HSPD1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery time may vary depending on your location and the shipping method you choose. For specific delivery time information, please consult your local distributor.
Synonyms
60 kDa chaperonin antibody; 60 kDa heat shock protein; mitochondrial antibody; CH60_HUMAN antibody; Chaperonin 60 antibody; Chaperonin; 60-KD antibody; CPN60 antibody; fa04a05 antibody; GROEL antibody; heat shock 60kDa protein 1 (chaperonin) antibody; Heat shock protein 1 (chaperonin) antibody; Heat shock protein 60 antibody; Heat shock protein 65 antibody; heat shock protein family D (Hsp60) member 1 antibody; HLD4 antibody; Hsp 60 antibody; HSP 65 antibody; HSP-60 antibody; HSP60 antibody; HSP65 antibody; HSPD1 antibody; HuCHA60 antibody; Mitochondrial matrix protein P1 antibody; P60 lymphocyte protein antibody; short heat shock protein 60 Hsp60s1 antibody; SPG13 antibody
Target Names
HSPD1
Uniprot No.

Target Background

Function
Hsp60 (Heat Shock Protein 60) is a molecular chaperone involved in mitochondrial protein import and macromolecular assembly. It functions in conjunction with Hsp10 to facilitate the correct folding of imported proteins. Hsp60 may also play a role in preventing protein misfolding and promoting the refolding and proper assembly of unfolded polypeptides, particularly in response to stress conditions within the mitochondrial matrix. The functional units of these chaperonins consist of heptameric rings of the large subunit Hsp60, which work together in a back-to-back double ring configuration. In a cyclical process, Hsp60 ring complexes bind one unfolded substrate protein per ring, followed by the binding of ATP and association with 2 heptameric rings of the co-chaperonin Hsp10. This leads to the encapsulation of the substrate protein within the inner cavity of Hsp60, where it can fold undisturbed by other cellular components for a specific period of time. Synchronous hydrolysis of ATP in all Hsp60 subunits results in the dissociation of the chaperonin rings and the release of ADP and the folded substrate protein (Probable).
Gene References Into Functions

