HSPB9 Human

Heat Shock Protein B9 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to HSPB9 Human

HSPB9 (Heat Shock Protein Family B (Small) Member 9) is a 20 kDa protein encoded by the HSPB9 gene located on chromosome 17q21.2 . As a member of the small heat shock protein (HSPB) family, it shares the conserved α-crystallin domain critical for oligomerization and chaperone activity . HSPB9 exhibits tissue-specific expression and emerging roles in cellular stress response and cancer biology.

Gene Characteristics

PropertyDetails
Gene SymbolHSPB9
Chromosomal Location17q21.2
Gene TypeProtein-coding
AliasesCT51, Cancer/Testis Antigen 51

Protein Structure

HSPB9 contains the α-crystallin domain, enabling interactions with misfolded proteins and cytoskeletal components . Its sequence homology to other HSPBs (e.g., HSPB1, HSPB5) suggests shared chaperone functions, though distinct tissue expression patterns distinguish it .

Tissue Expression and Cellular Localization

HSPB9 is predominantly expressed in testis germ cells, where it is involved in spermatogenesis . Notably, it is also detected in tumor tissues, classifying it as a cancer/testis antigen (CTA51) .

TissueExpression LevelCellular Localization
TestisHighGerm cells (spermatogenesis)
Tumors (e.g., breast, lung)VariableCytoplasm and nucleus

Key Functions

  1. Chaperone Activity: Protects proteins from stress-induced aggregation via α-crystallin domain-mediated binding .

  2. Cancer/Testis Antigen: Restricted expression in normal tissues but upregulated in tumors, making it a potential therapeutic target .

  3. Apoptosis Regulation: Overexpression induces early apoptosis in chicken DF-1 cells, suggesting dual roles in cell survival and stress response .

Interacting Proteins

PartnerInteraction ContextSource
TCTEL1 (dynein subunit)Cytoplasmic transport regulation
Dynein ComplexMicrotubule-based transport

Experimental Insights

  • Apoptosis Modulation: HSPB9 overexpression in DF-1 cells triggers early apoptosis but later enhances survival, indicating context-dependent effects .

  • Cancer Association: Detected in breast, lung, and other cancers, with tumor-specific expression patterns .

Clinical Relevance

  • Biomarker Potential: As a CTA, HSPB9 could serve as a target for immunotherapy or diagnostics in cancers .

  • Neurological and Muscular Disorders: Paralogs like HSPB1 and HSPB5 are linked to neuropathies and cardiomyopathies, suggesting potential roles for HSPB9 in related diseases .

Product Specs

Introduction
Heat Shock Protein B9 (HSPB9) is a small heat shock protein belonging to the HSP20 family. It is present in both the cytoplasm and nucleus of cells and is exclusively produced in the testes.
Description
Recombinant human HSPB9, expressed in E. coli, is a single, non-glycosylated polypeptide chain comprising 179 amino acids (residues 1-159) and possessing a molecular weight of 19.6 kDa. This protein includes a 20 amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
A clear, sterile-filtered solution.
Formulation
The HSPB9 solution is provided at a concentration of 0.25 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 50% glycerol, 0.2 M NaCl, and 2 mM DTT.
Stability
For short-term storage (2-4 weeks), the solution should be kept at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of this protein is greater than 85% as determined by SDS-PAGE analysis.
Synonyms
Heat shock protein beta-9, HspB9, Cancer/testis antigen 51, CT51, HSPB9.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MQRVGNTFSN ESRVASRCPS VGLAERNRVA TMPVRLLRDS PAAQEDNDHA RDGFQMKLDA HGFAPEELVV QVDGQWLMVT GQQQLDVRDP ERVSYRMSQK VHRKMLPSNL SPTAMTCCLT PSGQLWVRGQ CVALALPEAQ TGPSPRLGSL GSKASNLTR.

Q&A

What is HSPB9 and how was it initially identified?

HSPB9 represents the ninth member of the human small heat shock protein (sHsp) family, characterized by its tissue-specific expression pattern and specialized function. It was initially identified through investigations of EST (Expressed Sequence Tag) databases when researchers were searching for new members of the human small heat shock protein family . The protein contains the characteristic alpha-crystallin domain that is conserved across all small heat shock proteins, confirming its classification within this protein family . The initial discovery was reported by Kappé et al. in 2001 in Biochimica et Biophysica Acta, where they established its testis-specific expression through Northern blotting techniques . Subsequent research by the same team confirmed this expression pattern through RT-PCR analysis in a wider range of normal tissues, further solidifying HSPB9's classification as a testis-specific heat shock protein . The protein was formally named following the sequential numbering system for human small heat shock proteins, with HSPB9 representing the ninth identified member of this specialized chaperone family .

What is the tissue expression pattern of HSPB9 in normal human physiology?

HSPB9 demonstrates a highly restricted tissue expression pattern, with expression almost exclusively confined to the testis in normal human physiology . This distinct expression profile places HSPB9 in contrast with many other members of the small heat shock protein family, as approximately half of human HSPBs (specifically B1, B2, B5, B6, B7, and B8) are ubiquitously expressed throughout the body . Northern blotting techniques have consistently verified the testis-specific expression of HSPB9, and this finding has been further corroborated through more sensitive RT-PCR analysis across an extensive panel of normal human tissues . Within the testis, immunohistochemical staining has revealed that HSPB9 is specifically expressed in spermatogenic cells, with expression beginning at the late pachytene spermatocyte stage and continuing through to the elongate spermatid stage of sperm development . This highly regulated temporal expression pattern during spermatogenesis suggests a specialized function in sperm development and maturation processes. The remarkably restricted expression pattern of HSPB9 is relatively uncommon among heat shock proteins and indicates a highly specialized physiological role in male reproductive biology .

How does HSPB9 differ structurally from other members of the small heat shock protein family?

