1
|
Bieganowski P, Dalidowska I, Gazi O, Guzowska M, Przybylski M. Study of Hsp90α and Hsp90β role in virus replication using cell lines with Hsp90 gene knockouts. Virus Genes 2025; 61:277-283. [PMID: 39948206 DOI: 10.1007/s11262-025-02141-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/04/2025] [Indexed: 05/06/2025]
Abstract
Replication of the human Enterovirus 71 (EV71) and herpes simplex virus 1 (HSV-1) requires Hsp90 chaperone activity. Vertebrate cells express two cytosolic Hsp90 proteins, Hsp90α and Hsp90β. Earlier reports suggested that EV71 replication might depend solely on the Hsp90β, whereas HSV-1 replication depended on Hsp90α. Here, we describe construction of the cell line knockouts missing Hsp90α or Hsp90β protein. Using these cells, we found that HSV-1 and, another enterovirus, Coxsackievirus B5 (CVB5) replicate in both Hsp90α and Hsp90β knockout cells with equal efficiency. The presented results demonstrate that cell lines with a mutation inactivating the specific HSP90 gene might be an easy-to-use and robust system to study specific cellular functions of Hsp90α and Hsp90β.
Collapse
Affiliation(s)
- Pawel Bieganowski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Iga Dalidowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Olga Gazi
- Chair and Depatment of Medical Microbiology, Medical University of Warsaw, Warsaw, Poland
| | - Magdalena Guzowska
- Division of Biochemistry and Dietetics, Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Poland
| | - Maciej Przybylski
- Chair and Depatment of Medical Microbiology, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
2
|
Kaur J, Roy I. Pharmacological Regulation of Heat Shock Response via Aptamer-Antidote Couple. ACS Chem Neurosci 2025. [PMID: 40388587 DOI: 10.1021/acschemneuro.4c00865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025] Open
Abstract
Heat shock factor 1 (HSF1) orchestrates the cellular heat shock response (HSR) by binding to heat shock elements (HSEs) in the promoters of genes encoding heat shock proteins (HSPs). In a nonstressed state, HSF1 exists in a dormant complex with HSP90 and other chaperones. Upon cellular stress or upon inhibition of HSP90, HSF1 dissociates from the complex and activates the expression of HSPs to mitigate protein misfolding and aggregation. This study explores the potential of RNA aptamers selected against HSP90 to modulate HSF1 activity, with a role in Huntington's disease model characterized by protein aggregation. Selected aptamers disrupted the HSP90-HSF1 interaction, enhancing the binding of HSF1 with HSEs. This upregulated heat shock response (HSR) and reduced aggregation of Q74-huntingtin in Neuro 2a cells with improved cell survival. Designed antidote sequences could reverse the effect of the aptamers on the HSF1-HSE interaction, allowing for fine-tuning of HSR. Chronic activation of stress response pathways is deleterious for cellular fitness. Our findings suggest that coupling an antidote with an aptamer offers a novel therapeutic strategy to regulate cellular proteostasis under disease conditions.
Collapse
Affiliation(s)
- Jaskirat Kaur
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| |
Collapse
|
3
|
Shahana MV, Choudhary B. HSP90 and the cancer transcriptome: a comprehensive review of inhibitors and mechanistic insights. Int J Clin Oncol 2025:10.1007/s10147-025-02782-6. [PMID: 40383747 DOI: 10.1007/s10147-025-02782-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 05/01/2025] [Indexed: 05/20/2025]
Abstract
This review summarizes the structure, function, expression, and inhibitors of HSP90, the chaperone, in cancers. It systematically investigates the effects of HSP90 inhibitors, including AUY922, B11B021, CCT-018159, D7-gedunin, geldanamycin, and gedunin, across a range of cancer cell lines (HCC151, HT29, MCF7, PC3, VCAP, and A375) and a normal HA1E cell line, using data from the CLUE database. Our analysis reveals that treatment with these HSP90 inhibitors induces significant stress responses in tumor cells, initiating intrinsic and extrinsic apoptotic pathways. The HSP90AA1, HSP90AB1, HSP27, HSP70, VEGF, and NOTCH exhibited notable upregulation at 24 h post-treatment compared to 6 h, indicating a time-dependent increase in cellular stress (heat shock response) and activation of pro-survival signaling mechanisms. Additionally, the study highlights a significant upregulation of immune-related pathways, including those involving IL10, IL3, and IL7, following HSP90 inhibition, indicating that these inhibitors not only directly affect tumor cell viability but also modulate the tumor microenvironment by enhancing immune cell activation and cytokine release. The elevated levels of IL10 point to a dual role, where immune suppression mechanisms are also at play, potentially facilitating immune evasion by the tumor. The findings suggest that HSP90 inhibitors exhibit varying mechanisms of action across different cancer cell lines despite the presence of some common targets. These insights highlight the need for further investigation into the precise mechanisms of HSP90 inhibitors to optimize their therapeutic potential in different cancers.
Collapse
Affiliation(s)
- M V Shahana
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, 560100, India
- Research scholar, Manipal Academy of Higher Education (MAHE), Manipal, 576104, India
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Electronic City Phase 1, Bangalore, Karnataka, 560100, India.
| |
Collapse
|
4
|
Blatch GL, Edkins AL. New insights into Sti1/Hop's cochaperone function highlight the complexity of proteostatic regulation. FEBS J 2025. [PMID: 40259657 DOI: 10.1111/febs.70108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025]
Abstract
Sti1/Hop is a cochaperone that regulates Hsp70 and Hsp90 chaperones. Sti1/Hop function is perceived as limited to scaffolding chaperone complexes, although recent studies suggest a broader function. Rutledge et al. show that while Sti1/Hop functions within chaperone complexes under basal conditions, during high stress, it operates independently to sequester soluble misfolded protein in the cytoplasm, a function typically associated with chaperones rather than cochaperones. Furthermore, the localisation and levels of Sti1/Hop are finely tuned to ensure orderly sequestration and resolution of misfolded proteins. These data support a role for Sti1/Hop as a cochaperone specialised for stressed proteostasis networks.
Collapse
Affiliation(s)
- Gregory Lloyd Blatch
- The Vice Chancellery, The University of Notre Dame Australia, Fremantle, Australia
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda, South Africa
| | - Adrienne Lesley Edkins
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry, Microbiology and Bioinformatics, Rhodes University, Makhanda, South Africa
| |
Collapse
|
5
|
Osorio OA, Kluender CE, Raphael HE, Hassan GF, Cohen LS, Steinberg D, Katz-Kiriakos E, Payne MD, Luo EM, Hicks JL, Byers DE, Alexander-Brett J. HSP70 chaperones IL-33 in chronic airway disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635799. [PMID: 39974890 PMCID: PMC11838530 DOI: 10.1101/2025.01.30.635799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
RATIONALE IL-33 is a key driver of type 2 inflammation relevant to airway epithelial biology. However, the mechanisms for IL-33 secretion and regulation in the context of chronic airway disease is poorly understood. OBJECTIVES We sought to define how a disease associated isoform IL-33d34 that escapes nuclear sequestration and is tonically secreted from epithelial cells can be recruited to non-canonical secretory pathways. METHODS IL-33d34 interaction with HSP70 was assessed and validated by affinity purification, mass-spectrometry and miniTurboID proximity labeling. Secretion and activity reporter assays were used to probe the effect of HSP70 on epithelial IL-33d34 secretion and receptor binding. Human airway disease biospecimens were analyzed for dysregulation of heat shock pathways revealing modulation of TCP1 complex intermediates. MEASUREMENTS AND MAIN RESULTS We confirmed that HSP70 interacts directly with IL-33d34, recruits the cytokine to a vesicular compartment and enhances stability upon secretion. IL-33, HSP70 and other key mediators of proteostasis were found to be dysregulated in airway disease biospecimens and secreted extracellular vesicles. The IL-33d34 interactome was characterized and novel secretion modulators were identified. CONCLUSIONS This study confirms a role for HSP70 in non-canonical IL-33d34 secretion and function that may be amenable for therapeutic targeting in airway diseases.
Collapse
|
6
|
Saarensilta A, Chen J, Reitzner SM, Hart DA, Ahmed AS, Ackermann PW. Novel tissue biomarker candidates to predict both deep venous thrombosis and healing outcome after Achilles tendon rupture. Sci Rep 2025; 15:7318. [PMID: 40025102 PMCID: PMC11873306 DOI: 10.1038/s41598-025-91511-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Deep venous thrombosis (DVT) and poor long-term patient outcomes frequently occur in patients with Achilles tendon rupture (ATR). Biomarkers for DVT and their possible relationship to long-term healing outcomes remain unexplored. To identify DVT biomarkers from proteomic profiles during the inflammatory and proliferative healing stages and assess their associations with one-year healing outcomes after surgical repair of ATR. A cohort of 53 patients undergoing standardized ATR repair from previous clinical trials was investigated. Intraoperative inflammatory-stage tendon biopsies were obtained from 40 patients, and tendon microdialysates from 28 patients were collected two weeks later during the proliferative stage. Liquid chromatography-tandem mass spectrometry proteomic profiles were linked to DVT status at two weeks post-surgery using ultrasonography screening and to patient-reported outcomes at one-year post-surgery. Six candidate DVT biomarkers were identified from tendon biopsies, whereof four (ABI3BP, IGKV2-40/IGKV2D-40, PCYOX1, STIP1) were associated with one-year healing outcomes. In tendon microdialysates, 43 candidate DVT biomarkers were identified, but none were associated with healing outcomes. Bioinformatic analysis revealed pathways related to heat shock response, platelet signaling, collagen and extracellular matrix metabolism, and immunoglobulins. The results support shared inflammatory-stage protein pathways in regulating venous thrombosis and reported healing outcomes, where elements of individual hypoxic tolerance and platelet signaling emerge as potential key links.
Collapse
Affiliation(s)
- Annukka Saarensilta
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
- Department of Molecular Medicine and Surgery, K1 MMK Orthopaedics, Stockholm, 17176, Sweden.
| | - Junyu Chen
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Orthopaedic Surgery, West China Hosptial of Sichuan University, No.37 Guoxue Lane, Chengdu, 610041, Sichuan, P. R. China
| | - Stefan Markus Reitzner
- Department of Physiology & Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - David A Hart
- Department of Surgery, Faculty of Kinesiology and the McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB, Canada
| | - Aisha S Ahmed
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Paul W Ackermann
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Trauma, Acute Surgery and Orthopaedics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
7
|
Makhoba XH. Two sides of the same coin: heat shock proteins as biomarkers and therapeutic targets for some complex diseases. Front Mol Biosci 2025; 12:1491227. [PMID: 40051500 PMCID: PMC11882428 DOI: 10.3389/fmolb.2025.1491227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/31/2025] [Indexed: 03/09/2025] Open
Abstract
Heat shock proteins are molecular chaperones that play crucial roles in the folding and unfolding of complex polypeptides within the cellular system. These molecules are involved in various processes, including vesicular transport, prevention of protein aggregation in the cytosol, and cell signaling. They are also linked to autoimmunity, infection immunity, and tumor immunology. Stressors like heat shock, exposure to heavy metals, cytokines, reactive oxygen species, inflammation, and viruses can influence the production of these molecules. In complex diseases such as cancer, malaria, and COVID-19, heat shock proteins are considered both biomarkers and drug targets. The upregulation of small heat shock proteins like hsp27 and major heat shock proteins 70/90 has been recognized as crucial biomarkers and therapeutic targets for cancer. Additionally, it has been reported that the invasion of Plasmodium falciparum, the causative agent of malaria, leads to the upregulation of heat shock proteins such as hsp40, hsp70, and hsp90. This sudden increase is a protective mechanism from the human host and enhances the parasite's growth, making these proteins significant as biomarkers and malarial drug targets. The presence of the SARS-CoV-2 virus in the human cellular system correlates with a substantial increase in heat shock protein 70 production from host cells. Furthermore, our research group has demonstrated that SARS-CoV-2 hijacks the host's heat shock proteins, and we are currently developing tools to prevent the virus from utilizing the host's protein folding system. This review aims to highlight the role of heat shock proteins as biomarkers and therapeutic targets for selected refractory diseases, focusing on cancer, malaria, and COVID-19. A fundamental molecular docking study was performed to investigate the interaction between a non-structural complex from SARS-CoV-2 and chosen small molecules, which is emphasized in this review.
Collapse
Affiliation(s)
- Xolani Henry Makhoba
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa (UNISA), Florida Campus, Roodepoort, South Africa
| |
Collapse
|
8
|
Pokhrel S, Devi S, Gestwicki JE. Chaperone-dependent and chaperone-independent functions of carboxylate clamp tetratricopeptide repeat (CC-TPR) proteins. Trends Biochem Sci 2025; 50:121-133. [PMID: 39706778 PMCID: PMC12066812 DOI: 10.1016/j.tibs.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/23/2024]
Abstract
The molecular chaperones HSP70 and HSP90 play key roles in proteostasis by acting as adapters; they bind to a 'client' protein, often with the assistance of cochaperones, and then recruit additional cochaperones that promote specific fates (e.g., folding or degradation). One family of cochaperones contains a region termed the tetratricopeptide repeat with carboxylate clamps (CC-TPRs) domain. These domains bind to an EEVD motif at the C-termini of cytoplasmic HSP70 and HSP90 proteins, bringing them into proximity to chaperone-bound clients. It has recently become clear that CC-TPR proteins also bind to 'EEVD-like' motifs in non-chaperone proteins, circumventing the need for HSP70s or HSP90s. We provide an overview of the chaperone-dependent and -independent roles of CC-TPR proteins and discuss how, together, they shape proteostasis.
