SERPINE1 Human

Plasminogen Activator Inhibitor-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction and Overview

SERPINE1 (Serpin Peptidase Inhibitor, Clade E Member 1), also known as Plasminogen Activator Inhibitor-1 (PAI-1), is a multifunctional glycoprotein encoded by the SERPINE1 gene on human chromosome 7q22.1. It belongs to the serine protease inhibitor (serpin) superfamily and is a key regulator of fibrinolysis, cell migration, tissue remodeling, and senescence. Elevated SERPINE1 levels are linked to thrombotic disorders, cancer progression, metabolic syndromes, and aging-related pathologies .

Protein Characteristics

  • Molecular Weight: ~45 kDa (unprocessed precursor) .

  • Domains: Reactive center loop (RCL) critical for protease inhibition; vitronectin-binding domain for extracellular matrix interactions .

  • Mechanism: Inhibits tissue-type (tPA) and urokinase-type (uPA) plasminogen activators, preventing plasminogen conversion to plasmin and stabilizing blood clots .

Key Functional Roles

FunctionMechanismBiological Impact
Fibrinolysis RegulationBinds tPA/uPA, blocking plasmin generationPrevents premature clot dissolution
Cell MigrationModulates extracellular matrix (ECM) degradation via uPA inhibitionFacilitates tissue repair and cancer metastasis
SenescenceInduced by p53/p16 pathwaysPromotes aging phenotypes and organ dysfunction

Genomic Features

  • Chromosomal Location: 7q22.1 .

  • Promoter Polymorphisms: 4G/5G allele variants influence transcription; 4G allele increases SERPINE1 expression and thrombotic risk .

Tissue Expression

TissueExpression LevelClinical Relevance
Adipose TissueHighLinked to obesity-induced insulin resistance
PlacentaVery HighAssociated with gestational complications
Brain (Pons/Medulla)ModerateImplicated in glioblastoma progression

Role in Physiological Processes

  • Hemostasis: Maintains clot stability by inhibiting fibrinolysis .

  • Tissue Remodeling: Regulates ECM degradation during wound healing .

  • Circadian Rhythm: Modulates fibrinolytic activity diurnally, peaking in morning hours .

Disease Associations

DiseaseRole of SERPINE1Evidence
Plasminogen Activator Inhibitor-1 DeficiencyLoss-of-function mutations cause bleeding diathesisHomozygous mutations impair clot stabilization
Triple-Negative Breast Cancer (TNBC)Promotes chemoresistance via Akt/ERK signalingKnockdown reduces tumor growth and enhances apoptosis
Obesity-Related RadioresistanceUpregulated in adipocyte-rich microenvironmentsMediates DNA repair via ATM/MRE11 axis
Chronic RhinosinusitisDrives tissue remodeling and fibrosis4G allele correlates with disease severity

Inhibitors in Development

CompoundMechanismClinical Stage
TiplaxtininBinds SERPINE1, blocking protease inhibitionPreclinical (resensitizes TNBC to radiotherapy)
Nicotiflorin (Calotropis gigantea)Disrupts SERPINE1-FGF2 interactionComputational docking studies

Research Advances

  • Cancer Therapy: SERPINE1 knockdown reverses paclitaxel resistance in TNBC by suppressing anti-apoptotic pathways .

  • Aging Interventions: SERPINE1 deficiency extends lifespan in Klotho-deficient mice by reducing senescence .

Product Specs

Introduction

Plasminogen activator inhibitor-1 (PAI-1) is a key regulator of fibrinolysis, the process of breaking down blood clots. It acts by inhibiting tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA), which are enzymes that convert plasminogen to plasmin. Plasmin is responsible for degrading fibrin, the main component of blood clots. PAI-1 belongs to a family of proteins called serpins (serine protease inhibitors), specifically SERPINE1. While another PAI, plasminogen activator inhibitor-2 (PAI-2), is mainly present during pregnancy, SERPINE1 is the primary inhibitor of plasminogen activators in the body.

Description

This recombinant SERPINE1 protein is produced in E. coli and designed for research purposes. It is a single, non-glycosylated polypeptide chain consisting of 400 amino acids (residues 24-402) with a molecular weight of 45 kDa. A 21 amino acid His-Tag is fused to the N-terminus to facilitate purification, which is achieved using proprietary chromatographic techniques.

Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation

The protein is supplied in a buffer containing 50mM Sodium Acetate (pH 5.5), 10% glycerol, and 0.1M Sodium Chloride.

Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. Adding a carrier protein like HSA or BSA (0.1%) is advisable for long-term storage. To maintain protein integrity, avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 95% as assessed by SDS-PAGE analysis.
Biological Activity

The protein exhibits potent inhibitory activity against uPA, as evidenced by an IC50 value of less than 3nM. This measurement is based on the protein's ability to block uPA-mediated cleavage of the substrate Z-GGRAMC.

Synonyms
PAI-1, PAI1, PLANH1, SERPINE1, PAIE, PLASMINOGEN ACTIVATOR INHIBITOR, BETA-MIGRATING ENDOTHELIAL-CELL-DERIVED TYPE.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MVHHPPSYVA HLASDFGVRV FQQVAQASKD RNVVFSPYGVASVLAMLQLT TGGETQQQIQ AAMGFKIDDK GMAPALRHLY KELMGPWNKD EISTTDAIFVQRDLKLVQGF MPHFFRLFRS TVKQVDFSEV ERARFIINDW VKTHTKGMIS NLLGKGAVDQLTRLVLVNAL YFNGQWKTPF PDSSTHRRLF HKSDGSTVSV PMMAQTNKFN YTEFTTPDGHYYDILELPYH GDTLSMFIAA PYEKEVPLSA LTNILSAQLI SHWKGNMTRL PRLLVLPKFSLETEVDLRKP LENLGMTDMF RQFQADFTSL SDQEPLHVAQ ALQKVKIEVN ESGTVASSSTAVIVSARMAP EEIIMDRPFL FVVRHNPTGT VLFMGQVMEP.

Q&A

What detection methods are most reliable for quantifying SERPINE1 in human samples?

The solid-phase sandwich ELISA (Enzyme-Linked Immunosorbent Assay) is currently the gold standard for detecting and quantifying human SERPINE1 across various sample types. Modern ELISA kits detect both active and latent forms of SERPINE1, though they typically cannot detect SERPINE1 when complexed with uPA or tPA .

Current commercially available ELISA kits offer high sensitivity (as low as 0.16 ng/mL) with detection ranges typically spanning 0.312-20 ng/mL . When selecting a detection method, researchers should consider both intra-assay and inter-assay precision metrics:

Intra-Assay Precision:

SamplenMean (ng/mL)SDCV%
1201.00.054.8
2204.80.061.3
32017.60.432.4

Inter-Assay Precision:

SamplenMean (ng/mL)SDCV%
1241.40.042.6
2245.20.132.5
32418.40.361.9

These metrics demonstrate the high reliability of current ELISA methods for SERPINE1 quantification .

Which human sample types are optimal for SERPINE1 analysis?

Multiple human sample types can be effectively analyzed for SERPINE1 content, each with specific recovery characteristics:

Sample TypeAverage Recovery (%)Range (%)
Cell culture supernatants9084-98
Cell lysates8880-95
Human serum8677-98
Human plasma8572-99

For plasma samples specifically, multiple anticoagulant types can be used (EDTA, heparin, citrate, or CTAD), with recovery rates ranging from 83-115% . The choice of sample type should be guided by the specific research question, with plasma and serum being preferred for circulating SERPINE1 levels while cell lysates may be more appropriate for tissue-specific expression studies.

How can researchers effectively validate SERPINE1 knockdown in experimental models?

Effective validation of SERPINE1 knockdown requires both molecular verification and functional assessment. For initial molecular verification, both siRNA (transient) and shRNA (stable) approaches have proven effective. When establishing stable knockdown lines, researchers should generate and screen multiple clones as knockdown efficiency can vary considerably between clones .

For functional validation, cell invasion assays provide a reliable readout of SERPINE1 activity. The tumor spheroid invasion assay represents a more physiologically relevant 3D model that effectively demonstrates the functional consequences of SERPINE1 knockdown .

Notably, researchers should be aware that while mRNA quantification of SERPINE1 is straightforward, protein detection via immunoblotting has proven technically challenging even with commercially available antibodies, often requiring multiple optimization attempts .

How does the crosstalk between YAP and TGFβ pathways regulate SERPINE1 expression?

SERPINE1 expression is regulated through complex interactions between the YAP/Hippo and TGFβ signaling pathways. Pathway enrichment analysis of SMAD4-regulated genes reveals that the Hippo pathway (P=1.4×10^-4) is the second most enriched pathway after TGFβ signaling (P=0.2×10^-4) .

