1
|
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
|
2
|
Babi A, Menlibayeva K, Bex T, Doskaliev A, Akshulakov S, Shevtsov M. Targeting Heat Shock Proteins in Malignant Brain Tumors: From Basic Research to Clinical Trials. Cancers (Basel) 2022; 14:5435. [PMID: 36358853 PMCID: PMC9659111 DOI: 10.3390/cancers14215435] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 05/03/2024] Open
Abstract
Heat shock proteins (HSPs) are conservative and ubiquitous proteins that are expressed both in prokaryotic and eukaryotic organisms and play an important role in cellular homeostasis, including the regulation of proteostasis, apoptosis, autophagy, maintenance of signal pathways, protection from various stresses (e.g., hypoxia, ionizing radiation, etc.). Therefore, HSPs are highly expressed in tumor cells, including malignant brain tumors, where they also associate with cancer cell invasion, metastasis, and resistance to radiochemotherapy. In the current review, we aimed to assess the diagnostic and prognostic values of HSPs expression in CNS malignancies as well as the novel treatment approaches to modulate the chaperone levels through the application of inhibitors (as monotherapy or in combination with other treatment modalities). Indeed, for several proteins (i.e., HSP10, HSPB1, DNAJC10, HSPA7, HSP90), a direct correlation between the protein level expression and poor overall survival prognosis for patients was demonstrated that provides a possibility to employ them as prognostic markers in neuro-oncology. Although small molecular inhibitors for HSPs, particularly for HSP27, HSP70, and HSP90 families, were studied in various solid and hematological malignancies demonstrating therapeutic potential, still their potential was not yet fully explored in CNS tumors. Some newly synthesized agents (e.g., HSP40/DNAJ inhibitors) have not yet been evaluated in GBM. Nevertheless, reported preclinical studies provide evidence and rationale for the application of HSPs inhibitors for targeting brain tumors.
Collapse
Affiliation(s)
- Aisha Babi
- National Centre for Neurosurgery, Turan Ave., 34/1, Astana 010000, Kazakhstan
| | | | - Torekhan Bex
- National Centre for Neurosurgery, Turan Ave., 34/1, Astana 010000, Kazakhstan
| | - Aidos Doskaliev
- National Centre for Neurosurgery, Turan Ave., 34/1, Astana 010000, Kazakhstan
| | - Serik Akshulakov
- National Centre for Neurosurgery, Turan Ave., 34/1, Astana 010000, Kazakhstan
| | - Maxim Shevtsov
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
- Department of Radiation Oncology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
3
|
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: 11] [Impact Index Per Article: 5.5] [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
|
4
|
Matsui JK, Perlow HK, Ritter AR, Upadhyay R, Raval RR, Thomas EM, Beyer SJ, Pillainayagam C, Goranovich J, Ong S, Giglio P, Palmer JD. Small Molecules and Immunotherapy Agents for Enhancing Radiotherapy in Glioblastoma. Biomedicines 2022; 10:biomedicines10071763. [PMID: 35885067 PMCID: PMC9313399 DOI: 10.3390/biomedicines10071763] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor that is associated with a poor prognosis and quality of life. The standard of care has changed minimally over the past two decades and currently consists of surgery followed by radiotherapy (RT), concomitant and adjuvant temozolomide, and tumor treating fields (TTF). Factors such as tumor hypoxia and the presence of glioma stem cells contribute to the radioresistant nature of GBM. In this review, we discuss the current treatment modalities, mechanisms of radioresistance, and studies that have evaluated promising radiosensitizers. Specifically, we highlight small molecules and immunotherapy agents that have been studied in conjunction with RT in clinical trials. Recent preclinical studies involving GBM radiosensitizers are also discussed.
Collapse
Affiliation(s)
- Jennifer K. Matsui
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Haley K. Perlow
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.K.P.); (A.R.R.); (R.U.); (R.R.R.); (E.M.T.); (S.J.B.)
| | - Alex R. Ritter
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.K.P.); (A.R.R.); (R.U.); (R.R.R.); (E.M.T.); (S.J.B.)
| | - Rituraj Upadhyay
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.K.P.); (A.R.R.); (R.U.); (R.R.R.); (E.M.T.); (S.J.B.)
| | - Raju R. Raval
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.K.P.); (A.R.R.); (R.U.); (R.R.R.); (E.M.T.); (S.J.B.)
| | - Evan M. Thomas
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.K.P.); (A.R.R.); (R.U.); (R.R.R.); (E.M.T.); (S.J.B.)
| | - Sasha J. Beyer
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.K.P.); (A.R.R.); (R.U.); (R.R.R.); (E.M.T.); (S.J.B.)
| | - Clement Pillainayagam
- Department of Neuro-Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (C.P.); (J.G.); (S.O.); (P.G.)
| | - Justin Goranovich
- Department of Neuro-Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (C.P.); (J.G.); (S.O.); (P.G.)
| | - Shirley Ong
- Department of Neuro-Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (C.P.); (J.G.); (S.O.); (P.G.)
| | - Pierre Giglio
- Department of Neuro-Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (C.P.); (J.G.); (S.O.); (P.G.)
| | - Joshua D. Palmer
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (H.K.P.); (A.R.R.); (R.U.); (R.R.R.); (E.M.T.); (S.J.B.)
- Correspondence:
| |
Collapse
|
5
|
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: 0.5] [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]
|
6
|
Schnöller LE, Albrecht V, Brix N, Nieto AE, Fleischmann DF, Niyazi M, Hess J, Belka C, Unger K, Lauber K, Orth M. Integrative analysis of therapy resistance and transcriptomic profiling data in glioblastoma cells identifies sensitization vulnerabilities for combined modality radiochemotherapy. Radiat Oncol 2022; 17:79. [PMID: 35440003 PMCID: PMC9020080 DOI: 10.1186/s13014-022-02052-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
Background Inherent resistance to radio/chemotherapy is one of the major reasons for early recurrence, treatment failure, and dismal prognosis of glioblastoma. Thus, the identification of resistance driving regulators as prognostic and/or predictive markers as well as potential vulnerabilities for combined modality treatment approaches is of pivotal importance. Methods We performed an integrative analysis of treatment resistance and DNA damage response regulator expression in a panel of human glioblastoma cell lines. mRNA expression levels of 38 DNA damage response regulators were analyzed by qRT-PCR. Inherent resistance to radiotherapy (single-shot and fractionated mode) and/or temozolomide treatment was assessed by clonogenic survival assays. Resistance scores were extracted by dimensionality reduction and subjected to correlation analyses with the mRNA expression data. Top-hit candidates with positive correlation coefficients were validated by pharmacological inhibition in clonogenic survival assays and DNA repair analyses via residual γH2AX/53BP1-foci staining. Results Inherent resistance to single-shot and similarly also to fractionated radiotherapy showed strong positive correlations with mRNA expression levels of known vulnerabilities of GBM, including PARP1, NBN, and BLM, as well as ATR and LIG4—two so far underestimated targets. Inhibition of ATR by AZD-6738 resulted in robust and dose-dependent radiosensitization of glioblastoma cells, whereas LIG4 inhibition by L189 had no noticeable impact. Resistance against temozolomide showed strong positive correlation with mRNA expression levels of MGMT as to be expected. Interestingly, it also correlated with mRNA expression levels of ATM, suggesting a potential role of ATM in the context of temozolomide resistance in glioblastoma cells. ATM inhibition exhibited slight sensitization effects towards temozolomide treatment in MGMT low expressing glioblastoma cells, thus encouraging further characterization. Conclusions Here, we describe a systematic approach integrating clonogenic survival data with mRNA expression data of DNA damage response regulators in human glioblastoma cell lines to identify markers of inherent therapy resistance and potential vulnerabilities for targeted sensitization. Our results provide proof-of-concept for the feasibility of this approach, including its limitations. We consider this strategy to be adaptable to other cancer entities as well as other molecular data qualities, and its upscaling potential in terms of model systems and observational data levels deserves further investigation.
