SPINK1 Human

Serine Peptidase Inhibitor Kazal Type 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene Structure and Expression

  • The SPINK1 gene spans 7.5 kb, contains four exons, and encodes a 79-amino acid precursor protein with a 23-amino acid signal peptide .

  • Expression is highest in the pancreas but also occurs in extrapancreatic tissues (e.g., gastrointestinal and urinary tracts) and tumors .

Key Functional Domains

  • Three intramolecular disulfide bridges stabilize the structure: Cys9–Cys33, Cys16–Cys35, and Cys24–Cys56 .

  • The reactive site (Lys41-Ile42) binds trypsin, forming a covalent complex to inhibit proteolytic activity .

Pathological Relevance in Disease

Chronic Pancreatitis (CP)

  • Loss-of-function mutations (e.g., −53C>T, −142T>C, −147A>G) in the promoter region reduce SPINK1 expression by 20–46%, increasing CP risk .

  • The N34S mutation (c.101A>G) is strongly associated with hereditary and idiopathic CP (odds ratio ≥10) .

SPINK1 VariantFunctional ImpactClinical Association
−53C>T46% ↓ expressionChronic pancreatitis
N34SAltered secretionHereditary pancreatitis
c.194+2T>CExon skippingEarly-onset CP

Cancer Biology

SPINK1 overexpression is implicated in tumor progression via:

  • EGFR-ERK Signaling: Structural mimicry of EGF activates EGFR, driving proliferation in hepatocellular carcinoma (HCC) .

  • Angiogenesis: SPINK1 upregulates IL-8, ICAM-1, and VCAM-1 in endothelial cells, enhancing vascularization in acute lymphoblastic leukemia (ALL) .

  • Chemoresistance: In HCC, SPINK1 promotes CD133+ cancer stem cell survival via EGFR-ERK-CDK4/6-E2F2 signaling .

Cancer TypeSPINK1 RoleMechanism
ProstateTumor plasticityEGFR activation
HCCDedifferentiationCD133/ELF3 axis
ALLMetastasisIL-8/MAPK pathway

Mechanistic Insights from Recent Studies

Transcriptional Regulation

  • Promoter variants disrupt transcription factor binding (e.g., HNF1 and PTF1), altering SPINK1 expression .

  • The −215G>A variant increases luciferase activity by 30%, suggesting gain-of-function potential .

Autophagy and Cell Survival

  • In Spink3 knockout mice, SPINK1 deficiency induces autophagic cell death, linking it to pancreatic homeostasis .

Clinical and Therapeutic Implications

  • Diagnostic Biomarker: Elevated serum SPINK1 correlates with ALL progression and HCC dedifferentiation .

  • Therapeutic Targeting: Neutralizing antibodies against SPINK1 reduce tumor growth and chemoresistance in preclinical HCC models .

Product Specs

Introduction
Trypsin inhibitor SPINK1, secreted by pancreatic acinar cells, plays a crucial role in regulating trypsin activity within the pancreas. It prevents the premature activation of digestive enzymes, safeguarding the pancreas from autodigestion. Genetic variations in the SPINK1 gene are linked to pancreatic conditions like tropical calcific pancreatitis and hereditary pancreatitis.
Description
This product consists of a recombinant human SPINK1 protein produced in E. coli. It is a single, non-glycosylated polypeptide chain encompassing amino acids 24 to 79 of the native protein sequence, resulting in a molecular weight of 8.6 kDa. For purification and detection purposes, a 23 amino acid His-tag is attached to the N-terminus.
Physical Appearance
A clear solution free from particulate matter that has been sterilized by filtration.
Formulation
The SPINK1 protein is provided at a concentration of 0.5 mg/ml in a solution containing 20 mM Tris-HCl buffer (pH 8.0), 0.15 M NaCl, 1 mM DTT, and 20% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be kept at 4°C. For extended storage, it is recommended to store the product frozen at -20°C. To further enhance stability during long-term storage, consider adding a carrier protein such as HSA or BSA (0.1%). Repeated freezing and thawing of the product should be avoided.
Purity
The purity of this product exceeds 90% as determined by SDS-PAGE analysis.
Synonyms
Serine Peptidase Inhibitor Kazal Type 1, Pancreatic Secretory Trypsin Inhibitor, Tumor-Associated Trypsin Inhibitor, PCTT, TATI, Spink3, Serine Protease Inhibitor Kazal Type 1, TCP, PSTI.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSDSLGREA KCYNELNGCT KIYDPVCGTD GNTYPNECVL CFENRKRQTS ILIQKSGPC.