This crosstalk manifests through several key mechanisms:

  • TGFβ treatment rapidly enhances CTGF expression (a YAP-dependent gene) in mesenchymal lung cancer cells.

  • While SMAD4 knockdown does not affect basal SERPINE1 expression, it significantly attenuates TGFβ-mediated SERPINE1 expression.

  • YAP knockdown significantly decreases TGFβ-mediated SERPINE1 expression.

  • Simultaneous depletion of YAP and TAZ produces an even more pronounced reduction in TGFβ-mediated SERPINE1 expression.

These findings indicate that YAP and TAZ are required for TGFβ-SMAD4-dependent SERPINE1 expression in mesenchymal cancer cells . This interplay demonstrates how converging signaling pathways can synergistically regulate SERPINE1 transcription in specific cellular contexts.

Why do different cell types exhibit differential SERPINE1 expression patterns and functional consequences?

Cell-type specific SERPINE1 expression and function stem from fundamental differences in cellular phenotype and underlying regulatory networks. For example, mesenchymal lung cancer cells (TD cells) exhibit significantly higher SERPINE1 expression compared to epithelial lung cancer cells (A549) .

These differential expression patterns translate into functional consequences - SERPINE1 knockdown significantly reduces invasiveness in mesenchymal TD cells but has minimal effect in epithelial A549 cells . This suggests that SERPINE1's functional role is context-dependent and closely linked to cellular phenotype.

The mechanistic basis for these differences includes:

  • Differential activation of transcriptional regulators (YAP pathway is more active in mesenchymal cells)

  • Enhanced responsiveness to TGFβ stimulation in mesenchymal cells

  • Synergistic pathway interactions that are more pronounced in certain cell types

These observations underscore the importance of considering cellular context when designing SERPINE1-focused experiments and interpreting research findings .

How do SERPINE1 polymorphisms influence gene expression and protein levels?

Multiple SERPINE1 polymorphisms affect gene expression and circulating protein levels through alterations in transcriptional regulation. The extensively studied -675 4G/5G polymorphism (rs1799889) has demonstrated increased SERPINE1 transcription with the 4G allele compared to the 5G allele in vitro, correlating with increased plasma PAI-1 levels in vivo .

The -844A>G polymorphism (rs2227631) has also been associated with plasma PAI-1 levels, though it explains only approximately 1.25% of PAI-1 level variability. This SNP is implicated in SERPINE1 gene regulation as part of an Ets nuclear protein consensus sequence binding site .

Haplotype analysis reveals that six different SNPs are associated with various clinical and biological phenotypes, suggesting that modifications of SERPINE1 expression are regulated through different pathways. Comprehensive haplotype analysis, rather than focusing on single polymorphisms, provides more complete insight into how genetic variation affects SERPINE1 biology .

What mechanisms underlie SERPINE1's promotion of cancer cell invasiveness?

SERPINE1 promotes cancer cell invasiveness through mechanisms that are independent of its canonical plasminogen activator (PA) inhibitory function. In mesenchymal lung cancer cells, elevated SERPINE1 expression directly correlates with enhanced cellular invasiveness, as demonstrated by both conventional invasion assays and 3D tumor spheroid models .

The mechanistic basis involves:

  • Activation of pro-invasive signaling networks, particularly through YAP pathway activation

  • Synergistic interaction with the TGFβ pathway, which is well-established in promoting epithelial-to-mesenchymal transition

  • Modification of the tumor microenvironment through PA-independent mechanisms

Importantly, knockdown experiments demonstrate that SERPINE1 is not merely a biomarker but plays a functional role in driving invasiveness. YAP knockdown significantly attenuates invasiveness in mesenchymal cancer cells, mirroring the effects of SERPINE1 knockdown, further supporting the critical role of the YAP-SERPINE1 axis in cancer cell invasion .

How can researchers design experiments to elucidate SERPINE1's role in different cancer types?

Designing robust experiments to investigate SERPINE1's role in cancer requires multi-level approaches:

  • Expression profiling: Compare SERPINE1 expression between cancer cells with different phenotypes (e.g., epithelial vs. mesenchymal) and between tumor and matched normal tissues.

  • Genetic manipulation: Implement both loss-of-function (siRNA/shRNA knockdown) and gain-of-function (overexpression) approaches across different cancer cell types to establish cell-type specific effects .

