SPINT2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchase method and location. For specific delivery times, please consult your local distributors.
Synonyms
SPINT2; HAI2; KOP; Kunitz-type protease inhibitor 2; Hepatocyte growth factor activator inhibitor type 2; HAI-2; Placental bikunin
Target Names
SPINT2
Uniprot No.

Target Background

Function
SPINT2 Antibody is an inhibitor of HGF activator. It also inhibits plasmin, plasma and tissue kallikrein, and factor XIa.
Gene References Into Functions
  1. The expression of the SPINT2 gene is regulated by its methylation status. Notably, the methylation status of SPINT2 is altered by HPV infection. This suggests that aberrant methylation of the SPINT2 gene may play a significant role in the development of cervical cancer. PMID: 29843210
  2. Aberrant methylation of the SPINT2 gene is frequently observed in high-grade gliomas. This methylation may confer MET signaling in the glioma cells. PMID: 29987920
  3. A single heterozygous SPINT2 mutation was identified in 19 patients with isolated CA/GA. However, this finding was not statistically significant. PMID: 29499739
  4. Using homology modeling, molecular dynamic simulations, and free-energy calculations through MM/PBSA and MM/GBSA, KLK14 binding to either hepatocyte growth factor activator inhibitor type-1 (HAI-1) or type-2 (HAI-2) was successfully modeled. PMID: 28817220
  5. The limited role of HAI-2 in the inhibition of matriptase and prostasin is attributed to its primarily intracellular localization in basal and spinous layer keratinocytes. This localization likely prevents the Kunitz inhibitor from interacting with active prostasin or matriptase. PMID: 29438412
  6. Mutations in the HAI-2 Kunitz domain 1 influence the flux of matriptase to the plasma membrane by affecting the oligomerization, maturation, and/or folding of matriptase. PMID: 28371047
  7. Hypoxic inhibition of JMJD3 activity reduces demethylation of H3K27me3, nucleosome removal, and consequently, the induction of the STAT6 target gene CCL18. However, the induction of other STAT6-inducible genes, such as SPINT2, remained unaffected by JMJD3. PMID: 27737800
  8. The concomitant presence of TMPRSS13 with HAI-2 mediates phosphorylation of residues in the intracellular domain of the protease. This coincides with efficient transport of the protease to the cell surface and its subsequent shedding. PMID: 28710277
  9. A molecular characterization of congenital tufting enteropathy in Italian patients identified one mutation in the SPINT2 gene. PMID: 26684320
  10. Research aims to study the methylation status of the promoter region of Serine peptidase inhibitor/hepatocyte growth factor activator inhibitor type 2 (SPINT2/HAI-2). PMID: 26030814
  11. Findings suggest an involvement of SPINT2 in PCa tumorigenesis, potentially in association with a post-translational regulation of SPINT2. PMID: 26442953
  12. N-glycan branching regulates HAI-2 through different subcellular distribution, subsequently influencing its access to different target proteases. PMID: 26171609
  13. Matriptase inhibition by HAI-2 necessitates the translocation of HAI-2 to the cell surface. This process is observed in some breast cancer cells but not in mammary epithelial cells. PMID: 25786220
  14. Epigenetic silencing of SPINT2 promotes cancer cell motility in malignant melanoma. PMID: 25910030
  15. A missense mutation in the serine protease inhibitor SPINT2 may play a role in congenital sodium diarrhea. PMID: 24722141
  16. Results indicate that the SPINT2 underexpression in the MSC from MDS patients is likely involved in the adhesion of progenitors to the bone marrow niche through an increased HGF and SDF-1 signaling pathway. PMID: 24410667
  17. The role of hepatocyte growth factor activator inhibitor-2 (HAI-2) in prostate cancer cell migration, invasion, tumorigenicity, and metastasis is under investigation. PMID: 24121274
  18. The tumor suppressor gene SPINT2 is commonly silenced by promoter hypermethylation in human esophageal squamous cell carcinoma. Notably, SPINT2 hypermethylation correlates with poor overall survival. PMID: 24269829
  19. SPINT2 has been established as a second gene associated with congenital tufting enteropathy. PMID: 24142340
  20. HAI-2 is an inhibitor of matriptase-2 that modulates the synthesis of hepcidin. PMID: 23293962
  21. HAI-1 and -2 are overexpressed in the liver in cholangiopathies with ductular reactions, suggesting potential involvement in liver fibrosis and hepatic differentiation. PMID: 21898507
  22. Low levels of HAI-2 are associated with endometrial cancer. PMID: 20715109
