HSPA9 Human

Heat Shock 70kDa Protein 9 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Recombinant Human HSPA9 produced in E.Coli is a single,non-glycosylated polypeptide chain containing 654 amino acids (47-679) and having a molecular mass of 71 kDa.
HSP9A is expressed with a 20 amino acid His tag fused at N-Terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
HSPA9, a member of the heat shock protein 70 family, encompasses both heat-inducible and constitutively expressed proteins known as heat-shock cognate proteins. This gene encodes a heat-shock cognate protein that participates in regulating cell proliferation and functions as a chaperone. Notably, HSPA9 is localized to chromosome 5, band q31, a region frequently deleted in myeloid leukemias and myelodysplasia (MDS). This suggests its potential role as a tumor suppressor gene, aligning with the biological function observed in its murine counterpart. HSPA9 demonstrates its regulatory role by suppressing p53's nuclear translocation, transcriptional activation, and control over centrosome duplication.
Description
Recombinant Human HSPA9, expressed in E. coli, is a single, non-glycosylated polypeptide chain consisting of 654 amino acids (47-679). With a molecular weight of 71 kDa, it features a 20 amino acid His tag fused at the N-terminus. The protein is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterilized by filtration.
Formulation
The HSPA9 protein solution is formulated in 20mM Tris-HCl buffer at pH 8, with 10% glycerol and 0.5mM DTT.
Stability
For short-term storage (2-4 weeks), the protein can be stored at 4°C. For extended periods, storage at -20°C in a frozen state is recommended. To ensure long-term stability, adding a carrier protein like 0.1% HSA or BSA is advisable. Repeated freezing and thawing should be avoided.
Purity
The purity of the protein is determined to be greater than 95.0% using SDS-PAGE analysis.
Synonyms
Mortalin, GRP75, MOT2, GSPA9B, PBP74, MOT-2, MTHSP75, Stress-70 protein mitochondrial, 75 kDa glucose-regulated protein, GRP 75, Heat shock 70 kDa protein 9, Peptide-binding protein 74, MOT, HSPA9, HSPA9B, CSA, MGC4500.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MASEAIKGAV VGIDLGTTNS CVAVMEGKQA KVLENAEGAR TTPSVVAFTA DGERLVGMPA KRQAVTNPNN TFYATKRLIG RRYDDPEVQK DIKNVPFKIV RASNGDAWVE AHGKLYSPSQ IGAFVLMKMK ETAENYLGHT AKNAVITVPA YFNDSQRQAT KDAGQISGLN VLRVINEPTA AALAYGLDKS EDKVIAVYDL GGGTFDISIL EIQKGVFEVK STNGDTFLGG EDFDQALLRH IVKEFKRETG VDLTKDNMAL QRVREAAEKA KCELSSSVQT DINLPYLTMD SSGPKHLNMK LTRAQFEGIV TDLIRRTIAP CQKAMQDAEV SKSDIGEVIL VGGMTRMPKV QQTVQDLFGR APSKAVNPDE AVAIGAAIQG GVLAGDVTDV LLLDVTPLSL GIETLGGVFT KLINRNTTIP TKKSQVFSTA ADGQTQVEIK VCQGEREMAG DNKLLGQFTL IGIPPAPRGV PQIEVTFDID ANGIVHVSAK DKGTGREQQI VIQSSGGLSK DDIENMVKNA EKYAEEDRRK KERVEAVNMA EGIIHDTETK MEEFKDQLPA DECNKLKEEI SKMRELLARK DSETGENIRQ AASSLQQASL KLFEMAYKKM ASEREGSGSS GTGEQKEDQK EEKQ.

Q&A

What is HSPA9 and what are its primary functions in human cells?

