1
|
Gumilar KE, Chin Y, Ibrahim IH, Tjokroprawiro BA, Yang JY, Zhou M, Gassman NR, Tan M. Heat Shock Factor 1 Inhibition: A Novel Anti-Cancer Strategy with Promise for Precision Oncology. Cancers (Basel) 2023; 15:5167. [PMID: 37958341 PMCID: PMC10649344 DOI: 10.3390/cancers15215167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Heat shock factor 1 (HSF1) is a transcription factor crucial for regulating heat shock response (HSR), one of the significant cellular protective mechanisms. When cells are exposed to proteotoxic stress, HSF1 induces the expression of heat shock proteins (HSPs) to act as chaperones, correcting the protein-folding process and maintaining proteostasis. In addition to its role in HSR, HSF1 is overexpressed in multiple cancer cells, where its activation promotes malignancy and leads to poor prognosis. The mechanisms of HSF1-induced tumorigenesis are complex and involve diverse signaling pathways, dependent on cancer type. With its important roles in tumorigenesis and tumor progression, targeting HSF1 offers a novel cancer treatment strategy. In this article, we examine the basic function of HSF1 and its regulatory mechanisms, focus on the mechanisms involved in HSF1's roles in different cancer types, and examine current HSF1 inhibitors as novel therapeutics to treat cancers.
Collapse
Affiliation(s)
- Khanisyah Erza Gumilar
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan (Y.C.); (I.H.I.); (J.-Y.Y.)
- Department of Obstetrics and Gynecology, Faculty of Medicine, Airlangga University, Surabaya 60286, Indonesia;
| | - Yeh Chin
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan (Y.C.); (I.H.I.); (J.-Y.Y.)
| | - Ibrahim Haruna Ibrahim
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan (Y.C.); (I.H.I.); (J.-Y.Y.)
| | - Brahmana A. Tjokroprawiro
- Department of Obstetrics and Gynecology, Faculty of Medicine, Airlangga University, Surabaya 60286, Indonesia;
| | - Jer-Yen Yang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan (Y.C.); (I.H.I.); (J.-Y.Y.)
| | - Ming Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha 410013, China;
| | - Natalie R. Gassman
- Department of Pharmacology and Toxicology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Ming Tan
- Graduate Institute of Biomedical Science, China Medical University, Taichung 40402, Taiwan (Y.C.); (I.H.I.); (J.-Y.Y.)
- Institute of Biochemistry and Molecular Biology, Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan
| |
Collapse
|
2
|
Wang Y, Abazid A, Badendieck S, Mustea A, Stope MB. Impact of Non-Invasive Physical Plasma on Heat Shock Protein Functionality in Eukaryotic Cells. Biomedicines 2023; 11:biomedicines11051471. [PMID: 37239142 DOI: 10.3390/biomedicines11051471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/06/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Recently, biomedical research has increasingly investigated physical plasma as an innovative therapeutic approach with a number of therapeutic biomedical effects. It is known from radiation and chemotherapy that these applications can lead to the induction and activation of primarily cytoprotective heat shock proteins (HSP). HSP protect cells and tissues from physical, (bio)chemical, and physiological stress and, ultimately, along with other mechanisms, govern resistance and treatment failure. These mechanisms are well known and comparatively well studied in drug therapy. For therapies in the field of physical plasma medicine, however, extremely little data are available to date. In this review article, we provide an overview of the current studies on the interaction of physical plasma with the cellular HSP system.