  23. HAI-2 protein is significantly decreased in malignant prostate cancer lesions compared to normal and benign prostate hyperplasia lesions. Moreover, the most poorly differentiated tumors exhibit the lowest levels of HAI-2 expression. PMID: 20687215
  24. Epigenetic inactivation of HAI-2/SPINT2 contributes to gastric carcinogenesis and may serve as a potential biomarker for gastric cancer. PMID: 20063316
  25. Syndromic tufting enteropathy may be linked to a SPINT2 mutation observed in congenital sodium diarrhea [case report]. PMID: 20009592
  26. Bik-R can physically interact with the CD44v isoforms containing epitope v9 and function as a repressor to down-regulate PMA-stimulated uPA expression. This occurs, at least in part, by preventing clustering of CD44v isoforms containing epitope v9. PMID: 11777908
  27. Bikunin downregulates both constitutive and PMA-stimulated uPAR mRNA and protein. This occurs through suppression of upstream ERK cascade targets, regardless of whether cells were treated with exogenous bikunin or transfected with the bikunin gene. PMID: 12180971
  28. Urinary levels of bikunin provide insights into proteinase/proteinase inhibitor imbalance in patients with inflammatory diseases. PMID: 12211652
  29. Bikunin was colocalized with tryptase in dermal mast cells, and a small quantity of bikunin was also deposited in the intercellular spaces in FE and ADL. PMID: 12522575
  30. Research suggests a distinct regulation of hepatocyte growth factor activator inhibitor type 2 gene expression in the testis. This indicates that HAI-2 may play a role in spermatogenesis. PMID: 12553733
  31. Overexpression of bikunin is associated with suppression of invasion and peritoneal carcinomatosis of ovarian cancer cell lines. PMID: 12569552
  32. Studies have investigated the cDNA microarray analysis of gene expression regulated by bikunin in an ovarian cell line, including the pattern of gene expression leading to a block in cell invasion. PMID: 12571229
  33. Research demonstrates that the expression of HAI-2/PB is under methylation control to a variable extent in glioma cell lines, compared to other tested neoplasm cell lines (brain, breast, prostate, liver). PMID: 14558597
  34. Low levels of HAI-2 are associated with breast cancer. PMID: 14734471
  35. The use of high hydrostatic pressure (1000-3000 bar) for the refolding of bikunin aggregates has been investigated. PMID: 15388859
  36. Bikunin suppresses lipopolysaccharide-induced lethality through down-regulation of tumor necrosis factor-alpha and interleukin-1 beta in macrophages. PMID: 15717269
  37. Epigenetic inactivation of HAI-2/SPINT2 leads to loss of RCC tumor suppressor activity and implicates abnormalities of the MET pathway in clear cell and papillary sporadic RCC. PMID: 15930277
  38. There are no significant differences in serum HAI-2 levels among prostate cancer subgroups according to clinical stage. PMID: 16353247
  39. Hepatocyte growth factor activator inhibitor 2 (HAI-2) is regulated by hypoxia in breast cancer. PMID: 17255277
  40. HAI2 hypermethylation is associated with hepatocellular carcinoma. PMID: 18358501
  41. Low HAI-2 expression in cervical cancer may be associated with a poor prognosis. PMID: 18689863
  42. Unlike HAI-1 and matriptase, HAI-2 expression is detected in non-epithelial cells of the brain and lymph nodes. This suggests that HAI-2 may also be involved in the inhibition of serine proteases other than matriptase. PMID: 18713750
  43. Data supports the role of SPINT2 as a putative tumor suppressor gene in medulloblastoma. Further research implicates dysregulation of the HGF/MET signaling pathway in the pathogenesis of medulloblastoma. PMID: 19047176
  44. HAI-2 is a candidate tumor suppressor gene that is frequently hypermethylated and underexpressed in human HCCs. The KD-1 domain of HAI-2 is the key region responsible for its anti-invasive function. PMID: 19107935
  45. HAI-1 and HAI-2 may potentially be therapeutic targets for treatment approaches in ovarian cancer. PMID: 19148468
  46. Syndromic congenital sodium diarrhea is a distinct disease entity caused by SPINT2 loss-of-function mutations. PMID: 19185281
  47. The active form of HGFA is specifically complexed with the membrane-form HAI-1 (serine peptidase inhibitor, Kunitz type 1), but not with HAI-2/PB (serine peptidase inhibitor, Kunitz type 2), on the surface of epithelial cells expressing both inhibitors. PMID: 11013244
  48. Frequent hypermethylation of the SPINT2 gene is observed in human hepatocellular carcinoma. PMID: 14695180