Q&A

What is SPINK1 and what are its primary biological functions?

  • Inhibiting premature pancreatic enzyme activation

  • Participating in tissue regeneration processes

  • Contributing to cancer progression through multiple mechanisms

  • Functioning as a SASP (senescence-associated secretory phenotype) factor produced by stromal cells following DNA damage

  • Promoting angiogenesis through endothelial cell activation

The molecular weight of SPINK1 is approximately 6.5 kDa, and it contains a Kazal-type serine protease inhibitor domain that is critical for its protease inhibition functions. While initially characterized in pancreatic tissue, SPINK1 expression has been detected across multiple tissue types, suggesting context-dependent functions that extend beyond its canonical role.

What experimental models are appropriate for studying SPINK1 function?

When designing SPINK1 research, selecting appropriate experimental models is critical for generating valid and translatable results:

Cell Line Models:

  • Pancreatic acinar cell lines (AR42J, 266-6) - Useful for studying SPINK1's role in pancreatic physiology

  • HEK 293T cells - Often used for basic expression studies but have limitations for pancreas-specific functions

  • COLO-357 pancreatic cancer cells - Express pancreatic digestive enzymes and show strong SPINK1 promoter activity

  • HUVECs - Valuable for studying SPINK1's effects on angiogenesis and endothelial function

In Vivo Models:

  • Genetically modified mouse models (SPINK1 knockout, transgenic overexpression)

  • NOD/SCID mice for angiogenesis and cancer infiltration studies

  • Patient-derived xenograft models for cancer applications

Primary Human Samples:

  • Pancreatic tissue from pancreatectomy specimens

  • Serum/plasma samples from patients with pancreatitis or cancer

  • Patient tissues from total pancreatectomy with islet autotransplantation

Each model system has distinct advantages and limitations. For example, while cell lines offer experimental consistency and ease of manipulation, they may not fully recapitulate the complex in vivo environment. Conversely, clinical samples provide direct relevance to human pathophysiology but present challenges in experimental control and sample availability.

What methods are most effective for measuring SPINK1 expression?

Protein Detection Methods:

  • ELISA - Gold standard for quantifying SPINK1 in serum, plasma, and tissue homogenates

  • Western blot - For semi-quantitative detection and molecular weight verification

  • Immunohistochemistry - For spatial localization in tissue sections

  • Flow cytometry - For cell-specific expression analysis in mixed populations

Transcriptional Analysis:

  • RT-qPCR - For sensitive quantification of SPINK1 mRNA levels

  • RNA-seq - For comprehensive transcriptional profiling in context

  • In situ hybridization - For spatial mapping of transcript expression

Functional Analysis:

  • Luciferase reporter assays - Effective for studying SPINK1 promoter activity and regulation

  • Trypsin inhibition assays - Direct measurement of SPINK1's canonical function

Method Selection Considerations:
When selecting detection methods, consider experimental requirements for sensitivity, specificity, quantitative precision, and biological context. For example, luciferase reporter assays have proven particularly valuable for assessing SPINK1 promoter variants, with studies showing that different cell lines yield variable results that may be relevant to different physiological contexts .

How does SPINK1 contribute to chronic pancreatitis development?