Collapse
Affiliation(s)
- Leon Emanuel Schnöller
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Valerie Albrecht
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Nikko Brix
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Alexander Edward Nieto
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Daniel Felix Fleischmann
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany
| | - Julia Hess
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.
| |
Collapse
|
7
|
Wu Y, Shen S, Shi Y, Tian N, Zhou Y, Zhang X. Senolytics: Eliminating Senescent Cells and Alleviating Intervertebral Disc Degeneration. Front Bioeng Biotechnol 2022; 10:823945. [PMID: 35309994 PMCID: PMC8924288 DOI: 10.3389/fbioe.2022.823945] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/21/2022] [Indexed: 12/25/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) is the main cause of cervical and lumbar spondylosis. Over the past few years, the relevance between cellular senescence and IVDD has been widely studied, and the senescence-associated secretory phenotype (SASP) produced by senescent cells is found to remodel extracellular matrix (ECM) metabolism and destruct homeostasis. Elimination of senescent cells by senolytics and suppression of SASP production by senomorphics/senostatics are effective strategies to alleviate degenerative diseases including IVDD. Here, we review the involvement of senescence in the process of IVDD; we also discuss the potential of senolytics on eliminating senescent disc cells and alleviating IVDD; finally, we provide a table listing senolytic drugs and small molecules, aiming to propose potential drugs for IVDD therapy in the future.
Collapse
Affiliation(s)
- Yuhao Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Shiwei Shen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Shi
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| | - Yifei Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou, China
- Chinese Orthopaedic Regenerative Medicine Society, Hangzhou, China
- *Correspondence: Naifeng Tian, ; Yifei Zhou, ; Xiaolei Zhang,
| |
Collapse
|
8
|
Xu J, Wu PJ, Lai TH, Sharma P, Canella A, Welker AM, Beattie C, Timmers CD, Lang FF, Jacob NK, Elder JB, Lonser R, Easley M, Pietrzak M, Sampath D, Puduvalli VK. Disruption of DNA Repair and Survival Pathways through Heat Shock Protein inhibition by Onalespib to Sensitize Malignant Gliomas to Chemoradiation therapy. Clin Cancer Res 2022; 28:1979-1990. [PMID: 35140124 PMCID: PMC9064967 DOI: 10.1158/1078-0432.ccr-20-0468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/10/2021] [Accepted: 02/04/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Proficient DNA repair by homologous recombination (HR) facilitates resistance to chemo-radiation in glioma stem cells (GSCs). We evaluated whether compromising HR by targeting HSP90, a molecular chaperone required for the function of key HR proteins, using onalespib, a long-acting, brain-penetrant HSP90 inhibitor, would sensitize high-grade gliomas to chemo-radiation in vitro and in vivo Experimental Design: The ability of onalespib to deplete HR client proteins, impair HR repair capacity, and sensitize GBM to chemo-radiation was evaluated in vitro in GSCs, and in vivo using zebrafish and mouse intracranial glioma xenograft models. The effects of HSP90 inhibition on the transcriptome and cytoplasmic proteins was assessed in GSCs and in ex vivo organotypic human glioma slice cultures. RESULTS Treatment with onalespib depleted CHK1 and RAD51, two key proteins of the HR pathway, and attenuated HR repair, sensitizing GSCs to the combination of radiation and temozolomide (TMZ). HSP90 inhibition reprogrammed the transcriptome of GSCs and broadly altered expression of cytoplasmic proteins including known and novel client proteins relevant to GSCs. The combination of onalespib with radiation and TMZ extended survival in a zebra fish and a mouse xenograft model of GBM compared to the standard of care (radiation and TMZ) or onalespib with radiation. CONCLUSIONS The results of this study demonstrate that targeting HR by HSP90 inhibition sensitizes GSCs to radiation and chemotherapy and extends survival in zebrafish and mouse intracranial models of GBM. These results provide a preclinical rationale for assessment of HSP90 inhibitors in combination with chemoradiation in GBM patients.
Collapse
Affiliation(s)
- Jihong Xu
- Neuro-Oncology, The University of Texas MD Anderson Cancer Center
| | - Pei-Jung Wu
- Division of Neuro-oncology, The Ohio State University
| | - Tzung-Huei Lai
- Division of Hematology, Department of Medicine, The Ohio State University
| | - Pratibha Sharma
- Department of Neuro-oncology, The University of Texas MD Anderson Cancer Center
| | | | | | | | | | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center
| | - Naduparambil K Jacob
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center
| | - J Bradley Elder
- Dardinger Neuro-Oncology Center, Department of Neurosurgery, The Ohio State University
| | - Russell Lonser
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke
| | | | | | - Deepa Sampath
- Hematopoeitic Biology and Malignancy, The University of Texas MD Anderson Cancer Center
| | - Vinay K Puduvalli
- Department of Neuro-oncology, The University of Texas MD Anderson Cancer Center
| |
Collapse
|
9
|
Tani T, Tojo N, Ohnishi K. Preferential radiosensitization to glioblastoma cancer stem cell‑like cells by a Hsp90 inhibitor, N‑vinylpyrrolidone‑AUY922. Oncol Lett 2022; 23:102. [PMID: 35154433 PMCID: PMC8822487 DOI: 10.3892/ol.2022.13222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/22/2021] [Indexed: 11/06/2022] Open
Abstract
The present study examined the radiosensitization induced by a heat shock protein 90 inhibitor, N-vinylpyrrolidone (NVP)-AUY922, in CD133-positive cells in a hypoxic area of T98G spheroids. CD133-positive cells that are induced in the hypoxic microenvironment of spheroids have previously been reported to exhibit cancer stem cell-like properties. The present study used CD133-positive cells from a glioblastoma cell line (T98G) as cancer stem cell-like cells. CD133-positive and negative cells were sorted from T98G spheroids using fluorescence-activated cell sorting and used for colony formation assay. Colony formation assay results indicated that NVP-AUY922 enhanced radiosensitivity more strongly in CD133-positive cells compared with CD133-negative cells. This result showed that NVP-AUY922 was a preferential radiosensitization candidate targeting glioblastoma cancer stem cells. The mechanisms underlying radiosensitization by NVP-AUY922 are discussed in relation to the properties of cancer stem cells. Overall, HIF-1α inhibition by NVP-AUY922 may induce higher sensitization of cancer stem cells to radiation.
