HSP90 alpha is a 90 kDa protein composed of three domains:
N-terminal domain (NTD): Binds ATP and regulates ATPase activity.
Middle domain (MD): Modulates ATPase activity and interacts with co-chaperones (e.g., Aha1) and client proteins.
C-terminal domain (CTD): Mediates dimerization and binding to co-chaperones .
It chaperones signaling proteins (e.g., AKT, steroid hormone receptors) and regulates processes like apoptosis and cell cycle control. Overexpression is linked to cancer and neurodegenerative diseases .
Table 1 summarizes key antibodies and their applications:
Reactivity: Primarily human, mouse, and rat; cross-reactivity with other species varies .
Atopic Dermatitis (AD): Serum HSP90 alpha levels are 5.08-fold higher in AD patients vs. dermatitis herpetiformis (DH) patients, suggesting its role as a biomarker .
Cancer: Overexpression in tumors (e.g., breast, liver) correlates with poor prognosis. Antibodies like ab2928 and AF7247 validate HSP90 alpha in carcinoma cell lines .
HSP90 alpha interacts with misfolded proteins (e.g., tau, amyloid-beta) and modulates proteostasis. Antibodies enable tracking of its role in Alzheimer’s and Parkinson’s disease models .
Celiac Disease (CD): Anti-HSP90 IgA correlates with disease activity, detectable via ab79849 in CD patient sera .
AD vs. DH: HSP90 alpha distinguishes AD from DH, with 83.9% positivity in AD patients using a 2×SD cutoff .
Antibody | Knockout Model | Observation | Source |
---|---|---|---|
AF7247 | HSP90a knockout HEK293T | No signal in KO; 96 kDa band in parental | |
ab79849 | HSP90AA1 knockout HAP1 | Signal lost in KO; 90 kDa band in WT |
Antibody | Sample Tested | Detection | Source |
---|---|---|---|
ab2928 | HeLa, NIH/3T3, human tumors | 85 kDa band; cytoplasmic localization | |
AF0774 | Mouse testis, rat kidney | Cross-reactivity confirmed |
HSP90 Alpha (Heat shock protein 90-alpha) is one of the two major cytoplasmic isoforms of the HSP90 family, alongside HSP90 Beta. It functions primarily as a molecular chaperone, assisting in the proper folding and stabilization of numerous client proteins, including steroid hormone receptors and kinases critical for cellular signaling and stress responses. This chaperoning function helps maintain cellular homeostasis and protects cells from stress-induced damage, influencing processes such as cell growth, differentiation, and apoptosis . HSP90 Alpha forms complexes with various proteins, including the glucocorticoid receptor, which is rendered transcriptionally inactive in the absence of ligand, highlighting its role in regulating gene expression . It represents approximately 1-2% of total mammalian cellular proteins under non-stress conditions, underscoring its abundance and importance in cellular function . In humans, HSP90 encompasses multiple gene isoforms: HSP90AA1, HSP90AA2, HSP90AB1 (cytoplasmic); HSP90B1 (ER-localized); and TRAP1 (mitochondrial), with HSP90 Alpha specifically encoded by the HSP90AA1 gene . Its involvement in numerous diseases, including cancer, neurodegenerative disorders, and respiratory conditions, makes it a crucial target for both basic research and therapeutic development .
Researchers have access to several types of HSP90 Alpha antibodies that vary in their host species, clonality, and conjugation status. Monoclonal antibodies, such as the Mouse monoclonal HSP90 Alpha antibody (clone 2G5.G3), offer high specificity and reproducibility for applications including Western blot, immunofluorescence, immunohistochemistry, and flow cytometry . These antibodies typically recognize specific epitopes of HSP90 Alpha with minimal cross-reactivity. Polyclonal antibodies, such as Rabbit polyclonal antibodies to HSP90 Alpha, recognize multiple epitopes on the target protein and are often used for applications like Western blot and ELISA . These polyclonal antibodies frequently demonstrate broader reactivity across species, including human, monkey, chicken, mouse, Drosophila, and rat samples . Additionally, HSP90 Alpha/Beta antibodies that recognize both isoforms are available, such as the mouse monoclonal IgG2a kappa light chain antibody (F-8), which detects HSP90 Alpha/Beta protein of mouse, rat, and human origin . These antibodies come in various forms, including non-conjugated versions and conjugated formats with tags such as horseradish peroxidase (HRP), phycoerythrin (PE), fluorescein isothiocyanate (FITC), and multiple Alexa Fluor® conjugates, providing researchers flexibility based on their experimental needs .
