SPINT2 inhibits:
Hepatocyte growth factor activator (HGFA), blocking pro-HGF conversion into active HGF, a key regulator of cell migration and angiogenesis .
Matriptase and prostasin, serine proteases involved in epithelial integrity and cancer progression .
TMPRSS2, a host protease critical for SARS-CoV-2 viral entry .
SPINT2 downregulation is linked to tumor progression in oral squamous cell carcinoma (OSCC) and prostate cancer .
Overexpression reduces matrix metalloproteinase (MMP)-2/9 activity, suppressing cell invasion and metastasis .
In thoracic aortic dissection (TAD), SPINT2 overexpression inhibits smooth muscle cell (SMC) proliferation and migration by suppressing ERK pathway activation .
It maintains SMC contractile phenotypes, preventing pathological switching to synthetic states .
Elevated SPINT2 in maternal circulation correlates with placental insufficiency and small-for-gestational-age (SGA) infants .
Hypoxia upregulates SPINT2 in trophoblasts, suggesting a compensatory role in placental stress .
Model: PDGF-BB-induced aortic SMCs.
Outcomes:
SPINT2 overexpression reduced SMC proliferation by 40% (MTT assay) and migration by 35% (wound healing assay).
Suppressed MMP-2/9 expression by 50%, mitigating extracellular matrix degradation.
Reversed synthetic-to-contractile phenotypic switching via ERK inhibition.
Model: SARS-CoV-2 infection in Calu-3 lung cells.
Outcomes:
SPINT2 knockdown increased viral load by 3-fold, while overexpression reduced infection by 70%.
Coregulation with TMPRSS2 by transcription factors (e.g., ELF3, FOS) maintains protease-inhibitor balance.
Model: Ovarian cancer (OC) and OSCC cell lines.
Outcomes:
SPINT2, also known as hepatocyte growth factor activator inhibitor type-2, belongs to the Kunitz family of serine protease inhibitors. Its primary function is to bind to and inactivate HGFA, thereby impairing the conversion of inactive pro-HGF/SF (hepatocyte growth factor/scatter factor) into bioactive HGF/SF . This regulatory mechanism is significant because HGF can improve cell viability and invasiveness, stimulate angiogenesis, and function as a tumor progression factor .
To study SPINT2 function, researchers should employ gene expression analysis (RT-qPCR, RNA-seq), protein-protein interaction studies, and functional assays examining cell proliferation, migration, and invasion in various cell types with modulated SPINT2 expression. Western blotting combined with immunofluorescence provides valuable insights into protein expression levels and cellular localization patterns.
Multiple complementary approaches should be employed for comprehensive SPINT2 detection:
RNA-level detection:
Protein-level detection:
Bioinformatic approaches:
For optimal results, researchers should validate findings using multiple techniques, including appropriate positive and negative controls, and standardize protocols across experimental conditions.
SPINT2 serves as a critical upstream regulator of the HGF/Met signaling pathway through the following mechanisms:
Direct inhibition of HGFA: SPINT2 binds to and inactivates HGFA, which is responsible for converting pro-HGF to active HGF . This inhibitory action creates a rate-limiting step in the activation of HGF/SF.
Regulation of downstream signaling: By controlling HGF activation, SPINT2 indirectly modulates Met receptor signaling and subsequent cellular responses including proliferation, migration, survival, and angiogenesis .
Impact on tissue-specific functions: In prostate cancer cells, SPINT2 overexpression suppresses pro-HGF activation, extracellular matrix degradation, and cancer cell invasion . In smooth muscle cells, SPINT2 modulates PDGF receptor β expression, affecting phenotypic switching .
To experimentally investigate this interaction, researchers should employ:
Protein-protein interaction assays to detect SPINT2-HGFA binding
HGF activation assays measuring pro-HGF to HGF conversion
Phosphorylation analysis of Met and downstream signaling components
Functional readouts including migration, invasion, and proliferation assays
Gene Ontology (GO) and KEGG pathway analyses of SPINT2 co-expressed genes reveal enrichment in several key biological processes:
Cell adhesion and migration: SPINT2 co-expressed genes participate in regulating cell adhesion and migration pathways, suggesting a role in tissue architecture and cellular motility .
