PRSS3 Human, HEK

Protease Serine 3 Human Recombinant, HEK
Shipped with Ice Packs
In Stock

Description

Physiological and Pathological Roles

Normal Functions:

  • Pancreatic Digestion: PRSS3 processes dietary proteins into absorbable peptides .

  • Neuronal Activity: Expressed in the brain, it modulates neuropeptide signaling .

Disease Associations:

ConditionRole of PRSS3Clinical RelevanceSources
Pancreatic CancerOverexpression linked to tumor growthPotential therapeutic target
Hepatocellular Carcinoma (HCC)Hypermethylation silences PRSS3, promoting metastasisPrognostic biomarker for poor differentiation
Chronic PancreatitisNo direct genetic association foundExcluded as major susceptibility gene
Lung AdenocarcinomaHigh expression correlates with poor survival (HR = 2.22)Prognostic indicator

Research Applications of PRSS3 Human, HEK

Key Findings:

  • Viral Pathogenesis: PRSS3 interacts with enterovirus A71 3A protein, facilitating viral replication in neuronal cells .

  • Cancer Mechanisms:

    • Overexpression in HCC suppresses tumor growth by downregulating cyclin D1/CDK4 and MMP2, inhibiting ERK signaling .

    • Splice variants (PRSS3-V1/V2) show bipolar expression in HCC, with V2 serving as a diagnostic marker .

Functional Assays:

Assay TypeOutcomeSignificanceSources
Colony FormationPRSS3 overexpression reduces HCC cell proliferationHighlights tumor-suppressive role
Transwell InvasionPRSS3 inhibits metastasis via MMP2 downregulationLinks protease activity to metastasis
Kinase ActivityResistant to endogenous trypsin inhibitors (e.g., SPINK1)Explains pathological dominance

Comparative Analysis of PRSS3 Isoforms

PRSS3 has four splice variants (V1–V4), with distinct roles in cancer:

VariantExpression ProfileFunctional ImpactSources
V1High in Huh7/LM3 HCC cellsPromotes cell viability and invasion
V2Reduced in HCC tissuesTumor-suppressive; diagnostic marker
V3Low/absent in most cell linesNeuronal-specific interaction with EV-A71

Clinical and Therapeutic Implications

  • Biomarker Potential: PRSS3 methylation status in HCC correlates with tumor grade and patient survival .

  • Therapeutic Target: Small-molecule inhibitors of PRSS3 could mitigate metastasis in pancreatic and lung cancers .

Product Specs

Description
PRSS3 Human Recombinant produced in HEK293 Cells is a single, glycosylated polypeptide chain containing 238 amino acids (16-247 a.a.) and having a molecular mass of 26kDa. PRSS3 is fused to a 6 amino acid His-tag at C-terminus and is purified by proprietary chromatographic techniques.
Physical Appearance
Sterile, colorless, and filtered solution.
Formulation
PRSS3 protein solution at a concentration of 1mg/ml. It is supplied in a buffer containing 10% Glycerol and Phosphate-Buffered Saline with a pH of 7.4.
Stability
For short-term storage (up to 4 weeks), the product should be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the product is greater than 95.0% as determined by SDS-PAGE analysis.
Biological Activity
The specific activity of the enzyme is greater than 10,000 pmol/min/ug. This is defined as the amount of enzyme required to cleave 1 pmol of the substrate McaRPKPVE-Nval-WRK(Dnp)-NH2 per minute at a pH of 8.0 and a temperature of 37°C.
Synonyms

Protease, Serine, 3, Protease, Serine, 4 (Trypsin 4, Brain), Brain Trypsinogen, Mesotrypsinogen, Mesotrypsin, Trypsin III, EC 3.4.21.4 4, Trypsin IV, PRSS4, TRY3, TRY4 Protease, Serine, 3 (Mesotrypsin), Pancreatic Trypsinogen III, Serine Protease 3, Serine Protease 4, Trypsinogen IV, Trypsinogen 4, Trypsinogen 5, EC 3.4.21, MTG, T9.

Source

HEK293 Cells.

Amino Acid Sequence

VPFDDDDKIV GGYTCEENSL PYQVSLNSGS HFCGGSLISE QWVVSAAHCY KTRIQVRLGE HNIKVLEGNE QFINAAKIIR HPKYNRDTLD NDIMLIKLSS PAVINARVST ISLPTAPPAA GTECLISGWG NTLSFGADYP DELKCLDAPV LTQAECKASY PGKITNSMFC VGFLEGGKDS CQRDSGGPVV CNGQLQGVVS WGHGCAWKNR PGVYTKVYNY VDWIKDTIAA NSHHHHHH.