Related Research and Function

  1. HSP60 has been shown to possess pro-inflammatory properties in bronchial epithelial cells, mediated by activation of TLR-4-related molecules. PMID: 28976240
  2. Silencing HSP60 deactivates the mTOR pathway, contributing to the suppression of glioblastoma progression. PMID: 27325206
  3. The impact of Hsp60 on differentiation and invasion of hepatocellular carcinoma cells is potentially linked to mitochondrial biogenesis. PMID: 27677587
  4. Research has indicated that HSP60 participates in mitochondrial progesterone synthesis. These findings shed light on progesterone synthesis in the human placenta and its role in maintaining pregnancy. PMID: 28434777
  5. Clinical data suggests that upregulation of miR-382/3-NT and downregulation of HSPD1/Trx are observed in IgA nephropathy patients experiencing renal interstitial fibrosis. These findings support a novel mechanism where miR-382 targets HSPD1, contributing to redox imbalance during the development of renal fibrosis. PMID: 28680529
  6. Low HSP60 expression has been correlated with beta-cell hypertrophy and dysfunction. PMID: 27056903
  7. High levels of reactive oxygen species (ROS) are required for tumorigenesis and progression in tumors with low HSP60 expression. PMID: 27246978
  8. HSP60 regulation of SOX9 ubiquitination has been identified as a factor mitigating the development of knee osteoarthritis. PMID: 27118120
  9. Studies suggest that tumor cells may avoid apoptosis by utilizing Hsp60. Drugs like CubipyOXA, which interfere with Hsp60/pC3 complex formation, could potentially be used as anti-cancer agents by allowing the apoptotic cascade to proceed. PMID: 28212901
  10. The association of diabetes, combined with polymorphisms in the genes of fat mass and obesity-associated gene (FTO), interleukin 6 (IL-6), and heat shock protein 60 (HSPD1), with breast cancer risk and survival in a Chinese Han population, was investigated. PMID: 28591216
  11. 27-Hydroxycholesterol has been shown to upregulate the production of HSP60 in monocytic cells. PMID: 28549691
  12. Research indicates that HSP65 suppresses cholesterol efflux and increases cellular cholesterol content through an Lck-mediated pathway in T cells. PMID: 27742830
  13. Doxorubicin treatment of lung mucoepidermoid cells leads to Hsp60 post-translational modifications, resulting in the Hsp60/p53 complex dissociation and the initiation of replicative senescence. PMID: 27836734
  14. Phosphorylation and subsequent transient degradation of mitochondrial Hsp60 during the early hours of rotavirus-SA11 infection inhibits the premature import of nonstructural protein 4 into mitochondria, delaying early apoptosis. PMID: 27665089
  15. Data suggests that the interaction between cell cycle and apoptosis regulator 2 (CCAR2) and heat shock protein 60 (Hsp60) increases in the presence of rotenone. PMID: 28254432
  16. NIP-SNAP-1 and -2 localized in the mitochondrial inner membrane space, while HSP60 was found in the matrix. Knockdown of HSP60, but not HSP10, led to decreased expression levels of NIP-SNAP-1 and -2. This indicates that HSP60 promotes folding and maintains the stability of NIP-SNAP-1 and -2. PMID: 28011268
  17. Elevated expression of Hsp60 has been observed in lung adenocarcinoma tissue. PMID: 28178129
  18. Hsp10 and Hsp60 may be implicated in carcinogenesis from its very early stages in colorectal cancer. PMID: 27491302
  19. High HSP60 expression has been associated with gastric cancer. PMID: 26810190
  20. Elevated expression of HSPD1 in osteosarcoma tissues has been correlated with poor prognosis for osteosarcoma patients. PMID: 27259322
  21. Research suggests that tolerization with antigenic HSP60 protein or its peptides may arrest or even prevent atherosclerosis by increasing the production of regulatory T cells and/or anti-inflammatory cytokines. PMID: 26577462
  22. Data shows that Eclipta extract upregulates heat shock protein 60 (Hsp60), which is localized in the endoplasmic reticulum (ER). PMID: 26672742
  23. Studies indicate that the addition of the heat-shock proteins GroEL-GroES molecular chaperone system significantly enhanced the folding of the nascent chemokine receptor type 5 (CCR5). PMID: 26585937
  24. Immunohistochemistry for CD1a and Hsp60 can be useful in differentiating between Keratoacantomas and well-differentiated forms of squamous cell carcinomas. PMID: 26442925
  25. Anti-citrullinated protein antibodies promote apoptosis of mature human osteoblasts through cell-surface binding to citrullinated heat shock protein 60. PMID: 26275591
  26. Heat shock protein 60 stimulates the migration of vascular smooth muscle cells through Toll-like receptor 4 and ERK MAPK activation. PMID: 26477505
  27. While HSP60 is a target for pathogenic autoimmune inflammatory processes, it has also been shown to activate immunoregulatory pathways that may suppress these diseases. PMID: 26431161
  28. Biochemical and genetic data demonstrate that FUS interacts with a mitochondrial chaperonin, HSP60, and that FUS translocation to mitochondria is, at least in part, mediated by HSP60. PMID: 26335776
  29. Exposure of human promyelocytic HL-60 cells to a nontoxic concentration (10 μM) of 4-hydroxy-2-nonenal (HNE) yielded a HSP60 modified with HNE. PMID: 26078803
  30. HSP60 overexpression was associated with disease progression and prognosis in gastric cancer, and its expression significantly correlated with the expression of MMP-9. PMID: 25207654
  31. Hsp60 is elevated in both animals and patients with Temporal Lobe Epilepsy (TLE) in affected tissues, and in plasma in response to epileptic seizures. This points to Hsp60 as a potential biomarker of hippocampal stress, potentially useful for diagnosis and patient management. PMID: 25801186
  32. No significant relationship between anti-hsp60 antibodies and serological markers of infection was observed, suggesting an indirect role of infection in the assessment of breaking the immunological tolerance against autologous HSPs. PMID: 25654359
  33. Low levels of HSP60 may lead to a lack of anti-inflammatory response due to reduced Treg activation, potentially contributing to the pathogenesis of Immune Thrombocytopenia (ITP). PMID: 24749912
  34. Research has shown that HSPD1 interacts with IRF3, contributing to the induction of IFN-beta. PMID: 25506707
  35. Hsp60 has been found to be increased in cancerous tissue in patients with large bowel cancer. PMID: 26060090
  36. The immunological response to Hsp60/65 is elevated in the early clinical stages of ovarian cancer, and the level of anti-hsp60/65 antibodies could be a helpful diagnostic marker. PMID: 24618330
  37. An HLD4-associated (Asp-29-to-Gly) mutant of mitochondrial heat shock 60-kDa protein 1 (HSPD1) leads to shorter mitochondrial morphologies and increased mitochondrial numbers due to aberrant fission and fusion cycles. PMID: 25957474
  38. Structural analysis of the mutated human Hsp60-human Hsp10 complex has been conducted. PMID: 25918392
  39. Elevated levels of circulating autoantibodies against Hsp60, Hsp70, and Hsp90 were positively correlated with cutaneous disease activity in dermatitis herpetiformis. PMID: 24643797
  40. Research indicates that heat shock protein 60 (HSP60) interacts constitutively with NKG2D ligand ULBP2, and phosphatase of regenerating liver 3 (PRL-3) regulates HSP60 tyrosine phosphorylation. PMID: 25687758
  41. Increased levels of anti-heat-shock protein 60 (anti-Hsp60) indicate endothelial dysfunction, atherosclerosis, and cardiovascular diseases in patients with mixed connective tissue disease. PMID: 24838263
  42. Lysine biotinylation and methionine oxidation in the heat shock protein HSP60 synergize in the elimination of reactive oxygen species. PMID: 24582286
  43. Modified forms of LDL activate human T cells through dendritic cells, with HSP60 and 90 contributing to this T-cell activation. PMID: 25395618
  44. The Hsp60 mitochondrial import signal remains stable in solution. PMID: 24830947
  45. Seven proteins, particularly HSP 60, are potential biomarkers for the diagnosis of Rheumatic Heart Disease (RHD). PMID: 24738046
  46. Regions in the Hsp60 molecule exhibit structural similarity with thyroglobulin (TG) and thyroid peroxidase (TPO) molecules, supporting the idea that autoantibodies against TG and TPO may recognize Hsp60 on the plasma membrane of oncocytes. PMID: 24057177
  47. Data suggest that upregulation of HSP60/HSPD1 binding/reactivity leads to increased cytokine synthesis/secretion and other proinflammatory responses in adipocytes, particularly in mature visceral adipocytes. PMID: 24672802
  48. Citrullination of HSP60 has been linked to neoplasms. PMID: 24099319
  49. Antibodies to human HSP60 were detected in 19 (15.8%) of 120 patients with a history of recurrent miscarriages. PMID: 24680363
  50. The pathogenic variant of rs72466451 may play a role in a subgroup of sudden infant death syndrome cases with impaired Hsp60-mediated stress response. PMID: 23823174
Database Links

HGNC: 5261

OMIM: 118190

KEGG: hsa:3329

STRING: 9606.ENSP00000340019

UniGene: Hs.595053

Involvement In Disease
Spastic paraplegia 13, autosomal dominant (SPG13); Leukodystrophy, hypomyelinating, 4 (HLD4)
Protein Families
Chaperonin (HSP60) family
Subcellular Location
Mitochondrion matrix.

Q&A

What is HSPD1 and what are its primary functions in cells?

HSPD1, also known as HSP60, is a 60kDa chaperonin protein primarily localized in the mitochondrial matrix. It plays essential roles in mitochondrial protein import and macromolecular assembly. HSPD1 forms functional units consisting of heptameric rings that work as back-to-back double rings. Together with its co-chaperonin HSP10, it facilitates the correct folding of imported proteins and prevents misfolding of polypeptides under stress conditions . The protein functions through a cyclic reaction binding unfolded substrate proteins, followed by ATP binding and association with HSP10, which creates a protected environment for proper protein folding . Beyond its chaperone function, HSPD1 may also serve as a signaling molecule in the innate immune system, making it relevant to both cellular homeostasis and immunological research .

What types of HSPD1 antibodies are available for research?

