HTRA1 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

HTRA1 Antibody refers to monoclonal or polyclonal antibodies designed to target and modulate the activity of High-Temperature Requirement A1 (HTRA1), a secreted serine protease encoded by the HTRA1 gene. HTRA1 regulates extracellular matrix (ECM) remodeling, angiogenesis, and transforming growth factor-β (TGF-β) signaling pathways . Dysregulation of HTRA1 is implicated in cerebral small vessel disease (CSVD), age-related macular degeneration (AMD), cancer, and autoimmune disorders . Therapeutic HTRA1 antibodies aim to inhibit its proteolytic activity or neutralize its pathological effects.

Clinical Applications in AMD

Phase I/II Trials:

Trial PhaseDoseOutcomeReference
Phase I (NCT03295877)1–20 mg (single dose)No ocular/systemic adverse events; dose-dependent inhibition of HTRA1 activity for ≤8 weeks .
Phase II (GALLEGO)20 mg (Q4W/Q8W)No efficacy in reducing geographic atrophy (GA) progression vs. sham .

Biomarker Identification:

  • Dickkopf-related protein 3 (DKK3): Aqueous humor levels of cleaved DKK3 serve as a pharmacodynamic biomarker for HTRA1 inhibition .

Role in Cerebral Small Vessel Disease

  • HTRA1 mutations reduce protease activity, leading to uncontrolled TGF-β signaling and vascular fibrosis .

  • Anti-HtrA1 antibodies restore TGF-β regulation, mitigating ECM accumulation in cerebral arterioles .

Oncology and Autoimmunity

  • Cancer: HTRA1 overexpression in tumors (e.g., breast, ovarian) correlates with metastasis; antibodies block ECM degradation and angiogenesis .

  • Primary Membranous Nephropathy (MN): HTRA1 is a novel podocyte antigen targeted by IgG4 autoantibodies .

Preclinical Models

  • Fab15H6.v4.D221: Humanized Fab fragment with picomolar affinity for HTRA1. Inhibits vitreal HTRA1 activity in rabbits and cynomolgus monkeys for ≥14 days post-intravitreal injection .

Antibody CloneHostTarget DomainApplications
15H6.v4.D221HumanizedProtease domainAMD therapy
MAB2916MouseFull-length HTRA1Western blot, IHC
55011-1-APRabbitProtease domainWB, IF, IP

Technical Validation

  • Western Blot: Detects HTRA1 at ~51 kDa (non-reducing conditions) .

  • Immunohistochemistry (IHC): Localizes HTRA1 in vascular endothelia and podocytes .

Challenges and Future Directions

  • Clinical Limitations: Phase II trials showed no efficacy in GA, highlighting incomplete understanding of HTRA1’s role in AMD .

  • Biomarker Optimization: DKK3 validation requires larger cohorts to confirm sensitivity .

  • Therapeutic Expansion: Targeting HTRA1 in TGF-β-driven fibrosis and autoimmune MN is under exploration .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery times may vary depending on the purchasing method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
ARMD7 antibody; CARASIL antibody; High-temperature requirement A serine peptidase 1 antibody; HtrA antibody; HtrA serine peptidase 1 antibody; HTRA1 antibody; HTRA1_HUMAN antibody; IGFBP5 protease antibody; L56 antibody; ORF480 antibody; Protease serine 11 (IGF binding) antibody; protease serine 11 antibody; PRSS11 antibody; Serine protease 11 antibody; Serine protease HTRA1 antibody; Serine protease HTRA1 precursor antibody
Target Names
HTRA1
Uniprot No.