Q&A

What is PRSS3 and what cellular functions does it perform?

PRSS3 (mesotrypsinogen, also known as brain trypsinogen) is a serine protease that belongs to the trypsinogen family. It shares high sequence homology with other human trypsinogens like PRSS1 (cationic trypsinogen) . Unlike other trypsin isoforms, PRSS3 is specifically expressed in neuronal tissues, explaining its alternative designation as "brain trypsinogen" . In cellular contexts, PRSS3 functions as a proteolytic enzyme involved in protein processing and degradation. Recent research has uncovered its role in viral pathogenesis, specifically through interaction with enterovirus proteins . Additionally, PRSS3 has been implicated in malignant growth behavior in breast cancer cells, suggesting its potential involvement in cellular proliferation and tumor progression .

How does PRSS3 expression differ across human cell types?

Expression profiling of PRSS3 shows significant variation across human cell types. Neuroblastoma SH-SY5Y cells demonstrate strong PRSS3 expression while showing minimal PRSS1 expression . In contrast, HEK293T cells express all three PRSS subtypes (PRSS1, PRSS2, and PRSS3) . This differential expression pattern suggests tissue-specific regulation of PRSS genes. The expression of PRSS3 can be confirmed through multiple techniques including RT-PCR and immunofluorescence assays using PRSS3-specific antibodies . Research has identified at least four transcript variants of PRSS3, with variant-specific primers needed to distinguish between these isoforms . This expression diversity highlights the importance of cell type selection when designing PRSS3 research protocols.

What are the primary structural characteristics of PRSS3 protein?

PRSS3 is characterized as a serine protease with a molecular weight of approximately 22-26 kDa as determined by SDS-PAGE analysis . The protein contains multiple variants, with PRSS3 variant 3 being extensively studied in research contexts . Structurally, PRSS3 shares significant sequence homology with other trypsinogens, particularly PRSS1, which can present challenges in protein identification through mass spectrometry . When expressed in cells, PRSS3 typically displays a punctate distribution pattern in the cytosol as visualized by immunofluorescence microscopy . The protein can be tagged with epitopes such as Myc for detection and purification purposes without compromising its functional properties . Understanding these structural features is essential for experimental design involving protein isolation, detection, and functional characterization.

Why are HEK293T cells commonly used for PRSS3 research?

HEK293T cells represent an optimal model system for PRSS3 research due to several advantageous characteristics. First, they express all three PRSS subtypes (PRSS1, PRSS2, and PRSS3), making them suitable for comparative studies of trypsinogen isoforms . Second, HEK293T cells demonstrate high transfection efficiency compared to neuronal cell lines like SH-SY5Y, facilitating genetic manipulation studies including overexpression and knockdown experiments . Third, these cells are permissive to viral infection, including enterovirus A71 (EV-A71), enabling research on PRSS3 interactions with viral components . Although HEK293T cells are approximately 100 times less sensitive to EV-A71 than RD cells, this limitation can be overcome by using higher multiplicity of infection (MOI) . The combined attributes of robust PRSS3 expression, ease of genetic manipulation, and viral susceptibility make HEK293T cells a versatile platform for investigating PRSS3 function in both normal cellular processes and disease contexts.

What are the challenges in using HEK cells for neuronal PRSS3 studies?

While HEK293T cells offer numerous advantages for PRSS3 research, several challenges must be considered when extrapolating findings to neuronal contexts. The primary limitation stems from HEK293T cells' different expression profile of PRSS isoforms compared to neuronal cells . Unlike neuroblastoma SH-SY5Y cells that predominantly express PRSS3 with minimal PRSS1, HEK293T cells express all three PRSS subtypes, which may result in functional redundancy or compensation . This difference necessitates careful interpretation when studying PRSS3-specific functions. Additionally, HEK293T cells lack the specialized morphology and signaling pathways characteristic of neurons, limiting their utility for investigating PRSS3's neuronal-specific functions . Researchers must also consider the differential susceptibility to viral infection between HEK293T and neuronal cells when studying PRSS3's role in viral pathogenesis . To address these limitations, validation of key findings in neuronal cell lines or primary neurons is recommended, despite the technical challenges of working with these more specialized cell types.