Collapse
Affiliation(s)
- Yanqing Wang
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Alexander Abazid
- Department of General, Visceral and Thorax Surgery, Bundeswehr Hospital Berlin, Scharnhorststrasse 13, 10115 Berlin, Germany
| | - Steffen Badendieck
- Department of General, Visceral and Thorax Surgery, Bundeswehr Hospital Berlin, Scharnhorststrasse 13, 10115 Berlin, Germany
| | - Alexander Mustea
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Matthias B Stope
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| |
Collapse
|
3
|
Samant RS, Batista S, Larance M, Ozer B, Milton CI, Bludau I, Wu E, Biggins L, Andrews S, Hervieu A, Johnston HE, Al-Lazikhani B, Lamond AI, Clarke PA, Workman P. Native Size-Exclusion Chromatography-Based Mass Spectrometry Reveals New Components of the Early Heat Shock Protein 90 Inhibition Response Among Limited Global Changes. Mol Cell Proteomics 2023; 22:100485. [PMID: 36549590 PMCID: PMC9898794 DOI: 10.1016/j.mcpro.2022.100485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/16/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
The molecular chaperone heat shock protein 90 (HSP90) works in concert with co-chaperones to stabilize its client proteins, which include multiple drivers of oncogenesis and malignant progression. Pharmacologic inhibitors of HSP90 have been observed to exert a wide range of effects on the proteome, including depletion of client proteins, induction of heat shock proteins, dissociation of co-chaperones from HSP90, disruption of client protein signaling networks, and recruitment of the protein ubiquitylation and degradation machinery-suggesting widespread remodeling of cellular protein complexes. However, proteomics studies to date have focused on inhibitor-induced changes in total protein levels, often overlooking protein complex alterations. Here, we use size-exclusion chromatography in combination with mass spectrometry (SEC-MS) to characterize the early changes in native protein complexes following treatment with the HSP90 inhibitor tanespimycin (17-AAG) for 8 h in the HT29 colon adenocarcinoma cell line. After confirming the signature cellular response to HSP90 inhibition (e.g., induction of heat shock proteins, decreased total levels of client proteins), we were surprised to find only modest perturbations to the global distribution of protein elution profiles in inhibitor-treated HT29 cells at this relatively early time-point. Similarly, co-chaperones that co-eluted with HSP90 displayed no clear difference between control and treated conditions. However, two distinct analysis strategies identified multiple inhibitor-induced changes, including known and unknown components of the HSP90-dependent proteome. We validate two of these-the actin-binding protein Anillin and the mitochondrial isocitrate dehydrogenase 3 complex-as novel HSP90 inhibitor-modulated proteins. We present this dataset as a resource for the HSP90, proteostasis, and cancer communities (https://www.bioinformatics.babraham.ac.uk/shiny/HSP90/SEC-MS/), laying the groundwork for future mechanistic and therapeutic studies related to HSP90 pharmacology. Data are available via ProteomeXchange with identifier PXD033459.
Collapse
Affiliation(s)
- Rahul S Samant
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom; Signalling Programme, The Babraham Institute, Cambridge, United Kingdom.
| | - Silvia Batista
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Mark Larance
- Centre for Gene Regulation & Expression, University of Dundee, Dundee, United Kingdom
| | - Bugra Ozer
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Christopher I Milton
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Isabell Bludau
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Estelle Wu
- Signalling Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Laura Biggins
- Bioinformatics Group, The Babraham Institute, Cambridge, United Kingdom
| | - Simon Andrews
- Bioinformatics Group, The Babraham Institute, Cambridge, United Kingdom
| | - Alexia Hervieu
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Harvey E Johnston
- Signalling Programme, The Babraham Institute, Cambridge, United Kingdom
| | - Bissan Al-Lazikhani
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Angus I Lamond
- Centre for Gene Regulation & Expression, University of Dundee, Dundee, United Kingdom
| | - Paul A Clarke
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom
| | - Paul Workman
- Centre for Cancer Drug Discovery, The Institute of Cancer Research, London, United Kingdom.