Target Background

Function
HTRA1 is a serine protease with a broad range of targets, including extracellular matrix proteins like fibronectin. Fragments of fibronectin generated by HTRA1 stimulate synovial cells to upregulate the production of MMP1 and MMP3. HTRA1 can also degrade proteoglycans, such as aggrecan, decorin, and fibromodulin. Cleavage of proteoglycans by HTRA1 releases soluble FGF-glycosaminoglycan complexes, which enhance the range and intensity of FGF signaling in the extracellular environment. It regulates the availability of insulin-like growth factors (IGFs) by cleaving IGF-binding proteins. HTRA1 inhibits signaling mediated by TGF-beta family members. While the exact mechanism is unclear, this activity requires the integrity of the catalytic site and may not involve direct degradation of TGF-beta proteins. By modulating TGF-beta signaling, HTRA1 plays a role in various physiological processes, including retinal angiogenesis, and neuronal survival and maturation during development. Intracellularly, HTRA1 degrades TSC2, leading to the activation of downstream targets of TSC2.
Gene References Into Functions
  1. Nuclear downregulation of HtrA1 is associated with a better prognosis in women with high-grade serous ovarian carcinoma. PMID: 30131069
  2. Case Report: Novel compound heterozygous mutations in HTRA1 causing CARASIL in a Chinese patient. PMID: 30068478
  3. HTRA1 regulates odontoblastic differentiation of dental pulp cells through activation of the TGF-beta1/Smad signaling pathway. PMID: 29580722
  4. Aberrant expression of HTRA1 or HTRA4 may contribute to the onset of preeclampsia, and elevated HTRA1 or HTRA4 expression can potentially affect trophoblast functions. PMID: 30015931
  5. HtrA1 contributes to the development of keloid lesions as a matrix protease by remodeling keloid-specific ECM or cell surface molecules. PMID: 29695130
  6. Studies indicate significantly different high-temperature requirement factor A1 (HtrA1) expression in cancer and non-cancer tissue [Meta-analysis]. PMID: 29409460
  7. HtrA1 overexpression leads to impaired apical processes and reduced phagocytosis, a crucial function for photoreceptor survival. PMID: 29269042
  8. Research suggests that HTRA1 is involved in the pathogenesis of scars by regulating the activation of latent TGF-beta1 in keloid fibroblasts. PMID: 29412803
  9. The CADASIL-like family disease may result from heterozygous HTRA1 gene mutation, leading to autosomal dominant hereditary cerebral small vessel disease. PMID: 29561953
  10. Case Report/Review: A novel missense mutation in HTRA1 associated with the phenotype of CARASIL. PMID: 28628911
  11. The rs11200638-rs2672598 joint genotype AA-CC confers a higher risk of exudative age-related macular degeneration (AMD) compared to polypoidal choroidal vasculopathy (PCV). PMID: 27338780
  12. A possible proteolytic processing mechanism of mutant TGFBIp by HTRA1 and peptides generated by the mutant protein may form the beta-amyloid core of corneal aggregates in Corneal dystrophic patients. PMID: 28689406
  13. HtrA1 Proteolysis of ApoE In Vitro Is Allele Selective. PMID: 27379525
  14. The findings of this study further support the pathogenic role of heterozygous HTRA1 mutations in familial cerebral small vessel disease. PMID: 28782182
  15. These results suggest that the variation in the risk for age-related macular degeneration associated with chromosome 10q26 is likely due to variation in HTRA1 expression. PMID: 28659708
  16. HtrA1 role in cisplatin resistance in colon cancer. PMID: 28667026
  17. HtrA1 could serve as a marker to identify patients with ulcerative colitis of more than 10 years duration who are at a higher risk of developing colorectal cancer. PMID: 28586045
  18. To our knowledge, this is the first report of the association between rs11200638 and overall age-related macular degeneration in South America. PMID: 28846052
  19. Since the ARMS2/HTRA1 genes are located at a locus on chromosome 10q26 within a region with strong linkage disequilibrium, it is challenging to distinguish the functions of the individual genes. A review of recent epidemiological studies of Age-related macular degeneration (AMD) is provided. An argument for a definite correlation between the ARMS2 gene and AMD is presented. PMID: 28583181
  20. The expression of HtrA1 was strongly related to the T2 value, suggesting that HtrA1 plays a significant role in the pathological process of intervertebral disc degeneration. PMID: 28432852
  21. These findings suggest that the initiation stage of polypoidal choroidal vasculopathy is mediated by proteolytic degradation of extracellular matrix proteins due to increased HTRA1 activity. PMID: 28941979
  22. The findings of this study provide evidence that CFH gene variants and ARMS2/HTRA1 genes play a major role in the genetic susceptibility to AMD in a Greek population. These findings are directly relevant for disease management and help in mapping the genetic landscape of AMD. PMID: 26848857