HSPB9 shares the characteristic alpha-crystallin domain that defines all small heat shock proteins, yet exhibits several structural distinctions that set it apart from other family members . Like all HSPBs, HSPB9 contains the highly conserved central alpha-crystallin domain, flanked by less conserved N-terminal and C-terminal domains that contribute to its unique functional properties . Comparative protein sequence analysis reveals that HSPB9 displays considerable evolutionary divergence, particularly evident in the finding that mouse HSPB9 shows a remarkable 38% sequence difference from human HSPB9 - a degree of divergence that is unusually high among mammalian heat shock proteins, which are typically highly conserved across species . This substantial interspecies variation suggests that HSPB9 may have evolved specialized functions related to reproduction, possibly reflecting species-specific adaptations in spermatogenesis . In protein interaction networks, HSPB9 shows functional relationships with several other heat shock proteins, including HSPB7, CRYAA, HSPB2, HSPB6, and HSPB8, though with varying confidence scores for these interactions . The highest predicted functional partnership (scoring 0.787) is with HSPB7, suggesting possible cooperative functions despite their different tissue expression patterns .

What experimental methods are commonly used to detect and measure HSPB9 expression?

Researchers investigating HSPB9 employ various complementary molecular and cellular techniques to accurately detect and quantify its expression across different experimental contexts. Northern blotting was initially used to establish the testis-specific expression pattern of HSPB9, providing a foundation for understanding its tissue distribution . This technique continues to be valuable for examining HSPB9 mRNA expression across multiple tissue types. For more sensitive detection and quantification of HSPB9 transcript levels, reverse transcription polymerase chain reaction (RT-PCR) has been extensively utilized to verify and expand upon the initial Northern blotting findings . RT-PCR has proven particularly valuable for detecting potential low-level expression that might be missed by less sensitive methods, helping researchers establish the truly restricted expression pattern of HSPB9. At the protein level, immunohistochemical (IHC) staining using specific antibodies against HSPB9 has been instrumental in localizing its expression within testicular tissue and tumor samples . IHC analysis has revealed the specific developmental stages of spermatogenesis during which HSPB9 is expressed, providing critical insights into its potential biological functions . For protein interaction studies, yeast two-hybrid screening has been successfully employed to identify HSPB9-interacting proteins, most notably revealing its interaction with TCTEL1, a dynein light chain component .

What evidence supports HSPB9's classification as a cancer/testis antigen?

HSPB9 meets the defining criteria for classification as a cancer/testis antigen (CTA) based on multiple lines of experimental evidence across different detection methodologies. The fundamental characteristic of CTAs is their restricted expression in normal tissues (primarily testis) combined with aberrant expression in various cancer types, a pattern conclusively demonstrated for HSPB9 . During an examination of HSPB9 EST databases, researchers observed evidence of expression in tumor samples, prompting further investigation using more rigorous techniques . RT-PCR analysis subsequently confirmed HSPB9 expression across multiple tumor types, providing molecular validation of its presence in cancer tissues despite its absence in corresponding normal tissues . At the protein level, immunohistochemical staining has verified HSPB9 protein expression in various tumor samples, establishing concordance between transcript and protein expression in cancerous tissues . This combination of restricted normal tissue expression and aberrant activation in multiple cancer types conclusively positions HSPB9 within the cancer/testis antigen family, a group of proteins with significant implications for cancer immunotherapy due to their potential as tumor-specific targets . This classification has important implications for potential diagnostic and therapeutic applications in oncology research.

In which types of cancer has HSPB9 expression been detected?

HSPB9 expression has been detected across multiple cancer types, though comprehensive profiling of its expression across the full spectrum of human malignancies remains incomplete. Initially, the presence of HSPB9 was noted in tumor-derived EST sequences, prompting targeted investigation of its expression in cancer tissues . Subsequent RT-PCR analyses confirmed HSPB9 expression in various tumors, though the search results don't specify all particular cancer types examined . Immunohistochemical staining has further verified HSPB9 protein expression in tumor samples, providing visual confirmation of its presence in cancerous tissues . Recent research has suggested potential involvement of HSPB9 in chemoresistance mechanisms in ovarian cancer, indicating its expression and possible functional relevance in this gynecological malignancy . As a cancer/testis antigen, HSPB9 likely follows expression patterns similar to other members of this family, which are frequently detected in melanomas, lung carcinomas, breast cancers, bladder cancers, and various other malignancies . The expression of HSPB9 across different cancer stages, histological subtypes, and its correlation with clinical outcomes represents an area requiring further systematic investigation to fully understand its potential diagnostic, prognostic, and therapeutic implications in oncology.

What experimental approaches can be used to investigate HSPB9's role in cancer progression?

Investigating HSPB9's functional role in cancer progression requires a multi-faceted experimental approach combining molecular, cellular, and in vivo methodologies. Gene modulation techniques represent an essential starting point, wherein HSPB9 expression can be manipulated through RNA interference (siRNA or shRNA) for knockdown studies or overexpression vectors for gain-of-function experiments in cancer cell lines . These interventions allow researchers to assess the direct impact of HSPB9 on cellular phenotypes relevant to malignancy, including proliferation rates, resistance to apoptosis, migration, invasion, and chemoresistance . For mechanistic insights, protein interaction studies using co-immunoprecipitation, proximity ligation assays, or yeast two-hybrid screens can identify cancer-relevant binding partners of HSPB9, building upon the established interaction with TCTEL1 and potentially revealing connections to oncogenic signaling pathways . Cell stress response analyses can determine whether HSPB9 contributes to cancer cell survival under conditions relevant to the tumor microenvironment, such as hypoxia, nutrient deprivation, or exposure to therapeutic agents . In vivo studies using xenograft models with HSPB9-modulated cancer cells can evaluate its impact on tumor formation, growth kinetics, metastatic potential, and response to therapy . For translational relevance, correlative studies using tissue microarrays can connect HSPB9 expression levels with clinical parameters including disease stage, treatment response, and patient survival across multiple cancer types .

How might HSPB9's role as a cancer/testis antigen be exploited for cancer immunotherapy?

