1
|
Min L, Li X, Liang L, Ruan Z, Yu S. Targeting HSP90 in Gynecologic Cancer: Molecular Mechanisms and Therapeutic Approaches. Cell Biochem Biophys 2024:10.1007/s12013-024-01502-7. [PMID: 39249180 DOI: 10.1007/s12013-024-01502-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2024] [Indexed: 09/10/2024]
Abstract
One of the leading causes of mortality for women is gynecologic cancer (GC). Numerous molecules (tumor suppressor genes or oncogenes) are involved in this form of cancer's invasion, metastasis, tumorigenic process, and therapy resistance. Currently, there is a shortage of efficient methods to eliminate these diseases, hence it is crucial to carry out more extensive studies on GCs. Novel pharmaceuticals are required to surmount this predicament. Highly conserved molecular chaperon, heat shock protein (HSP) 90, is essential for the maturation of recently produced polypeptides and offers a refuge for misfolding or denatured proteins to be turned around. In cancer, the client proteins of HSP90 play a role in the entire process of oncogenesis, which is linked to all the characteristic features of cancer. In this study, we explore the various functions of HSPs in GC progression. We also discuss their potential as promising targets for pharmacological therapy.
Collapse
Affiliation(s)
- Lu Min
- Changchun University of Chinese Medicine Hospital, Changchun, 130000, China
| | - Xuewei Li
- Changchun University of Chinese Medicine Hospital, Changchun, 130000, China
| | - Lily Liang
- Changchun University of Chinese Medicine Hospital, Changchun, 130000, China
| | - Zheng Ruan
- Department of Traditional Chinese Medicine, 964th Hospital, Changchun, 130000, China
| | - Shaohui Yu
- Changchun University of Chinese Medicine Hospital, Changchun, 130000, China.
| |
Collapse
|
2
|
Rani K, Gotmare A, Maier A, Menghal R, Akhtar N, Fangaria N, Buchner J, Bhattacharyya S. Identification of a chaperone-code responsible for Rad51-mediated genome repair. J Biol Chem 2024; 300:107342. [PMID: 38705392 PMCID: PMC11154708 DOI: 10.1016/j.jbc.2024.107342] [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: 12/10/2023] [Revised: 04/15/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024] Open
Abstract
Posttranslational modifications of Hsp90 are known to regulate its in vivo chaperone functions. Here, we demonstrate that the lysine acetylation-deacetylation dynamics of Hsp82 is a major determinant in DNA repair mediated by Rad51. We uncover that the deacetylated lysine 27 in Hsp82 dictates the formation of the Hsp82-Aha1-Rad51 complex, which is crucial for client maturation. Intriguingly, Aha1-Rad51 complex formation is not dependent on Hsp82 or its acetylation status; implying that Aha1-Rad51 association precedes the interaction with Hsp82. The DNA damage sensitivity of Hsp82 (K27Q/K27R) mutants are epistatic to the loss of the (de)acetylase hda1Δ; reinforcing the importance of the reversible acetylation of Hsp82 at the K27 position. These findings underscore the significance of the cross talk between a specific Hsp82 chaperone modification code and the cognate cochaperones in a client-specific manner. Given the pivotal role that Rad51 plays during DNA repair in eukaryotes and particularly in cancer cells, targeting the Hda1-Hsp90 axis could be explored as a new therapeutic approach against cancer.
Collapse
Affiliation(s)
- Khushboo Rani
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Akanksha Gotmare
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Andreas Maier
- Department of Chemistry, Technical University of Munich, Garching, Germany
| | - Ruchira Menghal
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Nashat Akhtar
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Nupur Fangaria
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Johannes Buchner
- Department of Chemistry, Technical University of Munich, Garching, Germany
| | - Sunanda Bhattacharyya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India.
| |
Collapse
|
3
|
Huang H, Qiao Y, Chu L, Ye C, Lin L, Liao H, Meng X, Zou F, Zhao H, Zou M, Cai S, Dong H. Up-regulation of HSP90α in HDM-induced asthma causes pyroptosis of airway epithelial cells by activating the cGAS-STING-ER stress pathway. Int Immunopharmacol 2024; 131:111917. [PMID: 38527402 DOI: 10.1016/j.intimp.2024.111917] [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: 02/02/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Heat Shock protein 90 α (HSP90α), an main subtype of chaperone protein HSP90, involves important biological functions such as DNA damage repair, protein modification, innate immunity. However, the potential role of HSP90α in asthma occurrence and development is still unclear. This study aimed to elucidate the underlying mechanism of HSP90α in asthma by focusing on the cGAS-STING-Endoplasmic Reticulum stress pathway in inflammatory airway epithelial cell death (i.e., pyroptosis; inflammatory cell death). To accomplish that, we modeled allergen exposure in C57/6BL mice and bronchial epithelial cells with house dust mite. Protein technologies and immunofluorescence utilized to study the expression of HSP90α, activation of cGAS-STING pathway and pyroptosis. The effect of inhibitors on HDM-exposed mice detected by histological techniques and examination of bronchoalveolar lavage fluid. Results showed that HSP90α promotes asthma inflammation via pyroptosis and activation of the cGAS-STING-ER stress pathway. Treatment with the HSP90 inhibitor tanespimycin (17-AAG) significantly relieved airway inflammation and abrogated the effect of HSP90α on pyroptosis and cGAS-STING-ER stress in vitro and in vivo models of HDM. Further data indicated that up-regulation of HSP90α stabilized STING through interaction, which increased localization of STING on the ER. Activation of STING triggered ER stress and leaded to pyroptosis-related airway inflammation. The finding showed the potential role of pyroptosis caused by dysregulation of HSP90α on airway epithelial cells in allergic inflammation, suggested that targeting HSP90α in airway epithelial cells might prove to be a potential additional treatment strategy for asthma.
Collapse
Affiliation(s)
- Haohua Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yujie Qiao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lanhe Chu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Cuiping Ye
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lishan Lin
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hua Liao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaojing Meng
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Fei Zou
- School of Public Health, Southern Medical University, Guangzhou, China
| | - Haijin Zhao
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengchen Zou
- Department of Endocrinology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
4
|
Sesink A, Becerra M, Ruan JL, Leboucher S, Dubail M, Heinrich S, Jdey W, Petersson K, Fouillade C, Berthault N, Dutreix M, Girard PM. The AsiDNA™ decoy mimicking DSBs protects the normal tissue from radiation toxicity through a DNA-PK/p53/p21-dependent G1/S arrest. NAR Cancer 2024; 6:zcae011. [PMID: 38476631 PMCID: PMC10928987 DOI: 10.1093/narcan/zcae011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 02/01/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
AsiDNA™, a cholesterol-coupled oligonucleotide mimicking double-stranded DNA breaks, was developed to sensitize tumour cells to radio- and chemotherapy. This drug acts as a decoy hijacking the DNA damage response. Previous studies have demonstrated that standalone AsiDNA™ administration is well tolerated with no additional adverse effects when combined with chemo- and/or radiotherapy. The lack of normal tissue complication encouraged further examination into the role of AsiDNA™ in normal cells. This research demonstrates the radioprotective properties of AsiDNA™. In vitro, AsiDNA™ induces a DNA-PK/p53/p21-dependent G1/S arrest in normal epithelial cells and fibroblasts that is absent in p53 deficient and proficient tumour cells. This cell cycle arrest improved survival after irradiation only in p53 proficient normal cells. Combined administration of AsiDNA™ with conventional radiotherapy in mouse models of late and early radiation toxicity resulted in decreased onset of lung fibrosis and increased intestinal crypt survival. Similar results were observed following FLASH radiotherapy in standalone or combined with AsiDNA™. Mechanisms comparable to those identified in vitro were detected both in vivo, in the intestine and ex vivo, in precision cut lung slices. Collectively, the results suggest that AsiDNA™ can partially protect healthy tissues from radiation toxicity by triggering a G1/S arrest in normal cells.
Collapse
Affiliation(s)
- Anouk Sesink
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, 91405 Orsay, France
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, 91405 Orsay, France
| | - Margaux Becerra
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, 91405 Orsay, France
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, 91405 Orsay, France
| | - Jia-Ling Ruan
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, UK
| | - Sophie Leboucher
- Histology platform, Institut Curie, CNRS UMR3348, 91405 Orsay, France
| | - Maxime Dubail
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, 91405 Orsay, France
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, 91405 Orsay, France
| | - Sophie Heinrich
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, 91405 Orsay, France
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, 91405 Orsay, France
| | - Wael Jdey
- Valerio Therapeutics, 49 Bd du Général Martial Valin, 75015 Paris, France
| | - Kristoffer Petersson
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, UK
- Radiation Physics, Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| | - Charles Fouillade
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, 91405 Orsay, France
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, 91405 Orsay, France
| | - Nathalie Berthault
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, 91405 Orsay, France
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, 91405 Orsay, France
| | - Marie Dutreix
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, 91405 Orsay, France
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, 91405 Orsay, France
| | - Pierre-Marie Girard
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, 91405 Orsay, France
- Université Paris-Saclay, CNRS UMR 3347, INSERM U1021, 91405 Orsay, France
| |
Collapse
|
5
|
Sottile ML, Gómez LC, Redondo A, Ibarra J, García MB, Gonzalez L, Vargas-Roig LM, Nadin SB. Relevance of Comet Assay and Phosphorylated-Hsp90α in Cancer Patients' Peripheral Blood Leukocytes as Tools to Assess Cisplatin-based Chemotherapy Clinical Response and Disease Outcome. J Histochem Cytochem 2024; 72:173-188. [PMID: 38439738 PMCID: PMC10956442 DOI: 10.1369/00221554241236241] [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: 08/07/2023] [Accepted: 02/12/2024] [Indexed: 03/06/2024] Open
Abstract
Cisplatin (cPt) is a commonly used treatment for solid tumors. The main target of its cytotoxicity is the DNA molecule, which makes the DNA damage response (DDR) crucial for cPt-based chemotherapy. Therefore, it is essential to identify biomarkers that can accurately predict the individual clinical response and prognosis. Our goal was to assess the usefulness of alkaline comet assay and immunocytochemical staining of phosphorylated Hsp90α (p-Hsp90α), γH2AX, and 53BP1 as predictive/prognostic markers. Pre-chemotherapy peripheral blood leukocytes were exposed to cPt in vitro and collected at 0, 24 (T24), and 48 (T48) hr post-drug removal. Healthy subjects were also included. Baseline DNA damage was elevated in cancer patients (variability between individuals was observed). After cPt, patients showed increased γH2AX foci/nucleus (T24 and T48). Both in healthy persons and patients, the nuclear p-Hsp90α and N/C (nuclear/cytoplasmic) ratio augmented (T24), decreasing at T48. Favorable clinical response was associated with high DNA damage and p-Hsp90α N/C ratio following cPt. For the first time, p-Hsp90α significance as a predictive marker is highlighted. Post-cPt-DNA damage was associated with longer disease-free survival and overall survival. Our findings indicate that comet assay and p-Hsp90α (a marker of DDR) would be promising prognostic/predictive tools in cP-treated cancer patients.
