HMOX1 antibodies enable precise detection of HO-1 in diverse experimental models:
Use Case: Quantifying HO-1 expression in lysates from cell lines (e.g., A549, HeLa) or tissues (e.g., mouse spleen) .
Validation: Knockout (KO) cell lines confirm specificity (e.g., no band in HO-1 KO HeLa cells) .
Target Tissues: Liver, spleen, and tumor microenvironments (e.g., ovarian cancer stroma) .
Localization: Cytoplasmic, endoplasmic reticulum, and plasma membrane .
Knockdown/Overexpression: Assessing HO-1’s role in apoptosis, proliferation, and drug resistance .
Immune Cell Profiling: Correlating HO-1 with macrophage (M0/M1) and T cell infiltration in atherosclerosis or cancer .
Ischemia/Reperfusion Injury:
Ferroptosis: HO-1 limits iron-dependent oxidative damage in atherosclerosis .
Autoimmune Diseases:
Hemolytic Disorders:
Antibody | WB Band | IHC Localization | KO Validation |
---|---|---|---|
CAB11102 | 33 kDa | Cytoplasmic/membrane | No |
MAB3776 | 32 kDa | Not specified | Yes |
MA1-112 | 32 kDa | Cytoplasmic | No |
10701-1-AP | 28–33 kDa | Cytoplasmic | No |
HMOX1 (Heme Oxygenase 1), also known as HO-1 or HSP32, is an essential enzyme in heme catabolism that catalyzes the oxidative cleavage of heme at the alpha-methene bridge carbon. This reaction produces biliverdin IXalpha, carbon monoxide (CO), and releases ferrous iron from the heme structure . The enzyme plays a critical cytoprotective role by preventing programmed cell death through its ability to catabolize free heme, which would otherwise sensitize cells to undergo apoptosis . HMOX1 is expressed in numerous tissues, including vascular smooth muscle cells and endothelial cells, and can be induced by various stimuli such as hemolysis, inflammatory cytokines, oxidative stress, heat shock, heavy metals, and endotoxin . The protein typically appears at approximately 32 kDa in Western blot analyses, though sometimes it may be observed between 28-33 kDa due to potential post-translational modifications .
There are three known isoforms of heme oxygenase: HMOX1 (HO-1), HMOX2 (HO-2), and HMOX3 (HO-3). HMOX1 is highly inducible, whereas HMOX2 and HMOX3 are constitutively expressed . When designing experiments to distinguish between these isoforms, Western blot analysis can be particularly informative. For instance, recombinant human HO-1 and HO-2 can be included as reference standards (5 ng/lane) alongside your samples to confirm specificity . Another effective approach is to use knockout cell lines as negative controls, such as the HO-1/HMOX1/HSP32 knockout HeLa cell line alongside parental HeLa cells, where a specific band for HMOX1 should be detectable at approximately 32 kDa in the parental line but absent in the knockout line . Always include loading controls such as GAPDH to ensure equal protein loading across samples, which is crucial for accurate comparative analysis between isoforms .
Commercial HMOX1 antibodies exhibit various cross-reactivity profiles depending on the specific antibody clone and manufacturer. Based on available data, many HMOX1 antibodies demonstrate cross-reactivity with human, mouse, and rat samples . Some antibodies, like the rabbit polyclonal antibody from Proteintech (10701-1-AP), have been cited for reactivity with additional species including pig, monkey, chicken, bovine, and goat samples . When selecting an antibody for multi-species research, verify the validated reactivity provided by manufacturers and consider consulting literature citations where the antibody has been successfully used with your species of interest. For example, the Mouse Monoclonal Heme Oxygenase 1 antibody (HO-1-1) from Abcam has been cited in 328 publications and shows reactivity with human, rat, dog, cow, and mouse samples .
HMOX1 antibodies have been validated for multiple experimental applications with varying degrees of reliability. Western blot (WB) is the most extensively validated application, with over 1000 publications citing successful use with HMOX1 antibodies . Immunohistochemistry (IHC) and immunofluorescence (IF) are also well-established applications, with numerous citations supporting their reliability . Flow cytometry (intracellular staining), immunoprecipitation (IP), and co-immunoprecipitation (CoIP) have fewer citations but remain viable applications for specialized research needs . When designing experiments, consider that some antibodies have been specifically optimized for certain applications - for example, the Rabbit Polyclonal Antibody from OriGene is particularly recommended for IHC applications with human cervical cancer samples as positive controls . For the most reliable results, always follow manufacturer-specific protocols and recommended dilutions, which may vary significantly between applications and antibody clones.