HSPA9 is a 70kDa heat shock protein located primarily in mitochondria that functions as a molecular chaperone essential for mitochondrial protein import, folding, and degradation . The protein is highly conserved across species and plays crucial roles in multiple cellular processes including:

  • Protein folding and quality control within mitochondria

  • Regulation of apoptosis and cellular proliferation

  • Participation in erythrocyte differentiation and hematopoiesis

  • Protein export from the nucleus

  • Negative regulation of apoptotic processes

Research methodologies to study these functions typically include protein interaction studies, subcellular fractionation techniques, and functional assays measuring chaperone activity using purified recombinant proteins.

What is the genomic location and structure of the HSPA9 gene?

HSPA9 is located on chromosome 5q31.1, spanning from base pair 138,554,882 to 138,575,629 on the minus strand . To study the genomic structure and expression patterns, researchers typically employ:

  • Fluorescence in situ hybridization (FISH) to visualize the chromosomal location

  • Next-generation sequencing for detailed analysis of gene structure

  • PCR-based methods for expression analysis across different tissues

  • Chromatin immunoprecipitation (ChIP) to identify transcription factor binding sites

Understanding the genomic context is particularly important as deletions of chromosome 5q31.2 (where HSPA9 is located) are frequently observed in myelodysplastic syndromes and acute myeloid leukemia .

What protein interactions does HSPA9 participate in?

HSPA9 engages in numerous protein-protein interactions that facilitate its chaperone function and involvement in various cellular pathways. Key interaction partners include:

Interaction PartnerFunctional SignificanceDetection Method
TP53 (p53)Cell cycle regulation and apoptosisCo-immunoprecipitation, yeast two-hybrid
AKT1Cell survival signalingProximity ligation assay
EGFRGrowth signalingMass spectrometry-based proteomics
HSP90AA1Cooperative chaperone functionFRET-based interaction studies
SOD1Oxidative stress responsePull-down assays
SIRT1, SIRT3, SIRT7Metabolic regulation and agingImmunoprecipitation, protein arrays

These interactions can be studied using techniques such as co-immunoprecipitation, yeast two-hybrid screening, and proximity ligation assays to verify direct physical interactions in physiologically relevant contexts .

How does HSPA9 contribute to human hematopoiesis?

HSPA9 plays a critical role in hematopoiesis, particularly in erythroid development. Research methodologies to investigate this function include:

  • Knockdown studies: HSPA9 knockdown in human CD34+ hematopoietic progenitor cells results in:

    • Significantly delayed maturation of erythroid precursors

    • 6-fold suppression of cell growth due to increased apoptosis and decreased cell cycling

    • Greater reduction in burst-forming unit-erythroid (BFU-E) colonies (5.1-fold) compared to granulocyte/macrophage colonies (3.1-fold)

  • Flow cytometry analysis: Used to assess:

    • CD235a/GlyA and CD71 expression to track erythroid differentiation

    • Annexin V/7-AAD staining to quantify apoptosis rates

    • BrdU incorporation to analyze cell cycle status

  • Colony formation assays: Methylcellulose-based assays reveal that HSPA9 knockdown causes:

    • Reduced hemoglobin content in BFU-E colonies

    • Preferential impact on erythroid versus myeloid lineage development

Interestingly, HSPA9 knockdown does not significantly affect CD15+ myeloid cells or CD41a+ megakaryocytic cells, suggesting a lineage-specific role in erythropoiesis .

What evidence links HSPA9 to aging and longevity?

HSPA9 has been implicated in aging processes, though its role in human aging remains less defined than in model organisms:

  • Model organism studies:

    • Roundworms with extra copies of Hsp70F (HSPA9 homolog) live approximately 40% longer than controls

    • Research methodologies include lifespan assays, stress resistance tests, and genetic manipulation in C. elegans

  • Protein interaction studies reveal HSPA9 connections with:

    • SIRT1, SIRT3, and SIRT7 (longevity-associated proteins)

    • TP53 pathway components involved in cellular senescence

    • Proteins regulating mitochondrial function during aging

  • Expression analysis techniques:

    • qRT-PCR and western blotting to measure age-dependent changes in HSPA9 levels

    • Immunohistochemistry to assess tissue-specific expression patterns

    • Single-cell RNA sequencing to examine expression changes in aging cell populations

While a definitive role for HSPA9 in human aging has not been established, its conservation across species and functional relationship with known longevity pathways suggest potential significance that warrants further investigation .