  23. The function of the binding between MIF and HTRA1 is to inhibit the proteolytic activity of HTRA1. PMID: 28726057
  24. Results show that HTRA1 is epigenetically silenced in HCT116 colon carcinoma cells and during early stages of tumorigenesis in a mouse model of intestinal cancer. Downregulation of HTRA1 results in multiple phenotypes characteristic of cancer cells, including increased proliferation of mouse embryonic fibroblasts, as well as chromosome and centrosome amplifications. PMID: 27388476
  25. High HTRA1 expression is associated with cervical cell proliferation. PMID: 27809811
  26. Data indicate HtrA serine peptidase 1 (HTRA1) involvement in Age-related macular degeneration (AMD) pathogenesis. PMID: 27841854
  27. Variants in HTRA1 are not associated with age-related macular degeneration. PMID: 27879347
  28. Results show the heterozygous missense mutations p.G283E, p.P285L, p.R302Q, and p.T319I in the HTRA1 gene in 8 patients with symptomatic cerebral small vessel disease. Mutant HTRA1s observed in manifesting heterozygotes might lead to an impaired HTRA1 activation cascade of HTRA1 or be unable to form stable trimers. PMID: 27164673
  29. FN and HtrA1 are localized in the placental key growth zones, suggesting a crucial role in maintaining the balance among the molecules involved in placental development and differentiation. PMID: 28076935
  30. The correlation between TGFb1 and pSmad2, as well as between HtrA1 and TGFb1, and the very significant increase of Ki67 in the stromal compartment of eutopic endometrium suggest a potential involvement of HtrA1 in the pathogenesis of endometriosis. PMID: 26708185
  31. Two synonymous polymorphisms in exon 1 of the HTRA1 gene result in a protein with altered thermophoretic properties. PMID: 26310622
  32. A significant increase in serum HtrA1 in early-onset pre-eclampsia. PMID: 26187609
  33. A frameshift mutation in the HTRA1 gene detected in a CARASIL pedigree resulted in reduced HTRA1 protein and increased TGF-beta1 expression, which may cause severe CARASIL and peripheral small arterial disease. PMID: 25772074
  34. Human HTRA1 expression is enhanced by an Age-related macular degeneration-specific indel mutation in the promoter region of the HTRA1 gene, and this enhanced HTRA1 may be associated with inducing retinal neovascularization. PMID: 27125063
  35. Results do not conclusively support HTRA1's role as a tumor suppressor but suggest its possible prognostic role in many human tumors. PMID: 26035313
  36. Studies indicate that the high-risk allele of the 10q26 locus encompasses three genes, PLEKHA1, ARMS2, and HTRA1 with high linkage disequilibrium. PMID: 26427389
  37. Low HtrA1 expression is associated with gastric cancer. PMID: 25761858
  38. HtrA1 expression was closely related to EMT, which might be a potential mechanism underlying metastasis of HCC. PMID: 26403966
  39. Low HtrA1 expression is significantly associated with poor prognosis parameters in breast cancer, and HtrA1 loss in sentinel nodes is related to metastasis of non-sentinel nodes. PMID: 25530301
  40. It was concluded that high expression of HtrA1 could significantly reverse multidrug resistance of hepatoma cells by targeting XIAP. PMID: 25776486
  41. The growth of choroidal neovascularization in AMD would be affected by 2 genes: MMP20, a newly confirmed gene expressed in the retina, and ARMS2/HTRA1, a well-known susceptibility gene for AMD. PMID: 26337002
  42. HtrA1 emerged as an immunohistochemical marker to predict the behavior of meningioma, particularly recurrence. PMID: 25687108
  43. HtrA1 plays a positive role in human periodontal ligament cells osteogenic differentiation and may regulate this process through TGFB1. PMID: 25726184
  44. In this study, we found that the interaction of ARMS2 and ARMS2/HTRA1 is significantly associated with nAMD, and the interaction of CFH and ARMS2 is pronounced in PCV development in the Chinese population. PMID: 25771815
  45. HtrA1 expression in plasma cells could be correlated with the destruction of pathological periodontal tissue. PMID: 24979214
  46. Central, but not peripheral, drusen location was strongly associated with both [CFH HTRA1] and [CFH HTRA1]. Only [CFH HTRA1] was significantly associated with increased central drusen area. PMID: 25627090
  47. Heterozygous HTRA1 mutations are a significant cause of familial small vessel disease. PMID: 26063658
  48. Silence of the HTRA1 gene was associated with significantly higher levels of TGF-beta1, BMP4, and BMP2 mRNA and reduction in the proliferation and migration of ARPE-19 cells. PMID: 25550099
  49. Gene variants in CFH, ARMS2, and HTRA1 are related to an increased risk of age-related macular degeneration in a northern Chinese population. PMID: 24865190
  50. Alpha-1-antitrypsin as a substrate of HTRA1 synthetic. PMID: 25329061