HSP90 Alpha antibodies are versatile research tools employed across multiple experimental techniques. Western blotting (WB) represents one of the most common applications, with recommended dilutions ranging from 1:500 to 1:2500 for polyclonal antibodies and approximately 1:1000 for monoclonal antibodies . This technique allows researchers to detect and quantify HSP90 Alpha protein expression levels in various cell and tissue lysates. Immunohistochemistry with paraffin-embedded sections (IHC-P) enables visualization of HSP90 Alpha localization within tissue architecture, providing insights into its distribution across different cell types in physiological and pathological contexts . Immunofluorescence (IF) and immunocytochemistry (ICC) applications permit detailed subcellular localization studies, with some antibodies showing effective performance at dilutions around 1:100 . Flow cytometry applications facilitate the quantitative analysis of HSP90 Alpha expression at the single-cell level, allowing researchers to correlate its expression with other cellular parameters . Immunoprecipitation (IP) enables the isolation of HSP90 Alpha and its interacting partners, facilitating the investigation of protein-protein interactions and complex formation . Additionally, enzyme-linked immunosorbent assay (ELISA) applications allow for quantitative detection of HSP90 Alpha in solution, with polyclonal antibodies typically used at dilutions ranging from 1:5,000 to 1:25,000 . This diverse range of applications makes HSP90 Alpha antibodies indispensable tools for investigating the protein's expression, localization, interactions, and functions in various research contexts.
When selecting an HSP90 Alpha antibody, researchers should first consider the specific research question and experimental technique. For experiments requiring high specificity and reproducibility, such as quantitative analyses across multiple samples or antibody validation studies, monoclonal antibodies like the mouse monoclonal HSP90A antibody (clone 2G5.G3) offer advantages due to their consistent epitope recognition . Conversely, for techniques benefiting from signal amplification or when detecting potentially modified or partially degraded proteins, polyclonal antibodies may be preferable as they recognize multiple epitopes . Species reactivity is another crucial consideration; researchers should verify that the antibody recognizes HSP90 Alpha in their experimental model organism, whether human, mouse, rat, or other species . Importantly, researchers must determine whether their experiments require specific detection of HSP90 Alpha alone or if recognition of both HSP90 Alpha and Beta isoforms is acceptable or advantageous. For isoform-specific studies, antibodies like the rabbit polyclonal to HSP90 Alpha, which specifically recognizes the Alpha isoform without cross-reactivity to Beta, would be appropriate . Technical compatibility with downstream applications should also guide selection, as certain conjugated antibodies are optimized for specific techniques like flow cytometry (fluorochrome-conjugated) or Western blotting (HRP-conjugated) . Validation status should be considered, with preference given to antibodies validated in the intended application and cited in peer-reviewed publications. Finally, researchers should review the immunogen information to understand what region of HSP90 Alpha the antibody recognizes, which may be particularly important when studying specific domains, post-translational modifications, or truncated forms of the protein .
Sample preparation techniques for HSP90 Alpha detection vary depending on the experimental approach and sample type. For Western blotting, effective cell lysis is critical, with RIPA buffer containing protease inhibitors commonly used to extract total HSP90 Alpha from cultured cells or tissues. When studying HSP90 Alpha in its native conformational state or investigating its interactions with client proteins, milder lysis conditions using non-ionic detergents like NP-40 or Triton X-100 may be preferable to preserve protein-protein interactions . For immunohistochemistry applications, formalin fixation followed by paraffin embedding (FFPE) is a standard approach, though researchers should optimize antigen retrieval methods (typically heat-mediated in citrate buffer pH 6.0) to ensure effective epitope exposure after fixation . When preparing samples for immunofluorescence or immunocytochemistry, paraformaldehyde fixation (typically 4%) followed by permeabilization with 0.1-0.5% Triton X-100 generally yields good results for intracellular detection of HSP90 Alpha . For flow cytometry, researchers must choose between surface staining protocols (if examining potential membrane-associated HSP90 Alpha) or fixation and permeabilization approaches for intracellular detection . Importantly, regardless of the application, researchers should include appropriate positive controls (such as cell lines known to express HSP90 Alpha, like HEK-293T cells) and negative controls (such as HSP90 Alpha knockout cell lines) to validate antibody specificity . Additionally, when quantitative comparisons are needed, consistent sample handling, standardized protein quantification methods, and inclusion of loading controls (such as GAPDH for Western blotting) are essential for reliable and reproducible results .