Collapse
Affiliation(s)
- Toshiaki Tani
- Radiological Technology Section, QST Hospital, National Institutes for Quantum and Radiological Science and Technology, Chiba 263‑8555, Japan
| | - Naomi Tojo
- Department of Biology, Ibaraki Prefectural University of Health Sciences, Inashiki, Ibaraki 300‑0394, Japan
| | - Ken Ohnishi
- Department of Biology, Ibaraki Prefectural University of Health Sciences, Inashiki, Ibaraki 300‑0394, Japan
| |
Collapse
|
10
|
Chemotherapy of HER2- and MDM2-Enriched Breast Cancer Subtypes Induces Homologous Recombination DNA Repair and Chemoresistance. Cancers (Basel) 2021; 13:cancers13184501. [PMID: 34572735 PMCID: PMC8471926 DOI: 10.3390/cancers13184501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary MDM2 is a protein responsible for negative regulation of the p53 tumor suppressor. In addition, MDM2 exhibits chaperone-like properties similar to the HSP90 molecular chaperone. Multiple studies revealed that MDM2 is deeply involved in cancer development and progression. Some recently published results indicate that the role of MDM2 in DNA repair inhibition is more complex than previously thought. We show that MDM2 is directly involved in the homologous recombination DNA repair, and its chaperone-like activity is crucial for this function. The DNA repair inhibition is a result of inefficient MDM2 dissociation from the NBN protein complex. When cancer cells are treated with chemotherapy, MDM2 can be easily released from the interaction and degraded, resulting in effective homologous recombination DNA repair, which translates into the acquisition of a chemoresistant phenotype by the tumor. This knowledge may allow for identification of the patients that are at particular risk of tumor chemoresistance. Abstract Analyzing the TCGA breast cancer database, we discovered that patients with the HER2 cancer subtype and overexpression of MDM2 exhibited decreased post-treatment survival. Inhibition of MDM2 expression in the SKBR3 cell line (HER2 subtype) diminished the survival of cancer cells treated with doxorubicin, etoposide, and camptothecin. Moreover, we demonstrated that inhibition of MDM2 expression diminished DNA repair by homologous recombination (HR) and sensitized SKBR3 cells to a PARP inhibitor, olaparib. In H1299 (TP53−/−) cells treated with neocarzinostatin (NCS), overexpression of MDM2 WT or E3-dead MDM2 C478S variant stimulated the NCS-dependent phosphorylation of ATM, NBN, and BRCA1, proteins involved in HR DNA repair. However, overexpression of chaperone-dead MDM2 K454A variant diminished phosphorylation of these proteins as well as the HR DNA repair. Moreover, we demonstrated that, upon NCS treatment, MDM2 K454A interacted with NBN more efficiently than MDM2 WT and that MDM2 WT was degraded more efficiently than MDM2 K454A. Using a proliferation assay, we showed that overexpression of MDM2 WT, but not MDM2 K454A, led to acquisition of resistance to NCS. The presented results indicate that, following chemotherapy, MDM2 WT was released from MDM2-NBN complex and efficiently degraded, hence allowing extensive HR DNA repair leading to the acquisition of chemoresistance by cancer cells.
Collapse
|
11
|
Epp-Ducharme B, Dunne M, Fan L, Evans JC, Ahmed L, Bannigan P, Allen C. Heat-activated nanomedicine formulation improves the anticancer potential of the HSP90 inhibitor luminespib in vitro. Sci Rep 2021; 11:11103. [PMID: 34045581 PMCID: PMC8160139 DOI: 10.1038/s41598-021-90585-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 01/06/2023] Open
Abstract
The heat shock protein 90 inhibitor, luminespib, has demonstrated potent preclinical activity against numerous cancers. However, clinical translation has been impeded by dose-limiting toxicities that have necessitated dosing schedules which have reduced therapeutic efficacy. As such, luminespib is a prime candidate for reformulation using advanced drug delivery strategies that improve tumor delivery efficiency and limit off-target side effects. Specifically, thermosensitive liposomes are proposed as a drug delivery strategy capable of delivering high concentrations of drug to the tumor in combination with other chemotherapeutic molecules. Indeed, this work establishes that luminespib exhibits synergistic activity in lung cancer in combination with standard of care drugs such as cisplatin and vinorelbine. While our research team has previously developed thermosensitive liposomes containing cisplatin or vinorelbine, this work presents the first liposomal formulation of luminespib. The physico-chemical properties and heat-triggered release of the formulation were characterized. Cytotoxicity assays were used to determine the optimal drug ratios for treatment of luminespib in combination with cisplatin or vinorelbine in non-small cell lung cancer cells. The formulation and drug combination work presented in this paper offer the potential for resuscitation of the clinical prospects of a promising anticancer agent.
Collapse
Affiliation(s)
| | - Michael Dunne
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Linyu Fan
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - James C Evans
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Lubabah Ahmed
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Pauric Bannigan
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
12
|
MRE11 as a molecular signature and therapeutic target for cancer treatment with radiotherapy. Cancer Lett 2021; 514:1-11. [PMID: 34022282 DOI: 10.1016/j.canlet.2021.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 01/02/2023]
Abstract
MRE11, the core of the MRE11/RAD50/NBS1 complex, is one of key DNA damage response proteins. Increasing evidence suggests that its expression in cancer cells is critical to developing radioresistance; as such, MRE11 is an emerging marker for targeted radiosensitization strategies. Elevated MRE11 in tumor tissues has been associated with poor survival in patients undergoing radiotherapy, although in some cancer types, the opposite has been noted. The recent discovery of ionizing radiation-induced truncation of MRE11, which decreases its efficacy, may explain some of these paradoxical findings. The progress of research on the biological modulation of MRE11 expression is also discussed, with the potential application of small molecule or large molecule inhibitors of MRE11 for enhancing radiosensitivity. Current research has further highlighted both nuclease and non-nuclease activities of MRE11 in cancer cells treated with ionizing radiation, and differentiation between these is essential to verify the targeting effects of radiosensitizing agents. These updates clarify our understanding of how MRE11 expression may be utilized in future stratification of cancer patients for radiotherapy, and how it may be leveraged in shaping novel radiosensitization strategies.