Show More

Hide All

Database Links

HGNC: 9476

OMIM: 600142

KEGG: hsa:5654

STRING: 9606.ENSP00000357980

UniGene: Hs.501280

Involvement In Disease
Macular degeneration, age-related, 7 (ARMD7); Cerebral arteriopathy, autosomal recessive, with subcortical infarcts and leukoencephalopathy (CARASIL); Cerebral arteriopathy, autosomal dominant, with subcortical infarcts and leukoencephalopathy, 2 (CADASIL2)
Protein Families
Peptidase S1C family
Subcellular Location
Cell membrane. Secreted. Cytoplasm, cytosol.
Tissue Specificity
Widely expressed, with strongest expression in placenta (at protein level). Secreted by synovial fibroblasts. Up-regulated in osteoarthritis and rheumatoid arthritis synovial fluids and cartilage as compared with non-arthritic (at protein level).

Q&A

What is HTRA1 and what cell/tissue types express it?

HTRA1, also known as HTRA, PRSS11, and L56, belongs to the peptidase S1B family. It functions as a protease that regulates the availability of various proteins in cellular pathways. HTRA1 is expressed in multiple tissue types with significant expression observed in:

  • Human tissues: Placenta shows strong expression as demonstrated in immunohistochemistry studies

  • Cell lines: L02 cells, HAP1 cells, and HepG2 cells consistently show HTRA1 expression in Western blot analyses

  • Animal tissues: Mouse brain tissue exhibits detectable levels of HTRA1 protein

  • Vascular system: Vascular smooth muscle cells (VSMCs) express HTRA1, where it plays a crucial role in vascular maturation and homeostasis

  • Renal system: HTRA1 serves as a podocyte antigen in the glomerular system

HTRA1 has a calculated molecular weight of 51 kDa, though observed molecular weights in experimental settings may vary (35 kDa, 42 kDa, and 50 kDa) depending on post-translational modifications and experimental conditions .

What applications are validated for HTRA1 antibodies?

HTRA1 antibodies have been validated for multiple research applications with specific dilution recommendations:

ApplicationRecommended DilutionValidated Sample Types
Western Blot (WB)1:500-1:2000L02 cells, mouse brain tissue, HAP1 cells, HepG2 cells
Immunoprecipitation (IP)0.5-4.0 μg for 1.0-3.0 mg of total protein lysateL02 cells, HAP1 cells
Immunohistochemistry (IHC)1:50-1:500Human placenta tissue
Immunofluorescence (IF/ICC)1:10-1:100HepG2 cells
ELISAApplication-dependentVarious sample types

It is recommended to titrate the antibody in each testing system to obtain optimal results, as the effective concentration may be sample-dependent .

How should HTRA1 antibodies be stored for maximum stability?

For optimal preservation of antibody activity:

  • Store at -20°C in aliquots to avoid repeated freeze-thaw cycles

  • HTRA1 antibodies in PBS with 0.02% sodium azide and 50% glycerol (pH 7.3) are typically stable for one year after shipment when properly stored

  • Aliquoting is generally unnecessary for -20°C storage

  • Small volume formats (20μl) may contain 0.1% BSA as a stabilizing agent

What controls should be included when using HTRA1 antibodies?