Show More

Hide All

Database Links

HGNC: 11247

OMIM: 270420

KEGG: hsa:10653

STRING: 9606.ENSP00000301244

UniGene: Hs.31439

Involvement In Disease
Diarrhea 3, secretory sodium, congenital, with or without other congenital anomalies (DIAR3)
Subcellular Location
Membrane; Single-pass type I membrane protein.
Tissue Specificity
Expressed in placenta, kidney, pancreas, prostate, testis, thymus, and trachea.

Q&A

What is SPINT2 and what are its primary biological functions?

SPINT2 (Serine Peptidase Inhibitor, Kunitz Type 2) is a 252 amino acid protein with a molecular weight of approximately 28.2 kDa that functions as a Kunitz-type serine protease inhibitor . The protein is primarily membrane-localized and exhibits inhibitory activity against several serine proteases, most notably TMPRSS2 (Transmembrane Serine Protease 2) and HGF activator .

The biological significance of SPINT2 includes:

  • Regulation of epithelial cell barrier function through inhibition of matriptase

  • Modulation of HGF (Hepatocyte Growth Factor) signaling through inhibition of HGF activator

  • Control of TMPRSS2 activity, which has implications for viral entry mechanisms

  • Tissue-specific expression patterns, with notable presence in placenta, kidney, pancreas, prostate, testis, thymus, and trachea

The protein undergoes post-translational modification, particularly glycosylation, which may affect its inhibitory function . Up to two different isoforms have been reported, suggesting tissue-specific roles for different SPINT2 variants .

How is SPINT2 expression regulated at the transcriptional level?

SPINT2 expression exhibits a tightly regulated balance with its target proteases, particularly TMPRSS2. Research has revealed common transcription factors associated with genomic loci for both SPINT2 and TMPRSS2 genes, explaining their correlated expression patterns across different cell types .

Transcriptional regulation analysis shows:

  • Coregulation exists between SPINT2 and TMPRSS2, with consistent correlation across various cell types

  • The highest correlation between SPINT2 and TMPRSS2 expression is observed in tissues that are targets for SARS-CoV-2 infection

  • At single-cell resolution, both genes demonstrate specific co-expression in multiple cell types, corroborating the inferred coregulation

This transcriptional coregulation has significant implications for understanding how cells maintain protease/inhibitor balance in different physiological and pathological contexts.

What research methods are typically used to study SPINT2 protein interactions?