| |
Collapse
|
4
|
Shao H, Taguwa S, Gilbert L, Shkedi A, Sannino S, Guerriero CJ, Gale-Day ZJ, Young ZT, Brodsky JL, Weissman J, Gestwicki JE, Frydman J. A campaign targeting a conserved Hsp70 binding site uncovers how subcellular localization is linked to distinct biological activities. Cell Chem Biol 2022; 29:1303-1316.e3. [PMID: 35830852 PMCID: PMC9513760 DOI: 10.1016/j.chembiol.2022.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/20/2022] [Accepted: 06/20/2022] [Indexed: 12/14/2022]
Abstract
The potential of small molecules to localize within subcellular compartments is rarely explored. To probe this question, we measured the localization of Hsp70 inhibitors using fluorescence microscopy. We found that even closely related analogs had dramatically different distributions, with some residing predominantly in the mitochondria and others in the ER. CRISPRi screens supported this idea, showing that different compounds had distinct chemogenetic interactions with Hsp70s of the ER (HSPA5/BiP) and mitochondria (HSPA9/mortalin) and their co-chaperones. Moreover, localization seemed to determine function, even for molecules with conserved binding sites. Compounds with distinct partitioning have distinct anti-proliferative activity in breast cancer cells compared with anti-viral activity in cellular models of Dengue virus replication, likely because different sets of Hsp70s are required in these processes. These findings highlight the contributions of subcellular partitioning and chemogenetic interactions to small molecule activity, features that are rarely explored during medicinal chemistry campaigns.
Collapse
Affiliation(s)
- Hao Shao
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA; College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Shuhei Taguwa
- Department of Biology, Stanford University, Stanford, CA 94305, USA; Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan; Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Luke Gilbert
- Department of Urology and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Arielle Shkedi
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Sara Sannino
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Zachary J Gale-Day
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Zapporah T Young
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Weissman
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA.
| | - Judith Frydman
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
5
|
Omkar S, Wani TH, Zheng B, Mitchem MM, Truman AW. The APE2 Exonuclease Is a Client of the Hsp70–Hsp90 Axis in Yeast and Mammalian Cells. Biomolecules 2022; 12:biom12070864. [PMID: 35883419 PMCID: PMC9312491 DOI: 10.3390/biom12070864] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/18/2022] [Accepted: 06/18/2022] [Indexed: 12/10/2022] Open
Abstract
Molecular chaperones such as Hsp70 and Hsp90 help fold and activate proteins in important signal transduction pathways that include DNA damage response (DDR). Previous studies have suggested that the levels of the mammalian APE2 exonuclease, a protein critical for DNA repair, may be dependent on chaperone activity. In this study, we demonstrate that the budding yeast Apn2 exonuclease interacts with molecular chaperones Ssa1 and Hsp82 and the co-chaperone Ydj1. Although Apn2 does not display a binding preference for any specific cytosolic Hsp70 or Hsp90 paralog, Ssa1 is unable to support Apn2 stability when present as the sole Ssa in the cell. Demonstrating conservation of this mechanism, the exonuclease APE2 also binds to Hsp70 and Hsp90 in mammalian cells. Inhibition of chaperone function via specific small molecule inhibitors results in a rapid loss of APE2 in a range of cancer cell lines. Taken together, these data identify APE2 and Apn2 as clients of the chaperone system in yeast and mammalian cells and suggest that chaperone inhibition may form the basis of novel anticancer therapies that target APE2-mediated processes.
Collapse
|
6
|
Al-Bari AA. Inhibition of autolysosomes by repurposing drugs as a promising therapeutic strategy for the treatment of cancers. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2078894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- Abdul Alim Al-Bari
- Department of Pharmacy, Faculty of Science, University of Rajshahi, Rajshahi, Bangladesh
| |
Collapse
|
7
|
Antolin AA, Clarke PA, Collins I, Workman P, Al-Lazikani B. Evolution of kinase polypharmacology across HSP90 drug discovery. Cell Chem Biol 2021; 28:1433-1445.e3. [PMID: 34077750 PMCID: PMC8550792 DOI: 10.1016/j.chembiol.2021.05.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/12/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022]
Abstract
Most small molecules interact with several target proteins but this polypharmacology is seldom comprehensively investigated or explicitly exploited during drug discovery. Here, we use computational and experimental methods to identify and systematically characterize the kinase cross-pharmacology of representative HSP90 inhibitors. We demonstrate that the resorcinol clinical candidates ganetespib and, to a lesser extent, luminespib, display unique off-target kinase pharmacology as compared with other HSP90 inhibitors. We also demonstrate that polypharmacology evolved during the optimization to discover luminespib and that the hit, leads, and clinical candidate all have different polypharmacological profiles. We therefore recommend the computational and experimental characterization of polypharmacology earlier in drug discovery projects to unlock new multi-target drug design opportunities.