Collapse
Affiliation(s)
- Mayra L. Sottile
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Scientific and Technical Research Council, National University of Cuyo, Mendoza, Argentina
- Medical Sciences School, Mendoza University, Mendoza, Argentina
| | - Laura C. Gómez
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Scientific and Technical Research Council, National University of Cuyo, Mendoza, Argentina
- Medical Sciences School, Mendoza University, Mendoza, Argentina
| | - Analía Redondo
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Scientific and Technical Research Council, National University of Cuyo, Mendoza, Argentina
| | - Jorge Ibarra
- Regional Integration Cancer Center, Mendoza, Argentina
| | | | | | - Laura M. Vargas-Roig
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Scientific and Technical Research Council, National University of Cuyo, Mendoza, Argentina
| | - Silvina B. Nadin
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo, National Scientific and Technical Research Council, National University of Cuyo, Mendoza, Argentina
| |
Collapse
|
6
|
LIU F, WEI Y, WANG Z. β-D-Glucan promotes NF-κB activation and ameliorates high-LET carbon-ion irradiation-induced human umbilical vein endothelial cell injury. Turk J Med Sci 2023; 53:1621-1634. [PMID: 38813508 PMCID: PMC10760591 DOI: 10.55730/1300-0144.5731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/12/2023] [Accepted: 09/21/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim Heavy-ion irradiation seriously perturbs cellular homeostasis and thus damages cells. Vascular endothelial cells (ECs) play an important role in the pathological process of radiation damage. Protecting ECs from heavy-ion radiation is of great significance in the radioprotection of normal tissues. In this study, the radioprotective effect of β-D-glucan (BG) derived from Saccharomyces cerevisiae on human umbilical vein endothelial cell (EA.hy926) cytotoxicity produced by carbon-ion irradiation was examined and the probable mechanism was established. Materials and methods EA.hy926 cells were divided into seven groups: a control group; 1, 2, or 4 Gy radiation; and 10 μg/mL BG pretreatment for 24 h before 1, 2, or 4 Gy irradiation. Cell survival was assessed by colony formation assay. Cell cycles, apoptosis, DNA damage, and reactive oxygen species (ROS) levels were measured through flow cytometry. The level of malondialdehyde and antioxidant enzyme activities were analyzed using assay kits. The activation of NF-κB was analyzed using western blotting and a transcription factor assay kit. The expression of downstream target genes was detected by western blotting. Results BG pretreatment significantly increased the survival of irradiated cells, improved cell cycle progression, and decreased DNA damage and apoptosis. The levels of ROS and malondialdehyde were also decreased by BG. Further study indicated that BG increased the antioxidant enzyme activities, activated Src, and promoted NF-κB activation, especially for the p65, p50, and RelB subunits. The activated NF-κB upregulated the expression of antioxidant protein MnSOD, DNA damage-response and repair-related proteins BRCA2 and Hsp90α, and antiapoptotic protein Bcl-2. Conclusion Our results demonstrated that BG protects EA.hy926 cells from high linear-energy-transfer carbon-ion irradiation damage through the upregulation of prosurvival signaling triggered by the interaction of BG with its receptor. This confirms that BG is a promising radioprotective agent for heavy-ion exposure.
Collapse
Affiliation(s)
- Fang LIU
- International Genome Center, Jiangsu University, Zhenjiang, Jiangsu, P.R.
China
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, P.R.
China
| | - Yanting WEI
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, P.R.
China
| | - Zhuanzi WANG
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, P.R.
China
| |
Collapse
|
7
|
Fangaria N, Rani K, Singh P, Dey S, Kumar KA, Bhattacharyya S. DNA damage-induced nuclear import of HSP90α is promoted by Aha1. Mol Biol Cell 2022; 33:ar140. [PMID: 36260391 PMCID: PMC9727810 DOI: 10.1091/mbc.e21-11-0554] [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] [Indexed: 02/04/2023] Open
Abstract
The interplay between yHSP90α (Hsp82) and Rad51 has been implicated in the DNA double-strand break repair (DSB) pathway in yeast. Here we report that nuclear translocation of yHSP90α and its recruitment to the DSB end are essential for homologous recombination (HR)-mediated DNA repair in yeast. The HsHSP90α possesses an amino-terminal extension which is phosphorylated upon DNA damage. We find that the absence of the amino-terminal extension in yHSP90α does not compromise its nuclear import, and the nonphosphorylatable-mutant HsHSP90αT7A could be imported to the yeast nucleus upon DNA damage. Interestingly, the flexible charged-linker (CL) domains of both yHSP90α and HsHSP90α play a critical role during their nuclear translocation. The conformational restricted CL mutant yHSP90α∆(211-259), but not a shorter deletion version yHSP90α∆(211-242), fails to reach the nucleus. As the CL domain of yHSP90α is critical for its interaction with Aha1, we investigated whether Aha1 promotes the nuclear import of yHSP90α. We found that the nuclear import of yHSP90α is severely affected in ∆aha1 strain. Moreover, Aha1 is accumulated in the nucleus during DNA damage. Hence Aha1 may serve as a potential target for inhibiting nuclear function of yHSP90α. The increased sensitivity of ∆aha1 strain to genotoxic agents strengthens this notion.
Collapse
Affiliation(s)
- Nupur Fangaria
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Khushboo Rani
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Priyanka Singh
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Sandeep Dey
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Kota Arun Kumar
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Sunanda Bhattacharyya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India,*Address correspondence to: Sunanda Bhattacharyya (; )
| |
Collapse
|
8
|
Maiti S, Picard D. Cytosolic Hsp90 Isoform-Specific Functions and Clinical Significance. Biomolecules 2022; 12:1166. [PMID: 36139005 PMCID: PMC9496497 DOI: 10.3390/biom12091166] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
The heat shock protein 90 (Hsp90) is a molecular chaperone and a key regulator of proteostasis under both physiological and stress conditions. In mammals, there are two cytosolic Hsp90 isoforms: Hsp90α and Hsp90β. These two isoforms are 85% identical and encoded by two different genes. Hsp90β is constitutively expressed and essential for early mouse development, while Hsp90α is stress-inducible and not necessary for survivability. These two isoforms are known to have largely overlapping functions and to interact with a large fraction of the proteome. To what extent there are isoform-specific functions at the protein level has only relatively recently begun to emerge. There are studies indicating that one isoform is more involved in the functionality of a specific tissue or cell type. Moreover, in many diseases, functionally altered cells appear to be more dependent on one particular isoform. This leaves space for designing therapeutic strategies in an isoform-specific way, which may overcome the unfavorable outcome of pan-Hsp90 inhibition encountered in previous clinical trials. For this to succeed, isoform-specific functions must be understood in more detail. In this review, we summarize the available information on isoform-specific functions of mammalian Hsp90 and connect it to possible clinical applications.
Collapse
Affiliation(s)
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH-1211 Geneve, Switzerland
| |
Collapse
|
9
|
LncRNA-MEG3 attenuates hyperglycemia-induced damage by enhancing mitochondrial translocation of HSP90A in the primary hippocampal neurons. Exp Cell Res 2022; 419:113320. [PMID: 35998683 DOI: 10.1016/j.yexcr.2022.113320] [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: 04/20/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/20/2022]
Abstract
The diabetic cognitive impairments are associated with high-glucose (HG)-induced mitochondrial dysfunctions in the brain. Our previous studies demonstrated that long non-coding RNA (lncRNA)-MEG3 alleviates diabetic cognitive impairments. However, the underlying mechanism has still remained elusive. Therefore, this study was designed to investigate whether the mitochondrial translocation of HSP90A and its phosphorylation are involved in lncRNA-MEG3-mediated neuroprotective effects of mitochondrial functions in HG-treated primary hippocampal neurons and diabetic rats. The primary hippocampal neurons were exposed to 75 mM glucose for 72 h to establish a HG model in vitro. Firstly, the RNA pull-down and RNA immunoprecipitation (RIP) assays clearly indicated that lncRNA-MEG3-associated mitochondrial proteins were Annexin A2, HSP90A, and Plectin. Although HG promoted the mitochondrial translocation of HSP90A and Annexin A2, lncRNA-MEG3 over-expression only enhanced the mitochondrial translocation of HSP90A, rather than Annexin A2, in the primary hippocampal neurons treated with or without HG. Meanwhile, Plectin mediated the mitochondrial localization of lncRNA-MEG3 and HSP90A. Furthermore, HSP90A threonine phosphorylation participated in regulating mitochondrial translocation of HSP90A, and lncRNA-MEG3 also enhanced mitochondrial translocation of HSP90A through suppressing HSP90A threonine phosphorylation. Finally, the anti-apoptotic role of mitochondrial translocation of HSP90A was found to be associated with inhibiting death receptor 5 (DR5) in HG-treated primary hippocampal neurons and diabetic rats. Taken together, lncRNA-MEG3 could improve mitochondrial functions in HG-exposed primary hippocampal neurons, and the underlying mechanisms were involved in enhanced mitochondrial translocation of HSP90A via suppressing HSP90A threonine phosphorylation, which may reveal a potential therapeutic target for diabetic cognitive impairments.
Collapse
|
10
|
Ren X, Li T, Zhang W, Yang X. Targeting Heat-Shock Protein 90 in Cancer: An Update on Combination Therapy. Cells 2022; 11:cells11162556. [PMID: 36010632 PMCID: PMC9406578 DOI: 10.3390/cells11162556] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Heat-shock protein 90 (HSP90) is an important molecule chaperone associated with tumorigenesis and malignancy. HSP90 is involved in the folding and maturation of a wide range of oncogenic clients, including diverse kinases, transcription factors and oncogenic fusion proteins. Therefore, it could be argued that HSP90 facilitates the malignant behaviors of cancer cells, such as uncontrolled proliferation, chemo/radiotherapy resistance and immune evasion. The extensive associations between HSP90 and tumorigenesis indicate substantial therapeutic potential, and many HSP90 inhibitors have been developed. However, due to HSP90 inhibitor toxicity and limited efficiency, none have been approved for clinical use as single agents. Recent results suggest that combining HSP90 inhibitors with other anticancer therapies might be a more advisable strategy. This review illustrates the role of HSP90 in cancer biology and discusses the therapeutic value of Hsp90 inhibitors as complements to current anticancer therapies.
Collapse
Affiliation(s)
- Xiude Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin 300052, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin 300052, China
| | - Wei Zhang
- Departments of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC 27157, USA
- Correspondence: (W.Z.); (X.Y.)
| | - Xuejun Yang
- Department of Neurosurgery, Tsinghua University Beijing Tsinghua Changgung Hospital, Beijing 102218, China
- Correspondence: (W.Z.); (X.Y.)
| |
Collapse
|
11
|
The Role of Hsp90-R2TP in Macromolecular Complex Assembly and Stabilization. Biomolecules 2022; 12:biom12081045. [PMID: 36008939 PMCID: PMC9406135 DOI: 10.3390/biom12081045] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 01/27/2023] Open
Abstract
Hsp90 is a ubiquitous molecular chaperone involved in many cell signaling pathways, and its interactions with specific chaperones and cochaperones determines which client proteins to fold. Hsp90 has been shown to be involved in the promotion and maintenance of proper protein complex assembly either alone or in association with other chaperones such as the R2TP chaperone complex. Hsp90-R2TP acts through several mechanisms, such as by controlling the transcription of protein complex subunits, stabilizing protein subcomplexes before their incorporation into the entire complex, and by recruiting adaptors that facilitate complex assembly. Despite its many roles in protein complex assembly, detailed mechanisms of how Hsp90-R2TP assembles protein complexes have yet to be determined, with most findings restricted to proteomic analyses and in vitro interactions. This review will discuss our current understanding of the function of Hsp90-R2TP in the assembly, stabilization, and activity of the following seven classes of protein complexes: L7Ae snoRNPs, spliceosome snRNPs, RNA polymerases, PIKKs, MRN, TSC, and axonemal dynein arms.
Collapse
|
12
|
Berthault N, Bergam P, Pereira F, Girard PM, Dutreix M. Inhibition of DNA Repair by Inappropriate Activation of ATM, PARP, and DNA-PK with the Drug Agonist AsiDNA. Cells 2022; 11:cells11142149. [PMID: 35883597 PMCID: PMC9320633 DOI: 10.3390/cells11142149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 12/10/2022] Open
Abstract
AsiDNA is a DNA repair inhibitor mimicking DNA double-strand breaks (DSB) that was designed to disorganize DSB repair pathways to sensitize tumors to DNA damaging therapies such as radiotherapy and chemotherapy. We used the property of AsiDNA of triggering artificial DNA damage signaling to examine the activation of DSB repair pathways and to study the main steps of inhibition of DNA repair foci after irradiation. We show that, upon AsiDNA cellular uptake, cytoplasmic ATM and PARP are rapidly activated (within one hour) even in the absence of irradiation. ATM activation by AsiDNA leads to its transient autophosphorylation and sequestration in the cytoplasm, preventing the formation of ATM nuclear foci on irradiation-induced damage. In contrast, the activation of PARP did not seem to alter its ability to form DNA repair foci, but prevented 53BP1 and XRCC4 recruitment at the damage sites. In the nucleus, AsiDNA is essentially associated with DNA-PK, which triggers its activation leading to phosphorylation of H2AX all over chromatin. This pan-nuclear phosphorylation of H2AX correlates with the massive inhibition, at damage sites induced by irradiation, of the recruitment of repair enzymes involved in DSB repair by homologous recombination and nonhomologous end joining. These results highlight the interest in a new generation of DNA repair inhibitors targeting DNA damage signaling.