HSPB9's restricted expression pattern and classification as a cancer/testis antigen create several strategic opportunities for cancer immunotherapy development. The near-absence of HSPB9 expression in normal adult tissues outside the immune-privileged testis minimizes the risk of off-target effects, making it an attractive target for cancer-directed immunotherapeutic approaches . Peptide vaccine development represents one promising approach, wherein HSPB9-derived peptides that bind to common HLA molecules could be identified through epitope mapping and used to stimulate HSPB9-specific T-cell responses in cancer patients . Adoptive T-cell therapy approaches could involve isolating and expanding naturally occurring HSPB9-reactive T cells from patients or engineering T cells with HSPB9-specific T-cell receptors or chimeric antigen receptors . For antibody-based strategies, if HSPB9 demonstrates any cell-surface expression or protein secretion in cancer cells, it could be targeted using monoclonal antibodies, antibody-drug conjugates, or bispecific T-cell engagers . Checkpoint inhibitor combination strategies might enhance the efficacy of HSPB9-targeted approaches by simultaneously blocking immunosuppressive mechanisms that could otherwise limit anti-HSPB9 immune responses . Additionally, diagnostic applications could involve detecting HSPB9 expression in tumor samples or circulating tumor cells as a biomarker for certain cancer types or as a means of identifying patients who might benefit from HSPB9-targeted immunotherapies .

What is known about HSPB9's interaction with TCTEL1 and its functional significance?

HSPB9 has been conclusively shown to interact with TCTEL1 (T-Complex Testis Expressed Protein 1), a light chain component of both cytoplasmic and flagellar dynein, suggesting a potential role in cellular transport mechanisms . This interaction was originally identified through a yeast two-hybrid screen specifically designed to search for HSPB9-interacting proteins, representing a systematic approach to understanding HSPB9's functional network . The interaction was subsequently validated through complementary immunoprecipitation experiments, confirming that the association observed in the yeast system also occurs in a more physiologically relevant context . Immunohistochemical staining has revealed co-expression of HSPB9 and TCTEL1 in similar stages of spermatogenesis and in tumor cells, providing spatial and temporal evidence supporting the biological relevance of this interaction . From a functional perspective, this interaction may connect HSPB9 to dynein-mediated transport processes, potentially involving the movement of cellular components along microtubules during spermatogenesis . Given that proper cytoskeletal organization and intracellular transport are critical for sperm development and function, the HSPB9-TCTEL1 interaction may represent a specialized mechanism supporting these processes in male germ cells .

What other protein interactions have been identified for HSPB9?

Beyond its well-established interaction with TCTEL1, HSPB9 has been predicted to interact with several other proteins, primarily within the heat shock protein family and reproductive system-specific proteins. According to STRING database analysis, HSPB9 shows potential functional partnerships with several other heat shock proteins, most notably HSPB7 (with a confidence score of 0.787), suggesting possible cooperative functions or regulatory relationships within the heat shock protein network . Other predicted interacting partners include ODF1 (Outer Dense Fiber Protein 1), a component of the outer dense fibers of spermatozoa that helps maintain the structural integrity and elastic properties of the sperm tail, with this interaction scoring 0.743 in confidence metrics . Additional heat shock proteins showing potential functional relationships with HSPB9 include CRYAA (Alpha-crystallin A2, scoring 0.698), HSPB2 (Heat Shock Protein Beta-2, scoring 0.690), HSPB6 (Heat Shock Protein Beta-6, scoring 0.686), and HSPB8 (Heat Shock Protein Beta-8, scoring 0.683) . These interaction predictions are based on various evidence types including neighborhood analysis, gene fusion events, co-occurrence patterns, co-expression data, experimental evidence, database annotations, and text mining results . It is important to note that many of these interactions remain computationally predicted and require experimental validation through techniques such as co-immunoprecipitation, proximity ligation assays, or fluorescence resonance energy transfer to confirm their biological relevance.

What are the proposed molecular functions of HSPB9 based on current research?

Current research suggests that HSPB9 likely functions as a specialized molecular chaperone with particular relevance to spermatogenesis and possibly stress protection in certain cancer contexts. As a member of the small heat shock protein family, HSPB9 is expected to share the characteristic "holdase" function - the ability to interact with substrate proteins to prevent their aggregation under stress conditions, a foundational activity of all sHsps . Given its testis-specific expression pattern and temporal regulation during spermatogenesis, HSPB9 may provide chaperoning activity for proteins undergoing dramatic conformational changes during sperm development and maturation . The interaction with TCTEL1 suggests a potential role in dynein-mediated transport processes, which are crucial for proper spermatogenesis and sperm function, indicating HSPB9 may help coordinate protein transport along the cytoskeleton during male germ cell development . In embryonic development, particularly limb formation, research has indicated that HSPB9 may play important regulatory roles, as it has been implicated in both normal and abnormal development of embryonic forelimbs and hindlimbs . Within cancer contexts, preliminary evidence suggests HSPB9 may contribute to chemoresistance mechanisms in certain malignancies such as ovarian cancer, potentially by helping cancer cells tolerate cellular stress induced by chemotherapeutic agents .

How does HSPB9 contribute to spermatogenesis and male fertility?

HSPB9's contribution to spermatogenesis is suggested by its highly regulated expression pattern during specific stages of male germ cell development and its interaction with proteins involved in sperm structure and function. The protein's expression initiates at the late pachytene spermatocyte stage and continues through to the elongate spermatid stage, corresponding to critical periods of sperm differentiation and morphological transformation . This precisely timed expression pattern suggests HSPB9 may provide stage-specific chaperoning activity during dramatic cellular remodeling events that occur during spermiogenesis, including nuclear condensation, acrosome formation, and flagellum development . The interaction between HSPB9 and TCTEL1, a component of the dynein motor complex, indicates a potential role in intracellular transport processes that are essential for proper sperm development, particularly the organization of cytoskeletal elements during sperm tail formation . Additionally, HSPB9's predicted interaction with ODF1, a structural component of the sperm tail's outer dense fibers, further supports its involvement in the development of functional sperm ultrastructure . The substantial evolutionary divergence observed between human and mouse HSPB9 (38% sequence difference) is unusually high for heat shock proteins, suggesting species-specific adaptations in reproductive functions that may reflect different selective pressures on sperm competition or fertilization mechanisms across mammalian species .