What are the consequences of HSPA9 haploinsufficiency in mouse models?

Mouse models using lentivirally mediated gene silencing to achieve approximately 50% knockdown of Hspa9 (modeling haploinsufficiency) demonstrate:

  • Hematopoietic abnormalities:

    • Significant reduction in erythroid cells (Ter119High/CD71+) by 2.5-fold in bone marrow and 8.9-fold in spleen

    • Decreased erythroid cells in peripheral blood (3.8-fold reduction)

    • Reduced percentage of cells in S phase in bone marrow (21.3% vs. 27.2% in controls)

  • Progenitor cell effects:

    • Reduction in bone marrow progenitors including c-kit+/lineage-/Sca-1+ (KLS) cells

    • Decreased megakaryocyte/erythrocyte progenitor (MEP) populations

    • Diminished B lymphocytes

  • Competitive disadvantage:

    • Time-dependent loss of Hspa9-knockdown YFP+ peripheral blood cells

    • Hspa9-knockdown cells outcompeted by non-transduced normal cells

These findings are particularly relevant as they suggest that while Hspa9 haploinsufficiency causes significant hematopoietic abnormalities, cooperating gene mutations may be necessary for del(5q31.2) myelodysplastic syndrome cells to gain clonal dominance in the bone marrow .

How does HSPA9 dysfunction contribute to myelodysplastic syndromes and leukemia progression?

HSPA9 is located within the commonly deleted region at chromosome 5q31.2 in myelodysplastic syndromes (MDS). Advanced research methodologies reveal:

  • Mechanistic studies of hematopoietic dysfunction:

    • HSPA9 knockdown activates the intrinsic apoptosis pathway with increased:

      • Mitochondrial membrane depolarization

      • Cleaved caspase-3 expression

      • Cleavage of BAX to the apoptosis-inducing p18 form

    • Differentiating cells (CD34-) show higher susceptibility to apoptosis than progenitor cells (CD34+)

  • Leukemic transformation models suggest:

    • HSPA9 haploinsufficiency may contribute to ineffective hematopoiesis in early-stage MDS

    • Additional mutations (particularly in p53) may be required for progression to acute myeloid leukemia (AML)

    • Combined genetic alterations can be modeled using:

      • CRISPR/Cas9 genome editing of multiple loci

      • Retroviral overexpression of cooperating oncogenes

      • Patient-derived xenograft models

  • Single-cell analysis techniques:

    • RNA-seq to identify dysregulated pathways in HSPA9-deficient cells

    • ATAC-seq to assess chromatin accessibility changes

    • CyTOF to characterize altered signaling networks at the protein level

These findings suggest a model where HSPA9 haploinsufficiency creates a cellular context of ineffective hematopoiesis that, when combined with additional genetic alterations, may promote leukemic transformation .

What is the relationship between HSPA9 and other mitochondrial chaperones in human development and disease?

HSPA9 functions within a network of mitochondrial chaperones, with particularly important relationships to LONP1:

  • Comparative phenotypic analysis:

    • LONP1 mutations cause CODAS syndrome (cerebral, ocular, dental, auricular, and skeletal)

    • HSPA9 mutations cause EVEN-PLUS syndrome (epiphyseal, vertebral, ear, nose, plus associated findings)

    • Both syndromes share epiphyseal, vertebral, and ocular abnormalities but EVEN-PLUS also features severe microtia, nasal hypoplasia, and additional malformations

  • Functional interaction studies:

    • Both proteins participate in mitochondrial protein quality control

    • LONP1 has both chaperone and protease activity

    • HSPA9 (mHSP70/mortalin) is essential for mitochondrial protein import, folding, and degradation

  • Advanced research methodologies:

    • Protein co-localization studies using super-resolution microscopy

    • Proximity labeling techniques (BioID, APEX) to map the chaperone interactome

    • In vitro reconstitution of chaperone complexes

    • Cryo-EM structural studies of chaperone-substrate interactions

These overlapping phenotypes and functional relationships between HSPA9 and LONP1 have led to the concept of "mitochondrial chaperonopathies," pointing to an unexplored role of mitochondrial chaperones in human embryonic morphogenesis .