For optimal Western blot detection of HMOX1, several critical parameters must be considered. Use PVDF membranes, which have been successfully employed in published protocols . The antibody concentration should be carefully titrated; for example, with the Rat Anti-Human/Mouse HO-1/HMOX1/HSP32 Monoclonal Antibody (MAB3776), a concentration of 1 μg/mL has yielded specific bands , while the Goat Anti-Human/Mouse HO-1/HMOX1/HSP32 Antigen Affinity-purified Polyclonal Antibody (AF3776) has been effective at 0.5 μg/mL . Reducing conditions are generally recommended, with specific buffer systems such as Immunoblot Buffer Group 1 or 2 depending on the antibody used . For secondary antibody selection, choose one that corresponds to the host species of your primary antibody - for example, HRP-conjugated Anti-Rat IgG for rat monoclonal antibodies or HRP-conjugated Anti-Goat IgG for goat polyclonal antibodies . Include appropriate positive controls such as A549 human lung carcinoma cell line, DU145 human prostate carcinoma cell line, or A20 mouse B cell lymphoma cell line, which have been validated for HMOX1 expression .
To rigorously validate HMOX1 antibody specificity, implement a multi-layered control strategy. First, include positive control samples with known HMOX1 expression, such as A549 human lung carcinoma cells or A20 mouse B cell lymphoma cells, which consistently show detectable HMOX1 levels . Second, incorporate negative controls using HMOX1 knockout cell lines when available - the HMOX1 knockout HeLa cell line alongside parental HeLa cells provides an excellent system to confirm antibody specificity . Third, when analyzing both human and mouse samples, include recombinant HMOX1 protein standards (both human and mouse) to confirm proper molecular weight identification . Fourth, include loading controls such as GAPDH to normalize protein quantities across samples . For induced expression studies, compare baseline and stimulated conditions, as HMOX1 is highly inducible by LPS in macrophages and various stress conditions in other cell types . When possible, validate results with multiple antibodies targeting different epitopes of HMOX1 to strengthen confidence in your observations.
Successful immunohistochemistry for HMOX1 detection requires careful optimization of several parameters. Begin with appropriate tissue fixation and processing techniques - formalin-fixed, paraffin-embedded (FFPE) samples have been successfully used with commercial HMOX1 antibodies . For antigen retrieval, heat-induced epitope retrieval methods are generally recommended, though specific buffer conditions may vary between antibodies. When using the Rabbit Polyclonal Antibody from OriGene, the recommended dilution range for IHC is 1:50-1:200, with human cervical cancer tissue serving as an effective positive control . The antibody's predicted cellular localization is primarily cytoplasmic, which should guide your evaluation of staining patterns . Include appropriate positive and negative controls in each experiment - using tissue samples with known HMOX1 expression patterns and omitting primary antibody, respectively. For multiplexed IHC, carefully select antibodies raised in different host species to avoid cross-reactivity issues. Document and standardize all staining parameters including antibody concentration, incubation time, temperature, and detection system specifications to ensure reproducibility across experiments.
Multiple bands or unexpected molecular weights when detecting HMOX1 by Western blot may result from several biological and technical factors. The full-length HMOX1 protein is relatively unstable and susceptible to truncation, which can generate an inactive, soluble form at approximately 28 kDa instead of the expected 32-33 kDa band . This truncation may occur during sample processing or reflect biological reality in your samples. Post-translational modifications such as phosphorylation, ubiquitination, or glycosylation can also alter the apparent molecular weight of HMOX1. Another consideration is the specificity of your antibody - some antibodies may cross-react with related proteins like HMOX2, particularly if high antibody concentrations are used. To address these issues, include recombinant HMOX1 standards to confirm the expected molecular weight , use fresh samples with appropriate protease inhibitors during preparation, and validate results with HMOX1 knockout cells as negative controls . If multiple bands persist despite these precautions, consider using different antibodies targeting distinct epitopes to identify which bands represent genuine HMOX1 protein.