Collapse
Affiliation(s)
- Saugat Pokhrel
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Shweta Devi
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco (UCSF), San Francisco, CA 94158, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry and the Institute for Neurodegenerative Diseases, University of California San Francisco (UCSF), San Francisco, CA 94158, USA.
| |
Collapse
|
9
|
Akaree N, Secco V, Levy-Adam F, Younis A, Carra S, Shalgi R. Regulation of physiological and pathological condensates by molecular chaperones. FEBS J 2025. [PMID: 39756021 DOI: 10.1111/febs.17390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/17/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Biomolecular condensates are dynamic membraneless compartments that regulate a myriad of cellular functions. A particular type of physiological condensate called stress granules (SGs) has gained increasing interest due to its role in the cellular stress response and various diseases. SGs, composed of several hundred RNA-binding proteins, form transiently in response to stress to protect mRNAs from translation and disassemble when the stress subsides. Interestingly, SGs contain several aggregation-prone proteins, such as TDP-43, FUS, hnRNPA1, and others, which are typically found in pathological inclusions seen in autopsy tissues from amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) patients. Moreover, mutations in these genes lead to the familial form of ALS and FTD. This has led researchers to propose that pathological aggregation is seeded by aberrant SGs: SGs that fail to properly disassemble, lose their dynamic properties, and become pathological condensates which finally 'mature' into aggregates. Here, we discuss the evidence supporting this model for various ALS/FTD-associated proteins. We further continue to focus on molecular chaperone-mediated regulation of ALS/FTD-associated physiological condensates on one hand, and pathological condensates on the other. In addition to SGs, we review ALS/FTD-relevant nuclear condensates, namely paraspeckles, anisosomes, and nucleolar amyloid bodies, and discuss their emerging regulation by chaperones. As the majority of chaperoning mechanisms regulate physiological condensate disassembly, we highlight parallel themes of physiological and pathological condensation regulation across different chaperone families, underscoring the potential for early disease intervention.
Collapse
Affiliation(s)
- Nadeen Akaree
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Valentina Secco
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Flonia Levy-Adam
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amal Younis
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Reut Shalgi
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
10
|
Silbermann LM, Vermeer B, Schmid S, Tych K. The known unknowns of the Hsp90 chaperone. eLife 2024; 13:e102666. [PMID: 39737863 PMCID: PMC11687934 DOI: 10.7554/elife.102666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/17/2024] [Indexed: 01/01/2025] Open
Abstract
Molecular chaperones are vital proteins that maintain protein homeostasis by assisting in protein folding, activation, degradation, and stress protection. Among them, heat-shock protein 90 (Hsp90) stands out as an essential proteostasis hub in eukaryotes, chaperoning hundreds of 'clients' (substrates). After decades of research, several 'known unknowns' about the molecular function of Hsp90 remain unanswered, hampering rational drug design for the treatment of cancers, neurodegenerative, and other diseases. We highlight three fundamental open questions, reviewing the current state of the field for each, and discuss new opportunities, including single-molecule technologies, to answer the known unknowns of the Hsp90 chaperone.
Collapse
Affiliation(s)
- Laura-Marie Silbermann
- Groningen Biomolecular Sciences and Biotechnology Institute, University of GroningenGroningenNetherlands
| | - Benjamin Vermeer
- Laboratory of Biophysics, Wageningen University & ResearchWageningenNetherlands
| | - Sonja Schmid
- Laboratory of Biophysics, Wageningen University & ResearchWageningenNetherlands
| | - Katarzyna Tych
- Groningen Biomolecular Sciences and Biotechnology Institute, University of GroningenGroningenNetherlands
| |
Collapse
|
11
|
Rutledge BS, Kim YJ, McDonald DW, Jurado-Coronel JC, Prado MAM, Johnson JL, Choy WY, Duennwald ML. Stress-inducible phosphoprotein 1 (Sti1/Stip1/Hop) sequesters misfolded proteins during stress. FEBS J 2024. [PMID: 39739753 DOI: 10.1111/febs.17389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/16/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
Co-chaperones are key elements of cellular protein quality control. They cooperate with the major heat shock proteins Hsp70 and Hsp90 in folding proteins and preventing the toxic accumulation of misfolded proteins upon exposure to stress. Hsp90 interacts with the co-chaperone stress-inducible phosphoprotein 1 (Sti1/Stip1/Hop) and activator of Hsp90 ATPase protein 1 (Aha1) among many others. Sti1 and Aha1 control the ATPase activity of Hsp90, but Sti1 also facilitates the transfer of client proteins from Hsp70 to Hsp90, thus connecting these two major branches of protein quality control. We find that misbalanced expression of Sti1 and Aha1 in yeast and mammalian cells causes severe growth defects. Also, deletion of STI1 causes an accumulation of soluble misfolded ubiquitinated proteins and a strong activation of the heat shock response. We discover that, during proteostatic stress, Sti1 forms cytoplasmic inclusions in yeast and mammalian cells that overlap with misfolded proteins. Our work indicates a key role of Sti1 in proteostasis independent of its Hsp90 ATPase regulatory functions by sequestering misfolded proteins during stress.
Collapse
Affiliation(s)
- Benjamin S Rutledge
- Department of Biochemistry, The University of Western Ontario, London, Canada
| | - Young J Kim
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Canada
| | - Donovan W McDonald
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Canada
| | - Juan C Jurado-Coronel
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Canada
| | - Marco A M Prado
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Canada
- Robarts Research Institute and Department of Physiology and Pharmacology, The University of Western Ontario, London, Canada
| | - Jill L Johnson
- Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| | - Wing-Yiu Choy
- Department of Biochemistry, The University of Western Ontario, London, Canada
| | - Martin L Duennwald
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Canada
| |
Collapse
|
12
|
Garcia-Toscano L, Currey HN, Hincks JC, Stair JG, Lehrbach NJ, Liachko NF. Decreased Hsp90 activity protects against TDP-43 neurotoxicity in a C. elegans model of amyotrophic lateral sclerosis. PLoS Genet 2024; 20:e1011518. [PMID: 39724103 PMCID: PMC11709271 DOI: 10.1371/journal.pgen.1011518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 01/08/2025] [Accepted: 11/27/2024] [Indexed: 12/28/2024] Open
Abstract
Neuronal inclusions of hyperphosphorylated TDP-43 are hallmarks of disease for most patients with amyotrophic lateral sclerosis (ALS). Mutations in TARDBP, the gene coding for TDP-43, can cause some cases of familial inherited ALS (fALS), indicating dysfunction of TDP-43 drives disease. Aggregated, phosphorylated TDP-43 may contribute to disease phenotypes; alternatively, TDP-43 aggregation may be a protective cellular response sequestering toxic protein away from the rest of the cell. The heat shock responsive chaperone Hsp90 has been shown to interact with TDP-43 and stabilize its normal conformation; however, it is not known whether this interaction contributes to neurotoxicity in vivo. Using a C. elegans model of fALS mutant TDP-43 proteinopathy, we find that loss of function of HSP-90 protects against TDP-43 neurotoxicity and subsequent neurodegeneration in adult animals. This protection is accompanied by a decrease in both total and phosphorylated TDP-43 protein. We also find that hsp-90 mutation or inhibition upregulates key stress responsive heat shock pathway gene expression, including hsp-70 and hsp-16.1, and we demonstrate that normal levels of hsp-16.1 are required for hsp-90 mutation effects on TDP-43. We also observe that the neuroprotective effect due to HSP-90 dysfunction does not involve direct regulation of proteasome activity in C. elegans. Our data demonstrate for the first time that Hsp90 chaperone activity contributes to adverse outcomes in TDP-43 proteinopathies in vivo using a whole animal model of ALS.
Collapse
Affiliation(s)
- Laura Garcia-Toscano
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, United States of America
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Heather N. Currey
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, United States of America
| | - Joshua C. Hincks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, United States of America
| | - Jade G. Stair
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, United States of America
| | - Nicolas J. Lehrbach
- Basic Sciences Division, Fred Hutch Cancer Center, Seattle, Washington, United States of America
| | - Nicole F. Liachko
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, United States of America
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
13
|
Kärkkäinen V, Hannonen S, Rusanen M, Lehtola JM, Saari T, Uusitalo H, Leinonen V, Thiede B, Kaarniranta K, Koivisto AM, Utheim TP. Tear fluid reflects the altered protein expressions of Alzheimer's disease patients in proteins involved in protein repair and clearance system or the regulation of cytoskeleton. J Alzheimers Dis 2024:13872877241295315. [PMID: 39558606 DOI: 10.1177/13872877241295315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
BACKGROUND New biomarkers that improve diagnosis of Alzheimer's disease (AD) are warranted. Tear fluid (TF) containing variety of proteins that reflect pathophysiological changes of systemic diseases makes TF proteins potential biomarker candidates for AD. OBJECTIVE We investigated the expression levels of TF proteins in persons with mild AD and cognitively healthy controls (CO) to find out if altered proteins may link to the AD pathophysiology. METHODS We analyzed the data of the 53 study participants (34 COs, mean age 71 and Mini-Mental State Examination (MMSE) 28.9 ± 1.4 and 19 persons with AD, CDR 0.5-1, mean age 71 and MMSE 23.8 ± 2.8). All went through neurological status examination, cognitive tests, and ophthalmological examination. TF was collected using Schirmer strips. The TF protein content was evaluated via mass spectrometry-based proteomics and label-free quantification. RESULTS Eleven proteins having a role either in protein repair and clearance system, or regulation of cytoskeleton, showed altered expression in AD group compared to CO group. Seven of them were significantly (p ≤ 0.05) upregulated (Sti1, Twf1, Myl6, Otub1, Pls1 and Caza1) or, downregulated (HSP90) in AD group. CONCLUSIONS Altered expression of all these up- or downregulated proteins may be linked to AD pathophysiology. Thus, our results are encouraging for searching new biomarker candidates for AD. TF is potential biomarker candidate, because TF seems to reflect altered protein levels already in mild AD dementia.
Collapse
Affiliation(s)
- Virve Kärkkäinen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- NeuroCenter, Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Sanna Hannonen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Minna Rusanen
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Juha-Matti Lehtola
- Neurology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Toni Saari
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Hannu Uusitalo
- Faculty of Medicine and Health Technology, Eye and Vision Research, Tampere University, Tampere, Finland
| | - Ville Leinonen
- NeuroCenter, Neurosurgery, Kuopio University Hospital, Kuopio, Finland
- Neurosurgery, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Bernd Thiede
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, School of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Anne M Koivisto
- NeuroCenter, Neurology, Kuopio University Hospital, Kuopio, Finland
- Department of Geriatrics, Helsinki University Hospital and Department of Neurosciences, University of Helsinki, Helsinki, Finland
| | - Tor Paaske Utheim
- Faculty of Dentistry, Institute of Oral Biology, University of Oslo, Oslo, Norway|
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
14
|
Okpara M, Vaaltyn MC, Watson JL, Alhassan M, Albericio F, de la Torre BG, Clarke DJ, Veale CGL, Edkins AL. Modulators of the Hop-HSP90 Protein-Protein Interaction Disrupt KSHV Lytic Replication. ACS Infect Dis 2024; 10:3853-3867. [PMID: 39475219 PMCID: PMC11555673 DOI: 10.1021/acsinfecdis.4c00429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/09/2024]
Abstract
The central role of the chaperome in maintaining cellular proteostasis has seen numerous viral families evolve to parasitically exploit host chaperones in their life cycle. The HSP90 chaperone protein and its cochaperone Hop have both individually been shown to be essential factors for Kaposi sarcoma-associated herpesvirus (KSHV) lytic replication. Given the fundamental regulatory role that protein-protein interactions (PPIs) play in cellular biology, we reasoned that disrupting the Hop-HSP90 PPI may provide a new host-based target for inhibiting KSHV lytic replication. This study expands upon a previous report of non-natural peptides, which were found to disrupt the association between the HopTPR2A domain and its interacting HSP90CTD. Here, in addition to providing insight into the structure-activity relationships of PPI inhibition, we show disruption of the full-length Hop-HSP90 PPI. The inhibitory peptides selectively engaged the HopTPR2A domain in cell lysates and when tethered to a cell-penetrating peptide acted as noncytotoxic inhibitors of KSHV lytic replication by lowering the viral load, preventing the production of infectious virions, and reducing the expression of KSHV lytic genes. In addition to tentative evidence of Hop-HSP90 PPI as a much-needed target for KSHV drug discovery, this study represents an important step in understanding viral interactions with the host proteostasis machinery.