  • Functional readouts: Employ multiple functional assays including:

    • Conventional transwell invasion assays

    • 3D tumor spheroid invasion assays

    • Migration assays

    • Proliferation and viability assessments

  • Pathway analysis: Investigate interactions between SERPINE1 and key signaling pathways (YAP, TGFβ) using reporter assays, pathway inhibitors, and co-immunoprecipitation experiments .

  • Transcriptomic profiling: Implement RNA-seq to identify transcriptome signatures associated with SERPINE1 manipulation, which can inform drug-transcriptome data analysis approaches like Connectivity MAP .

This comprehensive approach can effectively distinguish between correlative and causative roles of SERPINE1 in cancer progression.

What therapeutic strategies could target SERPINE1-dependent cancer cell invasion?

Rather than directly inhibiting SERPINE1, more promising therapeutic approaches include:

  • Transcriptome-based drug repurposing: Using Connectivity MAP or similar drug-transcriptome data analysis methods to identify compounds that reverse SERPINE1-dependent invasive signatures .

  • Targeting upstream regulators: Inhibiting the YAP pathway or interfering with TGFβ-YAP crosstalk could indirectly modulate SERPINE1 expression and function.

  • Combinatorial approaches: Simultaneously targeting multiple nodes in the SERPINE1 regulatory network to overcome potential compensatory mechanisms.

Since SERPINE1's cancer-promoting effects occur largely independent of its PA-inhibitory function, traditional approaches targeting this canonical activity may prove ineffective. Instead, targeting the transcriptional networks and downstream effectors of SERPINE1 represents a more mechanistically sound strategy .

How should researchers design studies to investigate associations between SERPINE1 polymorphisms and disease risk?

Robust study design for investigating SERPINE1 polymorphism-disease associations requires several methodological considerations:

Why are there discrepancies in reported associations between SERPINE1 polymorphisms and metabolic syndrome?

Discrepancies in reported associations between SERPINE1 polymorphisms and metabolic syndrome stem from several factors:

  • Incomplete genetic characterization: Many studies focus solely on the -675 4G/5G polymorphism rather than analyzing multiple SNPs or haplotypes. Other SERPINE1 SNPs in linkage disequilibrium with the 4G/5G polymorphism may be more directly associated with metabolic syndrome components .

  • Population heterogeneity: Genetic associations often vary across different ethnic groups and populations due to differences in genetic background, linkage disequilibrium patterns, and environmental exposures.

  • Interaction with environmental factors: As demonstrated with smoking in MI risk, environmental factors may mask or modify genetic associations, leading to inconsistent findings across studies with different population characteristics .

  • Methodological differences: Variation in study design, phenotype definitions, genotyping methods, and statistical analyses contributes to discrepant results.

  • Complex regulation of SERPINE1: Different polymorphisms may influence SERPINE1 expression through distinct regulatory pathways, as suggested by the finding that six SNPs associate differentially with various clinical and biological phenotypes .

Comprehensive haplotype analysis across diverse populations with careful control for environmental factors is essential to reconcile these discrepancies.

What is the relationship between SERPINE1 polymorphisms and myocardial infarction risk?

The relationship between SERPINE1 polymorphisms and myocardial infarction (MI) risk exhibits considerable complexity:

  • The -675 4G/5G polymorphism (rs1799889) has been widely studied, with the 4G/4G genotype associated with a modest 1.2-fold increased risk of MI according to a large systematic review .

  • The effect of the 4G/5G polymorphism on MI risk is significantly influenced by the presence of underlying Metabolic Syndrome, demonstrating interaction with metabolic factors .

  • The -844A>G polymorphism (rs2227631) appears more relevant than the 4G/5G polymorphism for MI association in nonsmokers. Among nonsmokers, the frequency of haplotypes carrying the G allele was approximately three-fold lower in MI cases than in controls .

  • SERPINE1 haplotypes show mild associations with plasma PAI-1 levels, which may mediate some of the observed effects on MI risk .

  • The impact of SERPINE1 polymorphisms on MI risk is only observed in nonsmokers, suggesting that smoking's strong effect on risk overwhelms the relatively modest genetic influence .

These findings highlight the importance of considering both genetic and environmental factors when assessing MI risk associated with SERPINE1 variants.

What challenges might researchers encounter when measuring SERPINE1 protein and how can they be overcome?

