SPRR1A Antibody

Shipped with Ice Packs
In Stock

Description

Overview of SPRR1A Antibody

SPRR1A Antibody is a research reagent designed to detect the small proline-rich protein 1A (SPRR1A), a member of the SPRR family involved in diverse biological processes, including antimicrobial defense, cytoprotection, and cancer progression. This antibody is used in immunological assays such as Western blotting, immunofluorescence (IF), and immunohistochemistry (IHC) to study SPRR1A expression in tissues, cells, and disease models .

Applications in Research

SPRR1A Antibody is employed in various experimental contexts to elucidate SPRR1A’s roles in health and disease:

ApplicationMethodKey FindingsSource
Cancer PrognosisIHC, Western BlotHigh SPRR1A expression correlates with poor survival in colon, pancreatic, and osteosarcoma cancers .
Antimicrobial DefenseWestern Blot, IFSPRR1A binds bacterial membranes, disrupting pathogens like MRSA and P. aeruginosa .
Cardiomyocyte ProtectionImmunoblottingSPRR1A stabilizes myofibrils during ischemic stress, preventing cell death .
Nervous System InjuryIF, ImmunoblottingSPRR1A is upregulated in axotomized neurons, aiding axonal regeneration .

4.1. Prognostic Biomarker

High SPRR1A expression is linked to aggressive phenotypes and poor survival in multiple cancers:

Cancer TypeSPRR1A ExpressionClinical ImpactSource
Colon Cancer↑ mRNA in tumorsPoor OS and DFS; independent predictor of mortality .
Pancreatic Ductal Adenocarcinoma (PDAC)↑ in 35% of casesCorrelates with squamous differentiation markers .
Osteosarcoma↑ in MG-OKS cellsKnockdown reduces proliferation and migration .

4.2. Functional Studies

In osteosarcoma models, siRNA-mediated SPRR1A knockdown:

  • Reduced Proliferation: 79% decrease in cell viability (48 hr, WST-8 assay) .

  • Impaired Migration: 70% reduction in wound-healing ability .

  • Tumor Growth: 64% smaller xenograft volumes (4 weeks post-injection) .

5.1. Skin and Immune Defense

SPRR1A binds negatively charged bacterial membranes, inducing lysis of MRSA and P. aeruginosa . Antibodies confirm SPRR1A’s localization in sebocytes and keratinocytes, critical for antimicrobial activity .

5.2. Cardiac Stress Response

In ischemic cardiomyocytes, SPRR1A overexpression:

  • Mitigates Myofibril Disruption: Preserves cytoskeletal integrity .

  • Reduces Apoptosis: TUNEL staining shows fewer apoptotic nuclei .

Limitations and Future Directions

  • Specificity: Potential cross-reactivity with other SPRR family members (e.g., SPRR1B) requires validation .

  • Therapeutic Potential: SPRR1A inhibition could target aggressive cancers, but in vivo efficacy remains untested .

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Product shipment typically occurs within 1-3 business days of order receipt. Delivery times may vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
SPRR1A antibody; Cornifin-A antibody; 19 kDa pancornulin antibody; SPRK antibody; Small proline-rich protein IA antibody; SPR-IA antibody
Target Names
SPRR1A
Uniprot No.

Target Background

Function

This antibody targets the cross-linked envelope protein of keratinocytes. Specifically, it recognizes a keratinocyte protein initially present in the cell cytosol. This protein subsequently undergoes cross-linking to membrane proteins via transglutaminase, resulting in the formation of an insoluble envelope beneath the plasma membrane.

Gene References Into Functions

Related Research and SPRR1A Expression:

  1. Elevated mRNA levels of LELP1, SPRR1Av1, and SPRR1Av2 were observed in atopic dermatitis (AD) patients' skin compared to controls. PMID: 27304082
  2. SPRR1A expression may significantly influence the prognosis of postmenopausal breast carcinoma, particularly in progesterone receptor-positive patients. PMID: 25424702
  3. SPRR1A expression may serve as a prognostic marker for diffuse large B-cell lymphoma. PMID: 24886019
  4. Studies have revealed heterogeneity in the association between psoriasis and PGLYRP and SPRR genes at the PSORS4 locus (1q) across Finnish, Swedish, and Irish populations. PMID: 18643845
  5. Increased SPRR1A expression in chronic atopic dermatitis lesions suggests an insufficient rise in SPRR transcripts to compensate for the lack of LOR, contributing to the persistence of these lesions. PMID: 19672094
Database Links

HGNC: 11259

OMIM: 182265

KEGG: hsa:6698

STRING: 9606.ENSP00000307340

UniGene: Hs.46320

Protein Families
Cornifin (SPRR) family
Subcellular Location
Cytoplasm.