Collapse
Affiliation(s)
- Nathalie Berthault
- Institut Curie, PSL Research University, CNRS, INSERM, UMR 3347, 91405 Orsay, France; (N.B.); (F.P.); (P.-M.G.)
- Université Paris-Saclay, CNRS, UMR 3347, 91405 Orsay, France
| | - Ptissam Bergam
- Institut Curie, PSL Research University, CNRS, INSERM, UMS 2016, Multimodal Imaging Centre, 91405 Orsay, France;
- Université Paris-Saclay, CNRS, UMS 2016, 91405 Orsay, France
| | - Floriane Pereira
- Institut Curie, PSL Research University, CNRS, INSERM, UMR 3347, 91405 Orsay, France; (N.B.); (F.P.); (P.-M.G.)
- Université Paris-Saclay, CNRS, UMR 3347, 91405 Orsay, France
| | - Pierre-Marie Girard
- Institut Curie, PSL Research University, CNRS, INSERM, UMR 3347, 91405 Orsay, France; (N.B.); (F.P.); (P.-M.G.)
- Université Paris-Saclay, CNRS, UMR 3347, 91405 Orsay, France
| | - Marie Dutreix
- Institut Curie, PSL Research University, CNRS, INSERM, UMR 3347, 91405 Orsay, France; (N.B.); (F.P.); (P.-M.G.)
- Université Paris-Saclay, CNRS, UMR 3347, 91405 Orsay, France
- Correspondence:
| |
Collapse
|
13
|
Hasan A, Rizvi SF, Parveen S, Mir SS. Molecular chaperones in DNA repair mechanisms: Role in genomic instability and proteostasis in cancer. Life Sci 2022; 306:120852. [DOI: 10.1016/j.lfs.2022.120852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/14/2022] [Accepted: 07/27/2022] [Indexed: 01/09/2023]
|
14
|
Sottile ML, Cuello-Carrión FD, Gómez LC, Semino S, Ibarra J, García MB, Gonzalez L, Vargas-Roig LM, Nadin SB. DNA Damage Repair Proteins, HSP27, and Phosphorylated-HSP90α as Predictive/Prognostic Biomarkers of Platinum-based Cancer Chemotherapy: An Exploratory Study. Appl Immunohistochem Mol Morphol 2022; 30:425-434. [PMID: 35639358 DOI: 10.1097/pai.0000000000001037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 04/25/2022] [Indexed: 11/27/2022]
Abstract
Platinum analogs are commonly used for cancer treatment. There is increasing interest in finding biomarkers which could predict and overcome resistance, because to date there is no reliable predictive/prognostic marker for these compounds. Here we studied the immunohistochemical expression of proteins involved in DNA damage response and repair (γH2AX, 53BP1, ERCC1, MLH1, and MSH2) in primary tumor tissues from patients treated with platinum-based chemotherapy. Levels and localization of Heat Shock Protein (HSP)27 and phospho-(Thr5/7)-HSP90α (p-HSP90α) were also determined. The implications in clinical response, disease-free survival and overall survival were analyzed. High γH2AX and 53BP1 expressions were associated with poor clinical response. Nuclear p-HSP90α, as well as nuclear absence and low cytoplasmic expression of HSP27 correlated with good response. Patients with high γH2AX and high cytoplasmic HSP27 expressions had shorter overall survival and disease-free survival. MLH1, MSH2, or ERCC1 were not associated with clinical response or survival. We report the potential utility of p-HSP90α, HSP27, γH2AX, and 53BP1 as predictive/prognostic markers for platinum-based chemotherapy. We present the first study that evaluates the predictive and prognostic value of p-HSP90α in primary tumors. Our research opens new possibilities for clinical oncology and shows the usefulness of immunohistochemistry for predicting chemotherapy response and prognosis in cancer.
Collapse
Affiliation(s)
- Mayra L Sottile
- Tumor Biology Laboratory
- Medical Sciences School, Mendoza University
| | | | - Laura C Gómez
- Tumor Biology Laboratory
- Medical Sciences School, Mendoza University
| | | | - Jorge Ibarra
- Regional Integration Cancer Center, Mendoza, Argentina
| | | | | | | | | |
Collapse
|
15
|
Ijaz M, Li X, Zhang D, Bai Y, Hou C, Hussain Z, Zheng X, Huang C. Sarcoplasmic and myofibrillar phosphoproteins profile of beef M. longissimus thoracis with different pH u at different days postmortem. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:2464-2471. [PMID: 34642961 DOI: 10.1002/jsfa.11586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/27/2021] [Accepted: 10/12/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The abnormal ultimate pH (pHu ) in postmortem muscles affect the meat quality and results in substantial economic losses. Dark, firm, and dry (DFD) meat linked with the higher postmortem pHu values and exhibited many quality issues such as dark color, tough texture and shorter shelf life. This research aimed to investigate the effect of protein phosphorylation on variations in beef pHu in order to explore the possible mechanisms underlying DFD meat formation. RESULTS Glycogen and lactate contents were higher, while L* and a* were lower in high pHu beef. Shear force was higher in intermediate pHu group. Global phosphorylation of sarcoplasmic proteins was higher in low pHu samples on day 1 and of myofibrillar proteins was higher in intermediate pHu meat on days 1 and 2 postmortem. Sarcoplasmic protein bands with different phosphorylation levels were identified as containing some glycometabolism and stress response proteins and phosphorylated myofibrillar protein bands were identified sarcomeric and metabolic proteins. CONCLUSIONS Phosphorylation of multiple proteins of glycolytic pathway and contractile machinery may play critical roles in development of DFD beef. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Muawuz Ijaz
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-products Quality & Safety in Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing, P. R. China
- Department of Animal Sciences, CVAS-Jhang 35200, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Xin Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-products Quality & Safety in Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing, P. R. China
| | - Dequan Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-products Quality & Safety in Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing, P. R. China
| | - Yuqiang Bai
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-products Quality & Safety in Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing, P. R. China
| | - Chengli Hou
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-products Quality & Safety in Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing, P. R. China
| | - Zubair Hussain
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-products Quality & Safety in Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing, P. R. China
| | - Xiaochun Zheng
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-products Quality & Safety in Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing, P. R. China
| | - Caiyan Huang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-products Quality & Safety in Harvest, Storage, Transportation, Management and Control, Ministry of Agriculture and Rural Affairs, Beijing, P. R. China
| |
Collapse
|
16
|
Wang S, Zhu H, Li R, Mui D, Toan S, Chang X, Zhou H. DNA-PKcs interacts with and phosphorylates Fis1 to induce mitochondrial fragmentation in tubular cells during acute kidney injury. Sci Signal 2022; 15:eabh1121. [PMID: 35290083 DOI: 10.1126/scisignal.abh1121] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) regulates cell death. We sought to determine whether DNA-PKcs played a role in the tubular damage that occurs during acute kidney injury (AKI) induced by LPS injection (to mimic sepsis), cisplatin administration, or renal ischemia/reperfusion injury. Although DNA-PKcs normally localizes to the nucleus, we detected cytoplasmic DNA-PKcs in mouse kidney tissues and urinary sediments of human patients with septic AKI. Increased cytoplasmic amounts of DNA-PKcs correlated with renal dysfunction. Tubule cell-specific DNA-PKcs deletion attenuated AKI-mediated tubular cell death and changes in the abundance of various proteins with mitochondrial functions or roles in apoptotic pathways. DNA-PKcs interacted with Fis1 and phosphorylated it at Thr34 in its TQ motif, which increased the affinity of Fis1 for Drp1 and induced mitochondrial fragmentation. Knockin mice expressing a nonphosphorylatable T34A mutant exhibited improved renal function and histological features and reduced mitochondrial fragmentation upon induction of AKI. Phosphorylation of Thr34 in Fis1 was detectable in urinary sediments of human patients with septic AKI and correlated with renal dysfunction. Our findings provide insight into the role of cytoplasmic DNA-PKcs and phosphorylated Fis1 in AKI development.
Collapse
Affiliation(s)
- Shiyuan Wang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Hang Zhu
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Ruibing Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - David Mui
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sam Toan
- Department of Chemical Engineering, University of Minnesota-Duluth, Duluth, MN 55812, USA
| | - Xing Chang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China
| | - Hao Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, China.,Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| |
Collapse
|
17
|
Dylgjeri E, Knudsen KE. DNA-PKcs: A Targetable Protumorigenic Protein Kinase. Cancer Res 2022; 82:523-533. [PMID: 34893509 PMCID: PMC9306356 DOI: 10.1158/0008-5472.can-21-1756] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/17/2021] [Accepted: 11/10/2021] [Indexed: 01/07/2023]
Abstract
DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a pleiotropic protein kinase that plays critical roles in cellular processes fundamental to cancer. DNA-PKcs expression and activity are frequently deregulated in multiple hematologic and solid tumors and have been tightly linked to poor outcome. Given the potentially influential role of DNA-PKcs in cancer development and progression, therapeutic targeting of this kinase is being tested in preclinical and clinical settings. This review summarizes the latest advances in the field, providing a comprehensive discussion of DNA-PKcs functions in cancer and an update on the clinical assessment of DNA-PK inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Emanuela Dylgjeri
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Karen E. Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Urology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania.,Corresponding Author: Karen E. Knudsen, Thomas Jefferson University, 233 South 10th Street, BLSB 1050, Philadelphia, PA 19107. Phone: 215-503-5692; E-mail:
| |
Collapse
|
18
|
Unsymmetrical Trifluoromethyl Methoxyphenyl β-Diketones: Effect of the Position of Methoxy Group and Coordination at Cu(II) on Biological Activity. Molecules 2021; 26:molecules26216466. [PMID: 34770875 PMCID: PMC8588221 DOI: 10.3390/molecules26216466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/06/2021] [Accepted: 10/21/2021] [Indexed: 12/09/2022] Open
Abstract
Copper(II) complexes with 1,1,1-trifluoro-4-(4-methoxyphenyl)butan-2,4-dione (HL1) were synthesized and characterized by elemental analysis, FT-IR spectroscopy, and single crystal X-ray diffraction. The biological properties of HL1 and cis-[Cu(L1)2(DMSO)] (3) were examined against Gram-positive and Gram-negative bacteria and opportunistic unicellular fungi. The cytotoxicity was estimated towards the HeLa and Vero cell lines. Complex 3 demonstrated antibacterial activity towards S. aureus comparable to that of streptomycin, lower antifungal activity than the ligand HL1 and moderate cytotoxicity. The bioactivity was compared with the activity of compounds of similar structures. The effect of changing the position of the methoxy group at the aromatic ring in the ligand moiety of the complexes on their antimicrobial and cytotoxic activity was explored. We propose that complex 3 has lower bioavailability and reduced bioactivity than expected due to strong intermolecular contacts. In addition, molecular docking studies provided theoretical information on the interactions of tested compounds with ribonucleotide reductase subunit R2, as well as the chaperones Hsp70 and Hsp90, which are important biomolecular targets for antitumor and antimicrobial drug search and design. The obtained results revealed that the complexes displayed enhanced affinity over organic ligands. Taken together, the copper(II) complexes with the trifluoromethyl methoxyphenyl-substituted β-diketones could be considered as promising anticancer agents with antibacterial properties.