SPINK1 plays a critical role in protecting against premature trypsinogen activation in the pancreas. Loss-of-function mutations in the SPINK1 gene are associated with chronic pancreatitis through several mechanisms:

Pathophysiological Mechanisms:

  • Reduced inhibition of premature trypsin activation

  • Altered pancreatic enzyme homeostasis

  • Increased susceptibility to pancreatic injury

  • Progressive fibrosis rather than lipomatous atrophy, distinguishing it from other genetic forms of pancreatitis

Histopathological Characteristics:
Analysis of pancreatic specimens from patients with SPINK1-associated chronic pancreatitis reveals a distinct pattern characterized by:

  • Progressive exocrine parenchymal loss

  • Replacement by prominent fibrosis

  • Correlation between fibrosis severity and duration of abdominal pain

  • Divergent pathophysiology from PRSS1 and CFTR-associated pancreatitis, which typically show lipomatous atrophy without significant fibrosis

Importantly, SPINK1 mutations rarely act in isolation. Among patients with SPINK1-associated chronic pancreatitis, 86% have additional risk factors including:

  • Co-occurring mutations in other pancreatitis-associated genes (CFTR, CTRC)

  • Anatomic anomalies

  • Environmental factors such as tobacco use

These findings suggest that SPINK1 may function as a disease modifier gene rather than a primary causative factor in many cases of chronic pancreatitis.

What is the role of SPINK1 in cancer biology and the tumor microenvironment?

SPINK1 has emerged as a significant factor in cancer biology, particularly through its effects on the tumor microenvironment (TME) and therapy response:

Cancer-Promoting Mechanisms:

  • Promotes cancer cell invasion and metastasis in breast and colon cancers

  • Enhances cancer cell proliferation and migration

  • Functions as a SASP factor after genotoxic damage to stromal cells

  • Induces epithelial-endothelial transition (EET) via EGFR signaling

  • Promotes angiogenesis by activating endothelial cells

Tumor Microenvironment Effects:

  • Produced by senescent stromal cells following chemotherapy or radiation

  • Activates MAPK pathway in endothelial cells

  • Increases expression of IL-8, VCAM-1, and ICAM-1 in endothelial cells

  • Enhances endothelial cell motility and cancer cell trans-endothelial migration

  • Promotes in vivo vascularization and tissue infiltration

Clinical Relevance:

  • Detected in peripheral blood of cancer patients after chemotherapy

  • Potentially contributes to acquired treatment resistance

  • Represents a targetable factor in the damaged TME

  • May serve as a non-invasive biomarker for TME damage

This multifaceted role makes SPINK1 a promising target for cancer therapies, particularly those aimed at disrupting the tumor microenvironment or overcoming treatment resistance.

How do SPINK1 promoter variants affect gene expression and disease risk?

Variants in the SPINK1 promoter region can significantly impact gene expression, potentially altering disease susceptibility:

Functional Consequences of Promoter Variants:
Luciferase reporter assays have revealed that different SPINK1 promoter variants can cause:

  • Loss of function: Decreasing promoter activity, potentially predisposing to pancreatitis

  • Gain of function: Increasing promoter activity, potentially protective against pancreatitis or contributing to cancer risk

Cell-Type Specificity:
The functional effect of SPINK1 promoter variants depends on the cellular context:

  • Variants show different effects in pancreatic acinar cells versus non-pancreatic cells

  • Dexamethasone-treated AR42J cells show enhanced SPINK1 promoter activity compared to untreated cells

  • Effects observed in HEK 293T cells may be more relevant to extrapancreatic tissues and tumors than to pancreatic pathology

Clinically Significant Variants:
Research has identified several promoter variants with functional consequences:

  • Loss-of-function variants (c.-53C>T, c.-142T>C, c.-147A>G) - Likely pathogenic for pancreatitis

  • Gain-of-function variants (c.-81C>T, c.-215G>A) - Potentially protective against pancreatitis

  • Some variants (c.-142T>C, c.-164G>C, c.-215G>T) may act as risk factors for certain cancers

These findings highlight the importance of sequence evaluation of the SPINK1 promoter region as part of the etiological workup for patients with chronic pancreatitis.

What experimental approaches best capture SPINK1's role in the tumor microenvironment?