To ensure experimental validity:

  • Positive controls: Use L02 cells, HAP1 cells, HepG2 cells, or mouse brain tissue for Western blot applications

  • Negative controls: HTRA1 knockout or knockdown samples provide optimal negative controls, as demonstrated in studies using HtrA1-knockout mice for antibody development

  • Specificity verification: Compare HTRA1 wild-type samples with HtrA1 serine-to-alanine catalytically inactive mutants to confirm specificity in activity-based assays

  • Cross-reactivity assessment: Test against human, mouse, and rat samples, as the antibody shows documented reactivity with these species

How are HTRA1 antibodies utilized in therapeutic development?

HTRA1 antibodies have shown significant potential in therapeutic development, particularly for age-related macular degeneration (AMD):

  • Development approach: Anti-HtrA1 antibodies can be obtained by immunizing HtrA1-knockout mice with recombinant HtrA1 protease domain (HtrA1-PD)

  • Optimization process: The therapeutic development pipeline typically includes:

    • Initial hybridoma screening (identifying clone 15H6 with strong inhibition of HtrA1 enzymatic activity)

    • Humanization through CDR grafting into human consensus frameworks

    • Modification of problematic residues (N94A for cleavage, D55 for isomerization)

    • Affinity maturation via Fab phage display with deep-sequencing analysis

  • Clinical application: The development of HtrA1-blocking Fab fragments has enabled testing of the therapeutic hypothesis that HtrA1 protease activity contributes to AMD progression

  • Significance: Genome-wide association studies have identified genetic variation at the ARMS2/HTRA1 locus as a risk factor for AMD development and progression, making HtrA1 a relevant therapeutic target

What methodologies can confirm target engagement of anti-HTRA1 antibodies in vivo?

Confirming target engagement is crucial for validating anti-HTRA1 antibody efficacy:

  • Activity-based probes (ABPs):

    • Development of HtrA1-directed ABPs using diphenyl phosphonate as the reactive group targeting the nucleophilic active-site serine residue

    • Incorporation of Val and Leu at P1 and P2 positions to direct ABP reactivity against HtrA1

    • Addition of carboxytetramethylrhodamine (TAMRA) through a polyethylene glycol linker as a fluorescent reporter

  • N-terminomic proteomic profiling:

    • Compare amino termini of proteins in biological samples (e.g., rat vitreous humor) upon incubation with HtrA1 versus catalytically inactive HtrA1

    • Identify signature cleavage patterns specific to HtrA1 activity

    • Monitor changes in substrate processing upon antibody administration

  • Pharmacodynamic biomarkers:

    • Dickkopf-related protein 3 (DKK3) has been identified as a robust biomarker for anti-HtrA1 activity

    • This biomarker can be measured in both preclinical animal models and clinical samples

    • Successfully employed in a phase 1 study of geographic atrophy patients to demonstrate anti-HtrA1 Fab activity and duration

What role does HTRA1 play in cellular signaling pathways?

HTRA1 functions as a critical regulator of multiple signaling pathways:

  • TGFβ signaling regulation:

    • Loss of HTRA1 increases TGFβ1 availability and signaling

    • HTRA1 can cleave either pro-TGFβ1 or GFD6

    • HTRA1 silencing leads to elevated phosphorylated SMAD2/3 proteins

    • Increased SMAD-dependent luciferase activity is observed in co-culture experiments with HTRA1-silenced vascular smooth muscle cells

  • Notch signaling modulation:

    • The Notch ligand JAG1 is a substrate for HTRA1

    • HTRA1 cleaves JAG1 in the cytosol, leading to rapid degradation of the remaining JAG1 protein

    • JAG1/NOTCH3 signaling is crucial for differentiation, maintenance, and contractility of vascular smooth muscle cells

    • Loss of HTRA1 leads to over-activation of NOTCH3 signaling

  • Synergistic effects:

    • TGFβ and Notch pathways synergistically stimulate expression of HES and HEY transcriptional repressors

    • Moderate overexpression of NOTCH3-ICD increases SM22α protein levels

    • Higher NOTCH3-ICD expression decreases SM22α expression, indicating hyper-activation of a negative feedback loop

    • Combined overexpression of HES1 and HEYL represses transcription of α-SMA and SM22α in human umbilical artery smooth muscle cells

How can HTRA1 antibodies be used in studying pathological conditions?