To investigate SPINT2 interactions with target proteases like TMPRSS2, researchers employ several methodological approaches:

  • Co-immunoprecipitation assays: To detect physical interactions between SPINT2 and its target proteases in cell lysates

  • Enzymatic inhibition assays: Measuring protease activity in the presence of recombinant SPINT2 or cellular extracts with varying SPINT2 expression

  • Knockdown and overexpression studies: As demonstrated in the SARS-CoV-2 research, shRNA-mediated SPINT2 knockdown and overexpression systems help evaluate functional consequences of altered SPINT2 levels

  • Pharmacological inhibition: Using specific inhibitors like Camostat mesylate to block TMPRSS2 activity can help determine if SPINT2 effects are mediated through TMPRSS2 inhibition

  • Western blot analysis: To monitor expression levels of SPINT2 and potential interaction partners

In combination, these methods provide robust evidence for SPINT2's inhibitory functions and biological significance.

What criteria should researchers consider when selecting a SPINT2 antibody for their experiments?

When selecting a SPINT2 antibody for research applications, consider the following critical factors:

  • Application compatibility: Verify the antibody has been validated for your specific application (Western blot, ELISA, IHC, IF)

  • Species reactivity: Confirm reactivity with your experimental model species. Available SPINT2 antibodies may react with human, mouse, bovine, or other species

  • Epitope recognition: Consider whether you need an antibody targeting a specific region (e.g., C-terminal, middle region) which may be important if studying specific isoforms

  • Clonality: Monoclonal antibodies offer high specificity for a single epitope, while polyclonal antibodies recognize multiple epitopes, potentially providing stronger signals

  • Conjugation requirements: Determine if you need unconjugated antibodies or those with specific conjugates (HRP, biotin) based on your detection system

  • Validation evidence: Check for published citations demonstrating successful use in applications similar to yours

  • Isoform specificity: Since SPINT2 has up to two reported isoforms, ensure the antibody can detect your isoform of interest

What validation methods should be employed to confirm SPINT2 antibody specificity?

Thorough validation of SPINT2 antibodies is essential for reliable experimental results. Implement these validation approaches:

  • Positive and negative control samples: Use tissues or cell lines with known SPINT2 expression (placenta, kidney, pancreas) as positive controls, and non-expressing tissues as negative controls

  • Recombinant protein controls: Test antibody against purified recombinant SPINT2 protein in Western blot or ELISA

  • Knockdown/knockout validation: Compare antibody reactivity in wild-type cells versus SPINT2 knockdown cells, as demonstrated in the SARS-CoV-2 study where SPINT2 knockdown validation was performed at both transcript and protein levels

  • Cross-reactivity assessment: Test against related Kunitz-type inhibitors to ensure specificity

  • Peptide competition assays: Pre-incubate antibody with immunizing peptide to demonstrate signal specificity

  • Multiple antibody comparison: Use antibodies targeting different epitopes of SPINT2 to confirm consistent detection patterns

  • Molecular weight verification: Confirm detection at the expected molecular weight (approximately 28.2 kDa for the canonical form, with potential variation due to glycosylation)

What are the optimal protocols for SPINT2 detection in Western blot applications?

For optimal Western blot detection of SPINT2, follow these methodological considerations:

  • Sample preparation:

    • Use RIPA buffer supplemented with protease inhibitors

    • Include phosphatase inhibitors if studying phosphorylated forms

    • Denature samples at 95°C for 5 minutes in reducing conditions

  • Gel selection and transfer:

    • Use 10-12% SDS-PAGE gels for optimal separation

    • Transfer to PVDF membranes (preferred over nitrocellulose for glycosylated proteins)

    • Transfer at 100V for 60-90 minutes in cold transfer buffer containing 20% methanol

  • Blocking and antibody incubation:

    • Block with 5% non-fat milk in TBST for 1 hour at room temperature

    • Incubate with primary SPINT2 antibody at 1:500-1:1000 dilution overnight at 4°C