What methodologies can be used to investigate HSPB9's chaperone function and substrate specificity?

Investigating HSPB9's chaperone function and identifying its specific substrate proteins requires sophisticated biochemical and cellular approaches adapted for this testis-specific heat shock protein. In vitro aggregation assays represent a foundational approach, wherein purified recombinant HSPB9 can be tested for its ability to prevent the aggregation of model substrate proteins (such as citrate synthase, luciferase, or insulin) under stress conditions including elevated temperature, chemical denaturants, or oxidative stress . These assays can be quantified through light scattering, centrifugation-based sedimentation analysis, or fluorescence-based detection of protein aggregates. Thermal stability shift assays can evaluate whether HSPB9 binding increases the melting temperature of potential substrate proteins, providing evidence of stabilizing interactions . For identifying natural substrate proteins in testicular cells, proximity-based labeling methods such as BioID or APEX can be employed, wherein HSPB9 is fused to a promiscuous biotin ligase that biotinylates proteins in close proximity, allowing subsequent purification and mass spectrometry-based identification of interaction partners . Co-immunoprecipitation combined with mass spectrometry represents another powerful approach for identifying HSPB9-binding proteins in either testicular cell lysates or cancer cells expressing HSPB9 . Fluorescence recovery after photobleaching (FRAP) experiments with fluorescently tagged HSPB9 and potential substrates can assess the dynamics of these interactions in living cells . Hydrogen-deuterium exchange mass spectrometry can map the specific binding interfaces between HSPB9 and its substrate proteins, providing structural insights into these interactions .

How can researchers effectively study HSPB9 in developmental contexts, particularly embryonic limb formation?

Studying HSPB9 in developmental contexts, particularly embryonic limb formation, requires specialized approaches that account for its dynamic expression patterns and potential regulatory functions during embryogenesis. Spatiotemporal expression profiling using techniques such as in situ hybridization, immunohistochemistry, or single-cell RNA sequencing can map HSPB9 expression patterns throughout embryonic development with high resolution, identifying specific tissues, cell types, and developmental stages where HSPB9 is active . Gene perturbation studies using CRISPR/Cas9-mediated knockout or knockdown approaches in model organisms (particularly mouse models given the established role of HSPB9 in limb development) can reveal developmental phenotypes associated with HSPB9 deficiency . Conditional and inducible gene modulation systems are particularly valuable for studying embryonic development, allowing temporal control over HSPB9 disruption to pinpoint critical windows for its function . Three-dimensional limb bud cultures or organoid models can provide ex vivo systems for manipulating HSPB9 expression and observing effects on limb development under controlled conditions, with advanced imaging techniques to track morphological changes . For studying HSPB9's role in abnormal limb development, teratogen-induced models such as all-trans retinoic acid (atRA) treatment (which has been shown to cause limb malformations and alter HSPB9 expression) can be particularly informative . Comparative studies examining HSPB9 expression and function in normal versus malformed limbs can identify potential protective or pathogenic roles in response to developmental stressors or teratogens .

What bioinformatic approaches can help predict HSPB9 function and evolutionary significance?

Advanced bioinformatic approaches offer powerful tools for predicting HSPB9's function and understanding its evolutionary trajectory across species. Comparative genomic analysis examining HSPB9 sequence conservation, divergence patterns, and selection signatures across multiple species can provide insights into its evolutionary history and functional constraints, particularly interesting given the unusual 38% sequence divergence observed between human and mouse HSPB9 . Protein structure prediction using AlphaFold or similar deep learning algorithms can generate high-confidence three-dimensional models of HSPB9's structure, enabling visualization of its alpha-crystallin domain and identifying potential substrate-binding regions or interaction interfaces . Molecular dynamics simulations based on these structural models can investigate HSPB9's conformational dynamics, oligomerization properties, and potential mechanisms of substrate recognition . Gene co-expression network analysis using large-scale transcriptomic datasets from testicular tissue or cancer samples can identify genes with expression patterns correlated with HSPB9, suggesting functional relationships or shared regulatory mechanisms . Promoter analysis examining transcription factor binding sites in the HSPB9 gene regulatory regions can reveal potential regulatory mechanisms controlling its testis-specific expression pattern . Interactome prediction algorithms integrating multiple data types (co-expression, co-occurrence, experimental evidence) can expand upon the STRING database predictions to identify additional potential interaction partners . Pathway enrichment analysis for predicted HSPB9 interactors can suggest biological processes or cellular pathways in which HSPB9 may participate, generating hypotheses for functional studies .

What evidence links HSPB9 to embryonic limb development?

Several lines of evidence from developmental biology research have established connections between HSPB9 expression and embryonic limb formation, particularly in the context of normal and abnormal development. Studies examining gene expression patterns during mouse embryogenesis have identified HSPB9 as one of several heat shock proteins that demonstrate regulated expression during limb development . Specifically, research has shown that HSPB9 expression levels change significantly during critical periods of hindlimb development, typically occurring during gestational days 12-14 in mouse models . In experimental models of limb malformation induced by teratogens such as all-trans retinoic acid (atRA), HSPB9 expression patterns show significant alterations compared to control embryos, suggesting potential involvement in the response to developmental stressors affecting limb morphogenesis . Comparative analyses between normal and abnormal limb development have led researchers to conclude that HSPB9, along with several other heat shock proteins including HspB2, HspB10, and Grp78, "probably performed an important role in normal and abnormal development of embryonic forelimbs and hindlimbs" . The temporal expression profile of HSPB9 during limb development shows distinct patterns that appear dysregulated in atRA-induced limb malformations, with altered timing and magnitude of expression compared to normal development . These findings collectively suggest that HSPB9 may function as part of a protective response during embryonic limb development, potentially buffering against stressors that could lead to developmental abnormalities .