How can transfection efficiency be optimized for PRSS3 studies in HEK cells?

Optimizing transfection efficiency is crucial for successful PRSS3 studies in HEK293T cells, particularly when introducing expression constructs or RNA interference molecules. For PRSS3 expression studies, transfection protocols typically employ standard lipid-based transfection reagents with cells seeded at 50-60% confluency . When using lentiviral vectors for stable expression or knockdown, polybrene at a final concentration of 6 μg/ml significantly enhances transduction efficiency . For siRNA-mediated PRSS3 knockdown, pooled siRNAs targeting different regions of the PRSS3 transcript improve silencing efficiency and reduce off-target effects . Verification of successful transfection/transduction should include both mRNA quantification by qRT-PCR and protein expression analysis through Western blot or immunofluorescence . For co-expression studies involving PRSS3 and interacting partners, sequential transfection may yield better results than simultaneous introduction of multiple constructs . Researchers should also optimize the DNA:transfection reagent ratio specifically for PRSS3 constructs, as the size and nature of the expression vector can influence transfection efficiency.

How can protein-protein interactions between PRSS3 and viral proteins be identified and validated?

Identifying and validating protein-protein interactions between PRSS3 and viral proteins requires a multi-technique approach. Initial identification of PRSS3 as an interacting partner for viral proteins like EV-A71 3A employs pull-down assays coupled with liquid chromatography tandem mass spectrometry (LC-MS/MS) . This approach identified PRSS3 (mesotrypsinogen) as an EV-A71 3A-interacting protein with high Mascot scores . Once potential interactions are identified, validation proceeds through multiple complementary methods. Co-localization studies using fluorescently tagged proteins (such as PRSS3-Myc and FLAG-3A-mCherry) and confocal microscopy can visualize spatial associations within cells . These studies revealed that PRSS3 spreads in the cytosol as puncta and co-localizes with EV-A71 3A protein . Direct physical interaction is confirmed through co-immunoprecipitation assays, where PRSS3 and viral proteins are separately expressed, mixed, and then immunoprecipitated using antibody-conjugated beads . Western blot analysis of the precipitated complexes can verify the specific binding between PRSS3 and viral proteins . Functional validation using overexpression and knockdown approaches provides evidence for the biological significance of these interactions .

What experimental approaches are effective for studying PRSS3 variants?

Studying PRSS3 variants requires specialized experimental approaches to distinguish between closely related isoforms. RT-PCR using variant-specific primers is essential for identifying which PRSS3 transcript variants are expressed in different cell types . For PRSS3 variant 3, full-length cloning and expression strategies have been successfully employed using C-terminal Myc-tagged constructs (FL-PRSS3-Myc) . When investigating protein function, recombinant expression systems can generate purified PRSS3 variants for in vitro assays . For cellular studies, site-directed mutagenesis of key residues unique to specific variants helps elucidate their distinct functions. Comparative interaction studies between different PRSS3 variants and their binding partners can identify variant-specific associations, as demonstrated with EV-A71 3A protein . Functional assessments following selective knockdown of specific variants using variant-targeted siRNAs provide insights into their specialized roles . Due to the high sequence homology between trypsinogen family members, antibody selection is crucial—researchers should verify antibody specificity against different PRSS variants before experimental application .

What detection methods are most reliable for PRSS3 quantification in HEK cells?

Reliable quantification of PRSS3 in HEK cells requires selecting appropriate detection methods based on experimental objectives. For mRNA expression analysis, quantitative RT-PCR using TaqMan assays (Hs00605637_m1 for PRSS3) provides sensitive and specific detection . This approach can distinguish PRSS3 from other trypsinogen family members when using isoform-specific primers . For protein-level detection, Western blotting with validated anti-PRSS3 antibodies offers quantitative assessment of expression levels . Immunofluorescence microscopy enables visualization of PRSS3's subcellular localization, revealing its punctate distribution in the cytosol . For high-throughput screening applications, ELISA-based methods can quantify PRSS3 secretion. When identifying PRSS3 in complex protein mixtures, mass spectrometry following immunoprecipitation provides definitive identification, though careful analysis is required due to sequence homology with other trypsinogens . For functional studies, enzymatic activity assays using specific substrates can measure PRSS3 protease activity. Researchers should validate detection methods using positive and negative controls, including PRSS3 knockdown samples, to ensure specificity and sensitivity .