Collapse
Affiliation(s)
- Michael
O. Okpara
- Biomedical
Biotechnology Research Unit (BioBRU), Department of Biochemistry and
Microbiology, Rhodes University, Makhanda 6139, South Africa
| | - Michaelone C. Vaaltyn
- Biomedical
Biotechnology Research Unit (BioBRU), Department of Biochemistry and
Microbiology, Rhodes University, Makhanda 6139, South Africa
| | - Jessica L. Watson
- Biomedical
Biotechnology Research Unit (BioBRU), Department of Biochemistry and
Microbiology, Rhodes University, Makhanda 6139, South Africa
| | - Mahama Alhassan
- School
of Chemistry and Physics, University of
Kwa-Zulu Natal, Durban, Westville 4001, South Africa
| | - Fernando Albericio
- School
of Chemistry and Physics, University of
Kwa-Zulu Natal, Durban, Westville 4001, South Africa
| | - Beatriz G. de la Torre
- School
of Laboratory Medicine and Medical Sciences, University of Kwa-Zulu Natal, Durban 4041, South Africa
| | - David J. Clarke
- EaStCHEM,
School of Chemistry, University of Edinburgh, Joseph Black Building, David Brewster
Road, Edinburgh EH93FJ, United Kingdom
| | - Clinton G. L. Veale
- Department
of Chemistry, University of Cape Town, Rondebosch, Cape Town 7701, South Africa
| | - Adrienne L. Edkins
- Biomedical
Biotechnology Research Unit (BioBRU), Department of Biochemistry and
Microbiology, Rhodes University, Makhanda 6139, South Africa
| |
Collapse
|
15
|
Kirigin E, Okpara MO, Matandirotya L, Ruck JL, Weaver F, Jackson Z, Chakraborty A, Veale CGL, Whitehouse A, Edkins AL. Hsp70-Hsp90 organising protein (HOP/STIP1) is required for KSHV lytic replication. J Gen Virol 2024; 105. [PMID: 39607759 DOI: 10.1099/jgv.0.002053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is a DNA virus that causes Kaposi's sarcoma, a cancer of endothelial origin. KSHV uses the activity of host molecular chaperones like Hsp70 and Hsp90 for the folding of host and viral proteins required for productive infection. Hsp70 and Hsp90 chaperones form proteostasis networks with several regulatory proteins known as co-chaperones. Of these, Hsp90-Hsp70-organizing protein (HOP) is an early-stage co-chaperone that regulates the transfer of folding substrate proteins between the Hsp70 and Hsp90 chaperone systems. While the roles for Hsp90 and Hsp70 in KSHV biology have been described, HOP has not previously been studied in this context despite its prominent interaction with both chaperones. Here, we demonstrate a novel function for HOP as a new host factor required for effective lytic replication of KSHV in primary effusion cell lines.
Collapse
Affiliation(s)
- Elisa Kirigin
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6139, South Africa
| | - Michael Obinna Okpara
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6139, South Africa
| | - Lorraine Matandirotya
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6139, South Africa
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jamie-Lee Ruck
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6139, South Africa
| | - Frederick Weaver
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Zoe Jackson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Abir Chakraborty
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6139, South Africa
| | | | - Adrian Whitehouse
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6139, South Africa
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Adrienne Lesley Edkins
- Biomedical Biotechnology Research Unit (BioBRU), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6139, South Africa
- Centre for Chemico- and Biomedicinal Research (CCBR), Rhodes University, Makhanda, 6139, South Africa
| |
Collapse
|
16
|
Tanaka Y, Farkhondeh A, Yang W, Ueno H, Noda M, Hirokawa N. Kinesin-1 mediates proper ER folding of the Ca V1.2 channel and maintains mouse glucose homeostasis. EMBO Rep 2024; 25:4777-4802. [PMID: 39322740 PMCID: PMC11549326 DOI: 10.1038/s44319-024-00246-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 07/08/2024] [Accepted: 08/22/2024] [Indexed: 09/27/2024] Open
Abstract
Glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells is a principal mechanism for systemic glucose homeostasis, of which regulatory mechanisms are still unclear. Here we show that kinesin molecular motor KIF5B is essential for GSIS through maintaining the voltage-gated calcium channel CaV1.2 levels, by facilitating an Hsp70-to-Hsp90 chaperone exchange to pass through the quality control in the endoplasmic reticulum (ER). Phenotypic analyses of KIF5B conditional knockout (cKO) mouse beta cells revealed significant abolishment of glucose-stimulated calcium transients, which altered the behaviors of insulin granules via abnormally stabilized cortical F-actin. KIF5B and Hsp90 colocalize to microdroplets on ER sheets, where CaV1.2 but not Kir6.2 is accumulated. In the absence of KIF5B, CaV1.2 fails to be transferred from Hsp70 to Hsp90 via STIP1, and is likely degraded via the proteasomal pathway. KIF5B and Hsc70 overexpression increased CaV1.2 expression via enhancing its chaperone binding. Thus, ER sheets may serve as the place of KIF5B- and Hsp90-dependent chaperone exchange, which predominantly facilitates CaV1.2 production in beta cells and properly enterprises GSIS against diabetes.
Collapse
Affiliation(s)
- Yosuke Tanaka
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan
| | - Atena Farkhondeh
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan
| | - Wenxing Yang
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan
| | - Hitoshi Ueno
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan
| | - Mitsuhiko Noda
- Department of Diabetes, Metabolism and Endocrinology, Ichikawa Hospital, International University of Health and Welfare, Chiba, 272-0827, Japan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of Medicine, The University of Tokyo, Hongo, Tokyo, 113-0033, Japan.
- Department of Advanced Morphological Imaging, Graduate School of Medicine, Juntendo University, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| |
Collapse
|
17
|
Toribio R, Navarro A, Castellano MM. HOP stabilizes the HSFA1a and plays a main role in the onset of thermomorphogenesis. PLANT, CELL & ENVIRONMENT 2024; 47:4449-4463. [PMID: 39007522 DOI: 10.1111/pce.15036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Living organisms have the capacity to respond to environmental stimuli, including warm conditions. Upon sensing mild temperature, plants launch a transcriptional response that promotes morphological changes, globally known as thermomorphogenesis. This response is orchestrated by different hormonal networks and by the activity of different transcription factors, including the heat shock factor A1 (HSFA1) family. Members of this family interact with heat shock protein 70 (HSP70) and heat shock protein 90 (HSP90); however, the effect of this binding on the regulation of HSFA1 activity or of the role of cochaperones, such as the HSP70-HSP90 organizing protein (HOP) on HSFA1 regulation, remains unknown. Here, we show that AtHOPs are involved in the folding and stabilization of the HSFA1a and are required for the onset of the transcriptional response associated to thermomorphogenesis. Our results demonstrate that the three members of the AtHOP family bind in vivo to the HSFA1a and that the expression of multiple HSFA1a-responsive-responsive genes is altered in the hop1 hop2 hop3 mutant under warm temperature. Interestingly, HSFA1a is accumulated at lower levels in the hop1 hop2 hop3 mutant, while control levels are recovered in the presence of the proteasome inhibitor MG132 or the synthetic chaperone tauroursodeoxycholic acid (TUDCA). This uncovers the HSFA1a as a client of HOP complexes in plants and reveals the participation of HOPs in HSFA1a stability.
Collapse
Affiliation(s)
- René Toribio
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-CSIC (INIA/CSIC), Campus de Montegancedo, Pozuelo de Alarcón, Madrid, Spain
| | - Aurora Navarro
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-CSIC (INIA/CSIC), Campus de Montegancedo, Pozuelo de Alarcón, Madrid, Spain
| | - M Mar Castellano
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria-CSIC (INIA/CSIC), Campus de Montegancedo, Pozuelo de Alarcón, Madrid, Spain
| |
Collapse
|
18
|
Stewart M, Schisler JC. Targeting chaperone modifications: Innovative approaches to cancer treatment. J Biol Chem 2024; 300:107907. [PMID: 39433125 PMCID: PMC11599458 DOI: 10.1016/j.jbc.2024.107907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024] Open
Abstract
Cancer and other chronic diseases are marked by alterations in the protein quality control system, affecting the posttranslational destiny of various proteins that regulate, structure, and catalyze cellular processes. Cellular chaperones, also known as heat shock proteins (HSPs), are pivotal in this system, performing protein triage that often determines the fate of proteins they bind to. Grasping the regulatory mechanisms of HSPs and their associated cofactors is crucial for understanding protein quality control in both healthy and diseased states. Recent research has shed light on the interactions within the protein quality control system and how post-translational modification govern protein interactions, function, and localization, which can drive or inhibit cell proliferation. This body of work encompasses critical elements of the heat shock response, including heat shock protein 70, heat shock protein 90, carboxyl-terminus of HSC70 interacting protein, and heat shock protein organizing protein. This review aims to synthesize these advancements, offering a holistic understanding of the system and its response when commandeered by diseases like cancer. We focus on the mechanistic shift in co-chaperone engagement-transitioning from heat shock protein organizing protein to carboxyl-terminus of HSC70 interacting protein in association with heat shock protein 70 and heat shock protein 90-which could influence cellular growth and survival pathways. A comprehensive examination of posttranslational modification-driven regulation within the protein quality control network is presented, highlighting the roles of activation factors, chaperones, and co-chaperones. Our insights aim to inform new strategies for therapeutically targeting diseases by considering the entire heat shock response system.
Collapse
Affiliation(s)
- Mariah Stewart
- The McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jonathan C Schisler
- The McAllister Heart Institute and Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; The Department of Pathology and Lab Medicine and Computational Medicine Program, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| |
Collapse
|
19
|
Riedl S, Bilgen E, Agam G, Hirvonen V, Jussupow A, Tippl F, Riedl M, Maier A, Becker CFW, Kaila VRI, Lamb DC, Buchner J. Evolution of the conformational dynamics of the molecular chaperone Hsp90. Nat Commun 2024; 15:8627. [PMID: 39366960 PMCID: PMC11452706 DOI: 10.1038/s41467-024-52995-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 09/25/2024] [Indexed: 10/06/2024] Open
Abstract
Hsp90 is a molecular chaperone of central importance for protein homeostasis in the cytosol of eukaryotic cells, with key functional and structural traits conserved from yeast to man. During evolution, Hsp90 has gained additional functional importance, leading to an increased number of interacting co-chaperones and client proteins. Here, we show that the overall conformational transitions coupled to the ATPase cycle of Hsp90 are conserved from yeast to humans, but cycle timing as well as the dynamics are significantly altered. In contrast to yeast Hsp90, the human Hsp90 is characterized by broad ensembles of conformational states, irrespective of the absence or presence of ATP. The differences in the ATPase rate and conformational transitions between yeast and human Hsp90 are based on two residues in otherwise conserved structural elements that are involved in triggering structural changes in response to ATP binding. The exchange of these two mutations allows swapping of the ATPase rate and of the conformational transitions between human and yeast Hsp90. Our combined results show that Hsp90 evolved to a protein with increased conformational dynamics that populates ensembles of different states with strong preferences for the N-terminally open, client-accepting states.
Collapse
Affiliation(s)
- Stefan Riedl
- Center for Protein Assemblies, Department Bioscience, School of Natural Sciences, Technical University Munich, Garching, Germany
| | - Ecenaz Bilgen
- Department of Chemistry and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Ganesh Agam
- Department of Chemistry and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Viivi Hirvonen
- Department of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Alexander Jussupow
- Department of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Franziska Tippl
- Center for Protein Assemblies, Department Bioscience, School of Natural Sciences, Technical University Munich, Garching, Germany
| | - Maximilian Riedl
- Center for Protein Assemblies, Department Bioscience, School of Natural Sciences, Technical University Munich, Garching, Germany
| | - Andreas Maier
- Center for Protein Assemblies, Department Bioscience, School of Natural Sciences, Technical University Munich, Garching, Germany
| | - Christian F W Becker
- Institute of Biological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Ville R I Kaila
- Department of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural Sciences, Stockholm University, Stockholm, Sweden
| | - Don C Lamb
- Department of Chemistry and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | - Johannes Buchner
- Center for Protein Assemblies, Department Bioscience, School of Natural Sciences, Technical University Munich, Garching, Germany.
| |
Collapse
|
20
|
Castellano MM, Muñoz A, Okeke IC, Novo-Uzal E, Toribio R, Mangano S. The role of the co-chaperone HOP in plant homeostasis during development and stress. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:4274-4286. [PMID: 38330220 PMCID: PMC11263486 DOI: 10.1093/jxb/erae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/06/2024] [Indexed: 02/10/2024]
Abstract
Proteins need to acquire their native structure in order to become fully functional. In specific cases, the active conformation is obtained spontaneously; nevertheless, many proteins need the assistance of chaperones and co-chaperones to be properly folded. These proteins help to maintain protein homeostasis under control conditions and under different stresses. HOP (HSP70-HSP90 organizing protein) is a highly conserved family of co-chaperones that assist HSP70 and HSP90 in the folding of specific proteins. In the last few years, findings in mammals and yeast have revealed novel functions of HOP and re-defined the role of HOP in protein folding. Here, we provide an overview of the most important aspects of HOP regulation and function in other eukaryotes and analyse whether these aspects are conserved in plants. In addition, we highlight the HOP clients described in plants and the role of HOP in plant development and stress response.