Optimal dilutions and incubation conditions for HSP90 Alpha antibodies vary significantly across different experimental applications and specific antibody products. For Western blotting, rabbit polyclonal HSP90 Alpha antibodies typically perform well at dilutions ranging from 1:500 to 1:2500, while mouse monoclonal antibodies may be used at around 1:1000 to 1:2000 . Incubation is generally performed overnight at 4°C in blocking buffer containing 5% non-fat dry milk or bovine serum albumin (BSA) in TBST (Tris-buffered saline with 0.1% Tween-20). For enzyme-linked immunosorbent assay (ELISA) applications, significantly higher dilutions are recommended, with rabbit polyclonal antibodies typically used at 1:5,000 to 1:25,000 . For immunocytochemistry and immunofluorescence applications, more concentrated antibody solutions are often required, with effective dilutions reported around 1:100 . These applications generally involve incubation for 1-2 hours at room temperature or overnight at 4°C in BSA-containing buffer. Flow cytometry typically requires antibody dilutions between 1:50 and 1:200, with incubation times of 30-60 minutes at room temperature or on ice . For immunohistochemistry on paraffin-embedded sections, antibody dilutions similar to those used in immunofluorescence are common (approximately 1:100), with incubation times typically ranging from 1-2 hours at room temperature to overnight at 4°C . It is important to note that these recommendations serve as starting points, and researchers should perform dilution series to optimize conditions for their specific experimental systems, antibody lots, and detection methods. Additionally, including appropriate controls, such as isotype controls for flow cytometry or antibody omission controls for immunohistochemistry, is essential for interpreting results accurately.
Distinguishing between HSP90 Alpha and Beta isoforms presents a significant challenge due to their high sequence homology (approximately 86% identical at the amino acid level). For isoform-specific detection, researchers should prioritize antibodies raised against unique epitopes, such as the rabbit polyclonal antibody that recognizes amino acids 289-300 of human HSP90 Alpha, a region with sequence divergence from the Beta isoform . Validation of isoform specificity is crucial and can be accomplished through several complementary approaches. Researchers can utilize knockout cell lines, such as HSP90AA1 (Alpha) knockout HEK-293T cells, which should show signal loss with Alpha-specific antibodies while maintaining reactivity with Beta-specific or pan-HSP90 antibodies . Alternatively, siRNA-mediated knockdown of each isoform separately can provide further validation of antibody specificity. Western blotting may reveal subtle size differences between the isoforms, with HSP90 Alpha typically appearing at approximately 90 kDa . For molecular analyses, researchers can employ isoform-specific PCR primers or probes targeting unique regions of HSP90AA1 (Alpha) and HSP90AB1 (Beta) mRNAs to quantify expression at the transcript level. Mass spectrometry-based proteomics approaches can also identify isoform-specific peptides for unambiguous discrimination. When conducting functional studies, researchers should consider that HSP90 Alpha is generally more inducible under stress conditions, while HSP90 Beta tends to be more constitutively expressed, providing another parameter for distinction . Additionally, subcellular localization studies may reveal differential distribution patterns between the isoforms in certain cell types or under specific conditions. When absolute discrimination is not possible with available reagents, researchers can employ complementary approaches that combine different detection methods to build a more comprehensive understanding of isoform-specific expression and function.