What are effective strategies for PRSS3 knockdown in HEK cells?

Effective PRSS3 knockdown in HEK cells can be achieved through several RNA interference approaches. Short hairpin RNA (shRNA) delivered via lentiviral vectors provides stable long-term knockdown, with constructs such as NM_002771.2–302s1c1 (C23) and NM_002771.2–454s1c1 (C24) from the MISSION TRC-Hs1.0 library demonstrating efficacy . For transient knockdown, pooled siRNAs targeting different regions of the PRSS3 transcript achieve significant reduction in both mRNA and protein levels . Verification of knockdown efficiency should include qRT-PCR to measure transcript reduction and Western blot or immunofluorescence to confirm protein depletion . When designing RNA interference experiments, nontarget control (NTC) vectors containing short hairpins that do not recognize any human genes are essential negative controls . Additionally, knockdown of unrelated genes like GAPDH serves as a useful control for specificity . To maximize transduction efficiency with lentiviral vectors, HEK293T cells should be seeded at 50-60% confluency and treated with 6 μg/ml polybrene . For functional studies following knockdown, researchers should establish optimal timing that balances maximal PRSS3 depletion with minimal cellular stress responses.

How can recombinant PRSS3 be effectively expressed and purified for functional studies?

Expression and purification of recombinant PRSS3 for functional studies requires careful consideration of multiple factors. PRSS3 is typically expressed as the inactive zymogen (mesotrypsinogen) to prevent cellular toxicity and self-digestion during production . Expression systems for PRSS3 include bacterial (E. coli), mammalian (HEK293T), and insect cell systems, with mammalian systems providing proper post-translational modifications . For bacterial expression, codon optimization and inclusion of solubility tags such as MBP or SUMO can improve yield and solubility. Purification strategies typically employ affinity chromatography using tagged constructs (His, GST, or Myc) , followed by size exclusion chromatography to achieve high purity. Activation of recombinant mesotrypsinogen to active mesotrypsin requires controlled proteolytic processing, typically using enteropeptidase to cleave the N-terminal activation peptide . Purified recombinant mesotrypsin should be characterized by mass spectrometry, enzymatic activity assays, and circular dichroism to confirm identity, activity, and proper folding. For functional studies, stabilizing buffer conditions containing calcium ions and appropriate pH (7.5-8.5) maximize enzymatic stability. Storage at -80°C in small aliquots with glycerol prevents repeated freeze-thaw cycles and maintains enzymatic activity for extended periods.

How is PRSS3 implicated in cancer research beyond viral interactions?

PRSS3 demonstrates significant roles in cancer biology independent of its viral interactions, particularly in breast cancer progression. Research has shown that PRSS3 is upregulated in malignant T4-2 breast cancer cells compared to their nonmalignant progenitors . Functional studies demonstrate that knockdown of PRSS3 attenuates the malignant growth phenotype, while treatment with recombinant purified mesotrypsin enhances malignant behaviors . In 3D organotypic culture conditions, which better recapitulate tumor architecture than conventional 2D systems, serine protease inhibitors caused morphological reversion of malignant cells, with inhibition of proliferation and restoration of acinar structures with polarization of basal markers . This suggests PRSS3's involvement in maintaining the malignant phenotype. The cancer-promoting effects of PRSS3 appear to be mediated through its proteolytic activity, as treatment with protein protease inhibitors including aprotinin, soybean trypsin inhibitor, and Bowman-Birk inhibitor reversed malignant characteristics . These findings position PRSS3 as a potential therapeutic target in breast cancer, with naturally occurring serine protease inhibitors showing promise as anticancer therapeutics .

What technological advances are enhancing PRSS3 research in HEK systems?

Recent technological advances have significantly enhanced PRSS3 research capabilities in HEK cell systems. CRISPR-Cas9 genome editing now enables precise modification of endogenous PRSS3 genes, allowing introduction of point mutations, domain deletions, or reporter tags at the genomic level. Advanced imaging techniques, including super-resolution microscopy and live-cell imaging, provide unprecedented visualization of PRSS3 dynamics and interactions in real-time . Proximity labeling methods such as BioID and APEX2 are expanding the identification of PRSS3 interaction networks beyond traditional co-immunoprecipitation approaches . For structural studies, cryo-electron microscopy is revealing detailed conformational states of PRSS3 alone and in complex with binding partners. High-throughput screening platforms utilizing PRSS3 activity reporters in HEK cells facilitate rapid identification of modulators and interaction partners. Single-cell RNA sequencing technologies enable detailed analysis of PRSS3 expression heterogeneity within cell populations. Microfluidic systems coupled with HEK cell culture allow precise control of the cellular microenvironment for studying PRSS3 regulation under various conditions. These technological innovations are accelerating discovery in PRSS3 biology and opening new avenues for therapeutic development targeting PRSS3-dependent pathways.