    • Wash extensively with TBST (at least 3×10 minutes)

    • Incubate with HRP-conjugated secondary antibody at 1:5000 dilution for 1 hour

  • Detection considerations:

    • Due to glycosylation, SPINT2 may appear at a higher molecular weight than predicted

    • Use enhanced chemiluminescence (ECL) detection systems

    • Include positive control lysates from cells with known SPINT2 expression

  • Troubleshooting guidance:

    • If detecting multiple bands, confirm specificity with knockdown studies

    • For weak signals, extend primary antibody incubation time or increase concentration

    • For high background, increase washing steps or decrease antibody concentration

How can researchers optimize immunohistochemistry protocols for SPINT2 detection in tissue samples?

Optimizing immunohistochemistry (IHC) for SPINT2 detection requires specific attention to several parameters:

  • Tissue fixation and processing:

    • Use 10% neutral-buffered formalin for fixation (24-48 hours)

    • Paraffin embedding should follow standard protocols

    • Cut sections at 4-5 μm thickness for optimal staining

  • Antigen retrieval methods:

    • Heat-induced epitope retrieval (HIER) in citrate buffer (pH 6.0) is generally effective

    • Pressure cooking for 3-5 minutes typically provides sufficient retrieval

    • Allow slides to cool in retrieval solution for 20 minutes before proceeding

  • Blocking parameters:

    • Block endogenous peroxidase activity with 3% hydrogen peroxide for 10 minutes

    • Block non-specific binding with 5% normal serum from the species of secondary antibody

    • Include avidin-biotin blocking if using biotinylated detection systems

  • Antibody selection and dilution:

    • Test antibodies validated specifically for IHC applications

    • Start with manufacturer's recommended dilution and optimize as needed

    • Incubate primary antibody overnight at 4°C for best results

  • Detection systems:

    • Polymer-based detection systems often provide cleaner backgrounds than ABC methods

    • DAB (3,3'-diaminobenzidine) is the most common chromogen for visualization

    • Consider double staining with other markers if studying co-localization

Include positive control tissues known to express SPINT2 (placenta, kidney, pancreas) and implement appropriate negative controls (primary antibody omission, non-immune IgG substitution).

What methods should be used to quantify SPINT2 expression levels in cell and tissue samples?

Accurate quantification of SPINT2 expression requires selecting appropriate methods based on research questions:

  • Western blot quantification:

    • Use densitometry software (ImageJ, Image Lab) to analyze band intensity

    • Normalize SPINT2 signal to loading controls (β-actin, GAPDH, or total protein)

    • Include concentration standards if absolute quantification is needed

    • Present results as fold-change relative to control samples

  • qRT-PCR for transcript quantification:

    • Design primers spanning exon-exon junctions to avoid genomic DNA amplification

    • Normalize to validated reference genes stable in your experimental conditions

    • Calculate expression using the 2^-ΔΔCt method for relative quantification

  • ELISA-based quantification:

    • Commercial ELISA kits are available for SPINT2 quantification

    • Develop standard curves using recombinant SPINT2 protein

    • Consider sandwich ELISA format using two antibodies recognizing different epitopes

  • Immunohistochemistry quantification:

    • Use digital image analysis software for IHC staining quantification

    • Score based on staining intensity and percentage of positive cells

    • Consider H-score system (0-300) combining intensity and positivity percentage

    • Implement automated systems with machine learning algorithms for unbiased assessment

  • Single-cell analysis:

    • Flow cytometry can quantify SPINT2 at single-cell resolution

    • Single-cell RNA-seq can reveal expression heterogeneity within populations

How can researchers study the functional relationship between SPINT2 and TMPRSS2 in experimental systems?