Investigating HSP90 Alpha interactions with client proteins requires sophisticated experimental approaches that preserve physiologically relevant protein-protein interactions. Co-immunoprecipitation (Co-IP) using HSP90 Alpha-specific antibodies represents a foundational technique, where antibodies like the mouse monoclonal IgG2a (F-8) can effectively pull down HSP90 Alpha complexes from cell lysates prepared under mild conditions that preserve protein-protein interactions . Reciprocal Co-IP (immunoprecipitating the client protein and probing for HSP90 Alpha) can provide confirmation of the interaction. For more comprehensive analysis of the HSP90 Alpha interactome, immunoprecipitation followed by mass spectrometry enables unbiased identification of associated proteins. Proximity ligation assay (PLA) offers an in situ approach to visualize and quantify HSP90 Alpha-client interactions within cells with high sensitivity and spatial resolution. Förster resonance energy transfer (FRET) or bioluminescence resonance energy transfer (BRET) techniques can reveal direct protein-protein interactions and provide information about their dynamics in living cells. Pull-down assays using recombinant HSP90 Alpha (full-length or domain-specific) as bait can identify direct binding partners and characterize binding domains. Researchers can employ chemical crosslinking prior to immunoprecipitation to stabilize transient or weak interactions that might otherwise be lost during purification. To assess the functional significance of these interactions, combining HSP90 inhibition (using compounds that target the ATP-binding site) with functional readouts of client protein activity can reveal dependency relationships . Genetic approaches, including HSP90 Alpha knockdown/knockout followed by assessment of client protein stability, localization, or function, provide complementary evidence. Advanced techniques like hydrogen-deuterium exchange mass spectrometry (HDX-MS) or cryo-electron microscopy can provide structural insights into these interactions. When designing these experiments, researchers should consider that HSP90 interactions are often dynamic, ATP-dependent, and may involve additional co-chaperones that modulate client recognition and processing .
Post-translational modifications (PTMs) of HSP90 Alpha significantly impact both its detection with antibodies and its biological functions. HSP90 Alpha undergoes numerous PTMs, including phosphorylation, acetylation, nitrosylation, methylation, and ubiquitination, which can modulate its chaperoning activity, client protein interactions, and cellular localization. When selecting antibodies, researchers should consider whether the epitope recognized contains known modification sites, as PTMs may mask antibody binding sites or alter protein conformation, potentially reducing detection efficiency . For comprehensive analysis of HSP90 Alpha PTMs, researchers often employ phospho-specific or acetylation-specific antibodies in combination with standard HSP90 Alpha antibodies. Mass spectrometry-based proteomic approaches provide the most comprehensive characterization of PTMs, identifying modification sites and their relative abundances. Western blotting may reveal multiple bands or mobility shifts corresponding to differentially modified forms of HSP90 Alpha . When investigating functional implications, researchers should note that phosphorylation at specific sites (such as Ser263 by casein kinase 2) can modulate HSP90 Alpha's ATPase activity and client protein interactions. Acetylation, particularly at lysine residues in the middle domain, has been shown to reduce chaperoning activity. S-nitrosylation can inhibit HSP90 Alpha's ATPase activity, thereby affecting its chaperoning function. To experimentally manipulate these modifications, researchers can use phosphatase treatments to remove phosphorylation, deacetylase inhibitors to preserve acetylation, or introduce point mutations at modification sites (e.g., phosphomimetic or phospho-resistant mutations). When designing experiments to study HSP90 Alpha under different cellular conditions, researchers should consider that stress responses, cell cycle phases, and differentiation states can significantly alter the PTM profile of HSP90 Alpha, which in turn affects its functionality and detection . Additionally, the ATP-binding status of HSP90 Alpha, which is critical for its function and is targeted by inhibitors in therapeutic approaches, can influence its conformation and consequently the accessibility of certain epitopes or modification sites .
Researchers frequently encounter several challenges when working with HSP90 Alpha antibodies. One common issue is cross-reactivity with HSP90 Beta due to their high sequence homology, which can confound isoform-specific analyses. To address this, researchers should validate antibody specificity using HSP90 Alpha knockout cell lines or siRNA-mediated knockdown, comparing signal patterns with known pan-HSP90 antibodies . Non-specific background signals, particularly in immunohistochemistry and immunofluorescence applications, may occur due to high endogenous biotin, endogenous peroxidase activity, or non-specific binding of secondary antibodies. Researchers can mitigate these issues by including biotin and peroxidase blocking steps, using species-matched negative controls, and optimizing blocking conditions with bovine serum albumin or serum from the secondary antibody host species . Variable detection sensitivity across different sample types may arise from inconsistent fixation, epitope masking due to protein-protein interactions, or post-translational modifications affecting epitope accessibility. To overcome these limitations, researchers should optimize sample preparation protocols, consider alternative fixation methods, or employ multiple antibodies targeting different epitopes of HSP90 Alpha . For quantitative Western blot analyses, the high abundance of HSP90 (1-2% of cellular proteins) can lead to signal saturation, masking subtle expression changes . This can be addressed by reducing sample loading, shortening exposure times, or using gradient gels for better resolution. When investigating HSP90 Alpha in stress conditions, the rapid and dynamic changes in its expression, localization, and modification state can complicate interpretation. Time-course studies with appropriate controls are essential for capturing these dynamics accurately. Additionally, when studying HSP90 Alpha in disease models or patient samples, heterogeneity in expression patterns may reflect biological variability rather than technical issues, necessitating increased sample sizes and rigorous statistical analyses. To ensure reproducibility, researchers should maintain detailed records of antibody sources, lot numbers, and optimized protocols, particularly as antibody performance can vary between lots.