How might PRSS3 research translate to therapeutic applications?

PRSS3 research presents several promising avenues for therapeutic development across multiple disease contexts. In viral infections, particularly enterovirus A71-associated neurological complications, targeting the interaction between PRSS3 and viral 3A protein could disrupt viral replication organelle formation . Small molecule inhibitors or peptide-based drugs designed to prevent this protein-protein interaction represent potential antiviral strategies . For cancer applications, particularly in breast cancer where PRSS3 promotes malignant phenotypes, several therapeutic approaches emerge . Selective PRSS3 inhibitors could attenuate tumor growth and invasion, while naturally occurring serine protease inhibitors like aprotinin, soybean trypsin inhibitor, and Bowman-Birk inhibitor have already demonstrated anticancer potential in preclinical models . RNA interference-based therapeutics targeting PRSS3 mRNA could reduce protein expression in tumor cells . Additionally, understanding PRSS3's role in specific cellular pathways may reveal downstream targets that are more amenable to drug development. Combination therapies targeting PRSS3 alongside established treatments might enhance therapeutic efficacy. As research progresses, patient stratification based on PRSS3 expression or activity could enable personalized medicine approaches, directing PRSS3-targeted therapies to those most likely to benefit.

How can researchers address antibody cross-reactivity issues when studying PRSS3?

Antibody cross-reactivity represents a significant challenge in PRSS3 research due to high sequence homology between trypsinogen family members. To address this issue, researchers should implement a multi-faceted validation strategy before conducting definitive experiments. First, verify antibody specificity through Western blot analysis comparing lysates from cells with confirmed differential expression of trypsinogen isoforms; SH-SY5Y cells (predominantly expressing PRSS3) versus pancreatic cell lines (expressing multiple trypsinogens) provide useful comparison models . Second, validate antibody specificity using PRSS3 knockdown samples, which should show reduced signal intensity . Third, when possible, use epitope-tagged recombinant PRSS3 (such as PRSS3-Myc) and detect with anti-tag antibodies to circumvent specificity issues . Fourth, complement antibody-based detection with mRNA analysis using isoform-specific primers to confirm expression patterns . For immunoprecipitation studies, pre-clear lysates with protein A/G beads to reduce nonspecific binding. When studying protein interactions, reciprocal co-immunoprecipitation using antibodies against both PRSS3 and its binding partner provides stronger evidence of specific interaction . Finally, consider advanced proteomics approaches like selective reaction monitoring mass spectrometry for unambiguous identification and quantification of PRSS3 in complex samples.

How can researchers differentiate between direct and indirect effects of PRSS3 manipulation?

Distinguishing between direct and indirect effects following PRSS3 manipulation presents a significant challenge in functional studies. To address this issue, researchers should implement a comprehensive experimental strategy. First, establish temporal relationships by conducting time-course experiments after PRSS3 manipulation, as direct effects typically manifest earlier than secondary consequences . Second, employ multiple independent methods to modulate PRSS3 function, such as siRNA knockdown, CRISPR knockout, and specific inhibitors, as consistent results across different approaches strengthen evidence for direct causality . Third, perform rescue experiments where wild-type PRSS3 is reintroduced following knockdown; reversal of phenotypes supports direct involvement . Fourth, utilize catalytically inactive PRSS3 mutants to distinguish between scaffold functions and proteolytic activity-dependent effects . Fifth, identify direct substrates through techniques like TAILS (Terminal Amine Isotopic Labeling of Substrates) or proximity-based labeling. Sixth, employ pathway inhibitors to block potential downstream mediators; persistence of PRSS3-dependent effects despite pathway inhibition suggests direct action. Finally, corroborate findings across multiple cell types with varying levels of endogenous PRSS3 expression . This multi-faceted approach enables researchers to confidently differentiate between direct PRSS3-mediated effects and secondary consequences, enhancing mechanistic understanding and therapeutic targeting potential.