Investigating SPINK1's complex functions in the tumor microenvironment requires sophisticated experimental approaches:

Co-culture Systems:

  • HUVEC and cancer cell co-cultures can assess SPINK1's effects on:

    • Endothelial cell activation

    • Cancer cell transendothelial migration

    • Cellular reorganization and tube formation

  • 3D organoid co-cultures with stromal and cancer cells provide more physiologically relevant models

In Vivo Approaches:

  • NOD/SCID mice models for studying vascularization and tissue infiltration

  • Orthotopic xenograft models with manipulated SPINK1

  • Therapeutic intervention studies targeting SPINK1 in established tumors

Multi-omics Analysis:

  • Transcriptomic analysis of SPINK1-treated endothelial cells has revealed activation of MAPK pathway and alterations in several biological processes

  • Combined proteomics, transcriptomics, and metabolomics can provide comprehensive insights into SPINK1-mediated changes

Therapeutic Targeting Strategies:

  • Neutralizing antibodies against SPINK1

  • Small molecule inhibitors of downstream signaling

  • RNA interference approaches

When designing these experiments, researchers should consider the timing of SPINK1 expression/administration, dosage effects, cell-type specificity, and potential interactions with other factors in the microenvironment.

How can researchers effectively study the dual roles of SPINK1 in both protecting against pancreatitis and promoting cancer?

SPINK1's context-dependent functions present a research challenge that requires careful experimental design:

Comparative Studies:

  • Parallel experiments in pancreatic and cancer models to directly compare effects

  • Dose-response studies to identify potential threshold effects

  • Temporal studies to assess acute versus chronic effects

Mechanistic Dissection:

  • Domain-specific mutants to separate trypsin inhibition from growth-promoting functions

  • Receptor-binding studies to identify tissue-specific interaction partners

  • Signaling pathway analysis in different cellular contexts

Translational Approaches:

  • Patient-derived samples stratified by SPINK1 status and disease phenotype

  • Correlation of SPINK1 levels with clinical outcomes in both pancreatitis and cancer

  • Integration of genomic and expression data

Methodological Considerations:

  • Use multiple cell types relevant to both pathologies

  • Employ both genetic modulation and exogenous administration approaches

  • Consider systemic effects through in vivo models

This dual functionality highlights the importance of context in interpreting SPINK1 research findings and the need for comprehensive experimental approaches.

What are the best approaches for translating SPINK1 research findings into clinical applications?

Translating SPINK1 research into clinical applications requires careful consideration of several factors:

Biomarker Development:

  • Standardization of SPINK1 detection methods across laboratories

  • Establishment of reference ranges in different patient populations

  • Correlation with disease progression and treatment response

  • Combination with other biomarkers for improved specificity and sensitivity

Therapeutic Development:

  • Tissue-specific targeting strategies to avoid disrupting protective functions

  • Combination approaches targeting multiple aspects of disease pathophysiology

  • Consideration of temporal aspects of SPINK1 expression in treatment planning

Clinical Trial Design:

  • Stratification of patients based on SPINK1 genetic status or expression levels

  • Careful selection of endpoints relevant to SPINK1 biology

  • Monitoring of potential adverse effects related to SPINK1's dual functions

Challenges and Considerations:

  • The context-dependent nature of SPINK1 function may limit broad applicability

  • Genetic heterogeneity in patient populations may affect response to SPINK1-targeted interventions

  • Need for careful risk-benefit assessment given SPINK1's protective roles in some contexts

What methods should researchers use to comprehensively assess SPINK1 genetic variants?

Comprehensive genetic analysis of SPINK1 requires a methodical approach:

Sequencing Approaches:

  • Targeted gene sequencing for focused analysis of SPINK1 and known interacting genes

  • Whole exome sequencing for broader assessment of potential genetic interactions

  • Whole genome sequencing to capture regulatory regions and structural variants

Critical Regions for Analysis:

  • Coding exons for functional protein variants

  • Promoter region (at least 541 bp upstream of the start codon)

  • Intron-exon boundaries for splicing variants

  • Include examination of linked variant effects, as demonstrated by the interaction between the c.-215G>A promoter variant and the c.194+2T>C splice-site mutation

Variant Interpretation:

  • Functional validation using reporter assays in appropriate cell types

  • In silico prediction tools calibrated for SPINK1 variants

  • Population frequency data to assess rarity

  • Co-segregation in families with pancreatitis or cancer

Methodological Table: Recommended Approach for SPINK1 Genetic Analysis

Analysis TypeRegions to IncludeValidation MethodApplication
Clinical TestingCoding exons, canonical splice sites, promoter (c.-541 to c.35)Functional assays for novel variantsPatient diagnosis
Research SequencingEntire gene including 2kb upstream/downstreamReporter assays, protein expression studiesVariant discovery
Population StudiesTargeted hotspots and known pathogenic variantsStatistical association testingRisk assessment
Functional GenomicsEnhancers, repressors, 3'UTRCRISPR editing, luciferase assaysMechanistic insights

How should researchers interpret SPINK1 promoter variants of uncertain significance?