Collapse
|
19
|
Birbo B, Madu EE, Madu CO, Jain A, Lu Y. Role of HSP90 in Cancer. Int J Mol Sci 2021; 22:ijms221910317. [PMID: 34638658 PMCID: PMC8508648 DOI: 10.3390/ijms221910317] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 11/25/2022] Open
Abstract
HSP90 is a vital chaperone protein conserved across all organisms. As a chaperone protein, it correctly folds client proteins. Structurally, this protein is a dimer with monomer subunits that consist of three main conserved domains known as the N-terminal domain, middle domain, and the C-terminal domain. Multiple isoforms of HSP90 exist, and these isoforms share high homology. These isoforms are present both within the cell and outside the cell. Isoforms HSP90α and HSP90β are present in the cytoplasm; TRAP1 is present in the mitochondria; and GRP94 is present in the endoplasmic reticulum and is likely secreted due to post-translational modifications (PTM). HSP90 is also secreted into an extracellular environment via an exosome pathway that differs from the classic secretion pathway. Various co-chaperones are necessary for HSP90 to function. Elevated levels of HSP90 have been observed in patients with cancer. Despite this observation, the possible role of HSP90 in cancer was overlooked because the chaperone was also present in extreme amounts in normal cells and was vital to normal cell function, as observed when the drastic adverse effects resulting from gene knockout inhibited the production of this protein. Differences between normal HSP90 and HSP90 of the tumor phenotype have been better understood and have aided in making the chaperone protein a target for cancer drugs. One difference is in the conformation: HSP90 of the tumor phenotype is more susceptible to inhibitors. Since overexpression of HSP90 is a factor in tumorigenesis, HSP90 inhibitors have been studied to combat the adverse effects of HSP90 overexpression. Monotherapies using HSP90 inhibitors have shown some success; however, combination therapies have shown better results and are thus being studied for a more effective cancer treatment.
Collapse
Affiliation(s)
- Bereket Birbo
- Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Elechi E. Madu
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (E.E.M.); (C.O.M.); (A.J.)
| | - Chikezie O. Madu
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (E.E.M.); (C.O.M.); (A.J.)
| | - Aayush Jain
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (E.E.M.); (C.O.M.); (A.J.)
| | - Yi Lu
- Health Science Center, Department of Pathology and Laboratory Medicine, University of Tennessee, Memphis, TN 38163, USA
- Correspondence: ; Tel.: +1-(901)-448-5436; Fax: +1-(901)-448-5496
| |
Collapse
|
20
|
Gao JJ, Wang B, Peng RH, Li ZJ, Xu J, Tian YS, Yao QH. Phytoremediation of multiple persistent pollutants co-contaminated soil by HhSSB transformed plant. ENVIRONMENTAL RESEARCH 2021; 197:110959. [PMID: 33722526 DOI: 10.1016/j.envres.2021.110959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/28/2021] [Accepted: 02/27/2021] [Indexed: 06/12/2023]
Abstract
The high toxicity of persistent pollutants limits the phytoremediation of pollutants-contaminated soil. In this study, heterologous expressing Halorhodospira halophila single-stranded DNA binding protein gene (HhSSB) improves tolerance to 2,4,6-trinitrotoluene (TNT), 2,4,6-trichlorophenol (2,4,6-TCP), and thiocyanate (SCN-) in A. thaliana and tall fescue (Festuca arundinacea). The HhSSB transformed Arabidopsis, and tall fescue also exhibited enhanced phytoremediation of TNT, 2,4,6-TCP, and SCN- separately contaminated soil and co-contaminated soil compared to control plants. TNT assay was selected to explore the mechanism of how HhSSB enhances the phytoremediation of persistent pollutants. Our result indicates that HhSSB enhances the phytoremediation of TNT by enhancing the transformation of TNT in Arabidopsis. Moreover, transcriptomics and comet analysis revealed that HhSSB improves TNT tolerance through three pathways: strengthening the defense system, enhancing the ROS scavenging system, and reducing DNA damage. These results presented here would be particularly useful for further studies in the remediation of soil contaminated by organic and inorganic pollutants.
Collapse
Affiliation(s)
- Jian-Jie Gao
- Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China; Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China
| | - Bo Wang
- Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China; Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China
| | - Ri-He Peng
- Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China; Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China
| | - Zhen-Jun Li
- Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China; Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China
| | - Jing Xu
- Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China; Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China
| | - Yong-Sheng Tian
- Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China; Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China
| | - Quan-Hong Yao
- Biotechnology Research Institute, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China; Shanghai Key Laboratory of Agricultural Genetics and Breeding, Shanghai Academy of Agricultural Sciences, Shanghai, 201106, China.
| |
Collapse
|
21
|
Yu L, Lang Y, Guo J, Cai J, Shang ZF, Chen BPC. DNA-PKcs inhibition impairs HDAC6-mediated HSP90 chaperone function on Aurora A and enhances HDACs inhibitor-induced cell killing by increasing mitotic aberrant spindle assembly. Cell Cycle 2021; 20:211-224. [PMID: 33404279 DOI: 10.1080/15384101.2020.1867790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Combining targeted therapeutic agents is an attractive cancer treatment strategy associated with high efficacy and low toxicity. DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is an essential factor in DNA damage repair. Studies from us and others have revealed that DNA-PKcs also plays an important role in normal mitosis progression. Histone deacetylase (HDACs) inhibitors commonly lead to mitotic aberration and have been approved for treating various cancers in the clinic. We showed that DNA-PKcs depletion or kinase activity inhibition increases cancer cells' sensitivity to HDACs inhibitors in vitro and in vivo. DNA-PKcs deficiency significantly enhances HDACs inhibitors (HDACi)-induced mitotic arrest and is followed by apoptotic cell death. Mechanistically, we found that DNA-PKcs binds to HDAC6 and facilitates its acetylase activity. HDACi is more likely to impair HDAC6-induced deacetylation of HSP90 and abrogate HSP90's chaperone function on Aurora A, a critical mitotic kinase that regulates centrosome separation and mitotic spindle assembly in DNA-PKcs-deficient cells. Our current work indicates crosstalk between DNA-PKcs and HDACs signaling pathways, and highlights that the combined targeting of DNA-PKcs and HDACs can be used in cancer therapy. Abbreviations: DNA-PKcs, DNA-dependent protein kinase catalytic subunit, HDACs, Histone deacetylases, DSBs, DNA double-strand breaks, ATM, ataxia telangiectasia mutated, ATR, ATM-Rad3-related.
Collapse
Affiliation(s)
- Lan Yu
- Department of Radiation Oncology, Simmons Comprehensive Cancer Center at UT Southwestern Medical Center , Dallas, TX, USA
| | - Yue Lang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University , Suzhou, China
| | - Jiaming Guo
- Department of Radiation Oncology, Simmons Comprehensive Cancer Center at UT Southwestern Medical Center , Dallas, TX, USA.,Department of Radiation Medicine, College of Naval Medicine, Naval Medical University , Shanghai, China
| | - Jianming Cai
- Department of Radiation Medicine, College of Naval Medicine, Naval Medical University , Shanghai, China
| | - Zeng-Fu Shang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University , Suzhou, China
| | - Benjamin P C Chen
- Department of Radiation Oncology, Simmons Comprehensive Cancer Center at UT Southwestern Medical Center , Dallas, TX, USA
| |
Collapse
|
22
|
Meng F, Liang Z, Zhao K, Luo C. Drug design targeting active posttranslational modification protein isoforms. Med Res Rev 2020; 41:1701-1750. [PMID: 33355944 DOI: 10.1002/med.21774] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/29/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022]
Abstract
Modern drug design aims to discover novel lead compounds with attractable chemical profiles to enable further exploration of the intersection of chemical space and biological space. Identification of small molecules with good ligand efficiency, high activity, and selectivity is crucial toward developing effective and safe drugs. However, the intersection is one of the most challenging tasks in the pharmaceutical industry, as chemical space is almost infinity and continuous, whereas the biological space is very limited and discrete. This bottleneck potentially limits the discovery of molecules with desirable properties for lead optimization. Herein, we present a new direction leveraging posttranslational modification (PTM) protein isoforms target space to inspire drug design termed as "Post-translational Modification Inspired Drug Design (PTMI-DD)." PTMI-DD aims to extend the intersections of chemical space and biological space. We further rationalized and highlighted the importance of PTM protein isoforms and their roles in various diseases and biological functions. We then laid out a few directions to elaborate the PTMI-DD in drug design including discovering covalent binding inhibitors mimicking PTMs, targeting PTM protein isoforms with distinctive binding sites from that of wild-type counterpart, targeting protein-protein interactions involving PTMs, and hijacking protein degeneration by ubiquitination for PTM protein isoforms. These directions will lead to a significant expansion of the biological space and/or increase the tractability of compounds, primarily due to precisely targeting PTM protein isoforms or complexes which are highly relevant to biological functions. Importantly, this new avenue will further enrich the personalized treatment opportunity through precision medicine targeting PTM isoforms.
Collapse
Affiliation(s)
- Fanwang Meng
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Zhongjie Liang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Kehao Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Cheng Luo
- Drug Discovery and Design Center, the Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
23
|
Anisenko A, Kan M, Shadrina O, Brattseva A, Gottikh M. Phosphorylation Targets of DNA-PK and Their Role in HIV-1 Replication. Cells 2020; 9:E1907. [PMID: 32824372 PMCID: PMC7464883 DOI: 10.3390/cells9081907] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
The DNA dependent protein kinase (DNA-PK) is a trimeric nuclear complex consisting of a large protein kinase and the Ku heterodimer. The kinase activity of DNA-PK is required for efficient repair of DNA double-strand breaks (DSB) by non-homologous end joining (NHEJ). We also showed that the kinase activity of DNA-PK is essential for post-integrational DNA repair in the case of HIV-1 infection. Besides, DNA-PK is known to participate in such cellular processes as protection of mammalian telomeres, transcription, and some others where the need for its phosphorylating activity is not clearly elucidated. We carried out a systematic search and analysis of DNA-PK targets described in the literature and identified 67 unique DNA-PK targets phosphorylated in response to various in vitro and/or in vivo stimuli. A functional enrichment analysis of DNA-PK targets and determination of protein-protein associations among them were performed. For 27 proteins from these 67 DNA-PK targets, their participation in the HIV-1 life cycle was demonstrated. This information may be useful for studying the functioning of DNA-PK in various cellular processes, as well as in various stages of HIV-1 replication.
Collapse
Affiliation(s)
- Andrey Anisenko
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Marina Kan
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Olga Shadrina
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Anna Brattseva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119234, Russia;; (M.K.); (A.B.)
| | - Marina Gottikh
- Chemistry Department and Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (O.S.); (M.G.)
| |
Collapse
|
24
|
Girard P, Berthault N, Kozlac M, Ferreira S, Jdey W, Bhaskara S, Alekseev S, Thomas F, Dutreix M. Evolution of tumor cells during AsiDNA treatment results in energy exhaustion, decrease in responsiveness to signal, and higher sensitivity to the drug. Evol Appl 2020; 13:1673-1680. [PMID: 32821277 PMCID: PMC7428804 DOI: 10.1111/eva.12949] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/28/2020] [Accepted: 02/19/2020] [Indexed: 01/08/2023] Open
Abstract
It is increasingly suggested that ecological and evolutionary sciences could inspire novel therapies against cancer but medical evidence of this remains scarce at the moment. The Achilles heel of conventional and targeted anticancer treatments is intrinsic or acquired resistance following Darwinian selection; that is, treatment toxicity places the surviving cells under intense evolutionary selective pressure to develop resistance. Here, we review a set of data that demonstrate that Darwinian principles derived from the "smoke detector" principle can instead drive the evolution of malignant cells toward a different trajectory. Specifically, long-term exposure of cancer cells to a strong alarm signal, generated by the DNA repair inhibitor AsiDNA, induces a stable new state characterized by a down-regulation of the targeted pathways and does not generate resistant clones. This property is due to the original mechanism of action of AsiDNA, which acts by overactivating a "false" signaling of DNA damage through DNA-PK and PARP enzymes, and is not observed with classical DNA repair inhibitors such as the PARP inhibitors. Long-term treatment with AsiDNA induces a new "alarm down" state in the tumor cells with decrease in NAD level and reactiveness to it. These results suggest that agonist drugs such as AsiDNA could promote a state-dependent tumor cell evolution by lowering their ability to respond to high "danger" signal. This analysis provides a compelling argument that evolutionary ecology could help drug design development in overcoming fundamental limitation of novel therapies against cancer due to the modification of the targeted tumor cell population during treatment.