Collapse
Affiliation(s)
- M Mar Castellano
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223-Pozuelo de Alarcón, Madrid, Spain
| | - Alfonso Muñoz
- Departamento de Sistemas y Recursos Naturales, ETSI de Montes, Forestal y del Medio Natural, Universidad Politécnica de Madrid, 28040-Madrid, Spain
| | - Isabel C Okeke
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223-Pozuelo de Alarcón, Madrid, Spain
| | - Esther Novo-Uzal
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223-Pozuelo de Alarcón, Madrid, Spain
| | - René Toribio
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223-Pozuelo de Alarcón, Madrid, Spain
| | - Silvina Mangano
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM)-Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223-Pozuelo de Alarcón, Madrid, Spain
- INTECH, CONICET-UNSAM Avda. Intendente Marino KM 8.2, (7130), Chascomús, Provincia de Buenos Aires, Argentina
| |
Collapse
|
21
|
Wickramaratne AC, Wickner S, Kravats AN. Hsp90, a team player in protein quality control and the stress response in bacteria. Microbiol Mol Biol Rev 2024; 88:e0017622. [PMID: 38534118 PMCID: PMC11332350 DOI: 10.1128/mmbr.00176-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024] Open
Abstract
SUMMARYHeat shock protein 90 (Hsp90) participates in proteostasis by facilitating protein folding, activation, disaggregation, prevention of aggregation, degradation, and protection against degradation of various cellular proteins. It is highly conserved from bacteria to humans. In bacteria, protein remodeling by Hsp90 involves collaboration with the Hsp70 molecular chaperone and Hsp70 cochaperones. In eukaryotes, protein folding by Hsp90 is more complex and involves collaboration with many Hsp90 cochaperones as well as Hsp70 and Hsp70 cochaperones. This review focuses primarily on bacterial Hsp90 and highlights similarities and differences between bacterial and eukaryotic Hsp90. Seminal research findings that elucidate the structure and the mechanisms of protein folding, disaggregation, and reactivation promoted by Hsp90 are discussed. Understanding the mechanisms of bacterial Hsp90 will provide fundamental insight into the more complex eukaryotic chaperone systems.
Collapse
Affiliation(s)
- Anushka C. Wickramaratne
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sue Wickner
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrea N. Kravats
- Department of Chemistry and Biochemistry, Miami University, Oxford, Ohio, USA
| |
Collapse
|
22
|
Xing Z, Jiang H, Liu X, Chai Q, Xin Z, Zhu C, Bao Y, Chen H, Gao H, Ma D. Integrating DNA/RNA microbe detection and host response for accurate diagnosis, treatment and prognosis of childhood infectious meningitis and encephalitis. J Transl Med 2024; 22:583. [PMID: 38902725 PMCID: PMC11191231 DOI: 10.1186/s12967-024-05370-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 06/02/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Infectious meningitis/encephalitis (IM) is a severe neurological disease that can be caused by bacterial, viral, and fungal pathogens. IM suffers high morbidity, mortality, and sequelae in childhood. Metagenomic next-generation sequencing (mNGS) can potentially improve IM outcomes by sequencing both pathogen and host responses and increasing the diagnosis accuracy. METHODS Here we developed an optimized mNGS pipeline named comprehensive mNGS (c-mNGS) to monitor DNA/RNA pathogens and host responses simultaneously and applied it to 142 cerebrospinal fluid samples. According to retrospective diagnosis, these samples were classified into three categories: confirmed infectious meningitis/encephalitis (CIM), suspected infectious meningitis/encephalitis (SIM), and noninfectious controls (CTRL). RESULTS Our pipeline outperformed conventional methods and identified RNA viruses such as Echovirus E30 and etiologic pathogens such as HHV-7, which would not be clinically identified via conventional methods. Based on the results of the c-mNGS pipeline, we successfully detected antibiotic resistance genes related to common antibiotics for treating Escherichia coli, Acinetobacter baumannii, and Group B Streptococcus. Further, we identified differentially expressed genes in hosts of bacterial meningitis (BM) and viral meningitis/encephalitis (VM). We used these genes to build a machine-learning model to pinpoint sample contaminations. Similarly, we also built a model to predict poor prognosis in BM. CONCLUSIONS This study developed an mNGS-based pipeline for IM which measures both DNA/RNA pathogens and host gene expression in a single assay. The pipeline allows detecting more viruses, predicting antibiotic resistance, pinpointing contaminations, and evaluating prognosis. Given the comparable cost to conventional mNGS, our pipeline can become a routine test for IM.
Collapse
Affiliation(s)
- Zhihao Xing
- Biobank & Clinical laboratory & Department of Respiratory Medicine, Shenzhen Children's Hospital of Shantou University Medical College, Shenzhen, Guangdong, China
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Hanfang Jiang
- Clinical laboratory, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Xiaorong Liu
- Biobank & Clinical laboratory & Department of Respiratory Medicine, Shenzhen Children's Hospital of Shantou University Medical College, Shenzhen, Guangdong, China
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Qiang Chai
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Zefeng Xin
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Chunqing Zhu
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
- Clinical laboratory, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Yanmin Bao
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Hongyu Chen
- Clinical laboratory, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Hongdan Gao
- Medical Testing, Bengbu Medical College, Bengbu, Anhui, China
| | - Dongli Ma
- Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
23
|
Mansfield CR, Quan B, Chirgwin ME, Eduful B, Hughes PF, Neveu G, Sylvester K, Ryan DH, Kafsack BFC, Haystead TAJ, Leahy JW, Fitzgerald MC, Derbyshire ER. Selective targeting of Plasmodium falciparum Hsp90 disrupts the 26S proteasome. Cell Chem Biol 2024; 31:729-742.e13. [PMID: 38492573 PMCID: PMC11031320 DOI: 10.1016/j.chembiol.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The molecular chaperone heat shock protein 90 (Hsp90) has an essential but largely undefined role in maintaining proteostasis in Plasmodium falciparum, the most lethal malaria parasite. Herein, we identify BX-2819 and XL888 as potent P. falciparum (Pf)Hsp90 inhibitors. Derivatization of XL888's scaffold led to the development of Tropane 1, as a PfHsp90-selective binder with nanomolar affinity. Hsp90 inhibitors exhibit anti-Plasmodium activity against the liver, asexual blood, and early gametocyte life stages. Thermal proteome profiling was implemented to assess PfHsp90-dependent proteome stability, and the proteasome-the main site of cellular protein recycling-was enriched among proteins with perturbed stability upon PfHsp90 inhibition. Subsequent biochemical and cellular studies suggest that PfHsp90 directly promotes proteasome hydrolysis by chaperoning the active 26S complex. These findings expand our knowledge of the PfHsp90-dependent proteome and protein quality control mechanisms in these pathogenic parasites, as well as further characterize this chaperone as a potential antimalarial drug target.
Collapse
Affiliation(s)
- Christopher R Mansfield
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Baiyi Quan
- Department of Chemistry, Duke University, Durham, NC, USA
| | | | - Benjamin Eduful
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Gaëlle Neveu
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Daniel H Ryan
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Björn F C Kafsack
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - James W Leahy
- Department of Chemistry, University of South Florida, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Center for Drug Discovery and Innovation, University of South Florida, Tampa, FL, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
24
|
San Gil R, Pascovici D, Venturato J, Brown-Wright H, Mehta P, Madrid San Martin L, Wu J, Luan W, Chui YK, Bademosi AT, Swaminathan S, Naidoo S, Berning BA, Wright AL, Keating SS, Curtis MA, Faull RLM, Lee JD, Ngo ST, Lee A, Morsch M, Chung RS, Scotter E, Lisowski L, Mirzaei M, Walker AK. A transient protein folding response targets aggregation in the early phase of TDP-43-mediated neurodegeneration. Nat Commun 2024; 15:1508. [PMID: 38374041 PMCID: PMC10876645 DOI: 10.1038/s41467-024-45646-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/31/2024] [Indexed: 02/21/2024] Open
Abstract
Understanding the mechanisms that drive TDP-43 pathology is integral to combating amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD) and other neurodegenerative diseases. Here we generated a longitudinal quantitative proteomic map of the cortex from the cytoplasmic TDP-43 rNLS8 mouse model of ALS and FTLD, and developed a complementary open-access webtool, TDP-map ( https://shiny.rcc.uq.edu.au/TDP-map/ ). We identified distinct protein subsets enriched for diverse biological pathways with temporal alterations in protein abundance, including increases in protein folding factors prior to disease onset. This included increased levels of DnaJ homolog subfamily B member 5, DNAJB5, which also co-localized with TDP-43 pathology in diseased human motor cortex. DNAJB5 over-expression decreased TDP-43 aggregation in cell and cortical neuron cultures, and knockout of Dnajb5 exacerbated motor impairments caused by AAV-mediated cytoplasmic TDP-43 expression in mice. Together, these findings reveal molecular mechanisms at distinct stages of ALS and FTLD progression and suggest that protein folding factors could be protective in neurodegenerative diseases.
Collapse
Affiliation(s)
- Rebecca San Gil
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Dana Pascovici
- Insight Stats, Croydon Park, NSW, Australia
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, NSW, Australia
| | - Juliana Venturato
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Heledd Brown-Wright
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Prachi Mehta
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
- Motor Neuron Disease Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Lidia Madrid San Martin
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Jemma Wu
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, NSW, Australia
| | - Wei Luan
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Yi Kit Chui
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Adekunle T Bademosi
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Shilpa Swaminathan
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Serey Naidoo
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Britt A Berning
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Amanda L Wright
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Sean S Keating
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Maurice A Curtis
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Shyuan T Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Albert Lee
- Motor Neuron Disease Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Marco Morsch
- Motor Neuron Disease Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Roger S Chung
- Motor Neuron Disease Research Centre, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Emma Scotter
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Leszek Lisowski
- Vector and Genome Engineering Facility, Children's Medical Research Institute, Westmead, NSW, Australia
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine - National Research Institute, Warsaw, Poland
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde Sydney, NSW, Australia
| | - Adam K Walker
- Neurodegeneration Pathobiology Laboratory, Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
25
|
Castelli M, Magni A, Bonollo G, Pavoni S, Frigerio F, Oliveira ASF, Cinquini F, Serapian SA, Colombo G. Molecular mechanisms of chaperone-directed protein folding: Insights from atomistic simulations. Protein Sci 2023; 33:e4880. [PMID: 38145386 PMCID: PMC10895457 DOI: 10.1002/pro.4880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Molecular chaperones, a family of proteins of which Hsp90 and Hsp70 are integral members, form an essential machinery to maintain healthy proteomes by controlling the folding and activation of a plethora of substrate client proteins. This is achieved through cycles in which Hsp90 and Hsp70, regulated by task-specific co-chaperones, process ATP and become part of a complex network that undergoes extensive compositional and conformational variations. Despite impressive advances in structural knowledge, the mechanisms that regulate the dynamics of functional assemblies, their response to nucleotides, and their relevance for client remodeling are still elusive. Here, we focus on the glucocorticoid receptor (GR):Hsp90:Hsp70:co-chaperone Hop client-loading and the GR:Hsp90:co-chaperone p23 client-maturation complexes, key assemblies in the folding cycle of glucocorticoid receptor (GR), a client strictly dependent upon Hsp90/Hsp70 for activity. Using a combination of molecular dynamics simulation approaches, we unveil with unprecedented detail the mechanisms that underpin function in these chaperone machineries. Specifically, we dissect the processes by which the nucleotide-encoded message is relayed to the client and how the distinct partners of the assemblies cooperate to (pre)organize partially folded GR during Loading and Maturation. We show how different ligand states determine distinct dynamic profiles for the functional interfaces defining the interactions in the complexes and modulate their overall flexibility to facilitate progress along the chaperone cycle. Finally, we also show that the GR regions engaged by the chaperone machinery display peculiar energetic signatures in the folded state, which enhance the probability of partial unfolding fluctuations. From these results, we propose a model where a dynamic cross-talk emerges between the chaperone dynamics states and remodeling of client-interacting regions. This factor, coupled to the highly dynamic nature of the assemblies and the conformational heterogeneity of their interactions, provides the basis for regulating the functions of distinct assemblies during the chaperoning cycle.