Q&A

What is SPRR1A and why is it relevant to study in different disease models?

SPRR1A (Small Proline-Rich Protein 1A) is a stress-inducible protein that serves multiple biological functions depending on cell type and physiological context. In cardiac tissue, SPRR1A functions as a downstream mediator of gp130 cytokines and provides significant cardioprotection against ischemic injury . In the context of cancer, SPRR1A is recognized as a squamous differentiation marker but is also upregulated in certain non-squamous cancers, with high expression correlating with poor prognosis in pancreatic ductal adenocarcinoma . Additionally, SPRR1A has been identified as an antimicrobial peptide in the skin, exhibiting bactericidal activity against pathogens like MRSA and P. aeruginosa through bacterial membrane disruption . The diverse functions of SPRR1A across multiple tissue types and disease states make it a compelling target for research across cardiology, oncology, and dermatology fields.

What are the optimal sample preparation techniques for SPRR1A immunohistochemistry?

For optimal SPRR1A detection in immunohistochemistry applications, tissue samples should be properly fixed and processed according to standard histological protocols. Based on validated protocols for SPRR1A antibody applications, antigen retrieval is a critical step that significantly impacts staining quality. The recommended approach is to use TE buffer at pH 9.0 for antigen retrieval, although citrate buffer at pH 6.0 can serve as an alternative . For paraffin-embedded sections, deparaffinization and rehydration should be performed prior to antigen retrieval. The optimal antibody dilution range for immunohistochemical applications is 1:200-1:800, though this should be empirically determined for each specific application and tissue type . Positive control tissues should include human esophageal cancer tissue, where SPRR1A expression has been consistently demonstrated .

How can I confirm the specificity of SPRR1A antibody in my experimental system?

Confirming SPRR1A antibody specificity requires multiple validation approaches. First, include appropriate positive controls known to express SPRR1A, such as esophageal cancer tissue or stressed cardiomyocytes following ischemic injury . Negative controls should include tissues where SPRR1A is not normally expressed, such as untreated C57Bl/6 mouse hearts where SPRR1A mRNA is not detectable under normal conditions .

For more definitive validation, consider the following approaches:

  • Peptide competition assays where pre-incubation of the antibody with purified SPRR1A protein should abolish specific staining

  • Comparing staining patterns with multiple antibodies targeting different SPRR1A epitopes

  • Correlation of protein detection with mRNA expression using qRT-PCR

  • Using tissues or cells from SPRR1A knockout models (Sprr1a−/−) as negative controls

The specificity of SPRR1A staining can be confirmed as demonstrated in cardiac tissue studies where negative staining was observed after coincubation of antibody with absorbing antigen SPRR1A protein .

How should I design experiments to study SPRR1A induction in stress models?

Designing rigorous experiments to study SPRR1A induction in stress models requires careful consideration of temporal dynamics and tissue specificity. Based on cardiac research models, SPRR1A shows distinctive temporal expression patterns, with mRNA being tightly regulated at the transcriptional level while the protein demonstrates a remarkably long half-life in myocardial tissue .

When designing your experimental timeline, consider that in TAC (transverse aortic constriction) models, SPRR1A mRNA was detectable only 4 days after surgery, while the protein was continuously observed from 4 days to 2 weeks post-procedure . This suggests experiments should include multiple time points spanning acute and chronic phases of stress response.