Immune response: Functional enrichment analysis demonstrates significant involvement in immune-related processes, supporting SPINT2's role in modulating the tumor immune microenvironment .
Embryonic development: SPINT2 and its co-expressed genes are enriched in developmental processes, consistent with its known functions in embryogenesis .
Cancer-related pathways: KEGG analysis shows enrichment in pathways associated with cancer occurrence and metastasis .
To conduct similar analyses, researchers should:
Identify SPINT2 co-expressed genes using correlation analysis in large datasets
Employ tools like clusterProfiler for GO and KEGG enrichment analyses
Visualize results using chord plots and other graphical representations
Validate key pathway components through focused experimental studies
SPINT2 expression exhibits considerable tissue-specific and context-dependent regulation:
Age-related factors: In ovarian cancer, SPINT2 expression is significantly higher in patients above 40 years old compared to younger patients .
Tumor grade: Higher SPINT2 expression is observed in ovarian tumors above grade 2 compared to grade 1 tumors .
Tissue type: According to The Human Protein Atlas data, most cancer tissues display weak to moderate SPINT2 expression, while upregulated expression is observed in ovarian, prostate, pancreatic, breast, colorectal, and testis cancers .
Pathological conditions: In thoracic aortic dissection (TAD), SPINT2 is expressed at significantly lower levels compared to normal aortic tissues .
Growth factor stimulation: PDGF-BB treatment can modulate SPINT2 expression in smooth muscle cells, with concentration-dependent effects .
To study these influences, researchers should employ:
Stratified expression analysis across patient demographics and clinical features
In vitro stimulation with different growth factors and cytokines
Epigenetic profiling to identify potential regulatory mechanisms
Promoter analysis to identify tissue-specific transcription factor binding sites
SPINT2 significantly impacts immune cell infiltration in cancer, particularly affecting macrophages through multiple mechanisms:
Correlation with immune infiltration: TIMER analysis reveals SPINT2 expression positively correlates with macrophage infiltration in ovarian cancer (correlation coefficient r = 0.219, p = 1.29 × 10^-6) and neutrophil infiltration (r = 0.137, p = 2.57 × 10^-3) .
M2 macrophage recruitment: SPINT2 mediates the migration of M2 macrophages, potentially through chemotactic mechanisms involving cytokine or chemokine secretion .
Macrophage polarization: SPINT2 promotes polarization from M0 to M2 macrophages, as evidenced by increased expression of M2 markers (CD163 and CD206) and enhanced IL-10 release .
To experimentally investigate these mechanisms, researchers should:
Use THP-1 differentiation models with PMA stimulation to generate macrophage-like cells
Conduct migration assays to assess SPINT2's chemotactic effects
Measure M2 markers by western blotting and flow cytometry
Perform co-culture experiments with SPINT2-overexpressing cancer cells and monocytes
These findings explain how SPINT2 may contribute to creating an immunosuppressive tumor microenvironment, potentially contributing to its association with poor prognosis in certain cancers.
SPINT2 exhibits context-dependent roles in cancer, with evidence supporting both tumor-suppressive and oncogenic functions:
Tumor-suppressive evidence:
In glioma, SPINT2 decreases cell migration and invasion via downregulation of MMP-2 expression and activity .
In endometrial cancer, SPINT2 reduces vimentin levels, potentially inhibiting epithelial-to-mesenchymal transition .
In prostate cancer, SPINT2 overexpression suppresses pro-HGF activation, ECM degradation, and cancer cell invasion .
Oncogenic evidence:
To address these contradictions, researchers should:
Conduct comprehensive expression profiling across multiple cancer types
Perform functional studies in diverse cancer cell lines
Develop in vivo models to evaluate SPINT2's net impact on tumor growth
Investigate isoform-specific effects and post-translational modifications
Employ single-cell analysis to understand heterogeneous expression patterns
These contradictions likely reflect SPINT2's multifunctional nature and context-dependent interactions, emphasizing the need for cancer-specific therapeutic approaches.
To comprehensively investigate SPINT2's role in cell migration and invasion, researchers should employ multiple complementary methods:
Transwell assays:
Gene expression modulation:
Molecular pathway analysis:
HGF/Met pathway-specific assays:
Measurement of pro-HGF processing
Met phosphorylation analysis
Quantification of downstream signaling components
Advanced techniques:
For optimal results, researchers should validate findings using multiple approaches and include appropriate controls for each method.