When faced with novel or rare SPINK1 promoter variants, researchers should follow a systematic approach:

Functional Testing:

  • Luciferase reporter assays in multiple cell lines (AR42J, 266-6, COLO-357, HEK 293T)

  • Assessment of promoter activity with and without relevant stimuli (e.g., dexamethasone for AR42J cells)

  • Comparison with known pathogenic and benign variants

Bioinformatic Analysis:

  • Transcription factor binding site prediction

  • Conservation analysis across species

  • Allele frequency in control populations

  • Linkage analysis with known pathogenic variants

Classification Criteria:

  • 50% reduction in promoter activity suggests potential pathogenicity for pancreatitis

  • 2-fold increase may indicate protection against pancreatitis or increased cancer risk

  • Consider cell-type specificity of effects when interpreting results

  • Integrate with clinical and family history data

Research Example:
Studies have shown that when testing SPINK1 promoter variants, results can vary significantly between cell lines. For instance, the variant c.-215G>A showed 5.6-fold increased activity in HEK 293T cells but was linked to the deleterious splice-site mutation c.194+2T>C, complicating interpretation . This underscores the importance of comprehensive analysis rather than relying on single assays.

What statistical approaches are most appropriate for meta-analyses of SPINK1 genetic studies?

Meta-analyses of SPINK1 genetic studies require robust statistical approaches:

Model Selection:

  • Random-effects model (e.g., DerSimonian and Laird method) is generally preferred due to expected heterogeneity between studies

  • Fixed-effects models may be appropriate when study populations are homogeneous

Effect Size Calculation:

  • For binary outcomes (e.g., presence/absence of disease), calculate pooled odds ratios (OR) with 95% confidence intervals

  • For continuous outcomes, standardized mean differences may be more appropriate

Heterogeneity Assessment:

  • Use Cochrane Q test (significant at P < 0.05) to detect heterogeneity

  • Calculate I² statistic to quantify heterogeneity (25%, 50%, and 75% corresponding to low, moderate, and high degrees)

  • Explore sources of heterogeneity through subgroup analyses and meta-regression

Publication Bias Evaluation:

  • Visual inspection of funnel plots for asymmetry

  • Statistical tests such as Egger's test or Begg's test

  • Consider using trim-and-fill methods to adjust for potential publication bias

Software Considerations:

  • STATA, R, RevMan, or similar statistical software packages provide comprehensive tools for meta-analysis

  • Ensure transparency by clearly reporting all statistical methods, inclusion/exclusion criteria, and analytical decisions

What are the most promising approaches for targeting SPINK1 therapeutically?

Several innovative approaches show promise for SPINK1-targeted therapeutics:

Direct SPINK1 Inhibition:

  • Monoclonal antibodies against SPINK1 for extracellular neutralization

  • RNA interference (siRNA, shRNA) for expression knockdown

  • CRISPR-Cas9 gene editing for permanent modification in appropriate contexts

Pathway-Based Approaches:

  • EGFR inhibitors to block SPINK1-mediated signaling in cancer contexts

  • MAPK pathway modulators to prevent downstream effects in endothelial cells

  • Combinatorial approaches targeting multiple nodes in SPINK1-activated pathways

Context-Specific Targeting:

  • Tumor microenvironment-specific delivery systems

  • Temporal targeting strategies (e.g., post-chemotherapy administration)

  • Companion diagnostics to identify patients most likely to benefit

Challenges to Consider:

  • Need to preserve SPINK1's protective functions in pancreatic tissue

  • Potential for compensatory mechanisms

  • Identification of appropriate biomarkers for patient selection

  • Optimization of delivery to target tissues

Development of these therapeutic approaches should proceed with careful attention to SPINK1's dual roles and context-dependent functions to maximize efficacy while minimizing adverse effects.