HSPD1 antibodies are available in several formats to accommodate different research applications:

Antibody TypeHost OptionsClonalityConjugation OptionsSpecies ReactivityApplications
Full-length (AA 1-573)RabbitPolyclonalUnconjugatedHuman, Chicken, Cow, Dog, Mouse, Pig, RatWB, IHC, IP, ICC
Partial epitope (AA 27-240)RabbitPolyclonalUnconjugatedHumanWB, IHC, IF, IP
Middle region (AA 172-343)RabbitPolyclonalUnconjugatedHumanWB, IHC, IP, ICC
C-terminal (AA 316-573)RabbitPolyclonalUnconjugatedHumanWB, IHC, IP, ICC
C-terminal (AA 501-573)RabbitPolyclonalUnconjugatedHuman, Mouse, Rat, PigWB, ELISA, IHC, IF
LK1 cloneMouseMonoclonalUnconjugatedHumanWB, IHC, FACS, ELISA, IP
3G8 cloneMouseMonoclonalUnconjugatedHumanWB, IHC, FACS, ELISA, ICC
Detection antibodyMouseMonoclonalUnconjugated/ConjugatableHumanELISA, multiplex assays

Additionally, matched antibody pairs designed specifically for assays requiring capture and detection antibodies (such as ELISA and cytometric bead arrays) are available, including the pair 66041-2-PBS (capture) and 66041-4-PBS (detection) .

What applications are HSPD1 antibodies commonly used for?

HSPD1 antibodies support a wide range of experimental applications in cellular and molecular biology research:

  • Western Blotting (WB): For detecting HSPD1 protein expression levels in cell or tissue lysates with a typical detection at approximately 60-61 kDa .

  • Immunohistochemistry (IHC): For visualizing the distribution and localization of HSPD1 in tissue sections, including paraffin-embedded (IHC-P) and frozen sections (IHC-fro) .

  • Immunocytochemistry (ICC): For cellular localization studies in cultured cells .

  • Immunofluorescence (IF): For detailed subcellular localization studies using fluorescence microscopy .

  • Immunoprecipitation (IP): For isolating HSPD1 protein complexes to study interaction partners .

  • ELISA: For quantitative detection of HSPD1 in biological samples .

  • Flow Cytometry (FACS): For analyzing HSPD1 expression in individual cells within heterogeneous populations .

  • Multiplex Assays: Using matched antibody pairs for simultaneous detection of multiple targets .

  • Immunoelectron Microscopy (IEM): For ultrastructural localization studies at the electron microscope level .

The versatility of these applications allows researchers to investigate HSPD1 at different levels of biological organization, from molecular interactions to tissue distribution.

How should HSPD1 antibodies be stored and handled for optimal performance?

Proper storage and handling of HSPD1 antibodies is crucial for maintaining their activity and specificity:

  • Storage Temperature: Most HSPD1 antibodies should be stored at -80°C for long-term preservation of activity . Some products may be stored at -20°C, but always refer to product-specific guidelines.

  • Storage Buffer: Most antibodies are provided in PBS (phosphate-buffered saline) or similar stabilizing buffers. Some formulations are available as "PBS only" (BSA and azide-free) for applications requiring conjugation or those sensitive to additives .

  • Aliquoting: Upon receipt, aliquot antibodies into smaller volumes to avoid repeated freeze-thaw cycles, which can lead to protein denaturation and loss of activity.

  • Working Concentration: Always optimize the antibody concentration for your specific application and sample type. Starting dilutions are typically provided in product datasheets but may need adjustment.

  • Stability Assessment: The stability of antibodies can be evaluated through accelerated thermal degradation tests, where the protein is incubated at 37°C for 48 hours to assess degradation rates .

  • Reconstitution: If the antibody is lyophilized, reconstitute carefully according to manufacturer instructions using appropriate buffers.

  • Contamination Prevention: Use sterile techniques when handling antibodies to prevent microbial contamination.

Following these storage and handling guidelines will help ensure consistent, reliable results across experiments.

How can I validate the specificity of HSPD1 antibodies for my research?

Validating antibody specificity is a critical step in ensuring reliable research outcomes. For HSPD1 antibodies, consider these validation approaches:

  • Western Blot Analysis: Verify a single band at the expected molecular weight of 60-61 kDa in your experimental samples . The presence of additional bands may indicate cross-reactivity or protein modification.

  • Knockout/Knockdown Controls: Use HSPD1 knockout or knockdown samples as negative controls to confirm antibody specificity. The absence or reduction of signal in these samples supports antibody specificity.

  • Pre-absorption Tests: Pre-incubate the antibody with purified HSPD1 protein before application to your samples. This should abolish or significantly reduce specific staining.

  • Multiple Antibody Comparison: Use different HSPD1 antibodies that recognize distinct epitopes (e.g., N-terminal vs. C-terminal) to confirm consistent detection patterns .

  • Cross-Species Reactivity Analysis: If working with non-human samples, confirm the cross-reactivity of your chosen antibody with your species of interest. Many HSPD1 antibodies show reactivity across multiple species, including human, mouse, rat, chicken, cow, dog, and pig .

  • Peptide Competition Assay: Compete your antibody with the immunizing peptide to demonstrate specificity of binding.

  • Immunoprecipitation Followed by Mass Spectrometry: This approach can identify the specific proteins recognized by the antibody, confirming HSPD1 detection and revealing any off-target binding.

These validation steps should be documented and included in your research methods to strengthen the reliability of your findings.

What are the optimal conditions for immunoprecipitation of HSPD1 protein complexes?

Immunoprecipitation (IP) of HSPD1 requires careful consideration of experimental conditions to preserve protein-protein interactions while ensuring specific pulldown:

  • Antibody Selection: Choose an antibody specifically validated for IP applications . Consider antibodies targeting different epitopes of HSPD1 (e.g., N-terminal vs. C-terminal) as protein interactions may mask certain epitopes.

  • Lysis Buffer Optimization:

    • For studying HSPD1 interactions with other mitochondrial proteins, use a gentle lysis buffer containing 0.5-1% NP-40 or CHAPS to preserve native interactions.

    • Include protease inhibitors to prevent degradation during the IP procedure.

    • Consider including ATP (1-5 mM) in buffers when studying chaperonin-substrate interactions, as HSPD1 function is ATP-dependent .

  • Pre-clearing Step: Pre-clear lysates with appropriate control beads to reduce non-specific binding.