Inconsistent HMOX1 detection between experiments can stem from multiple sources of variability. First, consider the highly inducible nature of HMOX1 - expression levels can fluctuate dramatically in response to cellular stress, including seemingly minor variations in culture conditions, passage number, or handling procedures . Second, sample preparation techniques significantly impact protein integrity - HMOX1 is relatively unstable and susceptible to degradation, particularly its full-length form . Third, antibody quality can deteriorate over time through repeated freeze-thaw cycles or improper storage. Fourth, variations in blocking reagents, incubation times, and detection systems can affect signal intensity. To improve consistency, standardize all experimental variables including cell culture conditions, sample preparation protocols, and antibody handling procedures. Include positive controls in each experiment, such as recombinant HMOX1 protein or cell lines with stable HMOX1 expression . Consider preparing single-use aliquots of antibodies to avoid freeze-thaw cycles. Document and control for the time between sample preparation and analysis, as this can affect protein degradation rates and detection sensitivity.
Detecting low levels of HMOX1 expression requires strategies to enhance signal-to-noise ratio. Begin by optimizing antibody concentration through careful titration experiments - excess antibody increases background while insufficient antibody reduces signal sensitivity. For Western blot applications, enhance protein loading (up to 50-80 μg total protein, depending on sample type) and consider using high-sensitivity chemiluminescent substrates. Signal amplification systems, such as biotin-streptavidin detection methods, can significantly enhance signal without proportionately increasing background. For immunohistochemistry, optimize antigen retrieval conditions and consider using polymer-based detection systems that provide amplified signal with reduced background. When using flow cytometry for intracellular HMOX1 detection, thorough fixation and permeabilization are essential, as is careful optimization of antibody concentration and incubation time . For all applications, extend primary antibody incubation times (overnight at 4°C rather than 1-2 hours at room temperature) to maximize specific binding while using more stringent washing protocols to reduce non-specific background. Finally, consider inducing HMOX1 expression with known stimulants such as LPS in macrophages as positive controls to confirm that your detection system is functioning properly .
Distinguishing between membrane-bound and soluble forms of HMOX1 requires specialized experimental approaches. The full-length HMOX1 is unstable and susceptible to truncation, generating an inactive soluble form at approximately 28 kDa, while the membrane-bound form typically appears at 32-33 kDa . To differentiate between these forms, employ subcellular fractionation techniques to separate cytosolic and membrane fractions before Western blot analysis. Use ultracentrifugation protocols specifically designed to separate membrane-associated proteins from soluble proteins, followed by Western blotting with HMOX1 antibodies. Include fraction-specific markers such as Na+/K+ ATPase for membrane fractions and GAPDH for cytosolic fractions to confirm successful separation. For immunofluorescence approaches, co-staining with membrane markers can help visualize membrane-associated HMOX1. High-resolution microscopy techniques such as confocal or super-resolution microscopy provide superior visualization of subcellular localization. When performing flow cytometry, compare surface staining protocols (for membrane-bound form) with intracellular staining protocols (for total HMOX1) to distinguish between populations. For comprehensive characterization, combine these approaches with protease protection assays to determine the topology of membrane-associated HMOX1.
Studying HMOX1 induction requires methodologies that capture both temporal dynamics and magnitude of response. Design time-course experiments measuring HMOX1 protein levels at multiple timepoints (2, 4, 8, 12, 24 hours) after stimulus application, as induction kinetics vary by stimulus type and cell model. Western blot analysis remains the gold standard for quantifying protein induction, with densitometry analysis for quantification . Flow cytometry offers single-cell resolution of HMOX1 induction, revealing population heterogeneity in response to stimuli . Real-time monitoring can be achieved using HMOX1 promoter-reporter constructs (luciferase or fluorescent protein) transfected into target cells. For in vivo models, immunohistochemistry on tissue sections from treated and untreated animals can visualize tissue-specific induction patterns . Common inducers for positive controls include lipopolysaccharide (LPS) for macrophages, hemin for various cell types, and oxidative stressors like hydrogen peroxide . Always include both positive controls (known inducers) and negative controls (vehicle treatment) in each experiment. For comprehensive analysis, combine protein-level measurements with mRNA quantification using qRT-PCR to distinguish transcriptional induction from post-transcriptional regulation.