What experimental approaches are optimal for investigating HSPA9's role in erythropoiesis?

To rigorously investigate HSPA9's function in erythropoiesis, researchers employ sophisticated methodologies:

  • In vitro differentiation systems:

    • CD34+ hematopoietic progenitor cells cultured in erythroid differentiation media

    • Time-course analysis of differentiation markers (CD235a/GlyA, CD71)

    • Assessment of hemoglobinization and enucleation

    • Genome editing (CRISPR/Cas9) for precise genetic manipulation

  • In vivo mouse models:

    • Lentiviral shRNA knockdown in bone marrow followed by transplantation

    • Competitive repopulation assays using marker systems (e.g., Ly5.1/Ly5.2)

    • Analysis of multiple hematopoietic compartments (bone marrow, spleen, peripheral blood)

    • Flow cytometric quantification of specific cell populations

  • Molecular profiling techniques:

    • RNA-seq to identify transcriptional changes during erythroid differentiation

    • Proteomics to map protein interactions during different stages

    • Metabolomics to assess changes in mitochondrial function

    • ChIP-seq to identify transcription factor binding relevant to erythroid development

  • Functional mitochondrial assays:

    • Seahorse XF analysis to measure oxygen consumption and glycolytic function

    • JC-1 staining to assess mitochondrial membrane potential

    • MitoTracker staining to examine mitochondrial mass and morphology

    • Mitochondrial import assays using fluorescently labeled precursor proteins

These integrated approaches provide comprehensive insights into HSPA9's role in normal erythropoiesis and how its dysfunction contributes to disorders of red blood cell production .

How can HSPA9 dysfunction be targeted therapeutically in hematological disorders?

While direct therapeutic targeting of HSPA9 is still in early research stages, several approaches show promise:

  • Small molecule modulators:

    • Screening approaches using:

      • In silico structure-based drug design targeting HSPA9's ATPase domain

      • High-throughput screening of compound libraries

      • Fragment-based drug discovery

    • Validation in cell-based assays measuring:

      • Erythroid differentiation rescue

      • Apoptosis reduction

      • Cell cycle normalization

  • Gene therapy approaches:

    • AAV-mediated gene supplementation

    • CRISPR-based gene correction for specific mutations

    • Targeted increase of HSPA9 expression using epigenetic modulators

  • Mitochondrial function support:

    • Targeting downstream pathways affected by HSPA9 dysfunction:

      • Anti-apoptotic compounds

      • Cell cycle modulators

      • Mitochondrial biogenesis enhancers

  • Combination therapies:

    • Co-targeting multiple affected pathways in MDS/AML

    • Personalized approaches based on genetic profile

Early intervention focusing on enhancing erythropoiesis and reducing apoptosis may be particularly effective in MDS patients with 5q31.2 deletions affecting HSPA9 .

What is the significance of HSPA9 variants in human developmental disorders?

HSPA9 variants have been implicated in developmental disorders, particularly the EVEN-PLUS syndrome:

  • Genetic analysis methodologies:

    • Whole exome/genome sequencing to identify biallelic mutations

    • Segregation analysis in families

    • Functional characterization of variants using:

      • In vitro protein function assays

      • Patient-derived cells

      • Model organism studies

  • Phenotypic characterization techniques:

    • Detailed clinical assessment protocols

    • Radiological evaluation of skeletal abnormalities

    • Developmental milestone tracking

    • Comparison with related conditions (e.g., CODAS syndrome)

  • Translational approaches:

    • Development of patient registries

    • Natural history studies

    • Biomarker identification for disease progression

The characterization of EVEN-PLUS syndrome caused by HSPA9 mutations, alongside CODAS syndrome caused by LONP1 mutations, establishes the concept of "mitochondrial chaperonopathies" as an emerging category of developmental disorders .