Collapse
|
13
|
Orth M, Albrecht V, Seidl K, Kinzel L, Unger K, Hess J, Kreutzer L, Sun N, Stegen B, Nieto A, Maas J, Winssinger N, Friedl AA, Walch AK, Belka C, Zitzelsberger H, Niyazi M, Lauber K. Inhibition of HSP90 as a Strategy to Radiosensitize Glioblastoma: Targeting the DNA Damage Response and Beyond. Front Oncol 2021; 11:612354. [PMID: 33816244 PMCID: PMC8011354 DOI: 10.3389/fonc.2021.612354] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an essential component of multi-modality treatment of glioblastoma (GBM). However, treatment failure and recurrence are frequent and give rise to the dismal prognosis of this aggressive type of primary brain tumor. A high level of inherent treatment resistance is considered to be the major underlying reason, stemming from constantly activated DNA damage response (DDR) mechanisms as a consequence of oncogene overexpression, persistent replicative stress, and other so far unknown reasons. The molecular chaperone heat shock protein 90 (HSP90) plays an important role in the establishment and maintenance of treatment resistance, since it crucially assists the folding and stabilization of various DDR regulators. Accordingly, inhibition of HSP90 represents a multi-target strategy to interfere with DDR function and to sensitize cancer cells to radiotherapy. Using NW457, a pochoxime-based HSP90 inhibitor with favorable brain pharmacokinetic profile, we show here that HSP90 inhibition at low concentrations with per se limited cytotoxicity leads to downregulation of various DNA damage response factors on the protein level, distinct transcriptomic alterations, impaired DNA damage repair, and reduced clonogenic survival in response to ionizing irradiation in glioblastoma cells in vitro. In vivo, HSP90 inhibition by NW457 improved the therapeutic outcome of fractionated CBCT-based irradiation in an orthotopic, syngeneic GBM mouse model, both in terms of tumor progression and survival. Nevertheless, in view of the promising in vitro results the in vivo efficacy was not as strong as expected, although apart from the radiosensitizing effects HSP90 inhibition also reduced irradiation-induced GBM cell migration and tumor invasiveness. Hence, our findings identify the combination of HSP90 inhibition and radiotherapy in principle as a promising strategy for GBM treatment whose performance needs to be further optimized by improved inhibitor substances, better formulations and/or administration routes, and fine-tuned treatment sequences.
Collapse
Affiliation(s)
- Michael Orth
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Valerie Albrecht
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Karin Seidl
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Linda Kinzel
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Julia Hess
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Lisa Kreutzer
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Na Sun
- Research Unit Analytical Pathology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Benjamin Stegen
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium, Munich, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Alexander Nieto
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Jessica Maas
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Nicolas Winssinger
- Department of Organic Chemistry, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Anna A Friedl
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Axel K Walch
- Research Unit Analytical Pathology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium, Munich, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Center Munich, Neuherberg, Germany
| | - Horst Zitzelsberger
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Center Munich, Neuherberg, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium, Munich, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Center Munich, Neuherberg, Germany
| |
Collapse
|
14
|
Schrader L, Winter M, Errbii M, Delabie J, Oettler J, Gadau J. Inhibition of HSP90 causes morphological variation in the invasive ant
Cardiocondyla obscurior. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2021; 336:333-340. [DOI: 10.1002/jez.b.23035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/13/2021] [Accepted: 02/04/2021] [Indexed: 01/16/2023]
Affiliation(s)
- Lukas Schrader
- Institute for Evolution and Biodiversity University of Münster Münster Germany
| | - Miles Winter
- Institute for Evolution and Biodiversity University of Münster Münster Germany
| | - Mohammed Errbii
- Institute for Evolution and Biodiversity University of Münster Münster Germany
| | - Jacques Delabie
- Laboratório de Mirmecologia Cocoa Research Center‐CEPLAC & UESC‐DCAA Itabuna Bahia Brazil
| | - Jan Oettler
- Lehrstuhl für Zoologie/Evolutionsbiologie University of Regensburg Regensburg Germany
| | - Jürgen Gadau
- Institute for Evolution and Biodiversity University of Münster Münster Germany
| |
Collapse
|
15
|
Chan TG, O'Neill E, Habjan C, Cornelissen B. Combination Strategies to Improve Targeted Radionuclide Therapy. J Nucl Med 2020; 61:1544-1552. [PMID: 33037092 PMCID: PMC8679619 DOI: 10.2967/jnumed.120.248062] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 09/09/2020] [Indexed: 01/20/2023] Open
Abstract
In recent years, targeted radionuclide therapy (TRT) has emerged as a promising strategy for cancer treatment. In contrast to conventional radiotherapy, TRT delivers ionizing radiation to tumors in a targeted manner, reducing the dose that healthy tissues are exposed to. Existing TRT strategies include the use of 177Lu-DOTATATE, 131I-metaiodobenzylguanidine, Bexxar, and Zevalin, clinically approved agents for the treatment of neuroendocrine tumors, neuroblastoma, and non-Hodgkin lymphoma, respectively. Although promising results have been obtained with these agents, clinical evidence acquired to date suggests that only a small percentage of patients achieves complete response. Consequently, there have been attempts to improve TRT outcomes through combinations with other therapeutic agents; such strategies include administering concurrent TRT and chemotherapy, and the use of TRT with known or putative radiosensitizers such as poly(adenosine diphosphate ribose) polymerase and mammalian-target-of-rapamycin inhibitors. In addition to potentially achieving greater therapeutic effects than the respective monotherapies, these strategies may lead to lower dosages or numbers of cycles required and, in turn, reduce unwanted toxicities. As of now, several clinical trials have been conducted to assess the benefits of TRT-based combination therapies, sometimes despite limited preclinical evidence being available in the public domain to support their use. Although some clinical trials have yielded promising results, others have shown no clear survival benefit from particular combination treatments. Here, we present a comprehensive review of combination strategies with TRT reported in the literature to date and evaluate their therapeutic potential.