How does HSPB9 expression change during embryonic development?

HSPB9 expression during embryonic development follows complex spatiotemporal patterns that vary across different developmental stages and tissue types, particularly in the context of limb formation. During normal mouse embryonic hindlimb development, HSPB9 expression demonstrates a dynamic pattern that changes significantly across gestational days 12-18, a critical period for limb morphogenesis . When limb malformations are experimentally induced using teratogens such as all-trans retinoic acid (atRA), HSPB9 expression patterns become significantly altered compared to control embryos, suggesting its regulation is sensitive to developmental perturbations . The mRNA abundance of HSPB9 in atRA-treated embryos shows distinctive differences from control embryos across multiple developmental timepoints, indicating that expression changes may be either a cause or consequence of abnormal limb development . Research has identified HSPB9 as one of several heat shock proteins (including HspB2, HspB10, and others) that may be related to atRA-induced phocomelic and other limb abnormalities, suggesting coordinated regulation of multiple heat shock proteins during stress responses in developing limbs . Beyond limb development, HSPB9's expression in embryonic tissues appears to be relatively restricted compared to many other heat shock proteins, consistent with its generally tissue-specific expression pattern observed in adult organisms . These developmental expression patterns suggest that HSPB9 may play specialized roles during critical periods of embryogenesis, potentially providing chaperoning functions for specific developmental processes such as limb formation .

What experimental models are most suitable for studying HSPB9 in embryonic development?

Multiple experimental models offer complementary advantages for investigating HSPB9's role in embryonic development, each addressing different aspects of its function and regulation. Mouse models represent the predominant system for studying HSPB9 in mammalian development, allowing examination of expression patterns throughout embryogenesis and assessment of limb development under normal and perturbed conditions . Specifically, teratogen-induced mouse models using all-trans retinoic acid (atRA) have proven valuable for studying HSPB9's potential role in abnormal limb development, as atRA treatment reliably induces limb malformations and alters heat shock protein expression patterns . For higher-throughput developmental studies, zebrafish embryos offer advantages including external development, optical transparency, and amenability to genetic manipulation, though researchers must account for potential functional differences given the evolutionary distance from mammals . Ex vivo limb bud culture systems enable detailed manipulation of HSPB9 expression in developing limbs while maintaining three-dimensional tissue architecture, allowing real-time imaging of developmental processes and response to experimental perturbations . Embryonic stem cell differentiation models, particularly directed differentiation toward limb progenitors or organoid formation, can provide controlled systems for examining HSPB9's role in early developmental processes . For mechanistic studies, cell lines derived from embryonic tissues or engineered to express HSPB9 can facilitate detailed molecular analyses of protein interactions and signaling pathways . Genetic models including conditional knockout or knockin mice targeting HSPB9 specifically in developing limbs would provide the most definitive evidence regarding its developmental functions, though such models have not yet been reported in the literature .

How might HSPB9 contribute to normal versus abnormal limb development?

HSPB9's potential contributions to normal versus abnormal limb development likely involve its chaperoning functions and specific protein interactions during critical developmental windows. During normal limb development, HSPB9 may function as a specialized molecular chaperone protecting developmentally regulated proteins from misfolding or aggregation during the complex morphogenetic processes of limb formation . Its expression pattern changes significantly across gestational days 12-18 in mouse embryos, suggesting temporally regulated functions coinciding with critical stages of limb morphogenesis . In the context of abnormal limb development induced by teratogens such as all-trans retinoic acid (atRA), altered HSPB9 expression patterns may reflect either compensatory protective responses or disrupted regulatory mechanisms contributing to malformations . Research indicating that HSPB9, along with several other heat shock proteins, "may be related to atRA-induced phocomelic and other abnormalities" suggests it functions within a coordinated stress response network during limb development . The identification of HSPB9 as a gene that "probably performed an important role in normal and abnormal development of embryonic forelimbs and hindlimbs" based on expression analysis implies bidirectional involvement in both physiological development and pathological conditions . Mechanistically, HSPB9 might protect specific developmental signaling proteins or transcription factors from stress-induced misfolding, maintain cytoskeletal integrity during morphogenetic movements, or facilitate proper protein transport required for limb patterning and outgrowth .

How conserved is HSPB9 across different species?

HSPB9 demonstrates an unusually high degree of evolutionary divergence across species compared to most other heat shock proteins, suggesting specialized adaptive functions potentially related to reproduction. The most striking evidence of this divergence comes from comparative analysis between human and mouse HSPB9, which reveals a remarkable 38% sequence difference - a level of variation that is exceptionally high for heat shock proteins, which are typically highly conserved across mammalian species . This substantial interspecies variation stands in contrast to the general pattern observed for most small heat shock proteins, which tend to show high sequence conservation reflecting their fundamental cellular functions . The alpha-crystallin domain, which is the defining structural feature of all small heat shock proteins, is likely more conserved than the N-terminal and C-terminal regions of HSPB9, as this pattern of domain conservation is typical across the HSPB family . The exceptional sequence divergence observed for HSPB9 may reflect its testis-specific expression and potential role in reproduction, as reproductive proteins often evolve more rapidly than proteins with housekeeping functions due to sexual selection pressures or adaptive responses to sperm competition . This evolutionary pattern aligns with HSPB9's proposed roles in spermatogenesis, suggesting that its functions may have diverged to accommodate species-specific aspects of sperm development or function . The rapid evolution of HSPB9 is described as potentially "confirming its sex-related role," as reproductive proteins frequently display accelerated evolution compared to proteins with more generalized cellular functions .

What functional differences have been observed for HSPB9 across different species?