Researchers face several technical challenges when measuring SERPINE1 protein:

  • Antibody specificity issues: Immunoblotting for PAI-1 has proven technically difficult even with commercially available antibodies. Multiple attempts may be needed to detect PAI-1 protein expression, particularly in certain cell types .

  • Complex formation interference: Standard ELISA kits cannot detect SERPINE1 when complexed with uPA or tPA, potentially underestimating total SERPINE1 levels in samples where such complexes are prevalent .

  • Variable recovery across sample types: Recovery rates range from 72-99% depending on sample type, requiring appropriate validation for each specific sample matrix .

To overcome these challenges:

  • Validate multiple antibodies and optimize detection protocols specifically for your experimental system

  • Consider measuring both free and complexed forms when total SERPINE1 is of interest

  • Include appropriate recovery controls for your specific sample type

  • When possible, correlate protein measurements with mRNA expression data

  • Consider activity-based assays as complementary approaches to immunological detection methods

How can data from SERPINE1 genetic studies be appropriately analyzed to control for multiple testing?

Appropriate statistical analysis of SERPINE1 genetic studies requires careful consideration of multiple testing corrections:

  • Standard Bonferroni correction is overly conservative: Due to linkage disequilibrium between SNPs, standard Bonferroni correction (dividing the significance threshold by the total number of tests) is too stringent and increases type II error risk.

  • Determine effective number of independent tests: Use methods like that proposed by Li and Ji to estimate the number of independent components underlying the LD structure. In one study, 7 independent components were identified among the SERPINE1 SNPs tested .

  • Adjust significance threshold accordingly: Once the number of independent components is established, adjust the significance threshold (e.g., p<0.007 for 7 independent components rather than p<0.0056 for 9 total SNPs) .

  • Consider stratified analyses separately: When performing analyses stratified by factors like smoking status, clearly distinguish these as separate hypothesis tests rather than additional multiple tests.

  • Apply appropriate transformations: Ensure outcome variables meet statistical assumptions by applying transformations when necessary (e.g., square-root transformation for PAI-1 and log-transformation for insulin) .

This balanced approach maintains statistical rigor while acknowledging the correlated nature of genetic variants.

What are the most reliable reference standards for SERPINE1 quantification?

Selecting appropriate reference standards is crucial for accurate SERPINE1 quantification:

  • Recombinant human SERPINE1: The Sf21-expressed recombinant human SERPINE1 has been validated for accurately quantitating both active and latent forms as well as vitronectin complexes .

  • Comparison with natural human SERPINE1: Results using natural human SERPINE1 show linear curves parallel to standard curves obtained with recombinant standards, validating their use for determining relative mass values of naturally occurring human SERPINE1 .

  • Complex detection limitations: Current standards cannot reliably quantify SERPINE1 in complexes with uPA and tPA, representing an important limitation .

The precision of these standards is well-established:

Intra-Assay Precision:

SamplenMean (ng/mL)SDCV%
1202.60.124.4
2206.50.528.0
32012.60.987.8

Inter-Assay Precision:

SamplenMean (ng/mL)SDCV%
1402.30.29.5
2406.40.46.4
34013.10.86.1

These metrics demonstrate the high reliability of current reference standards for SERPINE1 quantification .

How might computational approaches advance SERPINE1-targeted therapeutic development?

Computational approaches offer promising avenues for SERPINE1-targeted therapeutic development:

  • Drug-transcriptome data analysis: Methods like Connectivity MAP can identify compounds that reverse SERPINE1-dependent transcriptional signatures, potentially repurposing existing drugs to target cancer invasion and other SERPINE1-driven pathologies .

  • Pathway-based virtual screening: Computational screening targeting the interaction interface between YAP and SMAD4 or other key nodes in the SERPINE1 regulatory network could identify novel small molecules that modulate SERPINE1 expression.

  • Genetic network analysis: Identifying nodes where SERPINE1 regulatory networks intersect with other disease-associated pathways could reveal novel therapeutic targets with potential synergistic effects.

  • Structure-based drug design: As SERPINE1 promotes cancer invasion through PA-independent mechanisms, structure-based approaches targeting non-canonical binding interfaces could yield inhibitors specific to these pathological functions while preserving normal physiological activity .

These computational approaches, combined with experimental validation, represent a promising strategy for developing therapeutics that selectively target pathological SERPINE1 functions.

What might explain the technical difficulties in detecting SERPINE1 protein despite high mRNA expression?