  • Antibody-Bead Coupling:

    • For polyclonal antibodies, Protein A-conjugated beads are generally suitable for rabbit-derived antibodies .

    • For mouse monoclonal antibodies, Protein G-conjugated beads may provide better binding .

    • Consider covalent coupling of antibodies to beads to avoid IgG contamination in downstream applications.

  • Incubation Conditions:

    • Perform antibody-antigen binding at 4°C overnight with gentle rotation to maximize specific interactions while minimizing degradation.

    • For studying transient interactions, consider shorter incubation times or crosslinking approaches.

  • Washing Stringency:

    • Use at least 3-5 washes with decreasing salt concentrations to remove non-specific binders.

    • Adjust washing stringency based on the stability of the protein complexes being studied.

  • Elution Methods:

    • For mass spectrometry applications, consider on-bead digestion to minimize contamination.

    • For western blot analysis, elute using SDS-PAGE loading buffer with heating.

    • For functional studies, consider gentle elution with excess immunizing peptide.

  • Controls: Always include appropriate negative controls (non-specific IgG or pre-immune serum) and input samples to assess IP efficiency and specificity.

By optimizing these conditions for your specific experimental goals, you can effectively study HSPD1's interactions with its co-chaperonin HSP10 and other binding partners or substrates.

What are the key considerations for detecting HSPD1 in tissue sections by immunohistochemistry?

Immunohistochemistry (IHC) for HSPD1 detection in tissue sections requires attention to several critical factors:

  • Fixation Protocol:

    • Formalin fixation may mask HSPD1 epitopes, requiring antigen retrieval optimization.

    • For mitochondrial proteins like HSPD1, overfixation can limit antibody accessibility to the inner mitochondrial structures.

    • Consider testing both frozen and paraffin-embedded sections to determine optimal preservation of antigenicity.

  • Antigen Retrieval:

    • Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or EDTA buffer (pH 9.0) is commonly effective for HSPD1.

    • Optimize retrieval time and temperature based on tissue type and fixation duration.

  • Antibody Selection:

    • Choose antibodies specifically validated for IHC applications .

    • Consider whether the antibody recognizes human, mouse, or both depending on your tissue origin.

    • Both monoclonal (e.g., 3G8 clone) and polyclonal antibodies have been validated for IHC .

  • Blocking and Permeabilization:

    • Thorough blocking with serum-based blockers reduces non-specific binding.

    • Adequate permeabilization is essential for accessing mitochondrial antigens like HSPD1.

  • Antibody Dilution and Incubation:

    • Optimize primary antibody dilutions through titration experiments.

    • Extended incubation times (overnight at 4°C) often improve specific staining for mitochondrial proteins.

  • Detection System:

    • Amplification systems like polymer-based detection may enhance sensitivity for detecting mitochondrial HSPD1.

    • Consider fluorescent secondary antibodies for co-localization studies with other mitochondrial markers.

  • Controls:

    • Positive controls: Include tissues known to express high levels of HSPD1 (e.g., heart, liver).

    • Negative controls: Omit primary antibody or use isotype control antibodies.

    • Specificity controls: Consider tissues from HSPD1-deficient models if available.

  • Counterstaining:

    • Choose counterstains that don't obscure the mitochondrial staining pattern.

    • For fluorescent detection, DAPI nuclear counterstain can provide cellular context without interfering with mitochondrial signal.

  • Image Analysis:

    • Due to HSPD1's mitochondrial localization, high-resolution imaging may be necessary to distinguish specific staining patterns.

    • Consider z-stack acquisitions for thicker tissue sections to capture the three-dimensional distribution of mitochondria.

By addressing these considerations, researchers can achieve consistent and specific detection of HSPD1 in various tissue types for comparative pathological studies or basic research applications.

Why might I observe multiple bands on Western blots when using HSPD1 antibodies?

Multiple bands on Western blots with HSPD1 antibodies can occur for several biological and technical reasons:

  • Post-translational Modifications:

    • HSPD1 undergoes various modifications including phosphorylation, acetylation, and ubiquitination that can alter its mobility.

    • Stress conditions may induce modifications that generate additional bands.

  • Processing Forms:

    • HSPD1 contains a mitochondrial targeting sequence that is cleaved upon import, potentially generating multiple forms.

    • The mature form is expected at approximately 60-61 kDa .

    • The precursor form with the targeting sequence may appear at a slightly higher molecular weight.

  • Proteolytic Degradation:

    • Inadequate protease inhibition during sample preparation can result in HSPD1 degradation products.

    • This is particularly common in samples from tissues with high protease content.

  • Antibody Cross-reactivity:

    • Some antibodies may cross-react with other members of the heat shock protein family with similar sequences.

    • Verify specificity by comparing results from antibodies targeting different epitopes of HSPD1 .

  • Alternative Splicing:

    • HSPD1 may have splice variants that generate proteins of different sizes.

    • Antibodies targeting different regions may detect specific splice variants.

  • Experimental Conditions:

    • Incomplete denaturation can result in partially folded forms with altered mobility.

    • Overloading of protein samples can cause band distortion.

    • Improper SDS-PAGE conditions, including buffer composition and pH, may affect protein migration.

  • Non-specific Binding:

    • Insufficient blocking or washing can lead to non-specific bands.

    • Using freshly prepared buffers and high-quality blocking agents can reduce this issue.

Troubleshooting steps should include:

  • Comparing results across different antibodies targeting different epitopes of HSPD1 .

  • Including appropriate positive and negative controls.

  • Optimizing sample preparation protocols to ensure complete denaturation and prevent degradation.

  • Performing peptide competition assays to identify specific versus non-specific bands.

Understanding the basis for multiple bands is important for accurate interpretation of HSPD1 expression studies.

How can I optimize Western blotting protocols for detecting HSPD1 in different sample types?

Optimizing Western blotting for HSPD1 detection across different sample types requires tailored approaches:

  • Cell Line Samples:

    • Lysis buffer: Use RIPA buffer supplemented with protease inhibitors.

    • Loading amount: 20-30 μg of total protein is typically sufficient.

    • Expected result: Clear single band at approximately 60-61 kDa .

  • Tissue Samples:

    • Homogenization: Use mechanical disruption (e.g., Dounce homogenizer) followed by brief sonication.

    • Buffer selection: For tissues rich in proteases (e.g., pancreas, spleen), increase protease inhibitor concentration.