While comparative studies specifically focused on HSPB9 functional differences across species remain limited, the significant sequence divergence observed between human and mouse HSPB9 strongly suggests the existence of species-specific functional adaptations. The 38% sequence difference between human and mouse HSPB9 is described as potentially "confirming its sex-related role," implying that these sequence variations likely reflect functional specializations related to reproduction in different mammalian lineages . In testicular expression patterns, HSPB9 appears consistently restricted to testicular tissue across the species examined, suggesting conservation of its tissue-specific regulatory mechanisms despite sequence divergence . The developmental expression patterns of HSPB9 during embryogenesis, particularly in limb development contexts, may vary across species, though detailed comparative developmental studies are lacking in the current literature . Based on knowledge of other rapidly evolving reproductive proteins, the substantial sequence divergence in HSPB9 may reflect adaptations to species-specific aspects of spermatogenesis, sperm competition, or fertilization mechanisms . The conservation of HSPB9's interaction with TCTEL1 or other binding partners across different species has not been specifically addressed in the available literature, representing an important area for future comparative studies . Understanding these functional differences would require comparative studies examining HSPB9 expression patterns, protein interactions, and phenotypic effects of HSPB9 manipulation across multiple species, potentially revealing evolutionary adaptations in its chaperoning functions or regulatory roles .

What research techniques are most effective for cross-species comparison of HSPB9?

Effective cross-species comparison of HSPB9 requires integrated approaches spanning genomic, biochemical, and functional analyses to understand its evolutionary trajectory and species-specific adaptations. Comparative genomics and phylogenetic analysis represent foundational approaches for tracing HSPB9 evolution across species lineages, identifying patterns of conservation and divergence, and detecting signatures of selection that might indicate functional adaptations . Protein sequence alignment and structural comparison can map conserved versus variable regions within HSPB9, potentially correlating structural features with functional domains and identifying species-specific structural adaptations . Recombinant protein expression and biochemical characterization of HSPB9 from multiple species can directly compare functional properties such as substrate binding specificity, chaperone activity, oligomerization behavior, and thermal stability . Cross-species protein interaction studies using techniques such as yeast two-hybrid assays or co-immunoprecipitation can determine whether HSPB9 interaction partners (such as TCTEL1) are conserved across species despite sequence divergence . Comparative expression analysis examining HSPB9 tissue distribution, developmental timing, and subcellular localization across species can reveal conserved versus divergent expression patterns . Functional complementation experiments introducing HSPB9 from one species into cells from another species can test the degree of functional conservation despite sequence differences . For understanding reproductive adaptations, comparative analyses of sperm development, structure, and function across species with known HSPB9 sequence variation could reveal correlations between HSPB9 divergence and species-specific reproductive strategies .

What evolutionary hypotheses might explain HSPB9's unusual divergence between human and mouse?

The substantial 38% sequence divergence between human and mouse HSPB9 has prompted several evolutionary hypotheses that might explain this unusually rapid evolution compared to most heat shock proteins. Sexual selection and reproductive adaptation represent primary evolutionary forces potentially driving HSPB9 divergence, as proteins involved in reproduction frequently evolve more rapidly than those with housekeeping functions due to selective pressures related to sperm competition, cryptic female choice, or male-female molecular co-evolution . The testis-specific expression pattern of HSPB9 places it in a tissue environment known for harboring rapidly evolving proteins, as testis-expressed genes generally show accelerated evolutionary rates compared to genes expressed in other tissues across mammalian species . Species-specific spermatogenesis requirements may necessitate adaptations in HSPB9 function to accommodate differences in sperm development, morphology, or function between rodents and primates, reflecting their distinct reproductive strategies and mating systems . Relaxed functional constraints could potentially allow greater sequence divergence if HSPB9 performs a more specialized or redundant function compared to other more broadly expressed heat shock proteins, permitting greater evolutionary flexibility . Molecular co-evolution with interaction partners may drive rapid sequence changes if HSPB9 must adapt to corresponding evolutionary changes in its binding partners such as TCTEL1 or other reproductive proteins . The alpha-crystallin domain likely remains more conserved while terminal domains evolve more rapidly, a pattern observed in other heat shock proteins where the functional core maintains structural conservation while peripheral regions diversify to accommodate species-specific adaptations .

What are best practices for generating and validating anti-HSPB9 antibodies for research?

Generating reliable anti-HSPB9 antibodies presents specific challenges due to potential cross-reactivity with other small heat shock proteins and the need for robust validation in testis-specific contexts. Antigen design requires careful consideration, with optimal approaches including the use of unique peptide sequences from HSPB9's N- or C-terminal regions that show minimal homology with other small heat shock proteins, or the use of full-length recombinant HSPB9 for immunization followed by affinity purification against specific HSPB9 epitopes . Multiple antibody production platforms should be considered, including polyclonal antibodies for broader epitope recognition or monoclonal antibodies for higher specificity, with antibodies generated in multiple host species to enable co-localization studies with other antigens of interest . Validation of antibody specificity must include multiple orthogonal approaches - Western blotting against recombinant HSPB9 and testicular lysates is essential, ideally with HSPB9 knockout or knockdown samples as negative controls . Immunohistochemistry validation should examine staining patterns in testicular tissues, confirming specific localization in expected spermatogenic cell stages (late pachytene spermatocytes through elongate spermatids) with comparison to established HSPB9 mRNA expression patterns . Cross-reactivity testing against other small heat shock proteins (particularly closely related family members) is crucial to ensure signal specificity, as the conserved alpha-crystallin domain could lead to false-positive detection . Additional validation approaches could include immunoprecipitation followed by mass spectrometry to confirm antibody specificity, and peptide competition assays to verify epitope-specific binding .

What expression systems are optimal for producing recombinant HSPB9 for biochemical studies?