What are the emerging techniques for studying HSPA9 function in human systems?

Cutting-edge methodologies are expanding our understanding of HSPA9:

  • Advanced genetic manipulation:

    • Base editing for precise introduction of patient-derived variants

    • Prime editing for flexible gene modification

    • Inducible knockdown/knockout systems for temporal control

    • Tissue-specific conditional models

  • Single-cell technologies:

    • Integrated multi-omics (scRNA-seq, scATAC-seq, scProteomics)

    • Spatial transcriptomics to understand HSPA9 function in tissue context

    • Lineage tracing to track cellular development

    • Single-cell metabolomics for mitochondrial function assessment

  • Organoid and iPSC models:

    • Patient-derived induced pluripotent stem cells

    • Hematopoietic organoids to model erythropoiesis

    • Brain organoids to investigate neurodevelopmental aspects

    • Multi-tissue organoids to study system-level effects

  • In vivo imaging techniques:

    • Intravital microscopy of hematopoietic stem cell niches

    • Mitochondrial dynamics visualization in living organisms

    • PET imaging with mitochondrial function tracers

These emerging technologies promise to provide unprecedented insights into the complex roles of HSPA9 in human development, aging, and disease processes .

How might HSPA9 research inform our understanding of mitochondrial contributions to human aging?

The connection between HSPA9 and aging opens several research avenues:

  • Comparative biology approaches:

    • Cross-species analysis of HSPA9 homologs and their effects on lifespan

    • Examination of naturally long-lived species for HSPA9 adaptations

    • Population genetics studies in human centenarians

  • Integrative aging models:

    • Investigation of HSPA9 in multiple hallmarks of aging:

      • Mitochondrial dysfunction

      • Cellular senescence

      • Stem cell exhaustion

      • Proteostasis disruption

    • Systems biology approaches to model interaction networks

  • Intervention testing:

    • Enhancement of HSPA9 function through genetic or pharmacological means

    • Evaluation of effects on healthspan markers

    • Integration with established anti-aging interventions

The significant lifespan extension (40%) observed in roundworms with extra copies of the HSPA9 homolog provides a compelling rationale for exploring similar approaches in mammalian systems .

Product Science Overview

Gene and Protein Structure

HSPA9 is encoded by the HSPA9 gene, which is located on chromosome 5, band q31.1 . This region is frequently deleted in myeloid leukemias and myelodysplasia (MDS), suggesting that HSPA9 may function as a tumor suppressor gene . The protein itself has a molecular weight of approximately 71 kDa and consists of 654 amino acids .

Function and Mechanism

HSPA9 functions primarily as a molecular chaperone. It assists in the proper folding of newly synthesized polypeptides, refolding of denatured proteins, and stabilization of native proteins . Additionally, HSPA9 is involved in the inhibition of nuclear translocation, transcriptional activation, and control of centrosome-duplication functions of the p53 protein .

Clinical Significance

Due to its role in protein folding and stress response, HSPA9 is implicated in various diseases, including cancer and autoimmune disorders . Its overexpression has been observed in inflamed tissues, and it has been shown to have immunosuppressive activity by downregulating nuclear factor-kappa B (NF-κB) activation . This makes HSPA9 a potential target for therapeutic interventions in conditions such as rheumatoid arthritis and other autoimmune diseases .

Recombinant HSPA9

Recombinant HSPA9 is produced using Escherichia coli (E. coli) expression systems and is purified using conventional chromatography techniques . The recombinant protein is often tagged with a His-tag at the N-terminus to facilitate purification and detection . It is typically formulated in a buffer containing Tris-HCl, DTT, and glycerol to maintain stability and activity .

Applications

Recombinant HSPA9 is widely used in research to study its function and role in various cellular processes. It is also used in drug discovery and development, particularly in the context of cancer and autoimmune diseases .

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.