Collapse
Affiliation(s)
| | - Andrea Magni
- Dipartimento di Chimica, Università di Pavia, Pavia, Italy
| | | | - Silvia Pavoni
- Department of Physical Chemistry, R&D Eni SpA, San Donato Milanese, Italy
| | - Francesco Frigerio
- Department of Physical Chemistry, R&D Eni SpA, San Donato Milanese, Italy
| | - A Sofia F Oliveira
- Centre for Computational Chemistry, School of Chemistry, University of Bristol, Bristol, UK
| | - Fabrizio Cinquini
- Upstream & Technical Services - TECS/STES - Eni Spa, San Donato Milanese, Italy
| | | | | |
Collapse
|
26
|
Maiti S, Bhattacharya K, Wider D, Hany D, Panasenko O, Bernasconi L, Hulo N, Picard D. Hsf1 and the molecular chaperone Hsp90 support a 'rewiring stress response' leading to an adaptive cell size increase in chronic stress. eLife 2023; 12:RP88658. [PMID: 38059913 PMCID: PMC10703448 DOI: 10.7554/elife.88658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
Cells are exposed to a wide variety of internal and external stresses. Although many studies have focused on cellular responses to acute and severe stresses, little is known about how cellular systems adapt to sublethal chronic stresses. Using mammalian cells in culture, we discovered that they adapt to chronic mild stresses of up to two weeks, notably proteotoxic stresses such as heat, by increasing their size and translation, thereby scaling the amount of total protein. These adaptations render them more resilient to persistent and subsequent stresses. We demonstrate that Hsf1, well known for its role in acute stress responses, is required for the cell size increase, and that the molecular chaperone Hsp90 is essential for coupling the cell size increase to augmented translation. We term this translational reprogramming the 'rewiring stress response', and propose that this protective process of chronic stress adaptation contributes to the increase in size as cells get older, and that its failure promotes aging.
Collapse
Affiliation(s)
- Samarpan Maiti
- Département de Biologie Moléculaire et Cellulaire, Université de GenèveGenèveSwitzerland
| | - Kaushik Bhattacharya
- Département de Biologie Moléculaire et Cellulaire, Université de GenèveGenèveSwitzerland
| | - Diana Wider
- Département de Biologie Moléculaire et Cellulaire, Université de GenèveGenèveSwitzerland
| | - Dina Hany
- Département de Biologie Moléculaire et Cellulaire, Université de GenèveGenèveSwitzerland
- On leave from: Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in AlexandriaAlexandriaEgypt
| | - Olesya Panasenko
- BioCode: RNA to Proteins Core Facility, Département de Microbiologie et Médecine Moléculaire, Faculté de Médecine, Université de GenèveGenèveSwitzerland
| | - Lilia Bernasconi
- Département de Biologie Moléculaire et Cellulaire, Université de GenèveGenèveSwitzerland
| | - Nicolas Hulo
- Institute of Genetics and Genomics of Geneva, Université de GenèveGenèveSwitzerland
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de GenèveGenèveSwitzerland
| |
Collapse
|
27
|
Canniff NP, Graham JB, Guay KP, Lubicki DA, Eyles SJ, Rauch JN, Hebert DN. TTC17 is an endoplasmic reticulum resident TPR-containing adaptor protein. J Biol Chem 2023; 299:105450. [PMID: 37949225 PMCID: PMC10783571 DOI: 10.1016/j.jbc.2023.105450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/11/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023] Open
Abstract
Protein folding, quality control, maturation, and trafficking are essential processes for proper cellular homeostasis. Around one-third of the human proteome is targeted to the endoplasmic reticulum (ER), the organelle that serves as entrance into the secretory pathway. Successful protein trafficking is paramount for proper cellular function and to that end there are many ER resident proteins that ensure efficient secretion. Here, biochemical and cell biological analysis was used to determine that TTC17 is a large, soluble, ER-localized protein that plays an important role in secretory trafficking. Transcriptional analysis identified the predominantly expressed protein isoform of TTC17 in various cell lines. Further, TTC17 localizes to the ER and interacts with a wide variety of chaperones and cochaperones normally associated with ER protein folding, quality control, and maturation processes. TTC17 was found to be significantly upregulated by ER stress and through the creation and use of TTC17-/- cell lines, quantitative mass spectrometry identified secretory pathway wide trafficking defects in the absence of TTC17. Notably, trafficking of insulin-like growth factor type 1 receptor, glycoprotein nonmetastatic melanoma protein B, clusterin, and UDP-glucose:glycoprotein glucosyltransferase 1 were significantly altered in H4 neuroglioma cells. This study defines a novel ER trafficking factor and provides insight into the protein-protein assisted trafficking in the early secretory pathway.
Collapse
Affiliation(s)
- Nathan P Canniff
- Program in Molecular and Cellular Biology, University of Massachusetts Amherst, USA
| | - Jill B Graham
- Program in Molecular and Cellular Biology, University of Massachusetts Amherst, USA
| | - Kevin P Guay
- Program in Molecular and Cellular Biology, University of Massachusetts Amherst, USA
| | - Daniel A Lubicki
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, USA
| | - Stephen J Eyles
- Program in Molecular and Cellular Biology, University of Massachusetts Amherst, USA; Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, USA; Institute for Applied Life Sciences, Mass Spectrometry Center, University of Massachusetts Amherst, USA
| | - Jennifer N Rauch
- Program in Molecular and Cellular Biology, University of Massachusetts Amherst, USA; Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, USA
| | - Daniel N Hebert
- Program in Molecular and Cellular Biology, University of Massachusetts Amherst, USA; Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst, USA.
| |
Collapse
|
28
|
Hua H, Su T, Han L, Zhang L, Huang Y, Zhang N, Yang M. LINC01226 promotes gastric cancer progression through enhancing cytoplasm-to-nucleus translocation of STIP1 and stabilizing β-catenin protein. Cancer Lett 2023; 577:216436. [PMID: 37806517 DOI: 10.1016/j.canlet.2023.216436] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
Gastric cancer (GC) remains one of the most common malignances and the leading cause of cancer-related mortality worldwide. Although the critical role of several long non-coding RNAs (lncRNAs) transcribed from several GC-risk loci has been established, we still know little about the biological significance of these lncRNAs at most gene loci and how they play in cell signaling. In the present study, we identified a novel oncogenic lncRNA LINC01226 transcribed from the 1p35.2 GC-risk locus. LINC01226 shows markedly higher expression levels in GC specimens compared with those in normal tissues. High expression of LINC01226 is evidently correlated with worse prognosis of GC cases. In line with these, oncogenic LINC01226 promotes proliferation, migration and metastasis of GC cells ex vivo and in vivo. Importantly, LINC01226 binds to STIP1 protein, leads to disassembly of the STIP1-HSP90 complex, elevates interactions between HSP90 and β-catenin, stabilizes β-catenin protein, activates the Wnt/β-catenin signaling and, thereby, promote GC progression. Together, our findings uncovered a novel layer regulating the Wnt signaling in cancers and uncovers a new epigenetic mode of GC tumorigenesis. These discoveries also shed new light on the importance of functional lncRNAs as innovative therapeutic targets through precisely controlling protein-protein interactions in cancers.
Collapse
Affiliation(s)
- Hui Hua
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Tao Su
- Shandong University Cancer Center, Jinan, Shandong Province, 250117, China
| | - Linyu Han
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Long Zhang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Yizhou Huang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China
| | - Nasha Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China.
| | - Ming Yang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, 250117, China; Shandong University Cancer Center, Jinan, Shandong Province, 250117, China; Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China.
| |
Collapse
|
29
|
Chiosis G, Digwal CS, Trepel JB, Neckers L. Structural and functional complexity of HSP90 in cellular homeostasis and disease. Nat Rev Mol Cell Biol 2023; 24:797-815. [PMID: 37524848 PMCID: PMC10592246 DOI: 10.1038/s41580-023-00640-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2023] [Indexed: 08/02/2023]
Abstract
Heat shock protein 90 (HSP90) is a chaperone with vital roles in regulating proteostasis, long recognized for its function in protein folding and maturation. A view is emerging that identifies HSP90 not as one protein that is structurally and functionally homogeneous but, rather, as a protein that is shaped by its environment. In this Review, we discuss evidence of multiple structural forms of HSP90 in health and disease, including homo-oligomers and hetero-oligomers, also termed epichaperomes, and examine the impact of stress, post-translational modifications and co-chaperones on their formation. We describe how these variations influence context-dependent functions of HSP90 as well as its interaction with other chaperones, co-chaperones and proteins, and how this structural complexity of HSP90 impacts and is impacted by its interaction with small molecule modulators. We close by discussing recent developments regarding the use of HSP90 inhibitors in cancer and how our new appreciation of the structural and functional heterogeneity of HSP90 invites a re-evaluation of how we discover and implement HSP90 therapeutics for disease treatment.
Collapse
Affiliation(s)
- Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Institute, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Institute, New York, NY, USA.
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Institute, New York, NY, USA
| | - Jane B Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Len Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
30
|
Chen Y, Yang Y, Cai W, Zeng J, Liu N, Wan Y, Fu G. Research progress of anti-environmental factor stress mechanism and anti-stress tolerance way of Saccharomyces cerevisiae during the brewing process. Crit Rev Food Sci Nutr 2023; 63:12308-12323. [PMID: 35848108 DOI: 10.1080/10408398.2022.2101090] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Saccharomyces cerevisiae plays a decisive role in the brewing of alcohol products, and the ideal growth and fermentation characteristics can give the pure flavor of alcohol products. However, S. cerevisiae can be affected profoundly by environmental factors during the brewing process, which have negative effects on the growth and fermentation characteristics of S. cerevisiae, and seriously hindered the development of brewing industry. Therefore, we summarized the environmental stress factors (ethanol, organic acids, temperature and osmotic pressure) that affect S. cerevisiae during the brewing process. Their impact mechanisms and the metabolic adaption of S. cerevisiae in response to these stress factors. Of note, S. cerevisiae can increase the ability to resist stress factors by changing the cell membrane components, expressing transcriptional regulatory factors, activating the anti-stress metabolic pathway and enhancing ROS scavenging ability. Meantime, the strategies and methods to improve the stress- tolerant ability of S. cerevisiae during the brewing process were also introduced. Compared with the addition of exogenous anti-stress substances, mutation breeding and protoplast fusion, it appears that adaptive evolution and genetic engineering are able to generate ideal environmental stress tolerance strains of S. cerevisiae and are more in line with the needs of the current brewing industry.
Collapse
Affiliation(s)
- Yanru Chen
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology & International Institute of Food Innovation, Nanchang University, Nanchang, PR China
| | - Yili Yang
- China Regional Research Centre, International Centre of Genetic Engineering & Biotechnology, Taizhou, PR China
| | - Wenqin Cai
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology & International Institute of Food Innovation, Nanchang University, Nanchang, PR China
| | - Jiali Zeng
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology & International Institute of Food Innovation, Nanchang University, Nanchang, PR China
| | - Na Liu
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology & International Institute of Food Innovation, Nanchang University, Nanchang, PR China
| | - Yin Wan
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology & International Institute of Food Innovation, Nanchang University, Nanchang, PR China
| | - Guiming Fu
- State Key Laboratory of Food Science and Technology & College of Food Science and Technology & International Institute of Food Innovation, Nanchang University, Nanchang, PR China
| |
Collapse
|
31
|
An L, Gao H, Zhong Y, Liu Y, Cao Y, Yi J, Huang X, Wen C, Tong R, Pan Z, Yan X, Liu M, Wang S, Wu H, Hu T. The potential roles of stress-induced phosphoprotein 1 and connexin 43 in rats with reperfusion arrhythmia. Immun Inflamm Dis 2023; 11:e852. [PMID: 37904692 PMCID: PMC10546868 DOI: 10.1002/iid3.852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 11/01/2023] Open
Abstract
OBJECTIVE Connexin 43 (Cx43) is a critical gene for maintaining myocardial homeostasis. Interestingly, Cx43 and stress-induced phosphoprotein 1 (STIP1) were recorded to be lowly expressed in ischemia/reperfusion (I/R). However, their impacts on reperfusion arrhythmia (RA) remain to be explored. Our study aimed to find out the related underlying mechanisms. METHODS After the establishment of an isolated heart model through Langendorff perfusion, the heart rate, conduction activation time, conduction velocity, and conduction direction of the left ventricle were evaluated, along with the apoptotic rate detection in the collected myocardial tissues. After the construction of a hypoxia/reoxygenation (H/R)-induced cellular model, cell apoptosis, intercellular communication, cell viability, and the content of reactive oxygen species, superoxide dismutase, malondialdehyde, and lactic dehydrogenase were measured. The expression of Cx43 and STIP1 was determined in both rat heart and cell models. The bindings of STIP3 and Cx43 to heat shock protein 90 (HSP90) and heat shock protein 70 (HSP70) were verified. RESULTS Relative to the corresponding controls, Cx43 and STIP1 were decreased in myocardial tissues of RA rats and H/R-stimulated H9C2 cells, where Cx43-binding HSP70 and HSP90 were respectively increased and decreased, and ubiquitination level of Cx43 was enhanced. STIP1 overexpression promoted protein expression of Cx43, intercellular communication, and cell viability, and reduced cell apoptosis and oxidative stress in H/R-stimulated H9C2 cells. CONCLUSION STIP1 promoted Cx43 expression to improve intercellular communication and reduce oxidative stress in H/R-stimulated H9C2 cells.