For stress induction models, consider:

  • Cardiac models: TAC surgery, myocardial infarction, or in vitro ischemia using 2-deoxyglucose treatment

  • Cancer models: Hypoxic conditions, nutrient deprivation, or chemotherapeutic stress

  • Skin models: Bacterial challenge with MRSA or P. aeruginosa

Control conditions should include physiological hypertrophy, cardiac hypertrophy related to the PI-3K pathway, and congenital cardiomyopathy models where SPRR1A is not normally induced . Analysis should combine mRNA quantification (qRT-PCR) with protein detection (Western blot and immunohistochemistry) to capture the full dynamics of SPRR1A regulation.

What are the technical challenges in detecting SPRR1A protein localization in tissue sections?

Detecting SPRR1A protein localization in tissue sections presents several technical challenges that researchers should anticipate and address. First, SPRR1A expression often occurs in highly localized, disseminated foci rather than homogeneously throughout tissue, as observed in ventricular myocardium after TAC . This patchy distribution necessitates comprehensive section sampling and analysis to avoid false negatives.

The relatively small size of SPRR1A protein (89 amino acids, approximately 10 kDa) may require optimized fixation protocols to prevent antigen loss during processing . Overfixation can mask epitopes, while underfixation may lead to tissue degradation and false localization.

Background staining can be problematic, particularly in tissues with high endogenous peroxidase activity. Effective blocking steps and careful titration of primary antibody (recommended range: 1:200-1:800) are essential . Additionally, autofluorescence can interfere with immunofluorescence detection, necessitating appropriate quenching steps.

In cardiac tissue specifically, the accumulation of SPRR1A protein along myofibrils has been observed in cardiomyocytes , requiring high-resolution imaging to accurately discern subcellular localization patterns. For dual labeling experiments to co-localize SPRR1A with other markers, careful antibody selection is necessary to avoid cross-reactivity while ensuring compatible species origin and detection systems.

How can I optimize SPRR1A antibody-based assays to study its antimicrobial functions?

Studying SPRR1A's antimicrobial functions requires specialized experimental approaches that integrate immunological techniques with microbiological methods. Based on research demonstrating SPRR1A's bactericidal activity against MRSA and P. aeruginosa , several optimization strategies are recommended:

For membrane disruption studies:

  • Propidium iodide (PI) uptake assay: Optimize bacterial concentration (108 CFU/ml recommended) and incubate with purified SPRR1A protein at various concentrations (effective range: low micromolar). Monitor PI fluorescence over time using flow cytometry or fluorescence microscopy to quantify membrane permeabilization .

  • Lipid binding assays: Use lipid strips dotted with different lipids to assess SPRR1A binding specificity. Include both negatively charged lipids (to which SPRR1A preferentially binds) and neutral lipids as controls .

  • Liposome dye release assay: Prepare liposomes composed of phosphatidylcholine (PC) and phosphatidylserine (PS) (85%:15% ratio to mimic bacterial membranes) loaded with carboxyfluorescein. Monitor dye efflux spectrofluorometrically after SPRR1A addition to quantify membrane disruption activity .

For in vivo antimicrobial function:

  • Bacterial challenge models using wild-type versus Sprr1a−/−;Sprr2a−/− mice to assess susceptibility to skin infection

  • Quantification of bacterial burden in skin using CFU counting

  • Histological assessment of infection severity and inflammatory response

When using antibodies to detect SPRR1A in infected tissue, adjust dilution factors (consider starting at 1:400) and be aware that inflammation may alter tissue architecture and potentially affect antibody access and specificity .

How does SPRR1A expression correlate with clinical outcomes in cancer studies?

Multivariate analysis confirmed that high SPRR1A expression is an independent prognostic factor in PDAC (HR 1.706, 95% CI 1.018 to 2.862, p = 0.0427), alongside residual tumor status . These findings were further validated through analysis of TCGA transcriptome data, which similarly demonstrated poorer prognosis in the high-SPRR1A-expression group .

When designing immunohistochemical studies to investigate SPRR1A as a prognostic marker, researchers should:

  • Establish clear scoring criteria for SPRR1A expression levels

  • Include appropriate controls, particularly squamous differentiation cases which naturally express SPRR1A

  • Correlate expression with comprehensive clinical data including treatment modalities

  • Consider combining SPRR1A with other prognostic markers for improved stratification

While SPRR1A overexpression did not significantly alter in vitro cancer cell phenotypes (proliferation, chemoresistance, EMT, migration), its strong association with clinical outcomes suggests it may serve as a valuable biomarker in PDAC .