Studying HMOX1 interactions with other proteins requires multiple complementary approaches. Immunoprecipitation (IP) and co-immunoprecipitation (Co-IP) have been validated for HMOX1 interaction studies, with specific antibodies shown to be effective for these applications . When performing Co-IP experiments, use gentle lysis conditions to preserve protein-protein interactions, and include appropriate controls such as IgG control immunoprecipitations and input samples. Proximity ligation assays (PLA) offer in situ visualization of protein interactions with high specificity and sensitivity, requiring fewer cells than biochemical approaches. Fluorescence resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) can reveal dynamic interactions in living cells when combined with fluorescently tagged HMOX1 and partner proteins. For higher-throughput interaction screening, consider affinity purification coupled with mass spectrometry (AP-MS) using HMOX1 as bait. Functional validation of identified interactions should include siRNA knockdown or CRISPR/Cas9 knockout of interaction partners, followed by assessment of HMOX1 activity, localization, or stability. Computational approaches such as molecular docking and protein-protein interaction prediction algorithms can guide experimental design by identifying potential interaction interfaces.
Interpreting changes in HMOX1 expression requires contextual understanding of its regulation and function. HMOX1 is highly inducible by various stimuli including inflammatory cytokines, oxidative stress, heat shock, heavy metals, and endotoxin . Therefore, increased HMOX1 expression typically indicates cellular stress response activation. In inflammatory contexts, elevated HMOX1 often represents an adaptive cytoprotective mechanism attempting to counteract pro-inflammatory processes and prevent apoptosis . The magnitude and kinetics of induction can provide insights into stress severity - moderate, transient increases may indicate successful adaptation, while sustained high expression might suggest ongoing stress that cells are unable to resolve. When interpreting HMOX1 expression in disease models, consider that both insufficient and excessive HMOX1 activity can be pathological depending on context. Comparative analysis across multiple experimental conditions and timepoints is essential for meaningful interpretation. Always normalize HMOX1 levels to appropriate housekeeping genes or proteins and include both positive controls (known HMOX1 inducers) and negative controls in analyses. For comprehensive interpretation, correlate HMOX1 expression with functional outcomes such as cell viability, inflammatory marker expression, or oxidative stress parameters.
Reliable quantification of HMOX1 protein levels requires rigorous methodological approaches. Western blot densitometry remains a standard approach but requires careful optimization - use multi-point standard curves with recombinant HMOX1 protein to ensure measurements fall within the linear range of detection . Include technical replicates and multiple biological replicates, with appropriate normalization to loading controls such as GAPDH that remain stable under your experimental conditions . Flow cytometry provides single-cell quantification of HMOX1 levels, allowing analysis of expression heterogeneity within populations and precise quantification of the percentage of cells expressing HMOX1 above threshold levels . ELISA-based quantification offers higher throughput than Western blotting with potentially greater sensitivity, though commercial HMOX1 ELISA kits should be validated with spike-and-recovery experiments for your sample types. For tissue section analysis, digital image analysis of immunohistochemistry staining using calibrated software can provide semi-quantitative data, particularly when using standardized staining protocols and including standard samples of known HMOX1 expression in each batch . Mass spectrometry-based approaches such as selected reaction monitoring (SRM) or parallel reaction monitoring (PRM) offer absolute quantification capabilities but require specialized equipment and expertise.
Knockout and knockdown models provide powerful approaches for studying HMOX1 function when properly implemented and interpreted. HMOX1 knockout cell lines, such as the HMOX1 knockout HeLa cell line described in the literature, serve as excellent negative controls for antibody validation and as experimental models to study HMOX1-dependent processes . When establishing new knockout models using CRISPR/Cas9 or similar technologies, verify complete protein loss by Western blot using antibodies targeting different epitopes of HMOX1. Include rescue experiments by re-expressing HMOX1 to confirm phenotype specificity. For transient knockdown studies using siRNA or shRNA, optimize transfection conditions to achieve at least 80% reduction in protein levels, and include non-targeting control siRNAs in all experiments. Temporal considerations are critical - acute HMOX1 knockdown may produce different phenotypes than stable knockout due to compensatory mechanisms. When using inducible knockout systems, carefully characterize the kinetics of protein depletion after inducer addition. For in vivo studies, consider that complete HMOX1 knockout is lethal in some genetic backgrounds, necessitating conditional knockout approaches. In all knockout/knockdown studies, comprehensively characterize phenotypes across multiple parameters including cell viability, morphology, stress responses, and relevant pathway activities to develop a complete understanding of HMOX1 function in your experimental system.