The reported difficulties in detecting SERPINE1 protein despite high mRNA expression suggest several possible mechanisms:

  • Post-transcriptional regulation: MicroRNAs or RNA-binding proteins may suppress translation without affecting mRNA levels.

  • Protein instability: SERPINE1 protein may undergo rapid degradation in certain cellular contexts, limiting steady-state protein levels despite robust transcription.

  • Complex formation masking: SERPINE1 may form complexes with other proteins that mask epitopes recognized by detection antibodies.

  • Subcellular localization: Different subcellular distribution patterns might affect extraction efficiency or epitope accessibility.

  • Technical factors: Buffer compositions, extraction methods, and antibody specificity issues could contribute to detection difficulties.

Addressing this discrepancy requires comparative analysis of multiple detection methods including:

  • Alternative antibodies targeting different epitopes

  • Native versus denaturing conditions

  • Various extraction protocols

  • Activity-based assays to complement immunological detection

Resolving this technical challenge would significantly advance SERPINE1 research by enabling more reliable protein quantification.

How might the complex interplay between SERPINE1 genetic variants and environmental factors be better understood?

Understanding the complex interplay between SERPINE1 genetic variants and environmental factors requires innovative approaches:

  • Large-scale gene-environment interaction studies: Prospective studies with sufficient power to detect interaction effects, particularly focusing on factors like smoking that have demonstrated modifying effects on SERPINE1 associations .

  • Epigenetic profiling: Analysis of DNA methylation, histone modifications, and chromatin accessibility at SERPINE1 regulatory regions to understand how environmental factors might modulate genetic effects through epigenetic mechanisms.

  • Tissue-specific expression quantitative trait loci (eQTL) studies: Investigating how SERPINE1 polymorphisms affect gene expression in different tissues and how these effects are modulated by environmental exposures.

  • Functional genomics approaches: Using CRISPR-based methods to introduce specific SERPINE1 variants in cellular models and assess their functional impact under various environmental conditions.

  • Systems biology integration: Developing computational models that integrate genetic, epigenetic, transcriptomic, and environmental data to predict SERPINE1 expression and function under various conditions.

These approaches would provide mechanistic insights into how genetic and environmental factors converge to influence SERPINE1-dependent pathophysiological processes, potentially guiding more personalized preventive and therapeutic strategies.

Product Science Overview

Genetic and Molecular Structure

The gene encoding PAI-1, known as SERPINE1, is located on chromosome 7 (7q21.3-q22) in humans . PAI-1 is a single-chain glycoprotein with a molecular weight of approximately 43 kDa . It acts as a “bait” for tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA), which are key enzymes in the conversion of plasminogen to plasmin . Plasmin is the principal enzyme responsible for the degradation of fibrin, a major component of blood clots .

Function and Mechanism of Action

PAI-1 functions by inhibiting the activity of tPA and uPA, thereby preventing the formation of plasmin and subsequently inhibiting fibrinolysis . This inhibition is achieved through active site binding, where PAI-1 forms a stable complex with tPA or uPA, rendering them inactive . Additionally, PAI-1 can bind to the uPA/uPA receptor complex, leading to its degradation .

Role in Disease

Elevated levels of PAI-1 are associated with an increased risk of thrombosis and atherosclerosis, as excessive inhibition of fibrinolysis can lead to the persistence of blood clots . Conversely, congenital deficiency of PAI-1 can result in a hemorrhagic diathesis, a condition characterized by an increased tendency to bleed due to insufficient suppression of fibrinolysis .

PAI-1 has also been implicated in various other physiological and pathological processes, including wound healing, organ fibrosis, aging, autophagy, immune responses, tumor invasion, and metastasis . Elevated PAI-1 levels are often observed in conditions such as insulin resistance, metabolic syndrome, and diabetes, making it a valuable biomarker for cardiovascular health and endothelial function .

Human Recombinant PAI-1

Human recombinant PAI-1 is a laboratory-produced form of the protein that is used in various research and clinical applications. It is synthesized using recombinant DNA technology, which involves inserting the human SERPINE1 gene into a suitable expression system, such as bacteria or mammalian cells, to produce the protein in large quantities . Recombinant PAI-1 is used as a marker for acute myocardial infarction and in the diagnosis of several thrombolytic disorders . It also protects platelets against the inhibitory effects of plasma and has a role complementary to that of α2-antiplasmin .

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.