Collapse
Affiliation(s)
- Tiffany G Chan
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Edward O'Neill
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Christine Habjan
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Bart Cornelissen
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
16
|
Hintelmann K, Kriegs M, Rothkamm K, Rieckmann T. Improving the Efficacy of Tumor Radiosensitization Through Combined Molecular Targeting. Front Oncol 2020; 10:1260. [PMID: 32903756 PMCID: PMC7438822 DOI: 10.3389/fonc.2020.01260] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022] Open
Abstract
Chemoradiation, either alone or in combination with surgery or induction chemotherapy, is the current standard of care for most locally advanced solid tumors. Though chemoradiation is usually performed at the maximum tolerated doses of both chemotherapy and radiation, current cure rates are not satisfactory for many tumor entities, since tumor heterogeneity and plasticity result in chemo- and radioresistance. Advances in the understanding of tumor biology, a rapidly growing number of molecular targeting agents and novel technologies enabling the in-depth characterization of individual tumors, have fuelled the hope of entering an era of precision oncology, where each tumor will be treated according to its individual characteristics and weaknesses. At present though, molecular targeting approaches in combination with radiotherapy or chemoradiation have not yet proven to be beneficial over standard chemoradiation treatment in the clinical setting. A promising approach to improve efficacy is the combined usage of two targeting agents in order to inhibit backup pathways or achieve a more complete pathway inhibition. Here we review preclinical attempts to utilize such dual targeting strategies for future tumor radiosensitization.
Collapse
Affiliation(s)
- Katharina Hintelmann
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.,Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Malte Kriegs
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Kai Rothkamm
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Thorsten Rieckmann
- Laboratory of Radiobiology & Experimental Radiation Oncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.,Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
17
|
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: 8.8] [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
|
18
|
Ashrafizadeh M, Farhood B, Eleojo Musa A, Taeb S, Najafi M. Damage-associated molecular patterns in tumor radiotherapy. Int Immunopharmacol 2020; 86:106761. [PMID: 32629409 DOI: 10.1016/j.intimp.2020.106761] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 02/06/2023]
Abstract
Radiotherapy is one of the most common modalities for the treatment of cancer. One of the most promising effects of radiotherapy is immunologic cell death and the release of danger alarms, which are known as damage-associated molecular patterns (DAMPs). DAMPs are able to trigger cancer cells and other cells within tumor microenvironment (TME), either for suppression or promotion of tumor growth. Heat shock proteins (HSPs) including HSP70 and HSP90, high mobility group box 1 (HMGB1), and adenosine triphosphate (ATP) and its metabolites such as adenosine are the most common danger alarms that are released after radiotherapy-induced immunologic cell death. Some DAMPs including adenosine is able to interact with both cancer cells as well as other cells in TME to promote tumor growth and resistance to radiotherapy. However, others are able to trigger anti-tumor immunity or both tumor suppressive and immunosuppressive mechanisms depending on affected cells. In this review, we explain the mechanisms behind the release of radiation-induced DAMPs, and its consequences on cells within tumor. Targeting of these mechanisms may be in favor of tumor control in combination with radiotherapy and radioimmunotherapy.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences (International Campus), Tehran, Iran
| | - Shahram Taeb
- Ionizing and Non-Ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
19
|
Advances in DNA Repair-Emerging Players in the Arena of Eukaryotic DNA Repair. Int J Mol Sci 2020; 21:ijms21113934. [PMID: 32486270 PMCID: PMC7313471 DOI: 10.3390/ijms21113934] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
Genomic DNA is constantly damaged by factors produced during natural metabolic processes as well as agents coming from the external environment. Considering such a wide array of damaging agents, eukaryotic cells have evolved a DNA damage response (DRR) that opposes the influence of deleterious factors. Despite the broad knowledge regarding DNA damage and repair, new areas of research are emerging. New players in the field of DDR are constantly being discovered. The aim of this study is to review current knowledge regarding the roles of sirtuins, heat shock proteins, long-noncoding RNAs and the circadian clock in DDR and distinguish new agents that may have a prominent role in DNA damage response and repair.
Collapse
|
20
|
Gvozdenov Z, Bendix LD, Kolhe J, Freeman BC. The Hsp90 Molecular Chaperone Regulates the Transcription Factor Network Controlling Chromatin Accessibility. J Mol Biol 2019; 431:4993-5003. [PMID: 31628945 PMCID: PMC6983977 DOI: 10.1016/j.jmb.2019.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 08/29/2019] [Accepted: 09/11/2019] [Indexed: 01/02/2023]
Abstract
Genomic events including gene regulation and chromatin status are controlled by transcription factors. Here we report that the Hsp90 molecular chaperone broadly regulates the transcription factor protein family. Our studies identified a biphasic use of Hsp90 in which early inactivation (15 min) of the chaperone triggered a wide reduction of DNA binding events along the genome with concurrent changes to chromatin structure. Long-term loss (6 h) of Hsp90 resulted in a decline of a divergent yet overlaying pool of transcription factors that produced a distinct chromatin pattern. Although both phases involve protein folding, the early point correlated with Hsp90 acting in a late folding step that is critical for DNA binding function, whereas prolonged Hsp90 inactivation led to a significant decrease in the steady-state transcription factor protein levels. Intriguingly, despite the broad chaperone impact on a variety of transcription factors, the operational influence of Hsp90 was at the level of chromatin with only a mild effect on gene regulation. Thus, Hsp90 selectively governs the transcription factor process overseeing local chromatin structure.
Collapse
Affiliation(s)
- Zlata Gvozdenov
- University of Illinois, Urbana-Champaign, Department of Cell and Developmental Biology, 601 S. Goodwin Avenue, Urbana, IL 61801, USA
| | - Lindsey D Bendix
- University of Illinois, Urbana-Champaign, Department of Cell and Developmental Biology, 601 S. Goodwin Avenue, Urbana, IL 61801, USA
| | - Janhavi Kolhe
- University of Illinois, Urbana-Champaign, Department of Cell and Developmental Biology, 601 S. Goodwin Avenue, Urbana, IL 61801, USA
| | - Brian C Freeman
- University of Illinois, Urbana-Champaign, Department of Cell and Developmental Biology, 601 S. Goodwin Avenue, Urbana, IL 61801, USA.