    • Loading amount: 40-50 μg may be needed for tissues with lower HSPD1 expression.

    • Consider subcellular fractionation to enrich for mitochondria in tissues with high background.

  • Primary Cell Cultures:

    • Harvest cells at 80-90% confluence to ensure consistent protein expression.

    • For sensitive primary cells, use gentler lysis buffers (0.5% NP-40 or Triton X-100).

    • Normalize loading based on housekeeping proteins appropriate for the cell type.

  • Mitochondrial Enriched Fractions:

    • Use specialized mitochondrial isolation kits to obtain enriched fractions.

    • Lower loading amounts (10-15 μg) may be sufficient due to enrichment.

    • Include mitochondrial markers (e.g., VDAC, COX IV) as loading controls rather than cytosolic markers.

  • Gel and Transfer Optimization:

    • Use 10-12% polyacrylamide gels for optimal resolution around 60 kDa.

    • Transfer conditions: 100V for 1 hour or 30V overnight at 4°C in Towbin buffer with 10-20% methanol.

    • PVDF membranes may provide better results than nitrocellulose for HSPD1 detection.

  • Antibody Selection and Dilution:

    • For human samples, multiple antibody options are available, including monoclonal antibodies like LK1 and 3G8 clones .

    • For cross-species studies, select antibodies with verified reactivity in your species of interest .

    • Optimize primary antibody concentration through titration (typical range: 1:500 to 1:5000).

    • Extend primary antibody incubation to overnight at 4°C for improved signal-to-noise ratio.

  • Detection System:

    • HRP-conjugated secondary antibodies with enhanced chemiluminescence (ECL) provide good sensitivity.

    • For low abundance samples, consider using higher sensitivity ECL substrates or fluorescence-based detection.

  • Stripping and Reprobing:

    • HSPD1 antibodies may be effectively stripped and membranes reprobed for other proteins.

    • Use mild stripping buffers to preserve membrane integrity if reprobing is planned.

By tailoring these parameters to your specific sample type, you can achieve consistent and specific detection of HSPD1 across diverse experimental conditions.

What could cause unexpected subcellular localization of HSPD1 in immunofluorescence studies?

While HSPD1 is primarily a mitochondrial protein, unexpected subcellular localization in immunofluorescence studies may occur due to various biological and technical factors:

  • Biological Factors:

    • Extramitochondrial HSPD1: Research suggests HSPD1 can localize to additional compartments including the cell surface, cytosol, and extracellular space under certain conditions.

    • Cellular Stress Response: Heat shock, oxidative stress, or inflammation can trigger relocalization of HSPD1 outside mitochondria as part of cellular stress responses.

    • Cell Cycle Variation: HSPD1 distribution may change during different phases of the cell cycle.

    • Pathological Conditions: In disease states, HSPD1 localization can be altered compared to healthy cells.

  • Technical Considerations:

    • Fixation Artifacts: Different fixation methods can affect apparent protein localization:

      • Paraformaldehyde (4%) may preserve mitochondrial morphology better than methanol.

      • Overfixation can cause epitope masking or non-specific binding.

      • Underfixation may allow protein redistribution during processing.

    • Permeabilization Effects: The choice of permeabilization agent impacts antibody accessibility:

      • Triton X-100 (0.1-0.5%) provides good accessibility but may extract some membrane proteins.

      • Saponin (0.1%) offers gentler permeabilization but may require continuous presence in buffers.

      • Digitonin at low concentrations can selectively permeabilize the plasma membrane while leaving mitochondrial membranes intact.

    • Antibody Specificity Issues:

      • Different epitopes may be differentially accessible in various cellular compartments.

      • Compare results using antibodies targeting different regions of HSPD1 .

      • Some antibodies may cross-react with related heat shock proteins in other compartments.

    • Sample Processing Time: Delays between fixation steps may allow redistribution of proteins.

  • Verification Approaches:

    • Co-localization Studies: Always co-stain with established mitochondrial markers (e.g., MitoTracker, TOMM20, COX IV).

    • Super-resolution Microscopy: Techniques like STED or STORM provide higher resolution to distinguish genuine localization from artifacts.

    • Biochemical Fractionation: Complement imaging with subcellular fractionation and Western blotting.

    • Live Cell Imaging: Where possible, use GFP-tagged HSPD1 in live cells to monitor localization without fixation artifacts.

    • Multiple Antibody Comparison: Use different antibodies targeting distinct epitopes to confirm unusual localization patterns .

  • Controls to Include:

    • Primary antibody omission controls to assess secondary antibody specificity.

    • Peptide competition controls to verify binding specificity.

    • Positive controls using cells known to express high levels of mitochondrial HSPD1.

Understanding whether unexpected localization represents genuine biological phenomena or technical artifacts is crucial for accurate data interpretation.

How can HSPD1 antibodies be used to investigate mitochondrial stress responses?

HSPD1 antibodies provide powerful tools for examining mitochondrial stress responses across multiple experimental platforms:

  • Quantitative Expression Analysis:

    • Western blotting with HSPD1 antibodies can quantify upregulation during mitochondrial stress conditions .

    • Compare HSPD1 levels against other mitochondrial stress markers like HSP10, ClpP, or LONP1 to establish stress response patterns.

    • Flow cytometry with permeabilized cells can assess HSPD1 expression changes at the single-cell level, revealing population heterogeneity in stress responses .

  • High-Content Imaging Approaches:

    • Immunofluorescence microscopy with HSPD1 antibodies can visualize changes in mitochondrial morphology under stress.

    • Quantitative image analysis can measure:

      • HSPD1 intensity changes relative to mitochondrial mass markers

      • Mitochondrial fragmentation or hyperfusion correlated with HSPD1 expression

      • Redistribution of HSPD1 within mitochondrial subcompartments during stress

    • Live-cell compatible immunolabeling techniques can track dynamic changes in HSPD1 distribution.

  • Protein-Protein Interaction Studies:

    • Immunoprecipitation with HSPD1 antibodies can identify stress-dependent interaction partners .

    • Proximity ligation assays can visualize in situ interactions between HSPD1 and substrate proteins under different stress conditions.

    • Mass spectrometry following HSPD1 immunoprecipitation can identify the "client repertoire" that changes during specific stresses.