How can SPRR1A antibodies be used to investigate cardioprotective mechanisms?

SPRR1A antibodies serve as critical tools for investigating cardioprotective mechanisms in ischemic heart disease models. Research has established SPRR1A as a stress-inducible downstream mediator of gp130 cytokines in cardiomyocytes with significant protective effects against ischemic stress . To effectively study these mechanisms, several methodological approaches utilizing SPRR1A antibodies are recommended:

For localization studies:

  • Immunohistochemical analysis of SPRR1A expression in cardiac tissue sections, focusing on regions adjacent to myocardial injuries. This reveals the spatial distribution pattern which typically appears as disseminated foci throughout the ventricles rather than homogeneous expression .

  • Immunofluorescence co-localization with myofibrillar markers to confirm accumulation of SPRR1A protein along myofibrils, which appears to be important for its protective function .

For functional mechanism studies:

  • Combine SPRR1A antibody detection with markers of apoptosis (TUNEL staining) and cell viability (calcein AM/ethidium homodimer) in cardiomyocytes subjected to ischemic stress models .

  • Use SPRR1A antibodies to track protein expression following in vivo gene transfer experiments, such as adenovirus-mediated SPRR1A overexpression, to confirm successful transduction prior to ischemia/reperfusion challenges .

  • Immunoblotting to quantify SPRR1A expression levels at different time points following ischemic injury, noting that while mRNA levels may fluctuate, protein persistence can extend significantly longer (from 4 days to 2 weeks after banding in TAC models) .

When analyzing cardiac protection, compare morphological preservation of myofibrils in SPRR1A-expressing cells versus controls, as SPRR1A appears to prevent myofibril disruption during ATP depletion . Additionally, quantify biomarkers of cardiac injury such as creatine kinase release to correlate with SPRR1A expression levels .

What are the key considerations for using SPRR1A antibodies in studying skin antimicrobial defense mechanisms?

When investigating SPRR1A's role in skin antimicrobial defense mechanisms, several key considerations should guide experimental design and interpretation. SPRR1A functions as an antimicrobial peptide in skin, with Sprr1a−/−;Sprr2a−/− mice showing increased susceptibility to MRSA and P. aeruginosa skin infections . These findings highlight SPRR1A's importance in cutaneous host defense.

For antimicrobial mechanism studies using SPRR1A antibodies:

  • Expression analysis in skin tissues:

    • Use immunohistochemistry with appropriate dilution (1:200-1:800) to detect SPRR1A in various skin layers

    • Consider differential expression between normal skin, infected skin, and sebaceous glands

    • LPS treatment significantly upregulates SPRR expression in sebocytes, suggesting a role in the response to Gram-negative bacteria

  • Functional studies:

    • Compare wild-type versus knockout models (Sprr1a−/−;Sprr2a−/−) for bacterial challenge responses

    • Quantify bacterial colonization and clearance rates

    • Analyze inflammatory responses in skin sections

  • Mechanistic investigations:

    • Use purified SPRR1A protein in conjunction with bacterial membrane permeability assays

    • Establish dose-dependent relationships between SPRR1A concentration and antimicrobial activity

    • Examine specificity against different pathogens (effective against MRSA and P. aeruginosa, but ineffective against E. coli)

  • Technical considerations:

    • Ensure antibody specificity through appropriate controls

    • For antimicrobial assays, purified SPRR1A protein rather than antibody detection should be used

    • Consider the interaction between SPRR1A and other antimicrobial peptides in skin

The antimicrobial function of SPRR1A operates through binding to negatively charged bacterial membrane lipids and subsequent membrane disruption . This mechanism suggests potential therapeutic applications for SPRR proteins as alternatives to traditional antibiotics for treating MRSA or P. aeruginosa skin infections .

How can I resolve inconsistent SPRR1A staining patterns in immunohistochemistry?

Inconsistent SPRR1A staining patterns in immunohistochemistry can arise from multiple sources and require systematic troubleshooting. First, consider the inherently heterogeneous expression pattern of SPRR1A, which appears in "disseminated foci" rather than uniform distribution, as documented in cardiac tissue . This natural distribution pattern may be mistaken for technical inconsistency.