Collapse
Affiliation(s)
- Li An
- School of AnesthesiologyGuizhou Medical UniversityGuiyangGuizhouChina
- Department of AnaesthesiologyAffiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
- Translational Medicine Research CenterGuizhou Medical UniversityGuiyangGuizhouChina
| | - Hong Gao
- Department of AnesthesiologyGuizhou Hospital of The 1st Affiliated Hospital, Sun Yat‐sen UniversityGuiyangGuizhouChina
| | - Yi Zhong
- Department of AnaesthesiologyAffiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Yanqiu Liu
- Department of AnesthesiologyGuiyang Fourth People's HospitalGuiyangGuizhouChina
| | - Ying Cao
- Department of AnesthesiologyGuiyang Second People's HospitalGuiyangGuizhouChina
| | - Jing Yi
- Department of AnaesthesiologyAffiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| | - Xiang Huang
- School of AnesthesiologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Chunlei Wen
- Department of AnesthesiologyChildren's Hospital of Guiyang Maternal and Child Health HospitalGuiyangGuizhouChina
| | - Rui Tong
- School of AnesthesiologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Zhijun Pan
- School of AnesthesiologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Xu Yan
- School of AnesthesiologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Meiyan Liu
- School of AnesthesiologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Shengzhao Wang
- School of AnesthesiologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Hao Wu
- School of AnesthesiologyGuizhou Medical UniversityGuiyangGuizhouChina
| | - Tingju Hu
- Department of AnaesthesiologyAffiliated Hospital of Guizhou Medical UniversityGuiyangGuizhouChina
| |
Collapse
|
32
|
Huang Z, Ito M, Zhang S, Toda T, Takeda JI, Ogi T, Ohno K. Extremely low-frequency electromagnetic field induces acetylation of heat shock proteins and enhances protein folding. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115482. [PMID: 37717354 DOI: 10.1016/j.ecoenv.2023.115482] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/21/2023] [Accepted: 09/12/2023] [Indexed: 09/19/2023]
Abstract
The pervasive weak electromagnetic fields (EMF) inundate the industrialized society, but the biological effects of EMF as weak as 10 µT have been scarcely analyzed. Heat shock proteins (HSPs) are molecular chaperones that mediate a sequential stress response. HSP70 and HSP90 provide cells under undesirable situations with either assisting covalent folding of proteins or degrading improperly folded proteins in an ATP-dependent manner. Here we examined the effect of extremely low-frequency (ELF)-EMF on AML12 and HEK293 cells. Although the protein expression levels of HSP70 and HSP90 were reduced after an exposure to ELF-EMF for 3 h, acetylations of HSP70 and HSP90 were increased, which was followed by an enhanced binding affinities of HSP70 and HSP90 for HSP70/HSP90-organizing protein (HOP/STIP1). After 3 h exposure to ELF-EMF, the amount of mitochondria was reduced but the ATP level and the maximal mitochondrial oxygen consumption were increased, which was followed by the reduced protein aggregates and the increased cell viability. Thus, ELF-EMF exposure for 3 h activated acetylation of HSPs to enhance protein folding, which was returned to the basal level at 12 h. The proteostatic effects of ELF-EMF will be able to be applied to treat pathological states in humans.
Collapse
Affiliation(s)
- Zhizhou Huang
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shaochuan Zhang
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takuro Toda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Jun-Ichi Takeda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine (RIeM), Nagoya University, Nagoya, Japan
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
33
|
Petrenko V, Vrublevskaya V, Bystrova M, Masulis I, Kopylova E, Skarga Y, Zhmurina M, Morenkov O. Proliferation, migration, and resistance to oxidative and thermal stresses of HT1080 cells with knocked out genes encoding Hsp90α and Hsp90β. Biochem Biophys Res Commun 2023; 674:62-68. [PMID: 37406487 DOI: 10.1016/j.bbrc.2023.06.076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/20/2023] [Accepted: 06/24/2023] [Indexed: 07/07/2023]
Abstract
Heat shock protein 90 (Hsp90) fulfils essential housekeeping functions in the cell associated with the folding, stabilization, and turnover of various proteins. In mammals, there exist two Hsp90 isoforms, stress-inducible Hsp90α and constitutively expressed Hsp90β. In an attempt to identify cellular processes dependent on Hsp90α and Hsp90β, we generated a panel of clones of human fibrosarcoma HT1080 cells with the knocked out HSP90AA1 or HSP90AB1 genes encoding, respectively, Hsp90α and Hsp90β. The knockout of the HSP90AA1 and HSP90AB1 genes practically did not affect cell proliferation and resistance to thermal shock and oxidative stress. The loss of Hsp90α in Hsp90α-null cell clones also did not impair cell migration, while the migration of the Hsp90β-null cell clones was prominently reduced as compared to parent HT1080 cells. This indicated the necessity of Hsp90β for efficient basal migration of HT1080 cells whereas Hsp90α seems to be dispensable for this process. The knockout of one Hsp90 isoform was invariably accompanied by an increase in the level of the other Hsp90 isoform by 30-50%, which partly or fully compensated for a decrease in the total level of Hsp90. Thus, we demonstrated the dispensability of Hsp90α and Hsp90β for HT1080 cells in several cellular processes under normal and stress conditions, which suggested the participation of the two Hsp90 isoforms in the same biological processes and full or at least partial functional substitution of one Hsp90 isoform by the other.
Collapse
Affiliation(s)
- Viktoria Petrenko
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, 1422290, Russia
| | - Veronika Vrublevskaya
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, 1422290, Russia
| | - Marina Bystrova
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, 1422290, Russia
| | - Irina Masulis
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, 1422290, Russia
| | - Elizaveta Kopylova
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, 1422290, Russia
| | - Yuri Skarga
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, 1422290, Russia
| | - Mariya Zhmurina
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, 1422290, Russia
| | - Oleg Morenkov
- Institute of Cell Biophysics, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Moscow Region, 1422290, Russia.
| |
Collapse
|
34
|
Hany D, Vafeiadou V, Picard D. CRISPR-Cas9 screen reveals a role of purine synthesis for estrogen receptor α activity and tamoxifen resistance of breast cancer cells. SCIENCE ADVANCES 2023; 9:eadd3685. [PMID: 37172090 PMCID: PMC10181187 DOI: 10.1126/sciadv.add3685] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
In breast cancer, resistance to endocrine therapies that target estrogen receptor α (ERα), such as tamoxifen and fulvestrant, remains a major clinical problem. Whether and how ERα+ breast cancers switch from being estrogen-dependent to estrogen-independent remains unclear. With a genome-wide CRISPR-Cas9 knockout screen, we identified previously unknown biomarkers and potential therapeutic targets of endocrine resistance. We demonstrate that high levels of PAICS, an enzyme involved in the de novo biosynthesis of purines, can shift the balance of ERα activity to be more estrogen-independent and tamoxifen-resistant. We find that this may be due to elevated activities of cAMP-activated protein kinase A and mTOR, kinases known to phosphorylate ERα specifically and to stimulate its activity. Genetic or pharmacological targeting of PAICS sensitizes tamoxifen-resistant cells to tamoxifen. Addition of purines renders them more resistant. On the basis of these findings, we propose the combined targeting of PAICS and ERα as a new, effective, and potentially safe therapeutic regimen.
Collapse
Affiliation(s)
- Dina Hany
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
- On leave from: Department of Pharmacology and Therapeutics Faculty of Pharmacy, Pharos University in Alexandria, Alexandria 21311, Egypt
| | - Vasiliki Vafeiadou
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH - 1211 Genève 4, Switzerland
| |
Collapse
|
35
|
Mangano S, Muñoz A, Fernández-Calvino L, Castellano MM. HOP co-chaperones contribute to GA signaling by promoting the accumulation of the F-box protein SNE in Arabidopsis. PLANT COMMUNICATIONS 2023; 4:100517. [PMID: 36597357 PMCID: PMC10203442 DOI: 10.1016/j.xplc.2023.100517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/11/2022] [Accepted: 12/31/2022] [Indexed: 05/11/2023]
Abstract
Gibberellins (GAs) play important roles in multiple developmental processes and in plant response to the environment. Within the GA pathway, a central regulatory step relies on GA-dependent degradation of the DELLA transcriptional regulators. Nevertheless, the relevance of the stability of other key proteins in this pathway, such as SLY1 and SNE (the F-box proteins involved in DELLA degradation), remains unknown. Here, we take advantage of mutants in the HSP70-HSP90 organizing protein (HOP) co-chaperones and reveal that these proteins contribute to the accumulation of SNE in Arabidopsis. Indeed, HOP proteins, along with HSP90 and HSP70, interact in vivo with SNE, and SNE accumulation is significantly reduced in the hop mutants. Concomitantly, greater accumulation of the DELLA protein RGA is observed in these plants. In agreement with these molecular phenotypes, hop mutants show a hypersensitive response to the GA inhibitor paclobutrazol and display a partial response to the ectopic addition of GA when GA-regulated processes are assayed. These mutants also display different phenotypes associated with alterations in the GA pathway, such as reduced germination rate, delayed bolting, and reduced hypocotyl elongation in response to warm temperatures. Remarkably, ectopic overexpression of SNE reverts the delay in germination and the thermally dependent hypocotyl elongation defect of the hop1 hop2 hop3 mutant, revealing that SNE accumulation is the key aspect of the hop mutant phenotypes. Together, these data reveal a pivotal role for HOP in SNE accumulation and GA signaling.
Collapse
Affiliation(s)
- Silvina Mangano
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223 Pozuelo de Alarcón (Madrid), Spain; Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBA, CONICET), Av. Patricias Argentinas 435, Buenos Aires C1405BWE, Argentina
| | - Alfonso Muñoz
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223 Pozuelo de Alarcón (Madrid), Spain; Departamento de Botánica, Ecología y Fisiología Vegetal, Campus de Rabanales, Edificio Severo Ochoa, Universidad de Córdoba, 14071 Córdoba, Spain
| | - Lourdes Fernández-Calvino
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223 Pozuelo de Alarcón (Madrid), Spain
| | - M Mar Castellano
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), Campus Montegancedo UPM, 28223 Pozuelo de Alarcón (Madrid), Spain.
| |
Collapse
|
36
|
Rong Y, Jensen SI, Lindorff-Larsen K, Nielsen AT. Folding of heterologous proteins in bacterial cell factories: Cellular mechanisms and engineering strategies. Biotechnol Adv 2023; 63:108079. [PMID: 36528238 DOI: 10.1016/j.biotechadv.2022.108079] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/20/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
The expression of correctly folded and functional heterologous proteins is important in many biotechnological production processes, whether it is enzymes, biopharmaceuticals or biosynthetic pathways for production of sustainable chemicals. For industrial applications, bacterial platform organisms, such as E. coli, are still broadly used due to the availability of tools and proven suitability at industrial scale. However, expression of heterologous proteins in these organisms can result in protein aggregation and low amounts of functional protein. This review provides an overview of the cellular mechanisms that can influence protein folding and expression, such as co-translational folding and assembly, chaperone binding, as well as protein quality control, across different model organisms. The knowledge of these mechanisms is then linked to different experimental methods that have been applied in order to improve functional heterologous protein folding, such as codon optimization, fusion tagging, chaperone co-production, as well as strain and protein engineering strategies.
Collapse
Affiliation(s)
- Yixin Rong
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | - Sheila Ingemann Jensen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | - Kresten Lindorff-Larsen
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaloes Vej 5, 2200 Copenhagen N, Denmark
| | - Alex Toftgaard Nielsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
37
|
Esfahanian N, Knoblich CD, Bowman GA, Rezvani K. Mortalin: Protein partners, biological impacts, pathological roles, and therapeutic opportunities. Front Cell Dev Biol 2023; 11:1028519. [PMID: 36819105 PMCID: PMC9932541 DOI: 10.3389/fcell.2023.1028519] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
Mortalin (GRP75, HSPA9A), a heat shock protein (HSP), regulates a wide range of cellular processes, including cell survival, growth, and metabolism. The regulatory functions of mortalin are mediated through a diverse set of protein partners associated with different cellular compartments, which allows mortalin to perform critical functions under physiological conditions, including mitochondrial protein quality control. However, alteration of mortalin's activities, its abnormal subcellular compartmentalization, and its protein partners turn mortalin into a disease-driving protein in different pathological conditions, including cancers. Here, mortalin's contributions to tumorigenic pathways are explained. Pathology information based on mortalin's RNA expression extracted from The Cancer Genome Atlas (TCGA) transcriptomic database indicates that mortalin has an independent prognostic value in common tumors, including lung, breast, and colorectal cancer (CRC). Subsequently, the binding partners of mortalin reported in different cellular models, from yeast to mammalian cells, and its regulation by post-translational modifications are discussed. Finally, we focus on colorectal cancer and discuss how mortalin and its tumorigenic downstream protein targets are regulated by a ubiquitin-like protein through the 26S proteasomal degradation machinery. A broader understanding of the function of mortalin and its positive and negative regulation in the formation and progression of human diseases, particularly cancer, is essential for developing new strategies to treat a diverse set of human diseases critically associated with dysregulated mortalin.