  • Mitochondrial Protein Folding Assessment:

    • Combine HSPD1 immunoprecipitation with analysis of bound unfolded proteins to assess chaperone loading during stress.

    • Crosslinking approaches can capture transient interactions between HSPD1 and substrates under stress conditions.

    • Correlate HSPD1 expression with measures of protein aggregation to assess chaperone function.

  • Experimental Design Considerations:

    • Stress Conditions: Different stressors affect HSPD1 differently:

      • Heat shock (42°C, 1-2 hours)

      • Oxidative stress (H₂O₂, paraquat)

      • Mitochondrial toxins (rotenone, antimycin A)

      • Protein folding stress (tunicamycin, MG132)

    • Time Course Analysis: HSPD1 response often follows a temporal pattern:

      • Early response (0-2 hours): translocation/activation of existing HSPD1

      • Intermediate response (2-8 hours): transcriptional upregulation

      • Late response (8-24 hours): adaptation or damage resolution

    • Cell Type Considerations: Mitochondria-rich cells (cardiomyocytes, neurons, hepatocytes) may show different HSPD1 dynamics compared to other cell types.

  • Analyzing the Unfolded Protein Response in Mitochondria (UPRmt):

    • HSPD1 is a key marker and effector of UPRmt.

    • Compare HSPD1 levels with activation of UPRmt transcription factors (ATFS-1/ATF5).

    • Correlate HSPD1 expression with mitochondrial proteostasis markers.

These approaches provide complementary data on how HSPD1 participates in the mitochondrial stress response, offering insights into both adaptive and maladaptive cellular reactions to mitochondrial dysfunction.

What roles does HSPD1 play in disease processes, and how can antibodies help investigate these roles?

HSPD1 has been implicated in various disease processes, and antibodies provide crucial tools for investigating these pathological roles:

  • Neurodegenerative Diseases:

    • HSPD1 expression changes have been observed in Alzheimer's, Parkinson's, and ALS.

    • Research applications:

      • Immunohistochemistry to assess HSPD1 distribution in brain tissue sections .

      • Co-localization studies with disease-specific protein aggregates (β-amyloid, α-synuclein, SOD1).

      • Analysis of HSPD1 post-translational modifications in disease models.

      • Investigation of HSPD1 interaction with disease-relevant proteins through co-immunoprecipitation .

  • Cardiovascular Diseases:

    • HSPD1 has been identified as both a potential biomarker and contributor to cardiovascular pathologies.

    • Research approaches:

      • Quantification of HSPD1 levels in cardiac tissue during ischemia-reperfusion injury.

      • Assessment of extracellular HSPD1 as a damage-associated molecular pattern (DAMP) in atherosclerosis.

      • Evaluation of HSPD1 autoantibodies in myocarditis and heart failure.

      • Analysis of HSPD1 chaperone function in cardiomyocyte mitochondrial proteostasis.

  • Cancer Biology:

    • HSPD1 is frequently overexpressed in various cancers and may contribute to tumor cell survival.

    • Investigation methods:

      • Immunohistochemical profiling of HSPD1 expression across tumor types and grades .

      • Correlation of HSPD1 levels with markers of mitochondrial metabolism in tumors.

      • Analysis of HSPD1 interaction with apoptotic regulators through protein-protein interaction studies.

      • Evaluation of HSPD1 as a biomarker for therapy response through longitudinal expression studies.

  • Autoimmune Disorders:

    • HSPD1 can act as an autoantigen in conditions like rheumatoid arthritis and multiple sclerosis.

    • Research applications:

      • Detection of HSPD1-specific autoantibodies in patient samples.

      • Analysis of HSPD1 presentation by antigen-presenting cells.

      • Evaluation of post-translational modifications that might increase immunogenicity.

      • Investigation of extracellular HSPD1 as an immunomodulator.

  • Metabolic Diseases:

    • HSPD1 dysfunction may contribute to metabolic disorders including diabetes and obesity.

    • Research approaches:

      • Assessment of HSPD1 chaperone activity in pancreatic β-cells under metabolic stress.

      • Analysis of HSPD1 involvement in mitochondrial quality control in insulin-responsive tissues.

      • Correlation of HSPD1 expression with markers of insulin resistance.

      • Investigation of HSPD1 interactions with metabolic enzymes through co-immunoprecipitation studies .

  • Methodological Considerations:

    • Tissue-specific Analysis: Different antibody dilutions and protocols may be needed for different tissue types .

    • Disease Stage Differentiation: Compare HSPD1 expression across disease progression stages.

    • Relationship with Clinical Parameters: Correlate HSPD1 patterns with clinical data and outcomes.

    • Multi-omics Integration: Combine antibody-based studies with transcriptomics and proteomics data.

  • Technical Approaches:

    • Multiplexed Immunofluorescence: Co-stain for HSPD1 alongside disease markers and mitochondrial indicators.

    • Tissue Microarrays: Enable high-throughput screening of HSPD1 expression across multiple patient samples.

    • Circulating HSPD1 Detection: Develop sandwich ELISA using matched antibody pairs for serum/plasma biomarker studies .

    • Single-cell Analysis: Combine flow cytometry with HSPD1 antibodies to assess heterogeneity in diseased tissues .

By applying these antibody-based approaches, researchers can elucidate the complex roles of HSPD1 in disease pathogenesis, potentially identifying new diagnostic markers or therapeutic targets.

How can HSPD1 antibodies be utilized in studying mitochondrial dynamics and quality control?

HSPD1 antibodies provide valuable tools for investigating the relationship between protein folding and broader aspects of mitochondrial quality control:

  • Mitochondrial Fission/Fusion Dynamics:

    • Immunofluorescence with HSPD1 antibodies paired with mitochondrial morphology markers can reveal correlations between chaperone activity and network restructuring .

    • Research applications:

      • Co-immunostaining with DRP1 (fission) or MFN1/2 (fusion) proteins to assess HSPD1 distribution during dynamic events.

      • Time-lapse imaging following stress induction to correlate HSPD1 redistribution with mitochondrial fragmentation or hyperfusion.

      • Super-resolution microscopy to visualize HSPD1 microdomains during network remodeling.

  • Mitophagy Processes:

    • HSPD1 antibodies can help track the fate of damaged mitochondria destined for autophagic degradation.