Investigating the SPINT2-TMPRSS2 regulatory axis requires sophisticated experimental approaches:

  • Co-expression analysis:

    • Quantify SPINT2 and TMPRSS2 expression correlation across tissues and cell types

    • Analyze single-cell RNA-seq data to identify co-expressing populations

    • Implement fluorescent dual-labeling to visualize co-expression at protein level

  • Genetic manipulation strategies:

    • Generate SPINT2 knockdown models using shRNA or siRNA approaches

    • Create SPINT2 overexpression systems with tagged constructs

    • Implement CRISPR/Cas9 for complete knockout studies

    • Develop inducible expression systems for temporal control of SPINT2 levels

  • Protease activity measurements:

    • Use fluorogenic peptide substrates specific for TMPRSS2 to measure activity

    • Compare protease activity in SPINT2-manipulated versus control cells

    • Implement live-cell imaging with protease-activated reporters

  • Binding interaction studies:

    • Perform co-immunoprecipitation to demonstrate physical interaction

    • Use surface plasmon resonance (SPR) to measure binding kinetics

    • Implement proximity ligation assays (PLA) to visualize interactions in situ

  • Functional readouts:

    • In viral infection models, measure viral entry and replication as functional readouts

    • For HGF signaling, assess downstream phosphorylation events (e.g., c-Met phosphorylation)

    • Monitor epithelial barrier integrity in cell models with SPINT2 manipulation

Research has shown that SPINT2 knockdown increases TMPRSS2 gene expression compared to wild-type or scramble control cells, suggesting a feedback regulatory mechanism .

What are the common challenges and solutions when working with SPINT2 antibodies in various applications?

Researchers frequently encounter these challenges when working with SPINT2 antibodies:

ChallengePotential CausesRecommended Solutions
Multiple bands in Western blot- Post-translational modifications
- Splice variants
- Non-specific binding
- Protein degradation
- Confirm with positive controls
- Perform peptide competition
- Use fresh samples with protease inhibitors
- Try antibodies targeting different epitopes
Weak or no signal in Western blot- Low SPINT2 expression
- Inefficient extraction
- Antibody sensitivity issues
- Increase protein loading (50-100μg)
- Try membrane enrichment protocols
- Extend exposure time
- Use signal enhancement systems
High background in IHC/IF- Non-specific binding
- Insufficient blocking
- Overfixation
- Increase blocking time/concentration
- Optimize antibody dilution
- Include detergent in wash buffers
- Try different antigen retrieval methods
Inconsistent results between applications- Epitope accessibility differs
- Application-specific requirements
- Select antibodies validated for specific applications
- Use different antibodies for different applications
- Optimize protocols for each application
Discrepancies between mRNA and protein levels- Post-transcriptional regulation
- Protein stability differences
- Analyze both mRNA and protein
- Include time-course studies
- Consider using proteasome inhibitors
Cross-reactivity with other Kunitz inhibitors- Epitope similarity
- Non-specific binding
- Perform specificity controls
- Use knockout/knockdown validation
- Select antibodies with verified specificity

How can researchers implement advanced imaging techniques to study SPINT2 localization and trafficking?

Advanced imaging approaches provide crucial insights into SPINT2 dynamics:

  • Super-resolution microscopy techniques:

    • Structured Illumination Microscopy (SIM) achieves 100nm resolution for detailed membrane localization

    • Stochastic Optical Reconstruction Microscopy (STORM) provides 20nm resolution for precise colocalization studies

    • Stimulated Emission Depletion (STED) microscopy allows for live-cell super-resolution imaging

  • Live-cell imaging strategies:

    • Generate SPINT2-fluorescent protein fusions (e.g., SPINT2-GFP) for real-time visualization

    • Implement photoactivatable or photoconvertible tags for pulse-chase imaging

    • Use fluorescence recovery after photobleaching (FRAP) to measure membrane dynamics

  • Proximity-based interaction studies:

    • Förster Resonance Energy Transfer (FRET) to study SPINT2-TMPRSS2 interactions

    • Proximity Ligation Assay (PLA) to visualize protein-protein interactions with standard microscopy