Collapse
Affiliation(s)
- Albert A Antolin
- Department of Data Science, The Institute of Cancer Research, London SM2 5NG, UK; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, UK.
| | - Paul A Clarke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| | - Ian Collins
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, UK
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, UK.
| | - Bissan Al-Lazikani
- Department of Data Science, The Institute of Cancer Research, London SM2 5NG, UK; Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London SM2 5NG, UK.
| |
Collapse
|
8
|
Aminzadeh Z, Ziamajidi N, Abbasalipourkabir R. Anticancer Effects of Cinnamaldehyde through Inhibition of ErbB2/HSF1/LDHA Pathway in 5637 Cell Line of Bladder Cancer. Anticancer Agents Med Chem 2021; 22:1139-1148. [PMID: 34315398 DOI: 10.2174/1871520621666210726142814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The growing prevalence of bladder cancer worldwide has become a major concern for the researchers, and the side effects of chemotherapy drugs have always been a major problem in cancer treatment. Cinnamaldehyde, the active ingredient in the Cinnamon plant, has long been considered with anti-oxidant and anti-inflammatory effects. METHODS Bladder cancer 5637 cell lines were treated with the different concentrations of Cinnamaldehyde. MTT assay was performed to evaluate cell viability at 24, 48, and 72 hours. The concentration of 0.02, 0.04, and 0.08 mg/ml of Cinnamaldehyde were selected. Apoptosis was assessed with Annexin V-FITC/PI and Hochest33258 staining. Cell migration was performed by the scratch test. To evaluate Cinnamaldehyde effect on glycolysis, the gene expression of epidermal growth factor receptor 2 (ErbB2), heat shock protein transcription factor-1 (HSF1) and lactate dehydrogenase A (LDHA), as well as the protein levels of HSF1 and LDHA, LDH activity and finally glucose consumption and lactate production, were measured. RESULTS Cinnamaldehyde significantly increased apoptosis rate in the 5637 cells (p<0.05). Furthermore, it significantly reduced the gene expression of ErbB2, HSF1, and LDHA, protein level of HSF1 and LDHA, LDH activity, as well as cell migration, glucose consumption, and lactate production (p<0.05). These changes were dose-dependent. CONCLUSION Thus, Cinnamaldehyde induced apoptosis and decreased growth in 5637 cells by reducing ErbB2-HSF1-LDHA pathway.
Collapse
Affiliation(s)
- Zeynab Aminzadeh
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nasrin Ziamajidi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Roghayeh Abbasalipourkabir
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
9
|
Heat Shock Factor 1 as a Prognostic and Diagnostic Biomarker of Gastric Cancer. Biomedicines 2021; 9:biomedicines9060586. [PMID: 34064083 PMCID: PMC8224319 DOI: 10.3390/biomedicines9060586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 01/09/2023] Open
Abstract
Identification of effective prognostic and diagnostic biomarkers is needed to improve the diagnosis and treatment of gastric cancer. Early detection of gastric cancer through diagnostic markers can help establish effective treatments. Heat shock factor 1 (HSF1), presented in this review, is known to be regulated by a broad range of transcription factors, including those characterized in various malignant tumors, including gastric cancer. Particularly, it has been demonstrated that HSF1 regulation in various cancers is correlated with different processes, such as cell death, proliferation, and metastasis. Due to the effect of HSF1 on the initiation, development, and progression of various tumors, it is considered as an important gene for understanding and treating tumors. Additionally, HSF1 exhibits high expression in various cancers, and its high expression adversely affects the prognosis of various cancer patients, thereby suggesting that it can be used as a novel, predictive, prognostic, and diagnostic biomarker for gastric cancer. In this review, we discuss the literature accumulated in recent years, which suggests that there is a correlation between the expression of HSF1 and prognosis of gastric cancer patients through public data. Consequently, this evidence also indicates that HSF1 can be established as a powerful biomarker for the prognosis and diagnosis of gastric cancer.