HTRA1 antibodies serve as valuable tools in investigating various disease states:

  • Age-related macular degeneration (AMD):

    • Anti-HtrA1 Fab fragments help test the hypothesis that HtrA1 protease activity contributes to AMD progression

    • DKK3 serves as a pharmacodynamic biomarker in aqueous humor samples from geographic atrophy patients, providing evidence of anti-HtrA1 Fab activity and duration in phase 1 clinical studies

  • Membranous nephropathy (MN):

    • HTRA1 has been identified as a novel podocyte antigen in a subset of patients with primary MN

    • Patient sera react by immunoblotting with a 51-kD protein within glomerular extracts

    • Anti-HTRA1 antibody titers appear to correlate with disease course

    • Serial monitoring of anti-HTRA1 antibodies could facilitate diagnostic and therapeutic decisions in this demographic group (mean age 67.3 years)

  • Vascular disorders:

    • Loss of HTRA1 in vascular smooth muscle cells (VSMCs) impairs their maturation and function

    • HTRA1 is essential for vascular homeostasis through its regulation of Notch and TGFβ signaling

    • HTRA1 antibodies can help investigate the role of this protease in familial small vessel disease, where blood vessel functions are impaired

What are the methodological considerations when designing experiments to identify HTRA1 substrates?

When investigating HTRA1 substrates, researchers should consider:

  • N-terminomics approach:

    • Compare amino termini of proteins in biological samples with and without active HTRA1

    • Incubate samples with HtrA1 or catalytically inactive HtrA1 (HtrA1-SA mutant)

    • Use mass spectrometry to identify cleaved peptides

  • Activity-based profiling:

    • Develop HtrA1-directed activity-based probes

    • Design probes based on consensus cleavage sites determined by N-terminomics

    • Include appropriate reactive groups (e.g., diphenyl phosphonate) and reporter tags (e.g., TAMRA)

  • Cross-species validation:

    • Perform experiments in multiple species to identify conserved substrates

    • This approach has successfully identified ocular substrates that were consistent across independent cross-species studies

  • Biomarker identification:

    • Test potential substrates as pharmacodynamic biomarkers

    • Assess correlation between substrate cleavage and HtrA1 activity

    • Validate biomarker utility in both preclinical models and clinical samples

    • Example: Dickkopf-related protein 3 was identified as a robust biomarker for anti-HtrA1 activity

What are the optimal antigen retrieval methods for HTRA1 immunohistochemistry?

For effective immunohistochemical detection of HTRA1:

  • Primary recommendation: Antigen retrieval with TE buffer at pH 9.0

  • Alternative method: Antigen retrieval with citrate buffer at pH 6.0

  • Validated tissue: Human placenta tissue shows positive IHC results

  • Recommended dilution: 1:50-1:500, with optimization for specific tissue types

How can researchers differentiate between HTRA1 isoforms?

When analyzing HTRA1 expression:

  • Expected molecular weights:

    • Calculated molecular weight: 51 kDa

    • Observed molecular weights: 35 kDa, 42 kDa, and 50 kDa

    • Variations likely represent alternative splicing, post-translational modifications, or proteolytic processing

  • Verification strategy:

    • Use HTRA1 knockout/knockdown controls to confirm specificity

    • Compare with recombinant HTRA1 proteins of known molecular weight

    • Perform immunoprecipitation followed by mass spectrometry to confirm identity of bands

How should experimental design be modified when studying HTRA1 in different species?

For cross-species HTRA1 research:

  • Validated reactivity: HTRA1 antibodies have been tested and confirmed to react with human, mouse, and rat samples

  • Cited reactivity: Published literature reports additional reactivity with zebrafish samples

  • Cross-species validation: Important substrates of HTRA1 have been identified through cross-species studies, suggesting conservation of function

  • Knockout models: HtrA1-knockout mice have been generated by traditional homologous recombination methods and serve as valuable tools for specificity controls and comparative studies

What controls should be included when measuring HTRA1 activity inhibition?

