone.

  • Immune modulation activities: Pan-cancer analysis demonstrates that SERPINH1 expression strongly correlates with immune cell infiltration patterns, immune regulatory molecules, and checkpoint expression, suggesting functions in immune system modulation that may be independent of its collagen chaperoning activity .

  • Stress response coordination: As a heat shock protein, SERPINH1 may participate in broader cellular stress response networks beyond collagen folding, potentially interacting with other heat shock proteins to coordinate integrated stress responses.

  • RNA binding capability: Gene Ontology annotations include RNA binding activity for SERPINH1, suggesting potential roles in RNA metabolism or post-transcriptional regulation that remain largely unexplored .

  • Potential intracellular signaling roles: The correlation between SERPINH1 expression and activation of specific cellular pathways in endothelial cells suggests it may function as a signaling node that integrates metabolic and stress signals into coordinated cellular responses .

Investigating these non-canonical functions requires experimental approaches that can distinguish collagen-dependent from collagen-independent activities. Promising strategies include structure-function studies with SERPINH1 mutants specifically deficient in collagen binding, proximity labeling approaches to identify the complete SERPINH1 interactome, and functional genomics screens in cellular contexts with minimal collagen expression.

What are the latest approaches for targeting SERPINH1 therapeutically in disease contexts?

SERPINH1 has emerged as a promising therapeutic target in multiple disease contexts, with several innovative targeting strategies in development:

  • Small molecule inhibitors: Structure-based drug design has yielded compounds that specifically bind the collagen-interaction pocket of SERPINH1, disrupting its chaperone function without affecting protein stability. These molecules show promise in preclinical fibrosis models and represent the most advanced approach for therapeutic intervention.

  • Aptamer-based targeting: RNA and DNA aptamers with high specificity for SERPINH1 offer an alternative to small molecules, with potential advantages in selectivity. Some aptamer candidates demonstrate the ability to modulate SERPINH1-collagen interactions without complete inhibition, potentially allowing finer control of activity.

  • Cell-specific delivery strategies: Given SERPINH1's importance in normal collagen biosynthesis, cell-targeted approaches using nanoparticle formulations, antibody-drug conjugates, or cell-penetrating peptide conjugates aim to restrict inhibition to pathological cell populations (e.g., cancer-associated fibroblasts, activated hepatic stellate cells).

  • Gene therapy approaches: For genetic disorders involving SERPINH1 mutations, AAV-mediated gene replacement or CRISPR-based gene editing strategies are being developed to restore normal SERPINH1 function in affected tissues.

  • Protein-protein interaction disruptors: Peptide mimetics designed to selectively disrupt interactions between SERPINH1 and specific collagen types offer the potential for subtype-specific modulation of collagen processing.

  • Antisense oligonucleotides (ASOs) and siRNA: Nucleic acid-based approaches targeting SERPINH1 mRNA provide highly specific knockdown capabilities with emerging tissue-targeting modifications to enhance delivery to relevant cell types.

When developing or evaluating SERPINH1-targeted therapeutics, researchers should consider:

  • The tissue-specific requirements for SERPINH1 function and potential off-target effects

  • The timing of intervention, as prevention of collagen deposition versus reversal of established fibrosis may require different strategies

  • The potential for compensatory mechanisms following SERPINH1 inhibition

  • The context-dependent role of SERPINH1 in different diseases, which may necessitate tailored targeting approaches

Preclinical evaluation should include careful assessment of effects on wound healing and tissue repair, given SERPINH1's physiological importance in these processes.

Product Science Overview

Introduction

Heat Shock Protein 47 (HSP47), also known as Serpin-H1, is a 47 kDa collagen-binding stress protein localized in the endoplasmic reticulum (ER) of collagen-secreting cells . It is a member of the heat shock protein (HSP) family, which are highly conserved proteins that play crucial roles in protein folding and protection against stress-induced damage.

Discovery and Function

Heat shock proteins were first discovered by Ferruccio Ritossa in 1962 when he observed chromosome “puffing” in Drosophila cells exposed to elevated temperatures . HSP47 specifically functions as a molecular chaperone essential for collagen biosynthesis . It binds to procollagen in the ER, ensuring proper folding and preventing premature aggregation .

Role in Collagen Biosynthesis

HSP47 is critical for the stability and secretion of collagen, a major structural protein in the extracellular matrix. It binds to the triple-helical region of procollagen, facilitating its proper folding and transport from the ER to the Golgi apparatus . This process is vital for maintaining the structural integrity of tissues such as skin, bones, and connective tissues.

Clinical Significance

HSP47 has been identified as a potential therapeutic target in various fibrotic conditions, where excessive collagen production leads to tissue scarring and organ dysfunction . Upregulation of HSP47 has been observed in collagen-producing cells in several fibrotic diseases, making it a promising candidate for drug development .

Recombinant HSP47

Recombinant human HSP47 is produced using Chinese Hamster Ovary (CHO) cells and is often tagged with a His-tag for purification purposes . This recombinant protein is used in research to study its role in collagen biosynthesis and its potential as a therapeutic target. It is also utilized in assays to enhance neurite outgrowth in neuronal cells .

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.