Collapse
Affiliation(s)
- Pierre‐Marie Girard
- Institut CurieCNRSINSERMUMR 3347PSL Research UniversityOrsayFrance
- CNRSINSERMUMR 3347Université Paris‐SudUniversité Paris‐SaclayFOrsayFrance
| | - Nathalie Berthault
- Institut CurieCNRSINSERMUMR 3347PSL Research UniversityOrsayFrance
- CNRSINSERMUMR 3347Université Paris‐SudUniversité Paris‐SaclayFOrsayFrance
| | - Maria Kozlac
- Institut CurieCNRSINSERMUMR 3347PSL Research UniversityOrsayFrance
- CNRSINSERMUMR 3347Université Paris‐SudUniversité Paris‐SaclayFOrsayFrance
| | - Sofia Ferreira
- Institut CurieCNRSINSERMUMR 3347PSL Research UniversityOrsayFrance
- CNRSINSERMUMR 3347Université Paris‐SudUniversité Paris‐SaclayFOrsayFrance
| | - Wael Jdey
- Institut CurieCNRSINSERMUMR 3347PSL Research UniversityOrsayFrance
- CNRSINSERMUMR 3347Université Paris‐SudUniversité Paris‐SaclayFOrsayFrance
- OnxeoParisFrance
| | - Srividya Bhaskara
- Huntsman Cancer InstituteUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Sergey Alekseev
- Institut CurieCNRSINSERMUMR 3347PSL Research UniversityOrsayFrance
- CNRSINSERMUMR 3347Université Paris‐SudUniversité Paris‐SaclayFOrsayFrance
| | - Frederic Thomas
- CREEC/MIVEGECUMR IRD 224‐CNRS 5290Université de MontpellierMontpellierFrance
| | - Marie Dutreix
- Institut CurieCNRSINSERMUMR 3347PSL Research UniversityOrsayFrance
- CNRSINSERMUMR 3347Université Paris‐SudUniversité Paris‐SaclayFOrsayFrance
| |
Collapse
|
25
|
Agarwal S, Ganesh S. Perinuclear mitochondrial clustering, increased ROS levels, and HIF1 are required for the activation of HSF1 by heat stress. J Cell Sci 2020; 133:jcs245589. [PMID: 32503939 DOI: 10.1242/jcs.245589] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/19/2020] [Indexed: 08/31/2023] Open
Abstract
The heat shock response (HSR) is a conserved cellular defensive response against stresses such as temperature, oxidative stress and heavy metals. A significant group of players in the HSR is the set of molecular chaperones known as heat shock proteins (HSPs), which assist in the refolding of unfolded proteins and prevent the accumulation of damaged proteins. HSP genes are activated by the HSF1 transcription factor, a master regulator of the HSR pathway. A variety of stressors activate HSF1, but the key molecular players and the processes that directly contribute to HSF1 activation remain unclear. In this study, we show that heat shock induces perinuclear clustering of mitochondria in mammalian cells, and this clustering is essential for activation of the HSR. We also show that this perinuclear clustering of mitochondria results in increased levels of reactive oxygen species in the nucleus, leading to the activation of hypoxia-inducible factor-1α (HIF-1α). To conclude, we provide evidence to suggest that HIF-1α is one of the crucial regulators of HSF1 and that HIF-1α is essential for activation of the HSR during heat shock.
Collapse
Affiliation(s)
- Saloni Agarwal
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology, Kanpur 208016, India
| |
Collapse
|
26
|
Backe SJ, Sager RA, Woodford MR, Makedon AM, Mollapour M. Post-translational modifications of Hsp90 and translating the chaperone code. J Biol Chem 2020; 295:11099-11117. [PMID: 32527727 DOI: 10.1074/jbc.rev120.011833] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cells have a remarkable ability to synthesize large amounts of protein in a very short period of time. Under these conditions, many hydrophobic surfaces on proteins may be transiently exposed, and the likelihood of deleterious interactions is quite high. To counter this threat to cell viability, molecular chaperones have evolved to help nascent polypeptides fold correctly and multimeric protein complexes assemble productively, while minimizing the danger of protein aggregation. Heat shock protein 90 (Hsp90) is an evolutionarily conserved molecular chaperone that is involved in the stability and activation of at least 300 proteins, also known as clients, under normal cellular conditions. The Hsp90 clients participate in the full breadth of cellular processes, including cell growth and cell cycle control, signal transduction, DNA repair, transcription, and many others. Hsp90 chaperone function is coupled to its ability to bind and hydrolyze ATP, which is tightly regulated both by co-chaperone proteins and post-translational modifications (PTMs). Many reported PTMs of Hsp90 alter chaperone function and consequently affect myriad cellular processes. Here, we review the contributions of PTMs, such as phosphorylation, acetylation, SUMOylation, methylation, O-GlcNAcylation, ubiquitination, and others, toward regulation of Hsp90 function. We also discuss how the Hsp90 modification state affects cellular sensitivity to Hsp90-targeted therapeutics that specifically bind and inhibit its chaperone activity. The ultimate challenge is to decipher the comprehensive and combinatorial array of PTMs that modulate Hsp90 chaperone function, a phenomenon termed the "chaperone code."
Collapse
Affiliation(s)
- Sarah J Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Rebecca A Sager
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA.,College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Mark R Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Alan M Makedon
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA .,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
27
|
Testosterone amplifies HSP70-2a, HSP90 and PCNA expression in experimental varicocele condition: Implication for DNA fragmentation. Reprod Biol 2020; 20:384-395. [PMID: 32461192 DOI: 10.1016/j.repbio.2020.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 01/10/2023]
Abstract
The DNA fragmentation and failure in post-meiotic maturation of the spermatozoa because of testosterone withdrawal can affect the fertilization potential in varicocele (VCL) patients. To find out the exact mechanism of VCL-induced failure in histone-protamine replacement process and DNA fragmentation, the correlations between the levels of expression of HSP70-2a, HSP90, PCNA, TP1/2 and PCNA genes and the patterns of DNA methylation were investigated before and after testosterone administration in rats. In total, 40 mature male Wistar rats (10 in each group) were assigned between control (with no intervention), control-sham (undergone a simple laparotomy), VCL-induced (VCL-sole), and testosterone-treated VCL-induced (VCLT) groups. The HSP70-2a, HSP90, PCNA, TP1, and TP2 genes expressions and the patterns of global DNA methylation were determined in all groups. A statistically significant (p < 0.05) reduction were found in the HSP70-2a, HSP90, PCNA, TP1 and TP2 genes expressions in VCL-sole group. In VCLT group, testosterone was shown to significantly (p < 0.05) up-regulate the HSP70-2a, HSP90, PCNA, and TP2expression levels, but TP1 expression has not been changed. Furthermore, the VCLT group exhibited higher DNA methylation rates compared to VCL-sole animals. In conclusion, testosterone, by up-regulating the HSP70-2a and HSP90 expressions and maintaining the pre-existing HSP70-2a and HSP90 proteins levels, may be the reason for the significant increment in TP2 expression during post-meiotic stage and can boost the global methylation rates of DNA via up-regulating the PCNA expression, suggesting that administration of testosterone can mitigate the VCL-impaired histone-protamine replacement and DNA methylation rates and protect the cellular DNA content from VCL-induced oxidative stress.
Collapse
|
28
|
Gvozdenov Z, Kolhe J, Freeman BC. The Nuclear and DNA-Associated Molecular Chaperone Network. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a034009. [PMID: 30745291 PMCID: PMC6771373 DOI: 10.1101/cshperspect.a034009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Maintenance of a healthy and functional proteome in all cellular compartments is critical to cell and organismal homeostasis. Yet, our understanding of the proteostasis process within the nucleus is limited. Here, we discuss the identified roles of the major molecular chaperones Hsp90, Hsp70, and Hsp60 with client proteins working in diverse DNA-associated pathways. The unique challenges facing proteins in the nucleus are considered as well as the conserved features of the molecular chaperone system in facilitating DNA-linked processes. As nuclear protein inclusions are a common feature of protein-aggregation diseases (e.g., neurodegeneration), a better understanding of nuclear proteostasis is warranted.
Collapse
Affiliation(s)
- Zlata Gvozdenov
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois 61801.,Department Chemie, Technische Universität München, Garching 85748, Germany
| | - Janhavi Kolhe
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois 61801
| | - Brian C Freeman
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois 61801
| |
Collapse
|
29
|
Dubrez L, Causse S, Borges Bonan N, Dumétier B, Garrido C. Heat-shock proteins: chaperoning DNA repair. Oncogene 2019; 39:516-529. [DOI: 10.1038/s41388-019-1016-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/04/2019] [Accepted: 09/06/2019] [Indexed: 02/08/2023]
|
30
|
Biau J, Chautard E, Berthault N, de Koning L, Court F, Pereira B, Verrelle P, Dutreix M. Combining the DNA Repair Inhibitor Dbait With Radiotherapy for the Treatment of High Grade Glioma: Efficacy and Protein Biomarkers of Resistance in Preclinical Models. Front Oncol 2019; 9:549. [PMID: 31275862 PMCID: PMC6593092 DOI: 10.3389/fonc.2019.00549] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/05/2019] [Indexed: 12/23/2022] Open
Abstract
High grade glioma relapses occur often within the irradiated volume mostly due to a high resistance to radiation therapy (RT). Dbait (which stands for DNA strand break bait) molecules mimic DSBs and trap DNA repair proteins, thereby inhibiting repair of DNA damage induced by RT. Here we evaluate the potential of Dbait to sensitize high grade glioma to RT. First, we demonstrated the radiosensitizer properties of Dbait in 6/9 tested cell lines. Then, we performed animal studies using six cell derived xenograft and five patient derived xenograft models, to show the clinical potential and applicability of combined Dbait+RT treatment for human high grade glioma. Using a RPPA approach, we showed that Phospho-H2AX/H2AX and Phospho-NBS1/NBS1 were predictive of Dbait efficacy in xenograft models. Our results provide the preclinical proof of concept that combining RT with Dbait inhibition of DNA repair could be of benefit to patients with high grade glioma.
Collapse
Affiliation(s)
- Julian Biau
- Centre de Recherche, Institut Curie, PSL Research University, Paris, France.,UMR3347, CNRS, Orsay, France.,U1021, INSERM, Orsay, France.,Research Department, Université Paris Sud, Orsay, France.,INSERM, U1240 IMoST, Université Clermont Auvergne, Clermont Ferrand, France.,Radiotherapy Department, Centre Jean Perrin, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Emmanuel Chautard
- INSERM, U1240 IMoST, Université Clermont Auvergne, Clermont Ferrand, France.,Pathology Department, Centre Jean Perrin, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Nathalie Berthault
- Centre de Recherche, Institut Curie, PSL Research University, Paris, France.,UMR3347, CNRS, Orsay, France.,U1021, INSERM, Orsay, France.,Research Department, Université Paris Sud, Orsay, France
| | - Leanne de Koning
- Laboratory of Proteomic Mass Spectrometry, Centre de Recherche, Institut Curie, Paris, France.,Department of Translational Research, Institut Curie, PSL Research University, Paris, France
| | - Frank Court
- GReD Laboratory, CNRS UMR 6293, INSERM U1103, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Bruno Pereira
- Biostatistics Department, DRCI, Clermont-Ferrand Hospital, Clermont-Ferrand, France
| | - Pierre Verrelle
- Centre de Recherche, Institut Curie, PSL Research University, Paris, France.,Radiotherapy Department, Centre Jean Perrin, Université Clermont Auvergne, Clermont-Ferrand, France.,U1196, INSERM, UMR9187, CNRS, Orsay, France.,Radiotherapy Department, Institut Curie Hospital, Paris, France
| | - Marie Dutreix
- Centre de Recherche, Institut Curie, PSL Research University, Paris, France.,UMR3347, CNRS, Orsay, France.,U1021, INSERM, Orsay, France.,Research Department, Université Paris Sud, Orsay, France
| |
Collapse
|
31
|
Abreu PL, Ferreira LMR, Cunha-Oliveira T, Alpoim MC, Urbano AM. HSP90: A Key Player in Metal-Induced Carcinogenesis? HEAT SHOCK PROTEINS 2019. [DOI: 10.1007/978-3-030-23158-3_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
32
|
Mahmoudi K, Bouras A, Bozec D, Ivkov R, Hadjipanayis C. Magnetic hyperthermia therapy for the treatment of glioblastoma: a review of the therapy's history, efficacy and application in humans. Int J Hyperthermia 2018; 34:1316-1328. [PMID: 29353516 PMCID: PMC6078833 DOI: 10.1080/02656736.2018.1430867] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/14/2018] [Accepted: 01/17/2018] [Indexed: 02/07/2023] Open
Abstract
Hyperthermia therapy (HT) is the exposure of a region of the body to elevated temperatures to achieve a therapeutic effect. HT anticancer properties and its potential as a cancer treatment have been studied for decades. Techniques used to achieve a localised hyperthermic effect include radiofrequency, ultrasound, microwave, laser and magnetic nanoparticles (MNPs). The use of MNPs for therapeutic hyperthermia generation is known as magnetic hyperthermia therapy (MHT) and was first attempted as a cancer therapy in 1957. However, despite more recent advancements, MHT has still not become part of the standard of care for cancer treatment. Certain challenges, such as accurate thermometry within the tumour mass and precise tumour heating, preclude its widespread application as a treatment modality for cancer. MHT is especially attractive for the treatment of glioblastoma (GBM), the most common and aggressive primary brain cancer in adults, which has no cure. In this review, the application of MHT as a therapeutic modality for GBM will be discussed. Its therapeutic efficacy, technical details, and major experimental and clinical findings will be reviewed and analysed. Finally, current limitations, areas of improvement, and future directions will be discussed in depth.