To systematically address inconsistent staining issues:

  • Optimize fixation protocols:

    • Duration of fixation significantly impacts epitope accessibility

    • For formalin-fixed tissues, limit fixation to 24 hours

    • Consider alternative fixatives if formalin consistently yields poor results

  • Refine antigen retrieval:

    • SPRR1A antibody detection is highly dependent on proper antigen retrieval

    • Primary recommendation: TE buffer at pH 9.0

    • Alternative approach: Citrate buffer at pH 6.0

    • Optimize heating time and temperature for consistent epitope exposure

  • Antibody optimization:

    • Titrate antibody concentrations within recommended range (1:200-1:800)

    • Test multiple primary antibody incubation times and temperatures

    • Consider testing different antibody clones if persistent issues occur

  • Background reduction:

    • Implement rigorous blocking steps (3-5% BSA or normal serum)

    • Include blocking of endogenous peroxidase activity

    • Consider tyramide signal amplification for low expression samples

  • Validate with alternative detection methods:

    • Confirm expression patterns with RT-PCR or Western blot

    • Use multiple antibodies targeting different SPRR1A epitopes

    • Consider RNAscope or in situ hybridization to validate protein expression locations

When analyzing staining patterns, remember that SPRR1A expression can be dramatically induced by stress conditions while remaining undetectable in untreated controls , resulting in seemingly "all-or-nothing" expression patterns that reflect biology rather than technical inconsistency.

What approaches can resolve contradictory findings between SPRR1A mRNA and protein detection?

Discrepancies between SPRR1A mRNA and protein detection are not uncommon and can reflect important biological regulatory mechanisms rather than technical errors. Research in cardiac models has demonstrated that SPRR1A mRNA is tightly regulated at the transcriptional level and may be transiently expressed, while the protein demonstrates remarkable stability and persistence in tissues .

To address and interpret these apparent contradictions:

  • Temporal analysis considerations:

    • In TAC models, SPRR1A mRNA was only detectable 4 days after surgery, yet the protein was continuously observed from 4 days to 2 weeks

    • Design experiments with sequential timepoints that capture both early and late phases of response

    • Consider pulse-chase experiments to determine protein half-life

  • Technical validation approaches:

    • For mRNA detection, use multiple primer sets targeting different regions of the transcript

    • Employ absolute quantification with standard curves for mRNA detection

    • For protein detection, use antibodies targeting different epitopes

    • Include appropriate positive and negative controls for both mRNA and protein detection

  • Regulatory mechanism investigation:

    • Examine post-transcriptional regulation (miRNAs, RNA-binding proteins)

    • Investigate post-translational modifications affecting protein stability

    • Consider protein compartmentalization that might affect extraction efficiency

  • Sample preparation considerations:

    • Extract RNA and protein from the same tissue samples when possible

    • Be aware that protein extraction protocols might not efficiently solubilize all SPRR1A protein due to its potential association with insoluble cellular components

    • For tissues with heterogeneous expression, laser capture microdissection might be necessary to correlate mRNA and protein from the same cell populations

  • Quantification methods:

    • Use digital PCR for low-abundance transcript detection

    • Consider mass spectrometry for protein quantification as an antibody-independent method

When reporting contradictory findings, carefully describe the temporal relationship between mRNA and protein detection, as this discrepancy likely reflects important regulatory mechanisms controlling SPRR1A function in stress responses.

How can SPRR1A antibodies be integrated with advanced imaging techniques for mechanistic studies?