    • Experimental approaches:

      • Co-localization analysis of HSPD1 with mitophagy receptors (PINK1, Parkin, BNIP3L).

      • Quantification of HSPD1 levels during mitophagy induction to assess degradation kinetics.

      • Differential extraction methods to distinguish HSPD1 in healthy versus mitophagy-targeted organelles.

  • Mitochondrial Unfolded Protein Response (UPRmt):

    • HSPD1 serves as both a marker and effector of UPRmt activation.

    • Investigation methods:

      • Quantitative Western blotting to assess HSPD1 upregulation during UPRmt activation .

      • Chromatin immunoprecipitation to study transcription factor binding at the HSPD1 promoter.

      • Pulse-chase experiments to measure HSPD1 synthesis rates during proteostatic stress.

      • RNA-protein co-immunoprecipitation to investigate translational regulation of HSPD1.

  • Mitochondrial Proteome Maintenance:

    • HSPD1 antibodies enable the study of protein folding networks within mitochondria.

    • Research applications:

      • Immunoprecipitation followed by mass spectrometry to identify HSPD1 "client proteins" .

      • Proximity labeling approaches to map the HSPD1 interactome under different conditions.

      • Analysis of HSPD1 cooperation with other chaperone systems (HSP70, HSP90).

      • Investigation of HSPD1-protease interactions in protein quality control.

  • Advanced Technical Approaches:

    • Live-cell Compatible Immunolabeling:

      • Use cell-permeable nanobodies derived from HSPD1 antibodies for real-time tracking.

      • Correlate dynamic HSPD1 redistribution with mitochondrial membrane potential fluctuations.

    • Cryo-Electron Microscopy:

      • Immunogold labeling with HSPD1 antibodies for ultrastructural localization.

      • Visualization of HSPD1 ring complexes in different functional states.

    • Functional Assays:

      • Combine HSPD1 immunodepletion with in vitro protein folding assays to assess chaperone capacity.

      • Microfluidic approaches to analyze single mitochondria with HSPD1 immunolabeling.

  • Disease Model Applications:

    • HSPD1 antibodies can reveal mitochondrial quality control defects in disease models:

      • Neurodegenerative disorders (Parkinson's, ALS) with impaired mitophagy.

      • Metabolic diseases with altered mitochondrial dynamics.

      • Cancer models with dysregulated mitochondrial biogenesis.

By applying these approaches, researchers can connect HSPD1's chaperone activity to broader mechanisms of mitochondrial homeostasis, providing insights into fundamental biological processes and disease pathogenesis.

What are the methodological considerations for studying post-translational modifications of HSPD1?

Post-translational modifications (PTMs) of HSPD1 significantly influence its function, localization, and interactions. Investigating these modifications requires specialized methodological approaches:

  • Detection Strategies for HSPD1 PTMs:

    • Phosphorylation:

      • Use phospho-specific antibodies when available.

      • Employ Phos-tag™ SDS-PAGE followed by Western blotting with general HSPD1 antibodies to separate phosphorylated forms .

      • Perform lambda phosphatase treatment as a control to confirm phosphorylation.

    • Acetylation:

      • Immunoprecipitate HSPD1 with general antibodies followed by Western blotting with anti-acetyllysine antibodies .

      • Treat samples with deacetylase inhibitors (TSA, nicotinamide) to preserve acetylation status.

    • Ubiquitination/SUMOylation:

      • Use denaturing conditions during immunoprecipitation to disrupt interacting proteins.

      • Perform sequential immunoprecipitation: first HSPD1, then blot for ubiquitin/SUMO or vice versa.

  • Mass Spectrometry-Based Approaches:

    • Sample Preparation Considerations:

      • Enrich for HSPD1 through immunoprecipitation using antibodies targeting non-modified regions .

      • Consider PTM-specific enrichment (e.g., TiO₂ for phosphopeptides, anti-acetyllysine antibodies for acetylated peptides).

      • Use proteases beyond trypsin (e.g., chymotrypsin, Glu-C) to increase coverage of modification sites.

    • Analysis Strategies:

      • Employ both collision-induced dissociation (CID) and electron-transfer dissociation (ETD) for complementary fragmentation patterns.

      • Implement targeted MS approaches (PRM, MRM) to increase sensitivity for known modification sites.

      • Use quantitative approaches (SILAC, TMT) to compare modification levels across conditions.

  • Site-Specific Modification Studies:

    • Generate site-specific antibodies for major HSPD1 modification sites if commercially unavailable.

    • Employ site-directed mutagenesis (Ser→Ala for phosphorylation, Lys→Arg for acetylation/ubiquitination) followed by functional assays.

    • Use CRISPR-Cas9 knock-in to introduce modification-mimicking mutations.

  • Functional Impact Assessment:

    • Chaperone Activity Assays:

      • Compare folding capacity of modified versus unmodified HSPD1 using purified components.

      • Assess the impact of modifications on ATP hydrolysis rates.

    • Structural Analysis:

      • Use hydrogen-deuterium exchange mass spectrometry to determine how modifications affect protein dynamics.

      • Apply cryo-EM to visualize structural changes induced by specific modifications.

    • Interaction Profiling:

      • Compare interactomes of modified versus unmodified HSPD1 through quantitative proteomics.

      • Assess the impact of modifications on HSPD1-HSP10 complex formation.

  • Spatiotemporal Dynamics of HSPD1 Modifications:

    • Develop workflows to track modification changes during:

      • Mitochondrial stress responses

      • Cell cycle progression

      • Differentiation processes

      • Disease development

    • Combine fractionation approaches with modification-specific detection to determine compartment-specific modification patterns.

  • Technical Challenges and Solutions:

    • Antibody Specificity:

      • Rigorously validate modification-specific antibodies using appropriate controls (phosphatase/deacetylase treatment, site-directed mutants).

      • Consider using multiple antibodies targeting the same modification for confirmation.

    • Low Abundance of Modified Forms:

      • Implement enrichment strategies before analysis.

      • Use cellular treatments that enhance specific modifications (kinase activators, deacetylase inhibitors).

    • Preservation of Labile Modifications:

      • Add inhibitors of modifying/demodifying enzymes immediately during cell/tissue lysis.

      • Maintain cold temperatures throughout processing.