Collapse
Affiliation(s)
- Keon Mahmoudi
- Department of Neurosurgery, Brain Tumor Nanotechnology Laboratory, Tisch Cancer Institute at Mount Sinai, New York, NY, USA
| | - Alexandros Bouras
- Department of Neurosurgery, Brain Tumor Nanotechnology Laboratory, Tisch Cancer Institute at Mount Sinai, New York, NY, USA
| | - Dominique Bozec
- Department of Neurosurgery, Brain Tumor Nanotechnology Laboratory, Tisch Cancer Institute at Mount Sinai, New York, NY, USA
| | - Robert Ivkov
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine Baltimore, MD, USA
| | - Constantinos Hadjipanayis
- Department of Neurosurgery, Brain Tumor Nanotechnology Laboratory, Tisch Cancer Institute at Mount Sinai, New York, NY, USA
- Department of Neurosurgery, Mount Sinai Beth Israel, New York, NY, USA
| |
Collapse
|
33
|
The Hsp70 co-chaperone Ydj1/HDJ2 regulates ribonucleotide reductase activity. PLoS Genet 2018; 14:e1007462. [PMID: 30452489 PMCID: PMC6277125 DOI: 10.1371/journal.pgen.1007462] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/03/2018] [Accepted: 10/11/2018] [Indexed: 11/19/2022] Open
Abstract
Hsp70 is a well-conserved molecular chaperone involved in the folding, stabilization, and eventual degradation of many “client” proteins. Hsp70 is regulated by a suite of co-chaperone molecules that assist in Hsp70-client interaction and stimulate the intrinsic ATPase activity of Hsp70. While previous studies have shown the anticancer target ribonucleotide reductase (RNR) is a client of Hsp70, the regulatory co-chaperones involved remain to be determined. To identify co-chaperone(s) involved in RNR activity, 28 yeast co-chaperone knockout mutants were screened for sensitivity to the RNR-perturbing agent Hydroxyurea. Ydj1, an important cytoplasmic Hsp70 co-chaperone was identified to be required for growth on HU. Ydj1 bound the RNR subunit Rnr2 and cells lacking Ydj1 showed a destabilized RNR complex. Suggesting broad conservation from yeast to human, HDJ2 binds R2B and regulates RNR stability in human cells. Perturbation of the Ssa1-Ydj1 interaction through mutation or Hsp70-HDJ2 via the small molecule 116-9e compromised RNR function, suggesting chaperone dependence of this novel role. Mammalian cells lacking HDJ2 were significantly more sensitive to RNR inhibiting drugs such as hydroxyurea, gemcitabine and triapine. Taken together, this work suggests a novel anticancer strategy-inhibition of RNR by targeting Hsp70 co-chaperone function. Ribonucleotide reductase (RNR) is a key enzyme in the synthesis of DNA and inhibition of RNR leads to cellular sensitivity to radiation. As such, RNR is a well-validated therapeutic target for a variety of diseases including cancer. Anti-RNR drugs are effective but are associated with a range of side effects in patients. Our previous work had identified that the Hsp90 and Hsp70 molecular chaperone proteins regulate RNR. The specificity and activity of Hsp70 and Hsp90 are regulated by “co-chaperone” proteins. We examined RNR activity in cells lacking individual co-chaperones and identified the Ydj1/HDJ2 protein as a novel regulator of RNR in yeast and human cells. Importantly, we demonstrate that inhibiting HDJ2 sensitizes cells to currently used anticancer drugs.
Collapse
|
34
|
Uehara Y, Temma K, Kobayashi Y, Irie N, Yamaguchi T. Reduction of Thermotolerance by Heat Shock Protein 90 Inhibitors in Murine Erythroleukemia Cells. Biol Pharm Bull 2018; 41:1393-1400. [PMID: 30175776 DOI: 10.1248/bpb.b18-00190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cells induce heat shock proteins (HSPs) against various stress. However, murine erythroleukemia (MEL) cells do not express HSP72, a heat-inducible member of HSP70 family. So, it is of interest to examine how MEL cells respond to heat stress (44°C, 30 min). Heat stress-induced apoptosis was suppressed by pretreatment of heat shock (44°C, 10 min). Such suppressive effects were maximal at 6 h after heat shock and remained up to 12 h. Interestingly, such effects of heat shock were abrogated by specific inhibitors of HSP90 such as 17-allylamino-17-demethoxygeldanamycin (17-AAG) and novobiocin. From flow cytometric analysis, it was found that MEL cells arrest in G2 phase at 6 h after heat shock, but restore original cell cycle at 12 h. High expression level of HSP90 was maintained before and after heat shock. Phosphorylation of HSP90α was observed in apoptotic cells induced by heat stress, but inhibited by pretreatment of heat shock. Such inhibition was abrogated by 17-AAG. Moreover, c-Jun NH2-terminal kinase (JNK) was activated in heat stress-induced apoptotic cells. Taken together, these results suggest that HSP90α in MEL cells plays an important role in the thermotolerance, i.e., suppression of heat stress-induced apoptosis by heat shock.
Collapse
Affiliation(s)
- Yousuke Uehara
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Kazunari Temma
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Yuuya Kobayashi
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Nobuyuki Irie
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Takeo Yamaguchi
- Department of Chemistry, Faculty of Science, Fukuoka University
| |
Collapse
|
35
|
Fuhrmann-Stroissnigg H, Niedernhofer LJ, Robbins PD. Hsp90 inhibitors as senolytic drugs to extend healthy aging. Cell Cycle 2018; 17:1048-1055. [PMID: 29886783 DOI: 10.1080/15384101.2018.1475828] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aging is characterized by progressive decay of biological systems and although it is not considered a disease, it is one of the main risk factors for chronic diseases and many types of cancers. The accumulation of senescent cells in various tissues is thought to be a major factor contributing to aging and age-related diseases. Removal of senescent cells during aging by either genetic or therapeutic methods have led to an improvement of several age related disease in mice. In this preview, we highlight the significance of developing senotherapeutic approaches to specifically kill senescent cells (senolytics) or suppress the senescence-associated secretory phenotype (SASP) that drives sterile inflammation (senomorphics) associated with aging to extend healthspan and potentially lifespan. Also, we provide an overview of the senotherapeutic drugs identified to date. In particular, we discuss and expand upon the recent identification of inhibitors of the HSP90 co-chaperone as a new class of senolytics.
Collapse
Affiliation(s)
- Heike Fuhrmann-Stroissnigg
- a Department of Molecular Medicine and The Center on Aging , The Scripps Research Institute , Jupiter , FL , USA
| | - Laura J Niedernhofer
- a Department of Molecular Medicine and The Center on Aging , The Scripps Research Institute , Jupiter , FL , USA
| | - Paul D Robbins
- a Department of Molecular Medicine and The Center on Aging , The Scripps Research Institute , Jupiter , FL , USA
| |
Collapse
|
36
|
Morishima Y, Mehta RK, Yoshimura M, Lau M, Southworth DR, Lawrence TS, Pratt WB, Nyati MK, Osawa Y. Chaperone Activity and Dimerization Properties of Hsp90 α and Hsp90 β in Glucocorticoid Receptor Activation by the Multiprotein Hsp90/Hsp70-Dependent Chaperone Machinery. Mol Pharmacol 2018; 94:984-991. [PMID: 29941666 DOI: 10.1124/mol.118.112516] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/20/2018] [Indexed: 11/22/2022] Open
Abstract
Several hundred proteins cycle into heterocomplexes with a dimer of the chaperone heat shock protein 90 (Hsp90), regulating their activity and turnover. There are two isoforms of Hsp90, Hsp90α and Hsp90β, and their relative chaperone activities and composition in these client protein•Hsp90 heterocomplexes has not been determined. Here, we examined the activity of human Hsp90α and Hsp90β in a purified five-protein chaperone machinery that assembles glucocorticoid receptor (GR)•Hsp90 heterocomplexes to generate high-affinity steroid-binding activity. We found that human Hsp90α and Hsp90β have equivalent chaperone activities, and when mixed together in this assay, they formed only GR•Hsp90αα and GR•Hsp90ββ homodimers and no GR•Hsp90αβ heterodimers. In contrast, GR•Hsp90 heterocomplexes formed in human embryonic kidney (HEK) cells also contain GR•Hsp90αβ heterodimers. The formation of GR•Hsp90αβ heterodimers in HEK cells probably reflects the longer time permitted for exchange to form Hsp90αβ heterodimers in the cell versus in the cell-free assembly conditions. This purified GR-activating chaperone machinery can be used to determine how modifications of Hsp90 affect its chaperone activity. To that effect, we have tested whether the unique phosphorylation of Hsp90α at threonines 5 and 7 that occurs during DNA damage repair affects its chaperone activity. We showed that the phosphomimetic mutant Hsp90α T5/7D has the same intrinsic chaperone activity as wild-type human Hsp90α in activation of GR steroid-binding activity by the five-protein machinery, supporting the conclusion that T5/7 phosphorylation does not affect Hsp90α chaperone activity.
Collapse
Affiliation(s)
- Yoshihiro Morishima
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Ranjit K Mehta
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Miyako Yoshimura
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Miranda Lau
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Daniel R Southworth
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Theodore S Lawrence
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - William B Pratt
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Mukesh K Nyati
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Yoichi Osawa
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| |
Collapse
|
37
|
Sottile ML, Nadin SB. Heat shock proteins and DNA repair mechanisms: an updated overview. Cell Stress Chaperones 2018; 23:303-315. [PMID: 28952019 PMCID: PMC5904076 DOI: 10.1007/s12192-017-0843-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/13/2017] [Indexed: 02/02/2023] Open
Abstract
Heat shock proteins (HSPs), also known as molecular chaperones, participate in important cellular processes, such as protein aggregation, disaggregation, folding, and unfolding. HSPs have cytoprotective functions that are commonly explained by their antiapoptotic role. Their involvement in anticancer drug resistance has been the focus of intense research efforts, and the relationship between HSP induction and DNA repair mechanisms has been in the spotlight during the past decades. Because DNA is permanently subject to damage, many DNA repair pathways are involved in the recognition and removal of a diverse array of DNA lesions. Hence, DNA repair mechanisms are key to maintain genome stability. In addition, the interactome network of HSPs with DNA repair proteins has become an exciting research field and so their use as emerging targets for cancer therapy. This article provides a historical overview of the participation of HSPs in DNA repair mechanisms as part of their molecular chaperone capabilities.