Comprehensive validation of HSP90 Alpha antibody specificity is essential for generating reliable and interpretable research data. A multi-faceted approach begins with genetic validation using HSP90 Alpha (HSP90AA1) knockout or knockdown systems. This can be accomplished using CRISPR/Cas9-engineered knockout cell lines, such as HSP90AA1 knockout HEK-293T cells, which should show complete loss of signal with Alpha-specific antibodies while maintaining reactivity with Beta-specific or pan-HSP90 antibodies . Similarly, siRNA or shRNA-mediated knockdown of HSP90AA1 should result in proportional signal reduction. Peptide competition assays provide another validation strategy, where pre-incubation of the antibody with the immunizing peptide (such as the synthetic peptide corresponding to amino acids 289-300 of human HSP90 Alpha) should abolish specific binding . Western blot analysis should reveal a single predominant band at the expected molecular weight (approximately 90 kDa), and researchers should compare the banding pattern across multiple cell lines with known HSP90 Alpha expression levels . Orthogonal detection methods employing alternative antibodies targeting different epitopes of HSP90 Alpha should yield consistent results regarding expression patterns and localization. Mass spectrometry confirmation, particularly for immunoprecipitation experiments, can provide unambiguous identification of the immunoprecipitated protein as HSP90 Alpha. For immunohistochemistry and immunofluorescence applications, researchers should observe staining patterns consistent with the known subcellular localization of HSP90 Alpha, predominantly cytoplasmic with potential nuclear localization under certain conditions . Additionally, researchers can leverage tissue or cell type-specific expression patterns as internal controls, as certain tissues exhibit characteristic HSP90 Alpha expression levels. When possible, antibody performance should be benchmarked against published results using the same antibody, particularly those from studies with rigorous validation approaches. Finally, researchers should confirm that the antibody response aligns with expected biological dynamics, such as increased HSP90 Alpha expression under heat shock or other stress conditions that are known to specifically upregulate this isoform .
Implementing rigorous quality control measures ensures reliable and reproducible results when working with HSP90 Alpha antibodies. Researchers should maintain detailed documentation of antibody specifications, including catalog numbers, lot numbers, host species, clonality, and immunogen information, as performance can vary between different lots of the same antibody . Upon receiving a new antibody, initial validation should include Western blotting with positive control lysates (e.g., HEK-293T cells) to confirm detection at the expected molecular weight of approximately 90 kDa . For each new experimental system or application, concentration optimization through titration experiments is essential, as optimal dilutions vary significantly between applications (e.g., 1:500-1:2500 for Western blot versus 1:5,000-1:25,000 for ELISA with polyclonal antibodies) . Every experiment should incorporate appropriate positive and negative controls: positive controls might include cell lines or tissues known to express HSP90 Alpha, while negative controls might include HSP90 Alpha knockout cells, isotype controls (for flow cytometry), or primary antibody omission controls (for immunohistochemistry) . For quantitative analyses, standard curves using recombinant HSP90 Alpha protein can help establish the linear detection range and absolute quantification capabilities. Regular antibody performance monitoring can detect degradation or variation over time; aliquoting antibodies and storing them according to manufacturer recommendations (typically at -20°C for long-term storage) helps maintain consistency . When publications or grant applications are being prepared, researchers should document the validation methods employed, following guidelines such as those from the International Working Group for Antibody Validation (IWGAV). For studies involving multiple antibodies or detection of both HSP90 Alpha and Beta isoforms, cross-reactivity testing is crucial to ensure specificity. Researchers should also implement experimental design controls such as randomization, blinding, and technical replicates to minimize bias and enhance reproducibility. Finally, when presenting or publishing results, transparent reporting of all antibody details, validation approaches, and experimental conditions enables proper evaluation and reproducibility by the scientific community.