Collapse
|
10
|
Meng C, Liu K, Cai X, Chen Y. Mechanism of miR-455-3 in suppressing epithelial-mesenchymal transition and angiogenesis of non-small cell lung cancer cells. Cell Stress Chaperones 2021; 27:107-117. [PMID: 35064898 PMCID: PMC8943084 DOI: 10.1007/s12192-022-01254-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/09/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
The tumor-suppressing role of miR-455-3p has been reported in lung cancer, but the working mechanism remains to be fully elucidated. This study aims to explore the possible mechanism of miR-455-3p in regulating epithelial-mesenchymal transition (EMT) progression and angiogenesis in non-small cell lung cancer (NSCLC) cells.The expressions of miR-455-3p, HSF1, GLS1, and EMT-related proteins (E-cadherin, N-cadherin, vimentin, and Snail-1) in both NSCLC tissues and cell lines were determined by RT-qPCR and western blot. After cell transfection, cell proliferation and angiogenesis ability on NSCLC cells were assessed by MTT and tube formation assay. The binding of miR-455-3p with HSF1 was measured by luciferase reporter gene assay, while the interaction between HSF1 and GLS1 was determined by co-immunoprecipitation assay (Co-IP).HSF1 was highly expressed in NSCLC tissues and cells. Inhibition of HSF1 expression or overexpression of miR-455-3p in NSCLC cells can suppress cell proliferation, angiogenesis ability, and EMT progression. miR-455-3p was found to negatively regulate HSF1 expression. Co-transfection of miR-455-3p overexpression and HSF1 inhibition in NSCLC cells showed that miR-455-3p can partially counteract the effect of HSF1 in NSCLC cells. HSF1 can interact with GLS1 and elevate the expression of GLS1. GLS1 can partially abolish the suppressive effect of miR-455-3p in NSCLC cells.miR-455-3p can bind HSF1 to suppress the GLS1 in NSCLC cells, therefore suppressing EMT progression and angiogenesis of NSCLC cells.
Collapse
Affiliation(s)
- Chong Meng
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Haikou, Hainan, 570311, People's Republic of China
| | - Kai Liu
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Haikou, Hainan, 570311, People's Republic of China
| | - Xingjun Cai
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Haikou, Hainan, 570311, People's Republic of China
| | - Yongxing Chen
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, No. 19, Xiuhua Road, Haikou, Hainan, 570311, People's Republic of China.
| |
Collapse
|
11
|
Shen Y, Zou Y, Li J, Chen F, Li H, Cai Y. CDK5RAP3, a Novel Nucleoplasmic Shuttle, Deeply Regulates HSF1-Mediated Heat Stress Response and Protects Mammary Epithelial Cells from Heat Injury. Int J Mol Sci 2020; 21:E8400. [PMID: 33182370 PMCID: PMC7664939 DOI: 10.3390/ijms21218400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/06/2020] [Accepted: 11/07/2020] [Indexed: 12/21/2022] Open
Abstract
CDK5RAP3 was regarded as the most significant regulator of cellular responses against heat stress, which is associated with dysfunctions of the immune system and animal susceptibility to disease. Despite this, little known about how CDK5RAP3 regulates heat stress response. In this study, CDK5RAP3 conditional Knockout (CKO) mice, CDK5RAP3-/- mouse embryo fibroblasts (MEFs) and bovine mammary epithelial cells (BMECs) were used as an in vitro and in vivo model, respectively to reveal the role of CDK5RAP3 in regulating the heat stress response. The deletion of CDK5RAP3 unexpectedly caused animal lethality after 1.5-h heat stimulations. Furthermore, BMECs were re-cultured for eight hours after heat stress and was found that the expression of CDK5RAP3 and HSPs showed a similar fluctuating pattern of increase (0-2, 4-6 h) and decrease (2-4, 6-8 h). In addition to the remarkably enhanced expression of heat shock protein, apoptosis rate and endoplasmic reticulum stress, the deletion of CDK5RAP3 also affected nucleoplasmic translocation and trimer formation of heat shock factor 1 (HSF1). These programs were further confirmed in the mammary gland of CDK5RAP3 CKO mice and CDK5RAP3-/- MEFs as well. Interestingly, genetic silencing of HSF1 downregulated CDK5RAP3 expression in BMECs. Immunostaining and immunoprecipitation studies suggested a physical interaction between CDK5RAP3 and HSF1 being co-localized in the cytoplasm and nucleus. Besides, CDK5RAP3 also interacted with HSP90, suggesting an operative machinery at both transcriptional level and protein functionality of HSP90 per se. Together, our findings suggested that CDK5RAP3 works like a novel nucleoplasmic shuttle or molecular chaperone, deeply participating in HSF1-mediated heat stress response and protecting cells from heat injury.