Collapse
Affiliation(s)
- Mayra L Sottile
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Adrián Ruiz Leal s/n Parque Gral. San Martín, 5500, Mendoza, Argentina
| | - Silvina B Nadin
- Tumor Biology Laboratory, Institute of Medicine and Experimental Biology of Cuyo (IMBECU), National Scientific and Technical Research Council (CONICET), Av. Adrián Ruiz Leal s/n Parque Gral. San Martín, 5500, Mendoza, Argentina.
| |
Collapse
|
38
|
Chung JH. The role of DNA-PK in aging and energy metabolism. FEBS J 2018; 285:1959-1972. [PMID: 29453899 DOI: 10.1111/febs.14410] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/15/2018] [Accepted: 02/12/2018] [Indexed: 12/17/2022]
Abstract
DNA-dependent protein kinase (DNA-PK) is a very large holoenzyme comprised of the p470 kDa DNA-PK catalytic subunit (DNA-PKcs ) and the Ku heterodimer consisting of the p86 (Ku 80) and p70 (Ku 70) subunits. It is best known for its nonhomologous end joining (NHEJ) activity, which repairs double-strand DNA (dsDNA) breaks (DSBs). As expected, the absence of DNA-PK activity results in sensitivity to ionizing radiation, which generates DSBs and defect in lymphocyte development, which requires NHEJ of the V(D)J region in the immunoglobulin and T-cell receptor loci. DNA-PK also has been reported to have functions seemingly unrelated to NHEJ. For example, DNA-PK responds to insulin signaling to facilitate the conversion of carbohydrates to fatty acids in the liver. More recent evidence indicates that DNA-PK activity increases with age in skeletal muscle, promoting mitochondrial loss and weight gain. These discoveries suggest that our understanding of DNA-PK is far from complete. As many excellent reviews have already been written about the role of DNA-PK in NHEJ, here we will review the non-NHEJ role of DNA-PK with a focus on its role in aging and energy metabolism.
Collapse
Affiliation(s)
- Jay H Chung
- Laboratory of Obesity and Aging Research, Genetics and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
39
|
Elaimy AL, Ahsan A, Marsh K, Pratt WB, Ray D, Lawrence TS, Nyati MK. ATM is the primary kinase responsible for phosphorylation of Hsp90α after ionizing radiation. Oncotarget 2018; 7:82450-82457. [PMID: 27738310 PMCID: PMC5347704 DOI: 10.18632/oncotarget.12557] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 10/05/2016] [Indexed: 01/01/2023] Open
Abstract
Heat shock protein 90 is a chaperone that plays an essential role in the stabilization of a large number of signal transduction molecules, many of which are associated with oncogenesis. An Hsp90 isoform (Hsp90α) has been shown to be selectively phosphorylated on two N-terminal threonine residues (threonine 5 and 7) and is involved in the DNA damage response and apoptosis. However, the kinase that phosphorylates Hsp90α after ionizing radiation (IR) and its role in post-radiation DNA repair remains unclear. Inasmuch as several proteins of the DNA damage response machinery are Hsp90 clients, the functional consequences of Hsp90α phosphorylation following IR have implications for the design of novel radiosensitizing agents that specifically target the Hsp90α isoform. Here we show that ATM phosphorylates Hsp90α at the T5/7 residues immediately after IR. The kinetics of Hsp90α T5/7 phosphorylation correlate with the kinetics of H2AX S139 phosphorylation (γH2AX). Although Hsp90α is located in both the cytoplasm and nucleus, only nuclear Hsp90α is phosphorylated by ATM after IR. The siRNA mediated knockdown of Hsp90α sensitizes head and neck squamous cell carcinoma cells, lung cancer cells and lung fibroblasts to IR. Furthermore, MEF cells that are Hsp90α null have reduced levels of γH2AX indicating that Hsp90α is important for the formation of γH2AX. Thus, this study provides evidence that Hsp90α is a component of the signal transduction events mediated by ATM following IR, and that Hsp90α loss decreases γH2AX levels. This work supports additional investigation into Hsp90α T5/7 phosphorylation with the goal of developing targeted radiosensitizing therapies.
Collapse
Affiliation(s)
- Ameer L Elaimy
- Department of Radiation Oncology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Aarif Ahsan
- Department of Radiation Oncology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Katherine Marsh
- Department of Radiation Oncology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - William B Pratt
- Department of Pharmacology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Dipankar Ray
- Department of Radiation Oncology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Theodore S Lawrence
- Department of Radiation Oncology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Mukesh K Nyati
- Department of Radiation Oncology, The University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
40
|
Karayazi Atici Ö, Urbanska A, Gopinathan SG, Boutillon F, Goffin V, Shemanko CS. ATM Is Required for the Prolactin-Induced HSP90-Mediated Increase in Cellular Viability and Clonogenic Growth After DNA Damage. Endocrinology 2018; 159:907-930. [PMID: 29186352 DOI: 10.1210/en.2017-00652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
Abstract
Prolactin (PRL) acts as a survival factor for breast cancer cells, but the PRL signaling pathway and the mechanism are unknown. Previously, we identified the master chaperone, heat shock protein 90 (HSP90) α, as a prolactin-Janus kinase 2 (JAK2)-signal transducer and activator of transcription 5 (STAT5) target gene involved in survival, and here we investigated the role of HSP90 in the mechanism of PRL-induced viability in response to DNA damage. The ataxia-telangiectasia mutated kinase (ATM) protein plays a critical role in the cellular response to double-strand DNA damage. We observed that PRL increased viability of breast cancer cells treated with doxorubicin or etoposide. The increase in cellular resistance is specific to the PRL receptor, because the PRL receptor antagonist, Δ1-9-G129R-hPRL, prevented the increase in viability. Two different HSP90 inhibitors, 17-allylamino-17-demethoxygeldanamycin and BIIB021, reduced the PRL-mediated increase in cell viability of doxorubicin-treated cells and led to a decrease in JAK2, ATM, and phosphorylated ATM protein levels. Inhibitors of JAK2 (G6) and ATM (KU55933) abolished the PRL-mediated increase in cell viability of DNA-damaged cells, supporting the involvement of each, as well as the crosstalk of ATM with the PRL pathway in the context of DNA damage. Drug synergism was detected between the ATM inhibitor (KU55933) and doxorubicin and between the HSP90 inhibitor (BIIB021) and doxorubicin. Short interfering RNA directed against ATM prevented the PRL-mediated increase in cell survival in two-dimensional cell culture, three-dimensional collagen gel cultures, and clonogenic cell survival, after doxorubicin treatment. Our results indicate that ATM contributes to the PRL-JAK2-STAT5-HSP90 pathway in mediating cellular resistance to DNA-damaging agents.
Collapse
Affiliation(s)
- Ödül Karayazi Atici
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Anna Urbanska
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sesha Gopal Gopinathan
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Florence Boutillon
- Inserm U1151, Institut Necker Enfants Malades, Team "PRL/GH Pathophysiology," Faculty of Medicine Paris Descartes, Sorbonne Paris Cité, Paris cedex 14, France
| | - Vincent Goffin
- Inserm U1151, Institut Necker Enfants Malades, Team "PRL/GH Pathophysiology," Faculty of Medicine Paris Descartes, Sorbonne Paris Cité, Paris cedex 14, France
| | - Carrie S Shemanko
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
41
|
Chaiswing L, Weiss HL, Jayswal RD, St. Clair DK, Kyprianou N. Profiles of Radioresistance Mechanisms in Prostate Cancer. Crit Rev Oncog 2018; 23:39-67. [PMID: 29953367 PMCID: PMC6231577 DOI: 10.1615/critrevoncog.2018025946] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Radiation therapy (RT) is commonly used for the treatment of localized prostate cancer (PCa). However, cancer cells often develop resistance to radiation through unknown mechanisms and pose an intractable challenge. Radiation resistance is highly unpredictable, rendering the treatment less effective in many patients and frequently causing metastasis and cancer recurrence. Understanding the molecular events that cause radioresistance in PCa will enable us to develop adjuvant treatments for enhancing the efficacy of RT. Radioresistant PCa depends on the elevated DNA repair system and the intracellular levels of reactive oxygen species (ROS) to proliferate, self-renew, and scavenge anti-cancer regimens, whereas the elevated heat shock protein 90 (HSP90) and the epithelial-mesenchymal transition (EMT) enable radioresistant PCa cells to metastasize after exposure to radiation. The up-regulation of the DNA repairing system, ROS, HSP90, and EMT effectors has been studied extensively, but not targeted by adjuvant therapy of radioresistant PCa. Here, we emphasize the effects of ionizing radiation and the mechanisms driving the emergence of radioresistant PCa. We also address the markers of radioresistance, the gene signatures for the predictive response to radiotherapy, and novel therapeutic platforms for targeting radioresistant PCa. This review provides significant insights into enhancing the current knowledge and the understanding toward optimization of these markers for the treatment of radioresistant PCa.
Collapse
Affiliation(s)
| | - Heidi L. Weiss
- The Markey Biostatistics and Bioinformatics Shared Resource Facility
| | - Rani D. Jayswal
- The Markey Biostatistics and Bioinformatics Shared Resource Facility
| | | | - Natasha Kyprianou
- Department of Toxicology and Cancer Biology
- Department of Urology
- Department of Biochemistry, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
42
|
Cdc7-Dbf4-mediated phosphorylation of HSP90-S164 stabilizes HSP90-HCLK2-MRN complex to enhance ATR/ATM signaling that overcomes replication stress in cancer. Sci Rep 2017; 7:17024. [PMID: 29209046 PMCID: PMC5717001 DOI: 10.1038/s41598-017-17126-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 10/09/2017] [Indexed: 12/31/2022] Open
Abstract
Cdc7-Dbf4 kinase plays a key role in the initiation of DNA replication and contributes to the replication stress in cancer. The activity of human Cdc7-Dbf4 kinase remains active and acts as an effector of checkpoint under replication stress. However, the downstream targets of Cdc7-Dbf4 contributed to checkpoint regulation and replication stress-support function in cancer are not fully identified. In this work, we showed that aberrant Cdc7-Dbf4 induces DNA lesions that activate ATM/ATR-mediated checkpoint and homologous recombination (HR) DNA repair. Using a phosphoproteome approach, we identified HSP90-S164 as a target of Cdc7-Dbf4 in vitro and in vivo. The phosphorylation of HSP90-S164 by Cdc7-Dbf4 is required for the stability of HSP90-HCLK2-MRN complex and the function of ATM/ATR signaling cascade and HR DNA repair. In clinically, the phosphorylation of HSP90-S164 indeed is increased in oral cancer patients. Our results indicate that aberrant Cdc7-Dbf4 enhances replication stress tolerance by rewiring ATR/ATM mediated HR repair through HSP90-S164 phosphorylation and by promoting recovery from replication stress. We provide a new solution to a subtyping of cancer patients with dominant ATR/HSP90 expression by combining inhibitors of ATR-Chk1, HSP90, or Cdc7 in cancer combination therapy.
Collapse
|
43
|
Pennisi R, Antoccia A, Leone S, Ascenzi P, di Masi A. Hsp90α regulates ATM and NBN functions in sensing and repair of DNA double-strand breaks. FEBS J 2017. [PMID: 28631426 DOI: 10.1111/febs.14145] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The molecular chaperone heat shock protein 90 (Hsp90α) regulates cell proteostasis and mitigates the harmful effects of endogenous and exogenous stressors on the proteome. Indeed, the inhibition of Hsp90α ATPase activity affects the cellular response to ionizing radiation (IR). Although the interplay between Hsp90α and several DNA damage response (DDR) proteins has been reported, its role in the DDR is still unclear. Here, we show that ataxia-telangiectasia-mutated kinase (ATM) and nibrin (NBN), but not 53BP1, RAD50, and MRE11, are Hsp90α clients as the Hsp90α inhibitor 17-(allylamino)-17-demethoxygeldanamycin (17-AAG) induces ATM and NBN polyubiquitination and proteosomal degradation in normal fibroblasts and lymphoblastoid cell lines. Hsp90α-ATM and Hsp90α-NBN complexes are present in unstressed and irradiated cells, allowing the maintenance of ATM and NBN stability that is required for the MRE11/RAD50/NBN complex-dependent ATM activation and the ATM-dependent phosphorylation of both NBN and Hsp90α in response to IR-induced DNA double-strand breaks (DSBs). Hsp90α forms a complex also with ph-Ser1981-ATM following IR. Upon phosphorylation, NBN dissociates from Hsp90α and translocates at the DSBs, while phThr5/7-Hsp90α is not recruited at the damaged sites. The inhibition of Hsp90α affects nuclear localization of MRE11 and RAD50, impairs DDR signaling (e.g., BRCA1 and CHK2 phosphorylation), and slows down DSBs repair. Hsp90α inhibition does not affect DNA-dependent protein kinase (DNA-PK) activity, which possibly phosphorylates Hsp90α and H2AX after IR. Notably, Hsp90α inhibition causes H2AX phosphorylation in proliferating cells, this possibly indicating replication stress events. Overall, present data shed light on the regulatory role of Hsp90α on the DDR, controlling ATM and NBN stability and influencing the DSBs signaling and repair.