Producing high-quality recombinant HSPB9 for biochemical studies requires careful selection of expression systems and purification strategies optimized for this testis-specific small heat shock protein. Bacterial expression systems, particularly Escherichia coli, offer advantages for producing substantial quantities of recombinant HSPB9, with BL21(DE3) strains or their derivatives commonly used for heat shock protein expression . Codon optimization for E. coli expression should be considered given the potential differences in codon usage between human testis-specific genes and bacterial systems . Various fusion tags can facilitate expression and purification, with His6 tags, GST fusion, or MBP fusion commonly employed for small heat shock proteins, though tag influence on oligomerization behavior should be assessed . For higher eukaryotic expression systems, insect cells using baculovirus vectors can provide more appropriate post-translational modifications and chaperone assistance for proper folding, potentially important if HSPB9 requires specific modifications for activity . Mammalian expression systems might be necessary if authentic post-translational modifications or binding partner co-expression is required, with HEK293 or testis-derived cell lines as potential hosts . Purification strategies typically involve affinity chromatography based on fusion tags, followed by size exclusion chromatography to separate different oligomeric states and remove aggregates . Quality control assessments should include verification of protein folding through circular dichroism spectroscopy, thermal stability analysis, oligomeric state determination via size exclusion chromatography with multi-angle light scattering, and functional validation through chaperone activity assays .

What cellular models are appropriate for studying HSPB9 function?

Given HSPB9's testis-specific expression pattern and potential roles in cancer, multiple cellular models offer complementary advantages for investigating its functions in different biological contexts. Testicular cell lines represent primary cellular models for studying HSPB9's physiological functions, with options including GC-1 spg and GC-2 spd(ts) mouse spermatogonial and spermatocyte cell lines, or TCam-2 human seminoma cells which might express HSPB9 endogenously or serve as appropriate hosts for exogenous expression . Primary spermatogenic cell cultures isolated from testicular tissue provide more physiologically relevant models, though they present challenges in terms of maintenance and manipulation . For developmental studies related to HSPB9's role in limb formation, primary cultures from embryonic limb buds or appropriate embryonic cell lines can be employed, particularly with manipulation of HSPB9 expression to assess its impact on differentiation or response to developmental stressors . Cancer cell models derived from tumors known to express HSPB9 can facilitate investigation of its potential roles in malignancy, particularly regarding chemoresistance mechanisms which have been suggested in ovarian cancer . For protein interaction studies, heterologous expression systems such as HEK293 cells co-expressing HSPB9 and candidate interacting proteins (such as TCTEL1) can enable detailed analysis of binding dynamics and functional consequences . For all cellular models, manipulation of HSPB9 expression through overexpression, knockdown, or CRISPR/Cas9-mediated gene editing is essential for establishing cause-effect relationships, with particular attention to creating physiologically relevant expression levels when studying overexpression phenotypes .

What mouse models can be developed to study HSPB9 in vivo?

Developing mouse models for studying HSPB9 requires consideration of its testis-specific expression pattern, potential developmental roles, and the significant sequence divergence between human and mouse orthologs. Conventional knockout models with complete deletion of the HSPB9 gene represent a fundamental approach for investigating its physiological functions, with phenotypic analysis focusing primarily on testicular development, spermatogenesis, and male fertility parameters . Conditional knockout models using testis-specific promoters (such as stimulated by retinoic acid gene 8 [Stra8] or protamine 1 [Prm1] promoters driving Cre recombinase) can provide temporal control over HSPB9 deletion specifically in male germ cells, avoiding potential developmental effects if HSPB9 has essential functions during embryogenesis . For developmental studies, conditional knockouts targeting HSPB9 in developing limb buds (using promoters such as Prx1-Cre) could elucidate its role in normal and abnormal limb formation without affecting other tissues . Humanized mouse models replacing mouse HSPB9 with the human ortholog can address the functional significance of the substantial sequence divergence (38%) between species, determining whether human HSPB9 can functionally compensate for the mouse protein despite sequence differences . Reporter mouse lines with fluorescent proteins or LacZ knocked into the HSPB9 locus can facilitate detailed visualization of its expression patterns during development and in adult tissues without disrupting protein function . For cancer-related studies, mouse models combining HSPB9 manipulation with oncogene activation or tumor suppressor deletion in relevant tissues could investigate its potential roles in tumorigenesis or as a cancer/testis antigen . Inducible expression systems allowing temporal control over HSPB9 upregulation can model its aberrant expression in cancer contexts or enable rescue experiments in knockout backgrounds .

What are the most promising therapeutic applications of HSPB9 research?

The unique expression pattern and biological properties of HSPB9 suggest several promising therapeutic applications that warrant further investigation. Cancer immunotherapy development represents perhaps the most promising application, leveraging HSPB9's status as a cancer/testis antigen with restricted normal tissue expression but aberrant activation in multiple tumor types . This characteristic makes HSPB9 an attractive target for cancer-specific interventions with potentially limited off-target effects, including peptide vaccines, adoptive T-cell therapies, or antibody-based approaches targeting tumors expressing HSPB9 . Diagnostic applications in oncology offer another translational direction, with HSPB9 potentially serving as a biomarker for certain cancer types or as a means of identifying patients who might benefit from specific immunotherapeutic approaches targeting this protein . The suggested involvement of HSPB9 in chemoresistance mechanisms, particularly in ovarian cancer, indicates potential applications in sensitizing resistant tumors to conventional chemotherapies through modulation of HSPB9 expression or function . In reproductive medicine, a deeper understanding of HSPB9's role in spermatogenesis could lead to novel approaches for addressing certain forms of male infertility or developing male contraceptive strategies targeting testis-specific proteins . For developmental disorders affecting limb formation, insights into HSPB9's role in normal and abnormal limb development might contribute to improved diagnostic approaches or therapeutic strategies for managing congenital limb malformations . Protein engineering applications could potentially utilize HSPB9's chaperoning properties in biotechnology contexts, particularly if it demonstrates unique substrate specificity or functional properties compared to other small heat shock proteins .

What key knowledge gaps remain in our understanding of HSPB9?