Integrating SPRR1A antibodies with advanced imaging techniques can provide unprecedented insights into this protein's cellular localization, molecular interactions, and functional dynamics. Several cutting-edge approaches can be implemented for comprehensive mechanistic studies:

  • Super-resolution microscopy:

    • Stimulated emission depletion (STED) microscopy or stochastic optical reconstruction microscopy (STORM) can resolve SPRR1A localization at nanometer resolution

    • Particularly valuable for visualizing SPRR1A accumulation along myofibrils in cardiomyocytes or interaction with bacterial membranes

    • Use directly conjugated SPRR1A antibodies (rather than secondary detection) to minimize spatial displacement

  • Live-cell imaging approaches:

    • Combine genetically encoded fluorescent SPRR1A fusions with antibody staining of fixed timepoints

    • Implementation of photoactivatable or photoconvertible SPRR1A fusions to track protein movement and turnover

    • Correlate with in situ SPRR1A antibody staining to validate fusion protein localization

  • Multi-modal correlative microscopy:

    • Correlative light and electron microscopy (CLEM) to visualize SPRR1A localization in relation to ultrastructural features

    • Particularly valuable for analyzing SPRR1A's interaction with bacterial membranes or myofibrillar structures

    • Implement immunogold labeling with SPRR1A antibodies for electron microscopy studies

  • Proximity labeling approaches:

    • BioID or APEX2 proximity labeling fused to SPRR1A to identify interacting proteins in living cells

    • Validate interactions using traditional co-immunoprecipitation with SPRR1A antibodies

    • Map interaction networks during stress response versus baseline conditions

  • Multiplexed imaging:

    • Cyclic immunofluorescence or mass cytometry imaging to simultaneously visualize SPRR1A and dozens of other proteins

    • Particularly valuable for analyzing SPRR1A in complex tissues like skin where multiple cell types contribute to antimicrobial defense

    • Implement machine learning approaches for pattern recognition in multiplexed datasets

When implementing these advanced techniques, careful validation with traditional SPRR1A antibody applications remains essential. The small size of SPRR1A (89 amino acids, 10 kDa) should be considered when designing fusion proteins to avoid interfering with native function and localization.

What emerging applications for SPRR1A antibodies show promise in translational research?

Several emerging applications for SPRR1A antibodies demonstrate significant translational potential across multiple disease areas. Based on current research findings, the following applications warrant further investigation:

  • Cancer biomarker development:

    • SPRR1A's association with poor prognosis in pancreatic ductal adenocarcinoma suggests potential as a clinical biomarker

    • Development of standardized immunohistochemistry protocols for prognostic stratification

    • Integration into multivariate prognostic models combining multiple biomarkers

    • Potential for SPRR1A antibody-based liquid biopsy approaches

  • Cardioprotective therapeutics:

    • SPRR1A's demonstrated cardioprotective effects suggest potential for targeted therapy development

    • Monitoring SPRR1A expression as a biomarker of cardiac stress response

    • Antibody-based imaging to identify at-risk myocardial regions following ischemic events

    • Development of SPRR1A-mimetic peptides for cardioprotection, with antibodies serving as validation tools

  • Antimicrobial therapeutic development:

    • SPRR1A's bactericidal activity against MRSA and P. aeruginosa presents opportunities for novel antimicrobial therapeutics

    • Antibody tools to monitor SPRR1A expression in skin infections and response to therapy

    • Development of SPRR1A-derived antimicrobial peptides, using antibodies to track biodistribution and stability

    • Potential for SPRR1A-based topical treatments for antibiotic-resistant skin infections

  • Tissue engineering applications:

    • Monitoring SPRR1A expression as a marker of stress response in engineered tissues

    • Antibody-based quality control for tissue-engineered products

    • SPRR1A incorporation into biomaterials with antimicrobial properties

  • Diagnostic imaging development:

    • SPRR1A antibody-based imaging agents to visualize stress responses in cardiac tissue

    • Potential for targeted delivery of therapeutics to stressed or damaged tissues expressing SPRR1A

The development of these applications requires further refinement of antibody specificity, sensitivity, and validation across diverse experimental and clinical contexts. Collaborative efforts between basic scientists and clinical researchers will be essential to translate these promising applications into clinical practice.

How might gene editing techniques be combined with SPRR1A antibodies to advance functional studies?