HMOX1 plays significant roles in infectious disease processes, particularly during viral infections like SARS-CoV-2. During SARS-CoV-2 infection, HMOX1 has been found to promote viral ORF3A-mediated autophagy, though it appears unlikely to be required for ORF3A-mediated induction of reticulophagy . This suggests HMOX1 may be involved in specific autophagic pathways during viral infection rather than general autophagy activation. The enzyme's well-established role in cytoprotection and prevention of programmed cell death becomes particularly relevant during infection, as it can modulate the balance between host cell survival and virus-induced cell death . Given that HMOX1 is inducible by inflammatory cytokines and oxidative stress - both prominent during infectious disease - its expression likely represents an adaptive response attempting to limit immunopathology during infection . To study HMOX1's role in infectious contexts, researchers should consider combined approaches including infection models in HMOX1 knockout cells, pharmacological modulation of HMOX1 activity during infection, and temporal analysis of HMOX1 expression throughout the course of infection. Time-course analyses that correlate HMOX1 expression with viral load, inflammatory markers, and cell survival outcomes are particularly informative for understanding its functional significance.
Studying HMOX1 in oxidative stress contexts requires specialized methodologies that capture both HMOX1 regulation and its impact on redox homeostasis. Design experiments that combine direct measurement of HMOX1 expression/activity with quantification of oxidative stress parameters such as reactive oxygen species (ROS) levels, glutathione depletion, lipid peroxidation products, and protein carbonylation. Time-course studies are essential, as HMOX1 induction typically follows oxidative stress rather than preceding it, with peak expression often occurring several hours after stress induction. Include gradient exposure to oxidative stressors (such as H₂O₂, paraquat, or tert-butyl hydroperoxide) to determine dose-response relationships. HMOX1 knockout or knockdown models allow direct assessment of how HMOX1 deficiency impacts cellular resistance to oxidative challenges . Complementary gain-of-function approaches using HMOX1 overexpression or pharmacological inducers can demonstrate protective effects. For mechanistic studies, measure the products of HMOX1 activity (CO, biliverdin/bilirubin, and free iron) and their individual contributions to cytoprotection using specific scavengers or inhibitors. In vivo models of oxidative stress-related pathologies provide translational relevance, particularly when combined with tissue-specific HMOX1 modulation. Advanced imaging techniques using redox-sensitive fluorescent proteins can provide real-time visualization of how HMOX1 expression correlates with subcellular redox dynamics.
Investigating HMOX1's therapeutic potential requires a systematic approach spanning from molecular mechanisms to preclinical models. Begin with high-throughput screening of compound libraries to identify novel HMOX1 inducers, using cell-based reporter assays where luciferase or fluorescent protein expression is driven by the HMOX1 promoter. Validate hits with orthogonal assays measuring endogenous HMOX1 protein levels by Western blot and enzymatic activity through bilirubin production assays. Structure-activity relationship studies can optimize lead compounds for potency, selectivity, and drug-like properties. For target validation, use genetic approaches including HMOX1 knockout models to confirm that observed therapeutic effects require HMOX1 expression . Investigate molecular mechanisms by which candidate therapeutics induce HMOX1, focusing on transcription factors such as Nrf2 and their regulatory pathways. Develop relevant disease models where HMOX1's cytoprotective effects would be beneficial, such as inflammatory, ischemic, or oxidative stress-related conditions. In preclinical efficacy studies, include both preventive and treatment paradigms, measuring not only HMOX1 induction but also functional outcomes and disease-relevant biomarkers. Pharmacokinetic/pharmacodynamic modeling should correlate drug exposure with both HMOX1 induction and therapeutic effects. Finally, consider potential limitations including tissue-specific effectiveness, dose-dependent effects (with possible hormetic responses), and safety concerns related to overactivation of heme catabolism.