Collapse
|
38
|
Castelli M, Bhattacharya K, Abboud E, Serapian SA, Picard D, Colombo G. Phosphorylation of the Hsp90 Co-Chaperone Hop Changes its Conformational Dynamics and Biological Function. J Mol Biol 2023; 435:167931. [PMID: 36572238 DOI: 10.1016/j.jmb.2022.167931] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/16/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
The molecular chaperones Hsp90 and Hsp70 and their regulatory co-chaperone Hop play a key role at the crossroads of the folding pathways of numerous client proteins by forming fine-tuned multiprotein complexes. Alterations of the biomolecules involved may functionally impact the chaperone machinery: here, we integrate simulations and experiments to unveil how Hop conformational fitness and interactions can be controlled by the perturbation of just one residue. Specifically, we unveil how mechanisms mediated by Hop residue Y354 control Hop open and closed states, which affect binding of Hsp70/Hsp90. Phosphorylation or mutation of Hop-Y354 are shown to favor structural ensembles that are indeed not optimal for stable interactions with Hsp90 and Hsp70. This disfavors cellular accumulation of the stringent Hsp90 clients glucocorticoid receptor and the viral tyrosine kinase v-Src, with detrimental effects on v-Src activity. Our results show how the post-translational modification of a specific residue in Hop provides a regulation mechanism for the larger chaperone complex of which it is part. In this framework, the effects of one single alteration are amplified at the cellular level through the perturbation of protein-interaction networks.
Collapse
Affiliation(s)
- Matteo Castelli
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy. https://twitter.com/mat_castelli
| | - Kaushik Bhattacharya
- Department of Molecular and Cellular Biology, Université de Genève, Sciences III, 1211 Genève 4, Switzerland. https://twitter.com/kaushik34371359
| | - Ernest Abboud
- Department of Molecular and Cellular Biology, Université de Genève, Sciences III, 1211 Genève 4, Switzerland
| | - Stefano A Serapian
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Didier Picard
- Department of Molecular and Cellular Biology, Université de Genève, Sciences III, 1211 Genève 4, Switzerland.
| | - Giorgio Colombo
- Department of Chemistry, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy.
| |
Collapse
|
39
|
Abstract
Protein homeostasis relies on a balance between protein folding and protein degradation. Molecular chaperones like Hsp70 and Hsp90 fulfill well-defined roles in protein folding and conformational stability via ATP-dependent reaction cycles. These folding cycles are controlled by associations with a cohort of non-client protein co-chaperones, such as Hop, p23, and Aha1. Pro-folding co-chaperones facilitate the transit of the client protein through the chaperone-mediated folding process. However, chaperones are also involved in proteasomal and lysosomal degradation of client proteins. Like folding complexes, the ability of chaperones to mediate protein degradation is regulated by co-chaperones, such as the C-terminal Hsp70-binding protein (CHIP/STUB1). CHIP binds to Hsp70 and Hsp90 chaperones through its tetratricopeptide repeat (TPR) domain and functions as an E3 ubiquitin ligase using a modified RING finger domain (U-box). This unique combination of domains effectively allows CHIP to network chaperone complexes to the ubiquitin-proteasome and autophagosome-lysosome systems. This chapter reviews the current understanding of CHIP as a co-chaperone that switches Hsp70/Hsp90 chaperone complexes from protein folding to protein degradation.
Collapse
Affiliation(s)
- Abantika Chakraborty
- Biomedical Biotechnology Research Unit, Department of Biochemistry and Microbiology, Rhodes University, Makhanda/Grahamstown, South Africa
| | - Adrienne L Edkins
- Biomedical Biotechnology Research Unit, Department of Biochemistry and Microbiology, Rhodes University, Makhanda/Grahamstown, South Africa.
| |
Collapse
|
40
|
Abstract
The Hsp70/Hsp90 organising protein (Hop, also known as stress-inducible protein 1/STI1/STIP1) has received considerable attention for diverse cellular functions in both healthy and diseased states. There is extensive evidence that intracellular Hop is a co-chaperone of the major chaperones Hsp70 and Hsp90, playing an important role in the productive folding of Hsp90 client proteins, although recent evidence suggests that eukaryotic Hop is regulatory within chaperone complexes rather than essential. Consequently, Hop is implicated in many key signalling pathways, including aberrant pathways leading to cancer. Hop is also secreted, and it is now well established that Hop interacts with the prion protein, PrPC, to mediate multiple signalling events. The intracellular and extracellular forms of Hop most likely represent two different isoforms, although the molecular determinants of these divergent functions are yet to be identified. There is also a growing body of research that reports the involvement of Hop in cellular activities that appear independent of either chaperones or PrPC. While the various cellular functions of Hop have been described, its biological function remains elusive. However, recent knockout studies in mammals suggest that Hop has an important role in embryonic development. This review provides a critical overview of the latest molecular, cellular and biological research on Hop, critically evaluating its function in healthy systems and how this function is adapted in diseased states.
Collapse
|
41
|
Prodromou C, Aran-Guiu X, Oberoi J, Perna L, Chapple JP, van der Spuy J. HSP70-HSP90 Chaperone Networking in Protein-Misfolding Disease. Subcell Biochem 2023; 101:389-425. [PMID: 36520314 DOI: 10.1007/978-3-031-14740-1_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Molecular chaperones and their associated co-chaperones are essential in health and disease as they are key facilitators of protein-folding, quality control and function. In particular, the heat-shock protein (HSP) 70 and HSP90 molecular chaperone networks have been associated with neurodegenerative diseases caused by aberrant protein-folding. The pathogenesis of these disorders usually includes the formation of deposits of misfolded, aggregated protein. HSP70 and HSP90, plus their co-chaperones, have been recognised as potent modulators of misfolded protein toxicity, inclusion formation and cell survival in cellular and animal models of neurodegenerative disease. Moreover, these chaperone machines function not only in folding but also in proteasome-mediated degradation of neurodegenerative disease proteins. This chapter gives an overview of the HSP70 and HSP90 chaperones, and their respective regulatory co-chaperones, and explores how the HSP70 and HSP90 chaperone systems form a larger functional network and its relevance to counteracting neurodegenerative disease associated with misfolded proteins and disruption of proteostasis.
Collapse
Affiliation(s)
| | - Xavi Aran-Guiu
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Jasmeen Oberoi
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton, UK
| | - Laura Perna
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - J Paul Chapple
- Centre for Endocrinology, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | | |
Collapse
|
42
|
Backe SJ, Woodford MR, Ahanin E, Sager RA, Bourboulia D, Mollapour M. Impact of Co-chaperones and Posttranslational Modifications Toward Hsp90 Drug Sensitivity. Subcell Biochem 2023; 101:319-350. [PMID: 36520312 PMCID: PMC10077965 DOI: 10.1007/978-3-031-14740-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Posttranslational modifications (PTMs) regulate myriad cellular processes by modulating protein function and protein-protein interaction. Heat shock protein 90 (Hsp90) is an ATP-dependent molecular chaperone whose activity is responsible for the stabilization and maturation of more than 300 client proteins. Hsp90 is a substrate for numerous PTMs, which have diverse effects on Hsp90 function. Interestingly, many Hsp90 clients are enzymes that catalyze PTM, demonstrating one of the several modes of regulation of Hsp90 activity. Approximately 25 co-chaperone regulatory proteins of Hsp90 impact structural rearrangements, ATP hydrolysis, and client interaction, representing a second layer of influence on Hsp90 activity. A growing body of literature has also established that PTM of these co-chaperones fine-tune their activity toward Hsp90; however, many of the identified PTMs remain uncharacterized. Given the critical role of Hsp90 in supporting signaling in cancer, clinical evaluation of Hsp90 inhibitors is an area of great interest. Interestingly, differential PTM and co-chaperone interaction have been shown to impact Hsp90 binding to its inhibitors. Therefore, understanding these layers of Hsp90 regulation will provide a more complete understanding of the chaperone code, facilitating the development of new biomarkers and combination therapies.
Collapse
Affiliation(s)
- Sarah J Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mark R Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Elham Ahanin
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Rebecca A Sager
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY, USA. .,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA. .,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
43
|
Tiwari S, Singh A, Gupta P, Singh S. UBA52 Is Crucial in HSP90 Ubiquitylation and Neurodegenerative Signaling during Early Phase of Parkinson's Disease. Cells 2022; 11:cells11233770. [PMID: 36497031 PMCID: PMC9738938 DOI: 10.3390/cells11233770] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/29/2022] Open
Abstract
Protein aggregation is one of the major pathological events in age-related Parkinson's disease (PD) pathology, predominantly regulated by the ubiquitin-proteasome system (UPS). UPS essentially requires core component ubiquitin; however, its role in PD pathology is obscure. This study aimed to investigate the role of ubiquitin-encoding genes in sporadic PD pathology. Both cellular and rat models of PD as well as SNCA C57BL/6J-Tg (Th-SNCA*A30P*A53T)39 Eric/J transgenic mice showed a decreased abundance of UBA52 in conjunction with significant downregulation of tyrosine hydroxylase (TH) and neuronal death. In silico predictions, mass spectrometric analysis, and co-immunoprecipitation findings suggested the protein-protein interaction of UBA52 with α-synuclein, HSP90 and E3-ubiquitin ligase CHIP, and its co-localization with α-synuclein in the mitochondrion. Next, in vitro ubiquitylation assay indicated an imperative requirement of the lysine-63 residue of UBA52 in CHIP-mediated HSP90 ubiquitylation. Myc-UBA52 expressed neurons inhibited alteration in PD-specific markers such as α-synuclein and TH protein along with increased proteasome activity in diseased conditions. Furthermore, Myc-UBA52 expression inhibited the altered protein abundance of HSP90 and its various client proteins, HSP75 (homolog of HSP90 in mitochondrion) and ER stress-related markers during early PD. Taken together, the data highlights the critical role of UBA52 in HSP90 ubiquitylation in parallel to its potential contribution to the modulation of various disease-related neurodegenerative signaling targets during the early phase of PD pathology.
Collapse
Affiliation(s)
- Shubhangini Tiwari
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Abhishek Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Parul Gupta
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sarika Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
- Correspondence:
| |
Collapse
|
44
|
Gaur D, Kumar N, Ghosh A, Singh P, Kumar P, Guleria J, Kaur S, Malik N, Saha S, Nystrom T, Sharma D. Ydj1 interaction at nucleotide-binding-domain of yeast Ssa1 impacts Hsp90 collaboration and client maturation. PLoS Genet 2022; 18:e1010442. [PMID: 36350833 PMCID: PMC9645627 DOI: 10.1371/journal.pgen.1010442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 09/24/2022] [Indexed: 11/11/2022] Open
Abstract
Hsp90 constitutes one of the major chaperone machinery in the cell. The Hsp70 assists Hsp90 in its client maturation though the underlying basis of the Hsp70 role remains to be explored. In the present study, using S. cerevisiae strain expressing Ssa1 as sole Ssa Hsp70, we identified novel mutations in the nucleotide-binding domain of yeast Ssa1 Hsp70 (Ssa1-T175N and Ssa1-D158N) that adversely affect the maturation of Hsp90 clients v-Src and Ste11. The identified Ssa1 amino acids critical for Hsp90 function were also found to be conserved across species such as in E.coli DnaK and the constitutive Hsp70 isoform (HspA8) in humans. These mutations are distal to the C-terminus of Hsp70, that primarily mediates Hsp90 interaction through the bridge protein Sti1, and proximal to Ydj1 (Hsp40 co-chaperone of Hsp70 family) binding region. Intriguingly, we found that the bridge protein Sti1 is critical for cellular viability in cells expressing Ssa1-T175N (A1-T175N) or Ssa1-D158N (A1-D158N) as sole Ssa Hsp70. The growth defect was specific for sti1Δ, as deletion of none of the other Hsp90 co-chaperones showed lethality in A1-T175N or A1-D158N. Mass-spectrometry based whole proteome analysis of A1-T175N cells lacking Sti1 showed an altered abundance of various kinases and transcription factors suggesting compromised Hsp90 activity. Further proteomic analysis showed that pathways involved in signaling, signal transduction, and protein phosphorylation are markedly downregulated in the A1-T175N upon repressing Sti1 expression using doxycycline regulatable promoter. In contrast to Ssa1, the homologous mutations in Ssa4 (Ssa4-T175N/D158N), the stress inducible Hsp70 isoform, supported cell growth even in the absence of Sti1. Overall, our data suggest that Ydj1 competes with Hsp90 for binding to Hsp70, and thus regulates Hsp90 interaction with the nucleotide-binding domain of Hsp70. The study thus provides new insight into the Hsp70-mediated regulation of Hsp90 and broadens our understanding of the intricate complexities of the Hsp70-Hsp90 network. Hsp70-Hsp90 constitutes major cellular chaperone machinery in cells. The Hsp70 plays critical role in Hsp90 chaperoning pathway. We have now identified novel mutations in the nucleotide-binding domain of yeast Ssa1 Hsp70 (Ssa1-T175N and Ssa1-D158N) that adversely affect Hsp90 client maturation. As compared to wt Ssa1, the identified Ssa1 mutants bind relatively better with Ydj1, and poorly support growth in the absence of Sti1, when present as the sole source of Ssa Hsp70 in S. cerevisiae. The cells expressing Ssa1-T175N as sole Ssa Hsp70 show downregulation of pathways involved in signaling, signal transduction, and protein phosphorylation upon repressing Sti1. The study shows that Ydj1 interaction at the nucleotide-binding domain of Ssa1 Hsp70 influences Hsp90 function.