Several strategies can be employed to modulate SPINT2 expression in cell culture models, each with specific advantages:
Adenoviral vector systems:
Methodology: SPINT2 sequence is synthesized, ligated to a vector (e.g., RedTrack-CMV), packaged in 293A cells, and used to infect target cells at optimized MOI (e.g., 100 for 24h) .
Advantages: High transduction efficiency in both dividing and non-dividing cells, temporal control of expression.
Validation: RT-qPCR and western blotting should confirm SPINT2 overexpression at both mRNA and protein levels .
CRISPR/Cas9 gene editing:
For creating stable knockout or knock-in cell lines.
Protocol involves designing guide RNAs targeting the SPINT2 locus, transfecting cells with Cas9 and guide RNA constructs, and screening for desired modifications.
Advantages: Permanent genetic modification, allows for complete knockout.
RNA interference:
siRNA for transient knockdown or shRNA for stable knockdown.
Include appropriate negative controls (scrambled sequences).
Validate knockdown efficiency at both mRNA and protein levels.
Growth factor modulation:
Inducible expression systems:
Tet-On/Tet-Off systems allow for controlled temporal expression.
Advantages: Adjustable expression levels by varying inducer concentration.
Method | Advantages | Limitations | Validation Approaches |
---|---|---|---|
Adenoviral vectors | High efficiency, works in non-dividing cells | Transient expression | RT-qPCR, Western blot |
CRISPR/Cas9 | Permanent modification, complete knockout | Time-consuming, potential off-targets | Sequencing, protein detection |
RNA interference | Relatively simple implementation | Incomplete knockdown | RT-qPCR, Western blot |
Growth factor modulation | Physiologically relevant | Indirect effects, multiple pathways | Expression analysis, pathway validation |
Inducible systems | Temporal control, adjustable levels | Requires stable cell lines | Expression analysis with/without inducer |
SPINT2 expression shows varied correlations with clinical outcomes across cancer types:
To study these correlations, researchers should:
Conduct Kaplan-Meier survival analysis stratified by SPINT2 expression levels
Mine public repositories (GEO, TCGA, UALCAN) for correlations with clinical features
Perform multivariate analysis to determine independent prognostic value
Validate findings in independent patient cohorts using tissue microarrays
These findings highlight the importance of tumor-specific analysis when evaluating SPINT2 as a prognostic biomarker or therapeutic target.
Serine Peptidase Inhibitor, Kunitz Type 2 (SPINT2), also known as Hepatocyte Growth Factor Activator Inhibitor Type 2 (HAI-2), is a protein encoded by the SPINT2 gene in humans. This protein plays a crucial role in inhibiting a variety of serine proteases, which are enzymes that cleave peptide bonds in proteins.
The SPINT2 gene is located on chromosome 19q13.2 and encodes a transmembrane protein with two extracellular Kunitz domains . These Kunitz domains are responsible for the inhibitory activity of the protein. The protein has a calculated molecular mass of approximately 28.2 kDa and contains an N-terminal signal peptide, a hydrophobic membrane-associated C-terminal region, and two potential N-glycosylation sites .
SPINT2 inhibits several serine proteases, including the Hepatocyte Growth Factor Activator (HGFAC), plasmin, and tissue kallikrein . By inhibiting HGFAC, SPINT2 prevents the formation of active hepatocyte growth factor, which is involved in cell growth, motility, and morphogenesis. This inhibition is crucial for regulating various physiological processes and maintaining tissue homeostasis.
SPINT2 is considered a putative tumor suppressor. Mutations in the SPINT2 gene have been associated with congenital sodium diarrhea, a condition characterized by severe diarrhea and electrolyte imbalance . Additionally, overexpression of SPINT2 has been observed in pancreatic cancer tissues and cell lines, suggesting its potential role in cancer biology .
Recombinant SPINT2 is produced using recombinant DNA technology, which involves inserting the SPINT2 gene into a suitable expression system, such as bacteria or mammalian cells, to produce the protein in large quantities. This recombinant protein is used in various research and clinical applications to study its inhibitory effects on serine proteases and its potential therapeutic uses.