Collapse
Affiliation(s)
- Rosa Pennisi
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Antonio Antoccia
- Department of Sciences, Roma Tre University, Roma, Italy.,Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Stefano Leone
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Alessandra di Masi
- Department of Sciences, Roma Tre University, Roma, Italy.,Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| |
Collapse
|
44
|
Hyttinen JMT, Błasiak J, Niittykoski M, Kinnunen K, Kauppinen A, Salminen A, Kaarniranta K. DNA damage response and autophagy in the degeneration of retinal pigment epithelial cells-Implications for age-related macular degeneration (AMD). Ageing Res Rev 2017; 36:64-77. [PMID: 28351686 DOI: 10.1016/j.arr.2017.03.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 12/14/2022]
Abstract
In this review we will discuss the links between autophagy, a mechanism involved in the maintenance of cellular homeostasis and controlling cellular waste management, and the DNA damage response (DDR), comprising various mechanisms preserving the integrity and stability of the genome. A reduced autophagy capacity in retinal pigment epithelium has been shown to be connected in the pathogenesis of age-related macular degeneration (AMD), an eye disease. This degenerative disease is a major and increasing cause of vision loss in the elderly in developed countries, primarily due to the profound accumulation of intra- and extracellular waste: lipofuscin and drusen. An abundance of reactive oxygen species is produced in the retina since this tissue has a high oxygen demand and contains mitochondria-rich cells. The retina is exposed to light and it also houses many photoactive molecules. These factors are clearly reflected in both the autophagy and DNA damage rates, and in both nuclear and mitochondrial genomes. It remains to be revealed whether DNA damage and DDR capacity have a more direct role in the development of AMD.
Collapse
Affiliation(s)
- Juha M T Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Janusz Błasiak
- Department of Molecular Genetics, University of Łódź, Pomorska 141/143, 90-236, Łódź, Poland
| | - Minna Niittykoski
- Institute of Biotechnology, Developmental Biology Program, University of Helsinki, P.O. Box 56, FI-00014, Finland
| | - Kati Kinnunen
- Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029, Finland
| |
Collapse
|
45
|
Bethea CL, Mueller K, Reddy AP, Kohama SG, Urbanski HF. Effects of obesogenic diet and estradiol on dorsal raphe gene expression in old female macaques. PLoS One 2017; 12:e0178788. [PMID: 28628658 PMCID: PMC5476244 DOI: 10.1371/journal.pone.0178788] [Citation(s) in RCA: 8] [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: 02/10/2017] [Accepted: 05/18/2017] [Indexed: 12/19/2022] Open
Abstract
The beneficial effects of bioidentical ovarian steroid hormone therapy (HT) during the perimenopause are gaining recognition. However, the positive effects of estrogen (E) plus or minus progesterone (P) administration to ovariectomized (Ovx) lab animals were recognized in multiple systems for years before clinical trials could adequately duplicate the results. Moreover, very large numbers of women are often needed to find statistically significant results in clinical trials of HT; and there are still opposing results being published, especially in neural and cardiovascular systems. One of the obvious differences between human and animal studies is diet. Laboratory animals are fed a diet that is low in fat and refined sugar, but high in micronutrients. In the US, a large portion of the population eats what is known as a "western style diet" or WSD that provides calories from 36% fat, 44% carbohydrates (includes 18.5% sugars) and 18% protein. Unfortunately, obesity and diabetes have reached epidemic proportions and the percentage of obese women in clinical trials may be overlooked. We questioned whether WSD and obesity could decrease the positive neural effects of estradiol (E) in the serotonin system of old macaques that were surgically menopausal. Old ovo-hysterectomized female monkeys were fed WSD for 2.5 years, and treated with placebo, Immediate E (ImE) or Delayed E (DE). Compared to old Ovx macaques on primate chow and treated with placebo or E, the WSD-fed monkeys exhibited greater individual variance and blunted responses to E-treatment in the expression of genes related to serotonin neurotransmission, CRH components in the midbrain, synapse assembly, DNA repair, protein folding, ubiquitylation, transport and neurodegeneration. For many of the genes examined, transcript abundance was lower in WSD-fed than chow-fed monkeys. In summary, an obesogenic diet for 2.5 years in old surgically menopausal macaques blunted or increased variability in E-induced gene expression in the dorsal raphe. These results suggest that with regard to function and viability in the dorsal raphe, HT may not be as beneficial for obese women as normal weight women.
Collapse
Affiliation(s)
- Cynthia L. Bethea
- Division of Reproductive and Developmental Science, Oregon National Primate Research Center, Beaverton, OR, United States of America
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States of America
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR, United States of America
| | - Kevin Mueller
- Division of Reproductive and Developmental Science, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Arubala P. Reddy
- Department of Internal Medicine, Texas Technical University Health Sciences Center School of Medicine, Lubbock, TX, United States of America
| | - Steven G. Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States of America
| | - Henryk F. Urbanski
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, United States of America
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, United States of America
| |
Collapse
|
46
|
Abstract
Heat shock protein 90 (Hsp90) is a molecular chaperone that is involved in the activation of disparate client proteins. This implicates Hsp90 in diverse biological processes that require a variety of co-ordinated regulatory mechanisms to control its activity. Perhaps the most important regulator is heat shock factor 1 (HSF1), which is primarily responsible for upregulating Hsp90 by binding heat shock elements (HSEs) within Hsp90 promoters. HSF1 is itself subject to a variety of regulatory processes and can directly respond to stress. HSF1 also interacts with a variety of transcriptional factors that help integrate biological signals, which in turn regulate Hsp90 appropriately. Because of the diverse clientele of Hsp90 a whole variety of co-chaperones also regulate its activity and some are directly responsible for delivery of client protein. Consequently, co-chaperones themselves, like Hsp90, are also subject to regulatory mechanisms such as post translational modification. This review, looks at the many different levels by which Hsp90 activity is ultimately regulated.
Collapse
|
47
|
Park SJ, Gavrilova O, Brown AL, Soto JE, Bremner S, Kim J, Xu X, Yang S, Um JH, Koch LG, Britton SL, Lieber RL, Philp A, Baar K, Kohama SG, Abel ED, Kim MK, Chung JH. DNA-PK Promotes the Mitochondrial, Metabolic, and Physical Decline that Occurs During Aging. Cell Metab 2017; 25:1135-1146.e7. [PMID: 28467930 PMCID: PMC5485859 DOI: 10.1016/j.cmet.2017.04.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 01/25/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022]
Abstract
Hallmarks of aging that negatively impact health include weight gain and reduced physical fitness, which can increase insulin resistance and risk for many diseases, including type 2 diabetes. The underlying mechanism(s) for these phenomena is poorly understood. Here we report that aging increases DNA breaks and activates DNA-dependent protein kinase (DNA-PK) in skeletal muscle, which suppresses mitochondrial function, energy metabolism, and physical fitness. DNA-PK phosphorylates threonines 5 and 7 of HSP90α, decreasing its chaperone function for clients such as AMP-activated protein kinase (AMPK), which is critical for mitochondrial biogenesis and energy metabolism. Decreasing DNA-PK activity increases AMPK activity and prevents weight gain, decline of mitochondrial function, and decline of physical fitness in middle-aged mice and protects against type 2 diabetes. In conclusion, DNA-PK is one of the drivers of the metabolic and fitness decline during aging, and therefore DNA-PK inhibitors may have therapeutic potential in obesity and low exercise capacity.
Collapse
Affiliation(s)
- Sung-Jun Park
- Laboratory of Obesity and Aging Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexandra L Brown
- Laboratory of Obesity and Aging Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jamie E Soto
- Program in Molecular Medicine and Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Shannon Bremner
- Department of Orthopedic Surgery, University of California and V.A. Medical Centers, San Diego, La Jolla, CA 92093, USA
| | - Jeonghan Kim
- Laboratory of Obesity and Aging Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xihui Xu
- Laboratory of Obesity and Aging Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Shutong Yang
- Laboratory of Obesity and Aging Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jee-Hyun Um
- Laboratory of Obesity and Aging Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lauren G Koch
- Department of Anesthesiology, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Steven L Britton
- Department of Anesthesiology, The University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular & Integrative Physiology, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard L Lieber
- Department of Orthopedic Surgery, University of California and V.A. Medical Centers, San Diego, La Jolla, CA 92093, USA
| | - Andrew Philp
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA USA 95616
| | - Keith Baar
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA USA 95616
| | - Steven G Kohama
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - E Dale Abel
- Program in Molecular Medicine and Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Myung K Kim
- Laboratory of Obesity and Aging Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jay H Chung
- Laboratory of Obesity and Aging Research, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
48
|
|
49
|
Kim Y, Kim A, Sharip A, Sharip A, Jiang J, Yang Q, Xie Y. Reverse the Resistance to PARP Inhibitors. Int J Biol Sci 2017; 13:198-208. [PMID: 28255272 PMCID: PMC5332874 DOI: 10.7150/ijbs.17240] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 09/09/2016] [Indexed: 12/24/2022] Open
Abstract
One of the DNA repair machineries is activated by Poly (ADP-ribose) Polymerase (PARP) enzyme. Particularly, this enzyme is involved in repair of damages to single-strand DNA, thus decreasing the chances of generating double-strand breaks in the genome. Therefore, the concept to block PARP enzymes by PARP inhibitor (PARPi) was appreciated in cancer treatment. PARPi has been designed and tested for many years and became a potential supplement for the conventional chemotherapy. However, increasing evidence indicates the appearance of the resistance to this treatment. Specifically, cancer cells may acquire new mutations or events that overcome the positive effect of these drugs. This paper describes several molecular mechanisms of PARPi resistance which were reported most recently, and summarizes some strategies to reverse this type of drug resistance.
Collapse
Affiliation(s)
- Yevgeniy Kim
- Department of Biology, Nazarbayev University, School of Science and Technology, Astana, 010000, Republic of Kazakhstan
| | - Aleksei Kim
- Department of Biology, Nazarbayev University, School of Science and Technology, Astana, 010000, Republic of Kazakhstan
| | - Ainur Sharip
- Department of Biology, Nazarbayev University, School of Science and Technology, Astana, 010000, Republic of Kazakhstan
| | - Aigul Sharip
- Department of Biology, Nazarbayev University, School of Science and Technology, Astana, 010000, Republic of Kazakhstan
| | - Juhong Jiang
- Department of Pathology, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qing Yang
- Department of Biology, Nazarbayev University, School of Science and Technology, Astana, 010000, Republic of Kazakhstan
| | - Yingqiu Xie
- Department of Biology, Nazarbayev University, School of Science and Technology, Astana, 010000, Republic of Kazakhstan
| |
Collapse
|
50
|
Localisation Microscopy of Breast Epithelial ErbB-2 Receptors and Gap Junctions: Trafficking after γ-Irradiation, Neuregulin-1β, and Trastuzumab Application. Int J Mol Sci 2017; 18:ijms18020362. [PMID: 28208769 PMCID: PMC5343897 DOI: 10.3390/ijms18020362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/28/2017] [Accepted: 01/30/2017] [Indexed: 12/28/2022] Open
Abstract
In cancer, vulnerable breast epithelium malignance tendency correlates with number and activation of ErbB receptor tyrosine kinases. In the presented work, we observe ErbB receptors activated by irradiation-induced DNA injury or neuregulin-1β application, or alternatively, attenuated by a therapeutic antibody using high resolution fluorescence localization microscopy. The gap junction turnover coinciding with ErbB receptor activation and co-transport is simultaneously recorded. DNA injury caused by 4 Gray of 6 MeV photon γ-irradiation or alternatively neuregulin-1β application mobilized ErbB receptors in a nucleograde fashion—a process attenuated by trastuzumab antibody application. This was accompanied by increased receptor density, indicating packing into transport units. Factors mobilizing ErbB receptors also mobilized plasma membrane resident gap junction channels. The time course of ErbB receptor activation and gap junction mobilization recapitulates the time course of non-homologous end-joining DNA repair. We explain our findings under terms of DNA injury-induced membrane receptor tyrosine kinase activation and retrograde trafficking. In addition, we interpret the phenomenon of retrograde co-trafficking of gap junction connexons stimulated by ErbB receptor activation.
Collapse
|