Collapse
Affiliation(s)
- Deepika Gaur
- Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh, India
| | - Navinder Kumar
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-Age Cap, University of Gothenburg, Gothenburg, Sweden
| | - Abhirupa Ghosh
- Division of Bioinformatics, Bose Institute, Kolkata, India
| | - Prashant Singh
- Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh, India
| | - Pradeep Kumar
- Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh, India
| | - Jyoti Guleria
- Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh, India
| | - Satinderdeep Kaur
- Pharmacology Department, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Nikhil Malik
- Department of Biochemistry, School of Interdisciplinary and Applied Life Sciences, Central University of Haryana, Mahendergarh, India
| | - Sudipto Saha
- Division of Bioinformatics, Bose Institute, Kolkata, India
| | - Thomas Nystrom
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-Age Cap, University of Gothenburg, Gothenburg, Sweden
| | - Deepak Sharma
- Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific & Innovative Research, Ghaziabad, India
- * E-mail:
| |
Collapse
|
45
|
Vaaltyn MC, Mateos‐Jimenez M, Müller R, Mackay CL, Edkins AL, Clarke DJ, Veale CGL. Native Mass Spectrometry-Guided Screening Identifies Hit Fragments for HOP-HSP90 PPI Inhibition. Chembiochem 2022; 23:e202200322. [PMID: 36017658 PMCID: PMC9826382 DOI: 10.1002/cbic.202200322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/25/2022] [Indexed: 01/11/2023]
Abstract
Contemporary medicinal chemistry considers fragment-based drug discovery (FBDD) and inhibition of protein-protein interactions (PPI) as important means of expanding the volume of druggable chemical space. However, the ability to robustly identify valid fragments and PPI inhibitors is an enormous challenge, requiring the application of sensitive biophysical methodology. Accordingly, in this study, we exploited the speed and sensitivity of nanoelectrospray (nano-ESI) native mass spectrometry to identify a small collection of fragments which bind to the TPR2AB domain of HOP. Follow-up biophysical assessment of a small selection of binding fragments confirmed binding to the single TPR2A domain, and that this binding translated into PPI inhibitory activity between TPR2A and the HSP90 C-terminal domain. An in-silico assessment of binding fragments at the PPI interfacial region, provided valuable structural insight for future fragment elaboration strategies, including the identification of losartan as a weak, albeit dose-dependent inhibitor of the target PPI.
Collapse
Affiliation(s)
- Michaelone C. Vaaltyn
- The Biomedical Biotechnology Research Unit (BioBRU) Department of Biochemistry and Microbiology DepartmentRhodes UniversityMakhanda6139South Africa
| | - Maria Mateos‐Jimenez
- EaStCHEM School of ChemistryJoseph Black Building, David Brewster RoadEdinburghEH93FJUK
| | - Ronel Müller
- School of Chemistry and PhysicsUniversity of KwaZulu-NatalScottsville3209South Africa
| | - C. Logan Mackay
- EaStCHEM School of ChemistryJoseph Black Building, David Brewster RoadEdinburghEH93FJUK
| | - Adrienne L. Edkins
- The Biomedical Biotechnology Research Unit (BioBRU) Department of Biochemistry and Microbiology DepartmentRhodes UniversityMakhanda6139South Africa
| | - David J. Clarke
- EaStCHEM School of ChemistryJoseph Black Building, David Brewster RoadEdinburghEH93FJUK
| | - Clinton G. L. Veale
- Department of ChemistryUniversity of Cape Town RondeboschCape Town7700South Africa
| |
Collapse
|
46
|
Bhattacharya K, Maiti S, Zahoran S, Weidenauer L, Hany D, Wider D, Bernasconi L, Quadroni M, Collart M, Picard D. Translational reprogramming in response to accumulating stressors ensures critical threshold levels of Hsp90 for mammalian life. Nat Commun 2022; 13:6271. [PMID: 36270993 PMCID: PMC9587034 DOI: 10.1038/s41467-022-33916-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 10/07/2022] [Indexed: 12/25/2022] Open
Abstract
The cytosolic molecular chaperone Hsp90 is essential for eukaryotic life. Although reduced Hsp90 levels correlate with aging, it was unknown whether eukaryotic cells and organisms can tune the basal Hsp90 levels to alleviate physiologically accumulated stress. We have investigated whether and how mice adapt to the deletion of three out of four alleles of the two genes encoding cytosolic Hsp90, with one Hsp90β allele being the only remaining one. While the vast majority of such mouse embryos die during gestation, survivors apparently manage to increase their Hsp90β protein to at least wild-type levels. Our studies reveal an internal ribosome entry site in the 5' untranslated region of the Hsp90β mRNA allowing translational reprogramming to compensate for the genetic loss of Hsp90 alleles and in response to stress. We find that the minimum amount of total Hsp90 required to support viability of mammalian cells and organisms is 50-70% of what is normally there. Those that fail to maintain a threshold level are subject to accelerated senescence, proteostatic collapse, and ultimately death. Therefore, considering that Hsp90 levels can be reduced ≥100-fold in the unicellular budding yeast, critical threshold levels of Hsp90 have markedly increased during eukaryotic evolution.
Collapse
Affiliation(s)
- Kaushik Bhattacharya
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Samarpan Maiti
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Szabolcs Zahoran
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Lorenz Weidenauer
- Protein Analysis Facility, Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Dina Hany
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Diana Wider
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Lilia Bernasconi
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland
| | - Manfredo Quadroni
- Protein Analysis Facility, Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Martine Collart
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Didier Picard
- Department of Molecular and Cellular Biology, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
47
|
Oroń M, Grochowski M, Jaiswar A, Legierska J, Jastrzębski K, Nowak-Niezgoda M, Kołos M, Kaźmierczak W, Olesiński T, Lenarcik M, Cybulska M, Mikula M, Żylicz A, Miączyńska M, Zettl K, Wiśniewski JR, Walerych D. The molecular network of the proteasome machinery inhibition response is orchestrated by HSP70, revealing vulnerabilities in cancer cells. Cell Rep 2022; 40:111428. [PMID: 36170818 DOI: 10.1016/j.celrep.2022.111428] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/15/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
Proteasome machinery is a major proteostasis control system in human cells, actively compensated upon its inhibition. To understand this compensation, we compared global protein landscapes upon the proteasome inhibition with carfilzomib, in normal fibroblasts, cells of multiple myeloma, and cancers of lung, colon, and pancreas. Molecular chaperones, autophagy, and endocytosis-related proteins are the most prominent vulnerabilities in combination with carfilzomib, while targeting of the HSP70 family chaperones HSPA1A/B most specifically sensitizes cancer cells to the proteasome inhibition. This suggests a central role of HSP70 in the suppression of the proteasome downregulation, allowing to identify pathways impinging on HSP70 upon the proteasome inhibition. HSPA1A/B indeed controls proteasome-inhibition-induced autophagy, unfolded protein response, and endocytic flux, and directly chaperones the proteasome machinery. However, it does not control the NRF1/2-driven proteasome subunit transcriptional bounce-back. Consequently, targeting of NRF1 proves effective in decreasing the viability of cancer cells with the inhibited proteasome and HSP70.
Collapse
Affiliation(s)
- Magdalena Oroń
- Mossakowski Medical Research Institute PAS, Warsaw, Poland
| | | | | | | | - Kamil Jastrzębski
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | | - Małgorzata Kołos
- Central Clinical Hospital of Ministry of Interior and Administration, Warsaw, Poland
| | | | | | | | | | | | - Alicja Żylicz
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Marta Miączyńska
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | | | | | - Dawid Walerych
- Mossakowski Medical Research Institute PAS, Warsaw, Poland.
| |
Collapse
|
48
|
Jamabo M, Bentley SJ, Macucule-Tinga P, Tembo P, Edkins AL, Boshoff A. In silico analysis of the HSP90 chaperone system from the African trypanosome, Trypanosoma brucei. Front Mol Biosci 2022; 9:947078. [PMID: 36213128 PMCID: PMC9538636 DOI: 10.3389/fmolb.2022.947078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
African trypanosomiasis is a neglected tropical disease caused by Trypanosoma brucei (T. brucei) and spread by the tsetse fly in sub-Saharan Africa. The trypanosome relies on heat shock proteins for survival in the insect vector and mammalian host. Heat shock protein 90 (HSP90) plays a crucial role in the stress response at the cellular level. Inhibition of its interactions with chaperones and co-chaperones is being explored as a potential therapeutic target for numerous diseases. This study provides an in silico overview of HSP90 and its co-chaperones in both T. brucei brucei and T. brucei gambiense in relation to human and other trypanosomal species, including non-parasitic Bodo saltans and the insect infecting Crithidia fasciculata. A structural analysis of T. brucei HSP90 revealed differences in the orientation of the linker and C-terminal domain in comparison to human HSP90. Phylogenetic analysis displayed the T. brucei HSP90 proteins clustering into three distinct groups based on subcellular localizations, namely, cytosol, mitochondria, and endoplasmic reticulum. Syntenic analysis of cytosolic HSP90 genes revealed that T. b. brucei encoded for 10 tandem copies, while T. b. gambiense encoded for three tandem copies; Leishmania major (L. major) had the highest gene copy number with 17 tandem copies. The updated information on HSP90 from recently published proteomics on T. brucei was examined for different life cycle stages and subcellular localizations. The results show a difference between T. b. brucei and T. b. gambiense with T. b. brucei encoding a total of twelve putative HSP90 genes, while T. b. gambiense encodes five HSP90 genes. Eighteen putative co-chaperones were identified with one notable absence being cell division cycle 37 (Cdc37). These results provide an updated framework on approaching HSP90 and its interactions as drug targets in the African trypanosome.
Collapse
Affiliation(s)
- Miebaka Jamabo
- Biotechnology Innovation Centre, Rhodes University, Grahamstown, South Africa
| | | | | | - Praise Tembo
- Biotechnology Innovation Centre, Rhodes University, Grahamstown, South Africa
| | - Adrienne Lesley Edkins
- Department of Biochemistry and Microbiology, Biomedical Biotechnology Research Unit (BioBRU), Rhodes University, Grahamstown, South Africa
| | - Aileen Boshoff
- Biotechnology Innovation Centre, Rhodes University, Grahamstown, South Africa
- *Correspondence: Aileen Boshoff,
| |
Collapse
|
49
|
Hoang TX, Kim JY. Cell Surface Hsp90- and αMβ2 Integrin-Mediated Uptake of Bacterial Flagellins to Activate Inflammasomes by Human Macrophages. Cells 2022; 11:cells11182878. [PMID: 36139453 PMCID: PMC9496951 DOI: 10.3390/cells11182878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
All-trans retinoic acid (ATRA) is an active metabolite of vitamin A, which plays an important role in the immune function. Here, we demonstrated that ATRA induces the heat shock protein (Hsp) 90 complex on the surface of THP-1 macrophages, which facilitates the internalization of exogenous bacterial flagellins to activate the inflammasome response. Mass spectrometric protein identification and co-immunoprecipitation revealed that the Hsp90 homodimer interacts with both Hsp70 and αMβ2 integrin. ATRA-induced complex formation was dependent on the retinoic acid receptor (RAR)/retinoid X receptor (RXR) pathway and intracellular calcium level and was essential for triggering the internalization of bacterial flagellin, which was clathrin dependent. Notably, in this process, αMβ2 integrin was found to act as a carrier to deliver flagellin to the cytosol to activate the inflammasome, leading to caspase-1 activity and secretion of interleukin (IL)-1β. Our study provides new insights into the underlying molecular mechanism by which exogenous bacterial flagellins are delivered into host cells without a bacterial transport system, as well as the mechanism by which vitamin A contributes to enhancing the human macrophage function to detect and respond to bacterial infection.
Collapse
|
50
|
Uncoupling the Hsp90 and DnaK chaperone activities revealed the in vivo relevance of their collaboration in bacteria. Proc Natl Acad Sci U S A 2022; 119:e2201779119. [PMID: 36070342 PMCID: PMC9478669 DOI: 10.1073/pnas.2201779119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Chaperone proteins are essential in all living cells to ensure protein homeostasis. Hsp90 is a major adenosine triphosphate (ATP)-dependent chaperone highly conserved from bacteria to eukaryotes. Recent studies have shown that bacterial Hsp90 is essential in some bacteria in stress conditions and that it participates in the virulence of pathogenic bacteria. In vitro, bacterial Hsp90 directly interacts and collaborates with the Hsp70 chaperone DnaK to reactivate model substrate proteins; however, it is still unknown whether this collaboration is relevant in vivo with physiological substrates. Here, we used site-directed mutagenesis on Hsp90 to impair DnaK binding, thereby uncoupling the chaperone activities. We tested the mutants in vivo in two bacterial models in which Hsp90 has known physiological functions. We found that the Hsp90 point mutants were defective to support (1) growth under heat stress and activation of an essential Hsp90 client in the aquatic bacterium Shewanella oneidensis and (2) biosynthesis of the colibactin toxin involved in the virulence of pathogenic Escherichia coli. Our study therefore demonstrates the essentiality of the direct collaboration between Hsp90 and DnaK in vivo in bacteria to support client folding. It also suggests that this collaboration already functional in bacteria has served as an evolutionary basis for a more complex Hsp70-Hsp90 collaboration found in eukaryotes.
Collapse
|