Optimizing flow cytometry for intracellular HMOX1 detection requires careful attention to fixation, permeabilization, and antibody selection. Begin with effective fixation using paraformaldehyde (2-4%) for 10-15 minutes at room temperature to preserve cellular structure while enabling subsequent antibody access. Follow with permeabilization using either saponin (0.1-0.5%) for reversible membrane permeabilization or methanol/Triton X-100 for more stringent permeabilization, optimizing conditions for your specific cell type. Select antibodies specifically validated for flow cytometry applications, such as those referenced in the literature with flow cytometry citations . Titrate antibodies carefully to determine optimal concentration, typically starting with manufacturer recommendations and adjusting as needed to maximize signal-to-noise ratio. Include appropriate controls in every experiment: isotype controls to assess non-specific binding, FMO (fluorescence minus one) controls for accurate gating, positive controls such as LPS-stimulated macrophages known to express high HMOX1 levels , and when possible, HMOX1 knockout cells as negative controls . For multiparameter analysis, choose fluorophores with minimal spectral overlap and perform proper compensation. When analyzing samples with potentially low HMOX1 expression, consider using bright fluorophores such as PE or APC rather than FITC. Allow sufficient incubation time (typically 30-60 minutes) for antibody binding, followed by thorough washing to remove unbound antibody.
Simultaneous detection of HMOX1 and its enzymatic products (biliverdin/bilirubin, CO, and iron) requires integrated multimodal approaches. For biliverdin/bilirubin detection alongside HMOX1 immunostaining, exploit the natural fluorescence properties of these tetrapyrroles, which emit in the green spectrum when excited with blue light. This can be combined with immunofluorescence for HMOX1 using red-emitting fluorophores to avoid spectral overlap. Alternatively, use chemical probes specific for bilirubin coupled with HMOX1 immunostaining. For carbon monoxide detection, employ CO-specific fluorescent probes based on palladium-porphyrin complexes, which can be used in combination with immunofluorescence techniques for HMOX1 visualization. Iron released during HMOX1 activity can be detected using iron-specific fluorescent sensors such as Phen Green SK or RhoNox-1, allowing correlation with HMOX1 expression in the same cells. For live cell applications, consider using genetically encoded sensors for both HMOX1 expression (fluorescent protein fusions) and its products. In tissue sections, sequential staining approaches may be necessary, first documenting the natural fluorescence of biliverdin/bilirubin followed by HMOX1 immunostaining. For quantitative correlation between HMOX1 expression and product formation, combine Western blot analysis of HMOX1 with biochemical assays measuring bilirubin production, CO release (using gas chromatography or myoglobin-based spectrophotometric assays), and iron release (using colorimetric ferrozine-based assays or inductively coupled plasma mass spectrometry).
Detecting HMOX1 in complex tissue microenvironments requires specialized approaches that preserve spatial context while enabling specific detection. Multiplex immunohistochemistry/immunofluorescence provides simultaneous visualization of HMOX1 along with cell type-specific markers, allowing precise identification of which cells express HMOX1 within heterogeneous tissues . Tyramide signal amplification can significantly enhance detection sensitivity for low-abundance HMOX1 expression without increasing background staining. For highly autofluorescent tissues such as liver or brain, consider spectral unmixing approaches or use of far-red fluorophores that minimize overlap with autofluorescence spectra. Laser capture microdissection combined with Western blot analysis allows quantitative HMOX1 detection from specific regions within complex tissues . RNAscope in situ hybridization for HMOX1 mRNA can complement protein detection methods, providing validation through an orthogonal approach. Tissue clearing techniques combined with light sheet microscopy enable three-dimensional visualization of HMOX1 expression patterns throughout intact tissue volumes. For quantitative spatial analysis, digital pathology approaches using trained algorithms can map HMOX1 expression intensity across tissue regions, allowing correlation with pathological features or specific microenvironmental factors. When analyzing tissues with potential induction of HMOX1, include both stressed/diseased samples and appropriate controls to establish baseline expression patterns, as HMOX1 is highly inducible and its expression patterns may change dramatically in response to pathological conditions .
HO-1 is highly inducible and is considered a key cytoprotective molecule against oxidative injury. The substrate, heme, is a potent pro-oxidant that can accelerate inflammation and cell death. Therefore, the activity of HO-1 is essential in mitigating these harmful effects . Research has shown that both humans and mice with defective HO-1 genes are highly susceptible to oxidative stress .
The Mouse Anti-Human Heme Oxygenase 1 antibody is a monoclonal antibody that specifically targets the HO-1 protein in human samples. This antibody is widely used in various applications, including Western Blot (WB), Immunohistochemistry (IHC), Flow Cytometry (Flow Cyt), and Enzyme-Linked Immunosorbent Assay (ELISA) .
The Mouse Anti-Human HO-1 antibody is extensively validated and cited in numerous publications. It is used in research to study the role of HO-1 in various physiological and pathological processes, including its anti-inflammatory and cytoprotective functions .