HSP90 Alpha antibodies serve as critical tools in cancer research, where HSP90 is frequently overexpressed and associated with tumor progression, metastasis, and treatment resistance. In fundamental cancer biology investigations, these antibodies enable researchers to assess HSP90 Alpha expression levels across various cancer types through immunohistochemistry of tissue microarrays, revealing correlations between expression patterns and clinical outcomes . When conducting such studies, researchers should implement standardized scoring systems and include pathologist validation to ensure consistent interpretation. For mechanistic studies examining HSP90 Alpha's role in oncogenic signaling, antibodies facilitate the investigation of interactions between HSP90 Alpha and client proteins critical in cancer progression, including HER2, EGFR, AKT, and mutant p53 . These studies typically employ co-immunoprecipitation approaches, which require careful optimization of lysis conditions to preserve protein-protein interactions. In the exploration of HSP90 Alpha's extracellular functions in cancer, which include stimulation of tumor cell migration and angiogenesis, researchers utilize antibodies in neutralization experiments to block these functions and assess phenotypic consequences. When studying the dynamic responses of HSP90 Alpha to anti-cancer treatments, time-course analyses with phospho-specific antibodies can reveal changes in post-translational modifications that may indicate adaptive resistance mechanisms . For translational research evaluating HSP90 Alpha as a biomarker, quantitative approaches like ELISA of patient serum samples can detect secreted HSP90 Alpha, which has been associated with aggressive disease in several cancer types . The development of HSP90 inhibitors as cancer therapeutics has spurred the use of antibodies in pharmacodynamic assays to monitor treatment effects on HSP90 client protein stability and HSP70 induction (a compensatory response to HSP90 inhibition) . Additionally, researchers investigating the potential of HSP90 Alpha-targeting antibodies as therapeutic agents themselves require extensive characterization of antibody specificity, binding kinetics, and functional effects on cancer cells. The differential expression and function of HSP90 Alpha versus Beta isoforms in cancer contexts necessitates isoform-specific antibodies for precise characterization of their distinct roles in tumorigenesis .
Investigating HSP90 Alpha in neurodegenerative diseases requires specialized methodological approaches that address the unique challenges of neural tissue analysis and the complex pathophysiology of these disorders. Researchers employ immunohistochemistry with HSP90 Alpha-specific antibodies to examine expression patterns in post-mortem brain tissue from patients with conditions such as Alzheimer's, Parkinson's, and Huntington's diseases, comparing these to age-matched controls . When conducting such studies, careful standardization of tissue procurement, fixation, and antigen retrieval protocols is essential to minimize variability introduced by post-mortem interval and fixation artifacts. Dual immunofluorescence labeling allows co-localization analyses of HSP90 Alpha with disease-specific protein aggregates (e.g., amyloid-β, tau, α-synuclein, or huntingtin), providing insights into potential chaperoning relationships . For mechanistic investigations in cellular models, researchers utilize primary neuronal cultures or induced pluripotent stem cell (iPSC)-derived neurons from patients, applying HSP90 Alpha antibodies in conjunction with markers of neuronal stress, protein aggregation, and cell death. In these models, temporal profiling of HSP90 Alpha expression and localization during disease progression can reveal critical intervention points. Biochemical fractionation of brain tissue followed by Western blotting enables assessment of HSP90 Alpha distribution between soluble and insoluble protein fractions, the latter being particularly relevant in protein misfolding disorders . Co-immunoprecipitation studies help identify interactions between HSP90 Alpha and neurodegeneration-associated client proteins, though these require careful optimization to preserve often weak or transient interactions in neural tissues . For functional studies, researchers combine HSP90 Alpha antibodies with live-cell imaging techniques to visualize the dynamics of protein aggregate formation and clearance in the presence of HSP90 modulators. Additionally, cerebrospinal fluid (CSF) analysis using highly sensitive ELISA methods can detect potential changes in extracellular HSP90 Alpha levels as biomarkers of disease progression or treatment response . In animal models of neurodegeneration, immunohistochemical mapping of HSP90 Alpha expression across different brain regions at various disease stages provides valuable spatial and temporal information. When designing these studies, researchers should consider the potential compensatory relationships between HSP90 Alpha and other chaperones, necessitating multiplexed detection approaches to comprehensively characterize the chaperone network dynamics in neurodegeneration contexts .