How can multi-omics approaches advance our understanding of SPINK1 biology?

Multi-omics integration offers powerful new insights into SPINK1 biology:

Integrative Approaches:

  • Combined genomics, transcriptomics, proteomics, and metabolomics

  • Single-cell multi-omics for cellular heterogeneity assessment

  • Spatial transcriptomics to map SPINK1 expression and effects in tissue context

  • Temporal multi-omics to capture dynamic responses

Key Research Applications:

  • Identifying novel SPINK1 interaction partners across different tissues

  • Mapping pathway alterations in response to SPINK1 modulation

  • Discovering biomarkers that correlate with SPINK1 activity

  • Characterizing differences between physiological and pathological SPINK1 function

Analytical Considerations:

  • Need for sophisticated computational methods to integrate diverse data types

  • Careful experimental design with appropriate controls and replicates

  • Validation of findings across multiple experimental systems

  • Translation of findings to human disease contexts

These multi-omics approaches have the potential to reveal new dimensions of SPINK1 biology, leading to novel therapeutic targets and more precise diagnostic approaches.

What are the critical unanswered questions in SPINK1 research?

Despite significant advances, several fundamental questions remain in SPINK1 research:

Molecular Mechanisms:

  • How does SPINK1 promote epithelial-endothelial transition at the molecular level?

  • What are the full spectrum of SPINK1 receptors and binding partners across different tissues?

  • How is SPINK1 expression regulated in different physiological and pathological contexts?

Clinical Translation:

  • Can SPINK1 serve as a reliable biomarker for therapeutic response in cancer?

  • What is the predictive value of SPINK1 genetic variants for pancreatitis progression?

  • How do SPINK1 levels correlate with specific disease phenotypes?

Therapeutic Development:

  • Can SPINK1-targeted therapies overcome treatment resistance in cancer?

  • Is it possible to selectively target SPINK1's pathological functions while preserving beneficial roles?

  • What combination strategies might enhance the efficacy of SPINK1-targeted interventions?

Evolutionary and Comparative Biology:

  • How has SPINK1 function evolved across species?

  • Are there functional differences in SPINK1 orthologs that might inform therapeutic approaches?

  • Do other Kazal-type inhibitors share SPINK1's dual functionality?

Addressing these questions will require collaborative efforts across disciplines and continued technological innovation in both experimental methods and analytical approaches.

Product Science Overview

Gene and Protein Structure

The SPINK1 gene is located on chromosome 5q32 in humans . The protein encoded by this gene is a trypsin inhibitor, which is secreted from pancreatic acinar cells into the pancreatic juice . The primary function of SPINK1 is to prevent the premature activation of trypsinogen, a precursor of the enzyme trypsin, within the pancreas and pancreatic ducts . This is essential to protect the pancreas from autodigestion and subsequent damage.

Biological Function

SPINK1 inhibits serine proteases, particularly trypsin, by binding to the active site of the enzyme and preventing it from cleaving its substrates . This inhibition is crucial for maintaining the balance of protease activity in the pancreas and preventing conditions such as pancreatitis . In the male reproductive tract, SPINK1 binds to sperm heads and modulates sperm capacitance by inhibiting calcium uptake and nitric oxide production .

Clinical Significance

Mutations in the SPINK1 gene have been associated with various forms of pancreatitis, including hereditary pancreatitis and tropical calcific pancreatitis . These mutations can lead to a loss of function of the SPINK1 protein, resulting in uncontrolled trypsin activity and subsequent pancreatic damage . Additionally, SPINK1 has been implicated in the progression of certain cancers, including prostate cancer .

Recombinant SPINK1

Recombinant SPINK1 refers to the protein produced through recombinant DNA technology, which involves inserting the SPINK1 gene into a suitable expression system to produce the protein in large quantities. This recombinant protein is used in various research and clinical applications, including studies on protease inhibition, pancreatitis, and cancer .

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.