| |
Collapse
|
21
|
Radiosensitization of HSF-1 Knockdown Lung Cancer Cells by Low Concentrations of Hsp90 Inhibitor NVP-AUY922. Cells 2019; 8:cells8101166. [PMID: 31569342 PMCID: PMC6829369 DOI: 10.3390/cells8101166] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 11/25/2022] Open
Abstract
The inhibition of heat shock protein 90 (Hsp90) a molecular chaperone for multiple oncogenic client proteins is considered as a promising approach to overcome radioresistance. Since most Hsp90 inhibitors activate HSF-1 that induces the transcription of cytoprotective and tumor-promoting stress proteins such as Hsp70 and Hsp27, a combined approach consisting of HSF-1 knockdown (k.d.) and Hsp90 inhibition was investigated. A specific HSF-1 k.d. was achieved in H1339 lung cancer cells using RNAi-Ready pSIRENRetroQ vectors with puromycin resistance. The Hsp90 inhibitor NVP-AUY922 was evaluated at low concentrations—ranging from 1–10 nM—in control and HSF-1 k.d. cells. Protein expression (i.e., Hsp27/Hsp70, HSF-1, pHSF-1, Akt, ß-actin) and transcriptional activity was assessed by western blot analysis and luciferase assays and radiosensitivity was measured by proliferation, apoptosis (Annexin V, active caspase 3), clonogenic cell survival, alkaline comet, γH2AX, 53BP1, and Rad51 foci assays. The k.d. of HSF-1 resulted in a significant reduction of basal and NVP-AUY922-induced Hsp70/Hsp27 expression levels. A combined approach consisting of HSF-1 k.d. and low concentrations of the Hsp90 inhibitor NVP-AUY922 reduces the Hsp90 client protein Akt and potentiates radiosensitization, which involves an impaired homologous recombination mediated by Rad51. Our findings are key for clinical applications of Hsp90 inhibitors with respect to adverse hepatotoxic effects.
Collapse
|
22
|
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: 11.0] [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]
|
23
|
Abstract
Radiation therapy is one of the most commonly used treatments for cancer. Radiation modifiers are agents that alter tumor or normal tissue response to radiation, such as radiation sensitizers and radiation protectors. Radiation sensitizers target aspects of tumor molecular biology or physiology to enhance tumor cell killing after irradiation. Radioprotectors prevent damage of normal tissues selectively. Radiation modifiers remain largely investigational at present, with the promise that molecular characterization of tumors may enhance the capacity for successful clinical development moving forward. A variety of radiation modifiers are described.
Collapse
Affiliation(s)
- Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Building 10 CRC, Room B2-3500, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
24
|
Wang X, An D, Liu X, Wang X, Li B. MicroRNA-27a downregulates the expression of Hsp90 and enhances the radiosensitivity in esophageal squamous cell carcinoma. Onco Targets Ther 2019; 12:5967-5977. [PMID: 31413593 PMCID: PMC6661375 DOI: 10.2147/ott.s197456] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/01/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose Accumulating evidence suggests that microRNAs (miRNAs) possess diverse cellular regulatory roles in radiation responses. In this study, we aimed to identify the role of miR-27a in esophageal squamous cell carcinoma (ESCC) radiosensitivity by exploring the relationship between miR-27a and heat shock protein 90 (Hsp90). Materials and methods We performed quantitative real-time polymerase chain reaction (qRT-PCR) to detect miR-27a expression in the plasma of ESCC patients and healthy volunteers. The expression of Hsp90 and its key client proteins associated with radioresistance were analyzed by Western blotting. Then, the effects of miR-27a on proliferation, apoptosis, cell cycle and radiosensitivity in ESCC cell lines were determined by CCK-8, flow cytometry, and clonogenic survival assay. We also generated subcutaneous tumors to explore whether miR-27a enhanced radiosensitivity in vivo. Results In our current study, we found that miR-27a expression was downregulated in the plasma of ESCC patients compared with that of healthy volunteers. Overexpression of miR-27a in ESCC cell lines caused a reduction of Hsp90 mRNA and protein. We also demonstrated that upregulation of miR-27a induced degradation of Hsp90 key client proteins associated with radioresistance. In related functional experiments, miR-27a significantly inhibited growth, increased radiation-induced apoptosis, induced cell cycle arrest in G0/G1 phase and enhanced ESCC radiosensitivity both in vitro and in vivo. Conclusion From these findings, we concluded that miR-27a may contribute to radiosensitivity by modulating Hsp90 expression. Moreover, miR-27a-based therapy utilized to target Hsp90 could be contemplated as a compelling alternative for sensitize ESCC to radiotherapy with fewer side effects.
Collapse
Affiliation(s)
- Xintong Wang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong, People's Republic of China.,Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Dianzheng An
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong, People's Republic of China
| | - Xiaomeng Liu
- University of Jinan, School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Xinlei Wang
- Department of Gastroenterology, Qingdao Hiser Medical Center, Qingdao, Shandong, People's Republic of China
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
25
|
Ma Y, Baltezor M, Rajewski L, Crow J, Samuel G, Staggs VS, Chastain KM, Toretsky JA, Weir SJ, Godwin AK. Targeted inhibition of histone deacetylase leads to suppression of Ewing sarcoma tumor growth through an unappreciated EWS-FLI1/HDAC3/HSP90 signaling axis. J Mol Med (Berl) 2019; 97:957-972. [PMID: 31025088 DOI: 10.1007/s00109-019-01782-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/17/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022]
Abstract
Ewing sarcoma (ES) are aggressive pediatric bone and soft tissue tumors driven by EWS-ETS fusion oncogenes, most commonly EWS-FLI1. Treatment of ES patients consists of up to 9 months of alternating courses of 2 chemotherapeutic regimens. Furthermore, EWS-ETS-targeted therapies have yet to demonstrate clinical benefit, thereby emphasizing a clinical responsibility to search for new therapeutic approaches. Our previous in silico drug screening identified entinostat as a drug hit that was predicted to reverse the ES disease signatures and EWS-FLI1-mediated gene signatures. Here, we establish preclinical proof of principle by investigating the in vitro and in vivo efficacy of entinostat in preclinical ES models, as well as characterizing the mechanisms of action and in vivo pharmacokinetics of entinostat. ES cells are preferentially sensitive to entinostat in an EWS-FLI1 or EWS-ERG-dependent manner. Entinostat induces apoptosis of ES cells through G0/G1 cell cycle arrest, intracellular reactive oxygen species (ROS) elevation, DNA damage, homologous recombination (HR) repair impairment, and caspase activation. Mechanistically, we demonstrate for the first time that HDAC3 is a transcriptional target of EWS-FLI1 and that entinostat inhibits growth of ES cells through suppressing a previously unexplored EWS-FLI1/HDAC3/HSP90 signaling axis. Importantly, entinostat significantly reduces tumor burden by 97.4% (89.5 vs. 3397.3 mm3 of vehicle, p < 0.001) and prolongs the median survival of mice (15.5 vs. 8.5 days of vehicle, p < 0.001), in two independent ES xenograft mouse models, respectively. Overall, our studies demonstrate promising activity of entinostat against ES, and support the clinical development of the entinostat-based therapies for children and young adults with metastatic/relapsed ES. KEY MESSAGES: • Entinostat potently inhibits ES both in vitro and in vivo. • EWS-FLI1 and EWS-ERG confer sensitivity to entinostat treatment. • Entinostat suppresses the EWS-FLI1/HDAC3/HSP90 signaling. • HDAC3 is a transcriptional target of EWS-FLI1. • HDAC3 is essential for ES cell viability and genomic stability maintenance.