What are the most promising unexplored aspects of PRSS3 biology in human cells?

Several unexplored aspects of PRSS3 biology in human cells represent promising avenues for future research. First, the tissue-specific regulation of PRSS3 expression, particularly in neuronal contexts where it's known as brain trypsinogen, remains poorly understood . Investigation of transcriptional and epigenetic regulatory mechanisms could reveal new insights into PRSS3's specialized functions across different tissues. Second, the identification of physiological substrates of PRSS3 in various cellular contexts would illuminate its biological roles beyond pathological conditions . Proteomics approaches such as degradomics could systematically identify PRSS3 targets in neuronal and non-neuronal cells. Third, the intracellular versus extracellular functions of PRSS3 warrant exploration, as most serine proteases function in extracellular spaces, yet PRSS3 demonstrates intracellular interactions with viral proteins . Fourth, the evolutionary biology of PRSS3, particularly its conservation and divergence across species, could provide insights into its fundamental biological importance. Fifth, the potential role of PRSS3 in other neurological diseases beyond viral encephalitis remains largely unexplored, despite its expression in neuronal tissues . Finally, investigation of PRSS3 post-translational modifications and their impact on activity, localization, and stability would enhance understanding of its regulation. These research directions collectively promise to uncover new dimensions of PRSS3 biology with implications for both basic science and therapeutic development.

How might integrative approaches advance PRSS3 research in disease contexts?

Integrative approaches combining multiple technologies and disciplines hold significant potential for advancing PRSS3 research in disease contexts. Systems biology frameworks incorporating transcriptomics, proteomics, and metabolomics can map PRSS3's position within broader cellular networks, identifying disease-relevant pathways and potential intervention points . Patient-derived models including organoids and iPSC-derived cells enable study of PRSS3 function in disease-relevant contexts with appropriate genetic backgrounds . Multi-omics approaches applied to clinical samples from conditions where PRSS3 is implicated, such as viral encephalitis or breast cancer, could reveal disease-specific alterations in PRSS3 expression, processing, or activity . Computational approaches including protein-protein interaction modeling and molecular dynamics simulations can predict structural details of PRSS3 interactions with viral proteins or potential inhibitors . Integrating animal models with cellular studies provides in vivo validation of mechanisms identified in vitro, particularly important for neurological disease contexts . Collaborative research networks bringing together virologists, cancer biologists, structural biologists, and clinicians can accelerate translation of basic PRSS3 findings into therapeutic applications. Implementation of these integrative approaches will address the complexities of PRSS3 biology across multiple disease contexts, potentially leading to novel diagnostic and therapeutic strategies targeting this versatile protease.

Product Science Overview

Structure and Production

PRSS3 is a single, glycosylated polypeptide chain consisting of 238 amino acids, with a molecular mass of approximately 26 kDa . The recombinant form of PRSS3 produced in HEK293 cells is fused to a 6 amino acid His-tag at the C-terminus, which facilitates its purification through chromatographic techniques .

Function and Mechanism

As a serine protease, PRSS3 plays a crucial role in the hydrolysis of peptide bonds in proteins, a process essential for protein digestion and metabolism. The enzyme operates by cleaving peptide bonds at the carboxyl side of lysine and arginine residues. This activity is vital for various physiological processes, including the activation of other proteases and the regulation of cellular functions.

Expression and Localization

PRSS3 is predominantly expressed in the pancreas, where it is secreted as an inactive zymogen (trypsinogen) and later activated in the small intestine. However, its expression is not limited to the pancreas. Studies have shown that PRSS3 is also expressed in other tissues such as the liver, heart, and testis, as well as in several tumor cell lines .

Clinical Significance

The activity of PRSS3 is significant in the context of various diseases. For instance, abnormal expression or mutations in the PRSS3 gene have been linked to pancreatic disorders and certain types of cancer. The enzyme’s ability to degrade fibrinogen and activate pro-urokinase-type plasminogen activator (pro-uPA) suggests its potential role in fibrinolysis and tumor progression .

Recombinant Production in HEK Cells

The production of recombinant PRSS3 in HEK293 cells offers several advantages. HEK cells provide a human-like post-translational modification system, ensuring that the recombinant protein is correctly folded and glycosylated. This is crucial for maintaining the enzyme’s activity and stability. The use of a His-tag facilitates the purification process, allowing for the isolation of high-purity PRSS3 for research and therapeutic applications .

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.