To properly assess HTRA1 inhibition by antibodies:

  • Enzymatic activity controls:

    • Positive control: Active recombinant HtrA1 protease domain

    • Negative control: Catalytically inactive version (HtrA1-SA mutant)

    • Dose-response measurements with varying antibody concentrations

  • Substrate processing:

    • Monitor cleavage of known HTRA1 substrates (e.g., DKK3)

    • Use activity-based probes to directly measure active HTRA1

    • Employ reporter systems like SMAD-dependent luciferase assays to measure downstream signaling effects

  • Signaling pathway analysis:

    • Measure phosphorylated SMAD2/3 levels to assess TGFβ pathway inhibition

    • Use gamma-secretase inhibitor DAPT as a control for Notch signaling inhibition

    • Use ALK5 inhibitor CAS 446859-33-2 as a control for TGFβ signaling inhibition

How can researchers enhance specificity when working with HTRA1 antibodies?

To improve specificity:

  • Antibody selection: Use antibodies derived from HtrA1-knockout animals for immunization, which reduces background reactivity

  • Purification method: Antigen affinity-purified antibodies offer higher specificity

  • Validation approach: Confirm antibody specificity using multiple techniques (WB, IP, IHC, IF/ICC) and positive/negative controls

  • Buffer optimization: PBS with 0.02% sodium azide and 50% glycerol at pH 7.3 provides optimal stability and specificity

What factors might influence HTRA1 antibody performance in different applications?

Performance variations may stem from:

  • Sample preparation: Different lysis buffers, fixation methods, or antigen retrieval techniques can affect epitope accessibility

  • Protein conformation: Native versus denatured conditions influence epitope exposure

  • Background reduction: Increasing blocking agent concentration or adjusting antibody dilution can improve signal-to-noise ratio

  • Application-specific considerations:

    • WB: Optimize transfer conditions and blocking agents

    • IHC: Test alternative antigen retrieval methods (TE buffer pH 9.0 versus citrate buffer pH 6.0)

    • IP: Adjust antibody-to-lysate ratio (0.5-4.0 μg antibody for 1.0-3.0 mg total protein)

    • IF/ICC: Consider permeabilization methods and fixation protocols

How can inconsistent results with HTRA1 antibodies be addressed?

When encountering variability:

  • Antibody titration: Test multiple dilutions to determine optimal concentration for each application and sample type

  • Controls expansion: Include both positive controls (L02 cells, HAP1 cells, HepG2 cells, mouse brain tissue) and negative controls (HTRA1 knockdown/knockout samples)

  • Cross-validation: Confirm results using alternative detection methods or antibody clones

  • Activity verification: For functional studies, use activity-based probes to confirm that observed effects correlate with HTRA1 enzymatic activity

How are HTRA1 antibodies advancing our understanding of disease mechanisms?

HTRA1 antibodies have facilitated significant discoveries:

  • AMD pathogenesis: Anti-HtrA1 Fab inhibitors have demonstrated that HtrA1 proteolytic activity contributes to AMD progression

  • Membranous nephropathy: Identification of HTRA1 as a novel podocyte antigen has expanded our understanding of this glomerular disease

  • Vascular disorders: HTRA1 inhibition studies have revealed its role in vascular maturation and homeostasis through Notch and TGFβ signaling regulation

What are the future prospects for HTRA1 antibodies in clinical applications?

The translational potential includes:

  • Diagnostic biomarkers: Anti-HTRA1 antibody titers may serve as biomarkers in membranous nephropathy, facilitating diagnosis and treatment decisions

  • Therapeutic development: HtrA1-blocking antibodies show promise for treating AMD, with pharmacodynamic biomarkers like DKK3 enabling monitoring of therapeutic efficacy

  • Personalized medicine: Genetic variation at the ARMS2/HTRA1 locus may inform patient selection for anti-HTRA1 therapies in AMD

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.