Collapse
Affiliation(s)
- Yangyang Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (Y.Z.); (F.C.)
| | - Yan Zou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (Y.Z.); (F.C.)
| | - Jun Li
- College of Life Sciences, Anhui Normal University, Wuhu 241000, China;
| | - Fanghui Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (Y.Z.); (F.C.)
| | - Honglin Li
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Yafei Cai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (Y.S.); (Y.Z.); (F.C.)
| |
Collapse
|
12
|
Darby JF, Vidler LR, Simpson PJ, Al-Lazikani B, Matthews SJ, Sharp SY, Pearl LH, Hoelder S, Workman P. Solution structure of the Hop TPR2A domain and investigation of target druggability by NMR, biochemical and in silico approaches. Sci Rep 2020; 10:16000. [PMID: 32994435 PMCID: PMC7524759 DOI: 10.1038/s41598-020-71969-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/24/2020] [Indexed: 02/08/2023] Open
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone that plays an important role in tumour biology by promoting the stabilisation and activity of oncogenic 'client' proteins. Inhibition of Hsp90 by small-molecule drugs, acting via its ATP hydrolysis site, has shown promise as a molecularly targeted cancer therapy. Owing to the importance of Hop and other tetratricopeptide repeat (TPR)-containing cochaperones in regulating Hsp90 activity, the Hsp90-TPR domain interface is an alternative site for inhibitors, which could result in effects distinct from ATP site binders. The TPR binding site of Hsp90 cochaperones includes a shallow, positively charged groove that poses a significant challenge for druggability. Herein, we report the apo, solution-state structure of Hop TPR2A which enables this target for NMR-based screening approaches. We have designed prototype TPR ligands that mimic key native 'carboxylate clamp' interactions between Hsp90 and its TPR cochaperones and show that they block binding between Hop TPR2A and the Hsp90 C-terminal MEEVD peptide. We confirm direct TPR-binding of these ligands by mapping 1H-15N HSQC chemical shift perturbations to our new NMR structure. Our work provides a novel structure, a thorough assessment of druggability and robust screening approaches that may offer a potential route, albeit difficult, to address the chemically challenging nature of the Hop TPR2A target, with relevance to other TPR domain interactors.
Collapse
Affiliation(s)
- John F Darby
- Division of Cancer Therapeutics, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Lewis R Vidler
- Division of Cancer Therapeutics, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Peter J Simpson
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
- Bruker UK Ltd, Banner Lane, Coventry, CV4 9GH, UK
| | - Bissan Al-Lazikani
- Division of Cancer Therapeutics, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Stephen J Matthews
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
| | - Swee Y Sharp
- Division of Cancer Therapeutics, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Laurence H Pearl
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Falmer, Brighton, UK
- Division of Structural Biology, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Swen Hoelder
- Division of Cancer Therapeutics, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Paul Workman
- Division of Cancer Therapeutics, Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK.
- Cancer Research UK Convergence Science Centre, The Institute of Cancer Research and Imperial College London, London, UK.
| |
Collapse
|