Collapse
Affiliation(s)
- Yan Ma
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, 4005B Wahl Hall East, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Michael Baltezor
- Lead Development Optimization Shared Resource, University of Kansas Cancer Center, Biotechnology Innovation and Optimization Center, Lawrence, KS, USA.,Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, KS, USA
| | - Lian Rajewski
- Lead Development Optimization Shared Resource, University of Kansas Cancer Center, Biotechnology Innovation and Optimization Center, Lawrence, KS, USA
| | - Jennifer Crow
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, 4005B Wahl Hall East, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA
| | - Glenson Samuel
- Division of Hematology/Oncology, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Vincent S Staggs
- Health Services & Outcomes Research, Children's Mercy Kansas City and School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Katherine M Chastain
- Division of Hematology/Oncology, Children's Mercy Kansas City, Kansas City, MO, USA
| | - Jeffrey A Toretsky
- Department of Oncology, Georgetown University Medical Center, Washington, D.C., USA
| | - Scott J Weir
- Institute for Advancing Medical Innovation, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA.,University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, 4005B Wahl Hall East, 3901 Rainbow Boulevard, Kansas City, KS, 66160, USA. .,University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
26
|
Hofving T, Sandblom V, Arvidsson Y, Shubbar E, Altiparmak G, Swanpalmer J, Almobarak B, Elf AK, Johanson V, Elias E, Kristiansson E, Forssell-Aronsson E, Nilsson O. 177Lu-octreotate therapy for neuroendocrine tumours is enhanced by Hsp90 inhibition. Endocr Relat Cancer 2019; 26:437-449. [PMID: 30730850 PMCID: PMC6391910 DOI: 10.1530/erc-18-0509] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/07/2019] [Indexed: 12/28/2022]
Abstract
177Lu-octreotate is an FDA-approved radionuclide therapy for patients with gastroenteropancreatic neuroendocrine tumours (NETs) expressing somatostatin receptors. The 177Lu-octreotate therapy has shown promising results in clinical trials by prolonging progression-free survival, but complete responses are still uncommon. The aim of this study was to improve the 177Lu-octreotate therapy by means of combination therapy. To identify radiosensitising inhibitors, two cell lines, GOT1 and P-STS, derived from small intestinal neuroendocrine tumours (SINETs), were screened with 1,224 inhibitors alone or in combination with external radiation. The screening revealed that inhibitors of Hsp90 can potentiate the tumour cell-killing effect of radiation in a synergistic fashion (GOT1; false discovery rate <3.2×10-11). The potential for Hsp90 inhibitor ganetespib to enhance the anti-tumour effect of 177Lu-octreotate in an in vivo setting was studied in the somatostatin receptor-expressing GOT1 xenograft model. The combination led to a larger decrease in tumour volume relative to monotherapies and the tumour-reducing effect was shown to be synergistic. Using patient-derived tumour cells from eight metastatic SINETs, we could show that ganetespib enhanced the effect of 177Lu-octreotate therapy for all investigated patient tumours. Levels of Hsp90 protein expression were evaluated in 767 SINETs from 379 patients. We found that Hsp90 expression was upregulated in tumour cells relative to tumour stroma in the vast majority of SINETs. We conclude that Hsp90 inhibitors enhance the tumour-killing effect of 177Lu-octreotate therapy synergistically in SINET tumour models and suggest that this potentially promising combination should be further evaluated.
Collapse
Affiliation(s)
- Tobias Hofving
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Correspondence should be addressed to T Hofving:
| | - Viktor Sandblom
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Yvonne Arvidsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Emman Shubbar
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Gülay Altiparmak
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - John Swanpalmer
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Bilal Almobarak
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin Elf
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Viktor Johanson
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Erik Elias
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Erik Kristiansson
- Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Eva Forssell-Aronsson
- Department of Radiation Physics, Institute of Clinical Sciences, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Ola Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Cancer Center, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
27
|
Chandra A, Park SS, Pignolo RJ. Potential role of senescence in radiation-induced damage of the aged skeleton. Bone 2019; 120:423-431. [PMID: 30543989 DOI: 10.1016/j.bone.2018.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/04/2018] [Accepted: 12/08/2018] [Indexed: 12/21/2022]
Abstract
Human aging-related changes are exacerbated in cases of disease and cancer, and conversely aging is a catalyst for the occurrence of disease and multimorbidity. For example, old age is the most significant risk factor for cancer and among people who suffer from cancer, >60% are above the age of 65. Oxidative stress and DNA damage, leading to genomic instability and telomere dysfunction, are prevalent in aging and radiation-induced damage and are major cellular events that lead to senescence. Human exposures from nuclear fallout, cosmic radiation and clinical radiotherapy (RT) are some common sources of irradiation that affect bone tissue. RT has been used to treat malignant tumors for over a century, but the effects of radiation damage on tumor-adjacent normal tissue has largely been overlooked. There is an increase in the percent survivorship among patients post-RT, and it is in older survivors where the deleterious synergy between aging and radiation exposure conspires to promote tissue deterioration and dysfunction which then negatively impacts their quality of life. Thus, an aging skeleton is already pre-disposed to architectural deterioration, which is further worsened by radiation-induced bone damage. Effects of senescence and the senescence associated secretory phenotype (SASP) have been implicated in age-associated bone loss, but their roles in radiation-associated bone damage are still elusive. RT is used in treatment for a variety of cancers and in different anatomical locations, the sequelae of which include long-term morbidity and lifelong discomfort. Therefore, consideration of the growing evidence that implicates the role of senescence in radiation-induced bone damage argues in favor of exploiting current senotherapeutic approaches as a possible prevention or treatment.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Medicine, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Sean S Park
- Department of Radiation Oncology, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Robert J Pignolo
- Department of Medicine, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA; Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
28
|
Lai TH, Mitchell S, Wu PJ, Orwick S, Liu C, Ravikrishnan J, Woyach J, Mims A, Plunkett W, Puduvalli VK, Byrd JC, Lapalombella R, Sampath D. HSP90 inhibition depletes DNA repair proteins to sensitize acute myelogenous leukemia to nucleoside analog chemotherapeutics. Leuk Lymphoma 2019; 60:2308-2311. [PMID: 30773117 DOI: 10.1080/10428194.2019.1571197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Tzung-Huei Lai
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| | - Shaneice Mitchell
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| | - Pei-Jung Wu
- Division of Neuro-oncology, Department of Neurosurgery, The Ohio State University , Columbus , OH , USA
| | - Shelley Orwick
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| | - Chaomei Liu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Janani Ravikrishnan
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| | - Jennifer Woyach
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| | - Alice Mims
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| | - William Plunkett
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Vinay K Puduvalli
- Division of Neuro-oncology, Department of Neurosurgery, The Ohio State University , Columbus , OH , USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| | - Rosa Lapalombella
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| | - Deepa Sampath
- Division of Hematology, Department of Internal Medicine, The Ohio State University , Columbus , OH , USA
| |
Collapse
|
29
|
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: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Abstract
From bacteria to humans, ancient stress responses enable organisms to contend with damage to both the genome and the proteome. These pathways have long been viewed as fundamentally separate responses. Yet recent discoveries from multiple fields have revealed surprising links between the two. Many DNA-damaging agents also target proteins, and mutagenesis induced by DNA damage produces variant proteins that are prone to misfolding, degradation, and aggregation. Likewise, recent studies have observed pervasive engagement of a p53-mediated response, and other factors linked to maintenance of genomic integrity, in response to misfolded protein stress. Perhaps most remarkably, protein aggregation and self-assembly has now been observed in multiple proteins that regulate the DNA damage response. The importance of these connections is highlighted by disease models of both cancer and neurodegeneration, in which compromised DNA repair machinery leads to profound defects in protein quality control, and vice versa.