Despite progress in characterizing HSPB9, significant knowledge gaps persist across multiple aspects of its biology that require targeted research efforts. The molecular mechanism of HSPB9's chaperone function remains poorly defined, including identification of its natural substrate proteins in testicular cells, the structural basis for substrate recognition, and the degree to which its chaperoning mechanism resembles or differs from other small heat shock proteins . The biological significance of HSPB9's interaction with TCTEL1 requires further elucidation, including how this interaction affects dynein motor function, what cellular processes it influences during spermatogenesis, and whether this interaction occurs in cancer contexts . The functional consequences of HSPB9's aberrant expression in cancer are incompletely understood, particularly regarding whether it actively contributes to tumor development, progression, or therapy resistance versus simply representing a byproduct of global cancer-associated gene dysregulation . The specific role of HSPB9 in embryonic development, particularly limb formation, remains largely correlative rather than mechanistic, with limited understanding of how it influences normal morphogenesis or responds to teratogenic insults . The physiological significance of the remarkable sequence divergence between human and mouse HSPB9 (38% difference) has not been functionally characterized, leaving open questions about species-specific adaptations in its function . The regulatory mechanisms controlling HSPB9's highly restricted expression pattern are not fully defined, including the transcription factors, epigenetic mechanisms, and signaling pathways that ensure its testis-specific expression in normal tissues and its reactivation in cancer contexts .

What emerging technologies could accelerate HSPB9 research?

Several cutting-edge technologies hold promise for addressing current knowledge gaps and accelerating research on HSPB9 across multiple domains. CRISPR/Cas9 genome editing technologies enable precise modification of HSPB9 in relevant cell types and model organisms, facilitating functional studies through knockout, knockin, or point mutation approaches with unprecedented efficiency and specificity . Single-cell transcriptomics and proteomics can provide high-resolution maps of HSPB9 expression across heterogeneous tissues such as testis or tumors, identifying specific cell populations expressing HSPB9 and correlating its expression with cell states or differentiation stages . Cryo-electron microscopy has revolutionized structural biology of protein complexes and could enable visualization of HSPB9 oligomeric assemblies or interactions with binding partners such as TCTEL1 at near-atomic resolution, providing mechanistic insights into its function . Proximity labeling methods (BioID, APEX) coupled with mass spectrometry enable comprehensive identification of proteins interacting with HSPB9 in living cells, potentially revealing previously unknown binding partners and cellular pathways . Advanced imaging techniques including super-resolution microscopy and live-cell imaging with fluorescent protein fusions can track HSPB9 localization and dynamics during spermatogenesis or in cancer cells with unprecedented spatial and temporal resolution . Organoid technologies modeling testicular tissue or embryonic limb development provide more physiologically relevant ex vivo systems for studying HSPB9 function than traditional cell culture approaches . CRISPR screening approaches can identify genes that synthetically interact with HSPB9, potentially revealing functional relationships and parallel pathways in both normal and cancer contexts .

How can collaborative approaches advance the field of HSPB9 research?

Advancing HSPB9 research requires multidisciplinary collaborative approaches that integrate diverse expertise and methodologies across multiple research domains. Cross-disciplinary collaborations between reproductive biologists, cancer researchers, and developmental biologists can provide complementary perspectives on HSPB9 function across different biological contexts, identifying common mechanisms and context-specific functions . Integration of basic and clinical research through collaborations between laboratory scientists and clinicians specializing in reproductive medicine, oncology, or developmental disorders can accelerate translational applications of HSPB9 research findings . Multi-omics research consortia combining genomics, transcriptomics, proteomics, and metabolomics approaches can provide comprehensive characterization of HSPB9's expression patterns, interactions, and functional impacts across multiple experimental systems . Structural biology collaborations utilizing complementary techniques such as X-ray crystallography, cryo-electron microscopy, and NMR spectroscopy can elucidate HSPB9's three-dimensional structure and dynamics at multiple scales . Computational biology partnerships leveraging expertise in bioinformatics, structural modeling, and systems biology can predict HSPB9 functions, identify potential binding partners, and simulate its behavior in cellular networks . Resource-sharing initiatives developing and distributing validated reagents such as antibodies, expression constructs, recombinant proteins, and cellular or animal models can accelerate research across multiple laboratories while ensuring methodological consistency . International research networks connecting labs studying different small heat shock proteins can facilitate comparative studies identifying shared and unique features of HSPB9 relative to other family members, potentially revealing functional specializations .

Product Science Overview

Introduction

Heat Shock Protein B9 (HSPB9), also known as CT51, is a member of the small heat shock protein (HSP20) family. These proteins are known for their role in protecting cells from stress by acting as molecular chaperones. HSPB9 is specifically expressed in the testis and localizes to both the cytoplasm and nucleus .

Structure and Function

HSPB9 is a small protein with a molecular weight of approximately 19.6 kDa. It consists of 159 amino acids and is characterized by the presence of a conserved alpha-crystallin domain, which is typical of small heat shock proteins . The primary function of HSPB9, like other heat shock proteins, is to assist in the proper folding of proteins, prevent protein aggregation, and protect cells from stress-induced damage.

Recombinant Production

Recombinant human HSPB9 is produced using bacterial expression systems, typically Escherichia coli (E. coli). The recombinant protein is often tagged with a His-tag at the N-terminus to facilitate purification. The production process involves cloning the HSPB9 gene into an expression vector, transforming the vector into E. coli, and inducing protein expression. The recombinant protein is then purified using affinity chromatography techniques .

Biological Significance

Heat shock proteins, including HSPB9, play crucial roles in cellular homeostasis and stress response. They are involved in various cellular processes such as protein folding, assembly, translocation, and degradation. HSPB9, in particular, has been implicated in the regulation of apoptosis and cellular stress responses. Its expression is upregulated in response to various stress conditions, including heat shock, oxidative stress, and exposure to toxic substances .

Clinical Relevance

HSPB9 and other heat shock proteins have gained significant attention in the field of cancer research. Tumor cells often exhibit elevated levels of heat shock proteins, which contribute to their survival, proliferation, and resistance to apoptosis. As a result, heat shock proteins, including HSPB9, are being explored as potential targets for cancer therapy. Additionally, recombinant HSPB9 is used in research to study its role in cellular stress responses and its potential therapeutic applications .

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 2024 Thebiotek. All Rights Reserved.