By addressing these methodological considerations, researchers can gain detailed insights into how PTMs regulate HSPD1 function in both normal physiology and disease states.

What are the emerging trends in HSPD1 antibody applications for research?

The research landscape for HSPD1 antibodies continues to evolve, with several emerging trends expanding their utility beyond traditional applications:

  • Integration with Advanced Imaging Technologies:

    • Super-resolution microscopy techniques (STED, STORM, PALM) are being combined with HSPD1 immunolabeling to visualize mitochondrial substructures at unprecedented resolution.

    • Correlative light and electron microscopy (CLEM) with HSPD1 antibodies enables linking functional observations to ultrastructural context.

    • Live-cell compatible immunolabeling approaches are developing to track dynamic changes in HSPD1 distribution.

  • Single-Cell Analysis Applications:

    • Mass cytometry (CyTOF) with metal-conjugated HSPD1 antibodies enables high-dimensional profiling of mitochondrial states in heterogeneous cell populations.

    • Imaging mass cytometry allows spatial mapping of HSPD1 expression in tissue contexts alongside dozens of other markers.

    • Single-cell Western blotting with HSPD1 antibodies can quantify expression variability within populations.

  • Spatially-Resolved Proteomics:

    • Proximity labeling approaches (BioID, APEX) combined with HSPD1 antibodies for validation are mapping the spatial organization of mitochondrial protein networks.

    • In situ protein interaction analysis methods like proximity ligation assays are being optimized for mitochondrial proteins including HSPD1.

    • Spatial transcriptomics combined with HSPD1 immunohistochemistry links protein expression to underlying transcriptional programs with spatial context.

  • Technical Advancements in Antibody Formats:

    • Conjugation-ready formats of HSPD1 antibodies without stabilizing proteins (BSA-free, azide-free) enable custom labeling for specialized applications .

    • Matched antibody pairs designed for multiplex detection systems allow simultaneous quantification of HSPD1 alongside other biomarkers .

    • Recombinant antibody technology is increasing consistency and reducing batch-to-batch variation in HSPD1 detection .

  • Clinical Research Applications:

    • Development of standardized immunohistochemistry protocols for HSPD1 assessment in pathology specimens .

    • Use of quantitative immunoassays with HSPD1 antibodies for biomarker studies in various diseases.

    • Digital pathology approaches incorporating HSPD1 staining patterns for automated diagnostic algorithms.

  • Systems Biology Integration:

    • Multi-omics approaches combining HSPD1 antibody-based protein detection with transcriptomics, metabolomics, and functional readouts.

    • Computational modeling of chaperone networks calibrated with quantitative HSPD1 expression data.

    • Large-scale screens combining HSPD1 immunodetection with genetic or pharmacological perturbations.

These emerging trends highlight the continuing importance of high-quality, well-characterized HSPD1 antibodies in advancing our understanding of mitochondrial biology, cellular stress responses, and disease mechanisms. As technology continues to evolve, antibody applications will likely expand further, particularly in areas requiring increased sensitivity, specificity, multiplexing capabilities, and compatibility with in vivo imaging approaches.

What future developments can we anticipate in HSPD1 antibody technology?

The field of HSPD1 antibody technology is poised for significant advancements that will expand research capabilities and clinical applications:

  • Next-Generation Antibody Engineering:

    • Development of single-domain antibodies (nanobodies) against HSPD1 epitopes for improved penetration of intact cells and mitochondria.

    • Creation of bispecific antibodies linking HSPD1 recognition with other mitochondrial markers for enhanced specificity.

    • Generation of conformation-specific antibodies that distinguish between different functional states of HSPD1 (ATP-bound, substrate-bound, HSP10-associated).

    • Engineering recombinant antibodies with reduced non-specific binding for challenging applications like brain tissue immunohistochemistry.

  • Enhanced Detection Technologies:

    • Development of more sensitive detection systems for low-abundance HSPD1 post-translational modifications.

    • Creation of FRET-based antibody pairs to detect HSPD1 conformational changes in live cells.

    • Implementation of amplification technologies that maintain spatial resolution while increasing detection sensitivity.

    • Design of antibody-based biosensors that report on HSPD1 chaperone activity rather than just protein levels.

  • Improved Spatial and Temporal Resolution:

    • Development of antibody fragments compatible with expanded microscopy techniques for super-resolution imaging of HSPD1 distribution.

    • Creation of photoactivatable antibody conjugates for pulse-chase tracking of HSPD1 dynamics.

    • Engineering of antibodies compatible with intravital imaging for in vivo tracking of HSPD1 responses.

    • Implementation of multiplexed imaging approaches that can simultaneously track dozens of proteins alongside HSPD1.

  • Clinical and Diagnostic Applications:

    • Development of standardized HSPD1 immunohistochemistry protocols for clinical pathology .

    • Creation of point-of-care diagnostic tools using HSPD1 antibodies for rapid biomarker assessment.

    • Implementation of digital pathology algorithms incorporating HSPD1 staining patterns for automated diagnosis.

    • Design of companion diagnostic tests using HSPD1 antibodies to guide personalized therapeutic approaches.

  • Technical Improvements:

    • Development of antibodies with enhanced stability for long-term storage at higher temperatures.

    • Creation of universal labeling platforms that enable modular attachment of detection moieties to HSPD1 antibodies.

    • Implementation of machine learning approaches to optimize antibody design and application-specific performance.

    • Engineering of antibodies with reduced cross-reactivity against homologous proteins in the heat shock family.

  • Research Tool Innovation:

    • Development of intrabodies that can track and potentially modulate HSPD1 function in live cells.

    • Creation of degron-tagged antibody fragments for acute inactivation of HSPD1 function.

    • Implementation of antibody-directed enzyme prodrug therapy (ADEPT) approaches using HSPD1 antibodies for targeted mitochondrial modulation.

    • Design of antibody-oligonucleotide conjugates for proximity-based detection of HSPD1-interacting RNAs.

  • Standardization and Reproducibility Initiatives:

    • Establishment of industry-wide standards for HSPD1 antibody validation across multiple applications.

    • Development of reference materials and protocols for quantitative calibration of HSPD1 immunoassays.

    • Creation of open-access databases documenting application-specific performance of different HSPD1 antibodies.

    • Implementation of automation in antibody production to enhance batch-to-batch consistency .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.