    • Bimolecular Fluorescence Complementation (BiFC) for direct visualization of interacting proteins

  • Correlative light and electron microscopy (CLEM):

    • Combine fluorescence imaging of SPINT2 with ultrastructural context

    • Implement immunogold labeling for electron microscopy visualization

    • Use cryo-electron microscopy for near-native state visualization

  • Multi-parametric imaging:

    • Co-stain with organelle markers to determine subcellular localization

    • Implement spectral unmixing for multi-protein localization studies

    • Use automated high-content imaging for quantitative localization analysis

These advanced techniques require careful validation with appropriate controls and may benefit from computational image analysis for quantitative assessment.

How is SPINT2 dysregulation implicated in viral infections, particularly SARS-CoV-2?

Research has revealed crucial roles for SPINT2 in viral infection processes:

  • SARS-CoV-2 infection mechanisms:

    • SPINT2 negatively correlates with SARS-CoV-2 expression in Calu-3 and Caco-2 cell lines

    • SPINT2 knockdown significantly increases viral load in experimental systems

    • SPINT2 overexpression leads to dramatic reduction in viral load

    • The mechanism appears to involve inhibition of TMPRSS2, which is required for S protein priming

  • Experimental evidence:

    • In Calu-3 cells, SPINT2 knockdown resulted in more than two-fold increase in SARS-CoV-2 positive cells

    • SPINT2 knockdown was associated with increased viral genome replication

    • Treatment with TMPRSS2 inhibitor (Camostat mesylate) abrogated the effects of SPINT2 knockdown

    • Similar results were observed in A549 cells overexpressing the SARS-CoV-2 receptor ACE2

  • Clinical correlations:

    • SPINT2 was found to be down-regulated in secretory cells from COVID-19 patients

    • This suggests SPINT2 could potentially serve as a biomarker for disease susceptibility

    • The relationship may explain some COVID-19 comorbidities in patients with conditions where SPINT2 is downregulated

These findings highlight SPINT2 as a potential therapeutic target for viral infections dependent on TMPRSS2 for entry.

What is the relationship between SPINT2 expression and various pathological conditions?

SPINT2 dysregulation has been associated with multiple pathological conditions:

  • Cancer associations:

    • SPINT2 is down-regulated across different types of tumors

    • Particularly evident in colon, kidney, and liver tumors

    • May function as a tumor suppressor through inhibition of proteases involved in invasion and metastasis

  • Metabolic disorders:

    • Down-regulation observed in alpha pancreatic islet cells from Type 2 diabetes patients

    • This may contribute to the comorbidity observed between diabetes and COVID-19

  • Gastrointestinal disorders:

    • SPINT2 gene has been associated with diarrheal diseases (DIAR3)

    • May involve dysregulation of epithelial barrier function

  • Infectious disease implications:

    • SPINT2 down-regulation may increase susceptibility to certain viral infections

    • This relationship is particularly evident in SARS-CoV-2 infection models

Research into these associations suggests SPINT2 expression could serve as both a biomarker and potential therapeutic target across multiple disease contexts.

How can researchers design experiments to investigate SPINT2 as a potential therapeutic target?

Developing SPINT2-focused therapeutic strategies requires systematic experimental approaches:

  • Target validation experiments:

    • Conduct dose-response studies with recombinant SPINT2 protein in disease models

    • Implement genetic rescue experiments in systems with SPINT2 deficiency

    • Use conditional knockout models to establish tissue-specific requirements

  • Therapeutic modulation strategies:

    • Design small molecule compounds that enhance SPINT2 stability or function

    • Develop peptide mimetics based on SPINT2 active domains

    • Explore gene therapy approaches to restore SPINT2 expression

    • Identify compounds that upregulate endogenous SPINT2 expression

  • Delivery system development:

    • Test tissue-specific delivery methods for SPINT2-based therapeutics

    • Explore nanoparticle formulations for targeted delivery

    • Investigate mRNA-based approaches for transient SPINT2 restoration

  • Efficacy assessment protocols:

    • Establish clear readouts for therapeutic efficacy in disease models

    • For viral infections, measure viral entry and replication metrics

    • For cancer applications, assess changes in invasion and metastasis

    • In metabolic disorders, monitor tissue-specific functional improvements

  • Safety and specificity assessment:

    • Evaluate off-target effects through protease activity profiling

    • Assess impact on related biological pathways

    • Implement toxicity screening in relevant cell and animal models

  • Combination therapy exploration:

    • Test SPINT2-based approaches with existing therapies

    • For SARS-CoV-2, combine with direct-acting antivirals or TMPRSS2 inhibitors

    • In cancer, combine with conventional chemotherapy or immunotherapy

These experimental approaches should be tailored to specific disease contexts while maintaining focus on SPINT2's established biological functions.

What emerging technologies could advance our understanding of SPINT2 biology and function?

Several cutting-edge technologies hold promise for deeper insights into SPINT2 biology:

  • Spatial transcriptomics and proteomics:

    • Map SPINT2 expression patterns with cellular resolution in intact tissues

    • Correlate with expression of target proteases and disease markers

    • Identify previously unknown tissue niches with significant SPINT2 activity

  • Protein structure determination:

    • Apply cryo-electron microscopy to resolve SPINT2-protease complexes

    • Implement AlphaFold or RoseTTAFold for computational structure prediction

    • Use hydrogen-deuterium exchange mass spectrometry to map interaction surfaces

  • Single-cell multi-omics approaches:

    • Combine transcriptomics, proteomics, and epigenomics at single-cell level

    • Identify regulatory networks controlling SPINT2 expression

    • Map trajectories of SPINT2 expression changes during disease progression

  • Organoid and microphysiological systems:

    • Develop tissue-specific organoids to study SPINT2 function in near-physiological contexts

    • Implement organ-on-chip technologies for dynamic functional studies

    • Create disease models incorporating patient-derived cells

  • CRISPR screening approaches:

    • Perform genome-wide CRISPR screens to identify modulators of SPINT2 expression

    • Use CRISPRi/CRISPRa for targeted modulation of SPINT2 regulatory elements

    • Implement base editing for precise modification of SPINT2 sequence

These technologies will enable more comprehensive understanding of SPINT2 biology and potentially reveal new therapeutic opportunities.

How can researchers better understand the tissue-specific roles of SPINT2 in normal physiology?

Investigating tissue-specific SPINT2 functions requires specialized methodological approaches:

  • Tissue-specific conditional knockout models:

    • Generate floxed SPINT2 alleles for Cre-mediated tissue-specific deletion

    • Implement inducible systems for temporal control of SPINT2 deletion

    • Analyze phenotypic consequences in multiple organ systems

  • Cell type-specific transcriptomic profiling:

    • Apply single-cell RNA sequencing to tissues with significant SPINT2 expression

    • Identify cell populations with highest SPINT2 expression

    • Map co-expression networks to infer tissue-specific functions

  • Organotypic culture systems:

    • Develop 3D culture models recapitulating tissue architecture

    • Manipulate SPINT2 expression in these systems

    • Assess functional outcomes relevant to specific tissues

  • In vivo imaging approaches:

    • Generate SPINT2 reporter mouse models for in vivo visualization

    • Implement intravital microscopy to observe dynamic changes

    • Correlate SPINT2 expression with tissue function in real-time

  • Targeted proteomics:

    • Identify tissue-specific SPINT2 interacting partners

    • Characterize post-translational modifications across tissues

    • Determine if different SPINT2 isoforms predominate in specific tissues

These approaches will help define the physiological roles of SPINT2 beyond its known pathological associations, potentially revealing new therapeutic opportunities.

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 2025 TheBiotek. All Rights Reserved.