The integration of gene editing technologies with SPRR1A antibody applications represents a powerful combination for advanced functional studies. Several strategic approaches can significantly enhance our understanding of SPRR1A biology:

  • CRISPR/Cas9 endogenous tagging:

    • Introduction of fluorescent or epitope tags at the endogenous SPRR1A locus

    • Enables live-cell imaging while maintaining physiological expression regulation

    • Validation of tag functionality using established SPRR1A antibodies

    • Allows for chromatin immunoprecipitation studies to identify transcriptional regulators of SPRR1A

  • Domain-specific functional analysis:

    • Generation of precise deletion mutants targeting specific SPRR1A functional domains

    • Use SPRR1A antibodies to confirm expression and localization of mutant proteins

    • Particularly valuable for dissecting domains responsible for antimicrobial function versus cardioprotection

    • Structure-function analysis of SPRR1A's ability to bind bacterial membranes

  • Conditional expression systems:

    • Development of cell type-specific and temporally controlled SPRR1A expression models

    • Integration of inducible CRISPR interference or activation systems to modulate SPRR1A expression

    • Combine with SPRR1A antibody detection to validate system efficiency and monitor expression dynamics

  • In vivo gene editing approaches:

    • Development of tissue-specific SPRR1A knockout or overexpression models

    • Extension of existing Sprr1a−/−;Sprr2a−/− models to create tissue-specific or inducible systems

    • SPRR1A antibodies essential for validating editing efficiency and specificity in complex tissues

    • Analysis of compensatory mechanisms in response to SPRR1A deletion

  • High-throughput screening applications:

    • CRISPR library screens to identify regulators of SPRR1A expression

    • Antibody-based detection as readout for screening success

    • Development of reporter systems validated by correlation with antibody staining

    • Identification of compounds that modulate SPRR1A expression or activity

These combined approaches address key questions about SPRR1A function that cannot be resolved through either technique alone. For example, while gene editing can establish causality, antibodies provide crucial validation and spatial information about protein expression and localization. The temporal dynamics of SPRR1A expression—with transient mRNA but persistent protein presence —make this combined approach particularly valuable for comprehensive functional analysis.

What are the implications of SPRR1A's multiple biological functions for antibody-based therapeutic development?

The multifunctional nature of SPRR1A presents both unique opportunities and significant challenges for antibody-based therapeutic development. SPRR1A's diverse roles—cardioprotection against ischemic stress , correlation with cancer prognosis , and antimicrobial activity —require careful consideration when developing targeted therapeutic strategies.

Key implications and strategic approaches include:

  • Tissue-specific targeting considerations:

    • Therapeutic antibodies must distinguish between beneficial SPRR1A functions (cardioprotection, antimicrobial activity) and potentially detrimental roles (cancer progression)

    • Development of tissue-restricted delivery systems for antibody-based therapeutics

    • Consideration of local versus systemic administration routes

  • Function-specific epitope targeting:

    • Different SPRR1A domains likely mediate distinct biological functions

    • Antibodies targeting specific epitopes could selectively inhibit or enhance particular functions

    • Structure-function mapping studies essential to identify these functional domains

    • For example, antibodies targeting the bacterial membrane-binding region might inhibit antimicrobial function while preserving cardioprotection

  • Contextual expression considerations:

    • SPRR1A expression is tightly regulated and often induced only under stress conditions

    • Therapeutic antibodies must account for this dynamic expression pattern

    • Time-limited or condition-responsive therapeutic approaches may be necessary

    • Companion diagnostics to identify patients with relevant SPRR1A expression patterns

  • Therapeutic modality options:

    • Direct SPRR1A-targeting antibodies for function inhibition

    • Antibody-drug conjugates for targeted delivery to SPRR1A-expressing cells

    • SPRR1A-mimetic peptides with antibodies used for pharmacokinetic/pharmacodynamic studies

    • Bispecific antibodies linking SPRR1A with effector functions or target cells

  • Potential therapeutic applications:

    • Cardioprotection: SPRR1A-mimetic therapeutics to protect against ischemic damage

    • Cancer: Monitoring SPRR1A as a prognostic biomarker in pancreatic cancer

    • Antimicrobial: Development of SPRR1A-derived peptides for antibiotic-resistant infections

The development pathway should include comprehensive safety assessments addressing the risk of interfering with SPRR1A's beneficial functions while targeting its pathological roles. Additionally, the relatively low expression of SPRR1A in normal tissues under non-stress conditions may provide a favorable therapeutic window for targeting stress-induced expression.

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.