HSP90 Alpha antibodies offer valuable tools for investigating this chaperone's emerging roles across diverse disease contexts beyond cancer and neurodegeneration. In cardiovascular disease research, these antibodies enable the examination of HSP90 Alpha's interactions with nitric oxide synthase and soluble guanylate cyclase, proteins critical for vascular function . When designing such studies, researchers should employ co-immunoprecipitation followed by activity assays to assess both physical interactions and functional consequences. For investigations in pulmonary arterial hypertension (PAH) and asthma, where HSP90 has been implicated in disease pathophysiology, immunohistochemical analyses of lung tissue using validated antibodies can reveal altered expression patterns in affected versus healthy tissues . These studies benefit from dual-staining approaches to identify the specific cell types exhibiting dysregulated HSP90 Alpha expression. In inflammatory and autoimmune conditions, researchers can utilize flow cytometry with fluorochrome-conjugated HSP90 Alpha antibodies to quantitatively assess expression in different immune cell populations, correlating levels with activation status and functional parameters . When investigating potential extracellular functions of HSP90 Alpha in wound healing and tissue repair, researchers employ neutralizing antibodies in functional assays to block HSP90 Alpha-mediated effects on cell migration and extracellular matrix remodeling. For exploring HSP90 Alpha's emerging roles in metabolic disorders, Western blotting and immunoprecipitation approaches help characterize interactions with metabolic enzymes and insulin signaling components . In studying the involvement of HSP90 Alpha in hematopoietic disorders, researchers can leverage antibodies to investigate its role in globin maturation, potentially revealing insights relevant to conditions like sickle cell anemia and thalassemia . The optimization of chromatin immunoprecipitation (ChIP) protocols with HSP90 Alpha antibodies enables exploration of its potential epigenetic functions, as HSP90 has been shown to affect the epigenetic state at specific loci . When designing therapeutic interventions targeting HSP90 in these diverse disease contexts, researchers utilize antibodies in pharmacodynamic assays to monitor the effects of HSP90 inhibitors on client protein stability . Additionally, for diseases where extracellular HSP90 Alpha may play a pathogenic role, the development of therapeutic antibodies requires extensive characterization of binding specificity, affinity, and neutralizing capacity. Throughout these investigations, researchers should consider the differential regulation and potentially distinct functions of HSP90 Alpha versus Beta isoforms in different tissue and disease contexts, necessitating isoform-specific detection approaches .
Heat shock proteins (HSPs) are a family of proteins that play a crucial role in protecting cells from stress. Among them, Heat Shock Protein 90-alpha (HSP90α) is a highly conserved molecular chaperone involved in the proper folding, stabilization, and degradation of many proteins. This article delves into the background of HSP90α, particularly focusing on the mouse anti-human HSP90α antibody.
HSP90α is one of the isoforms of the HSP90 family, which also includes HSP90β. It is a cytosolic protein that is highly expressed in eukaryotic cells and accounts for 1-2% of total cellular proteins under non-stressed conditions. This percentage can increase to 4-6% under stress conditions .
HSP90α is composed of three main domains:
The primary function of HSP90α is to act as a molecular chaperone, assisting in the proper folding of nascent proteins, stabilization of proteins against heat stress, and aiding in protein degradation. It also plays a significant role in the maturation and activation of various client proteins, including steroid hormone receptors, kinases, and transcription factors .
HSP90α is implicated in various diseases, particularly cancer. It stabilizes several oncoproteins, making it a target for anti-cancer therapies. HSP90 inhibitors are being investigated for their potential to disrupt the function of HSP90α, thereby destabilizing its client proteins and inhibiting tumor growth .
Mouse anti-human HSP90α antibodies are monoclonal or polyclonal antibodies developed in mice to specifically target the human HSP90α protein. These antibodies are widely used in research and diagnostic applications, including Western blotting, immunoprecipitation, immunohistochemistry, and flow cytometry .
The development of mouse anti-human HSP90α antibodies involves immunizing mice with human HSP90α protein or peptides, followed by the collection and purification of the antibodies produced. These antibodies are then validated for their specificity and reactivity with human HSP90α .
In research, these antibodies are used to study the expression, localization, and function of HSP90α in various biological contexts. They are also employed in diagnostic assays to detect HSP90α levels in clinical samples, aiding in the diagnosis and prognosis of diseases such as cancer .