Collapse
|
31
|
Zabinsky RA, Mason GA, Queitsch C, Jarosz DF. It's not magic - Hsp90 and its effects on genetic and epigenetic variation. Semin Cell Dev Biol 2018; 88:21-35. [PMID: 29807130 DOI: 10.1016/j.semcdb.2018.05.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/15/2018] [Accepted: 05/15/2018] [Indexed: 10/14/2022]
Abstract
Canalization, or phenotypic robustness in the face of environmental and genetic perturbation, is an emergent property of living systems. Although this phenomenon has long been recognized, its molecular underpinnings have remained enigmatic until recently. Here, we review the contributions of the molecular chaperone Hsp90, a protein that facilitates the folding of many key regulators of growth and development, to canalization of phenotype - and de-canalization in times of stress - drawing on studies in eukaryotes as diverse as baker's yeast, mouse ear cress, and blind Mexican cavefish. Hsp90 is a hub of hubs that interacts with many so-called 'client proteins,' which affect virtually every aspect of cell signaling and physiology. As Hsp90 facilitates client folding and stability, it can epistatically suppress or enable the expression of genetic variants in its clients and other proteins that acquire client status through mutation. Hsp90's vast interaction network explains the breadth of its phenotypic reach, including Hsp90-dependent de novo mutations and epigenetic effects on gene regulation. Intrinsic links between environmental stress and Hsp90 function thus endow living systems with phenotypic plasticity in fluctuating environments. As environmental perturbations alter Hsp90 function, they also alter Hsp90's interaction with its client proteins, thereby re-wiring networks that determine the genotype-to-phenotype map. Ensuing de-canalization of phenotype creates phenotypic diversity that is not simply stochastic, but often has an underlying genetic basis. Thus, extreme phenotypes can be selected, and assimilated so that they no longer require environmental stress to manifest. In addition to acting on standing genetic variation, Hsp90 perturbation has also been linked to increased frequency of de novo variation and several epigenetic phenomena, all with the potential to generate heritable phenotypic change. Here, we aim to clarify and discuss the multiple means by which Hsp90 can affect phenotype and possibly evolutionary change, and identify their underlying common feature: at its core, Hsp90 interacts epistatically through its chaperone function with many other genes and their gene products. Its influence on phenotypic diversification is thus not magic but rather a fundamental property of genetics.
Collapse
Affiliation(s)
- Rebecca A Zabinsky
- Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford, CA, United States
| | | | - Christine Queitsch
- Department of Genome Sciences, University of Washington, Seattle, WA, United States.
| | - Daniel F Jarosz
- Department of Chemical and Systems Biology, Stanford School of Medicine, Stanford, CA, United States; Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
32
|
Hsp90 Is Essential for Chl1-Mediated Chromosome Segregation and Sister Chromatid Cohesion. mSphere 2018; 3:3/3/e00225-18. [PMID: 29875144 PMCID: PMC5990887 DOI: 10.1128/msphere.00225-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/07/2018] [Indexed: 11/20/2022] Open
Abstract
Recently, Hsp90 functional loss has been linked to aneuploidy; however, until now none of the components of sister chromatid cohesion (SCC) have been demonstrated as the putative clients of Hsp90. In this study, we have established that Chl1, the protein which is involved in maintaining sister chromatid cohesion as well as in preventing chromosome loss, is a direct client of Hsp90. Thus, with understanding of the molecular mechanism, how Hsp90 controls the cohesion machinery might reveal new insights which can be exploited further for attenuation of tumorigenesis. Recent studies have demonstrated that aberrant sister chromatid cohesion causes genomic instability and hence is responsible for the development of a tumor. The Chl1 (chromosome loss 1) protein (homolog of human ChlRl/DDX11 helicase) plays an essential role in the proper segregation of chromosomes during mitosis. The helicase activity of Chl1 is critical for sister chromatid cohesion. Our study demonstrates that Hsp90 interacts with Chl1 and is necessary for its stability. We observe that the Hsp90 nonfunctional condition (temperature-sensitive iG170Dhsp82 strain at restrictive temperature) induces proteasomal degradation of Chl1. We have mapped the domains of Chl1 and identified that the presence of domains II, III, and IV is essential for efficient interaction with Hsp90. We have demonstrated that Hsp90 inhibitor 17-AAG (17-allylamino-geldenamycin) causes destabilization of Chl1 protein and enhances significant disruption of sister chromatid cohesion, which is comparable to that observed under the Δchl1 condition. Our study also revealed that 17-AAG treatment causes an increased frequency of chromosome loss to a similar extent as that of the Δchl1 cells. Hsp90 functional loss has been earlier linked to aneuploidy with very poor mechanistic insight. Our result identifies Chl1 as a novel client of Hsp90, which could be further explored to gain mechanistic insight into aneuploidy. IMPORTANCE Recently, Hsp90 functional loss has been linked to aneuploidy; however, until now none of the components of sister chromatid cohesion (SCC) have been demonstrated as the putative clients of Hsp90. In this study, we have established that Chl1, the protein which is involved in maintaining sister chromatid cohesion as well as in preventing chromosome loss, is a direct client of Hsp90. Thus, with understanding of the molecular mechanism, how Hsp90 controls the cohesion machinery might reveal new insights which can be exploited further for attenuation of tumorigenesis.
Collapse
|
33
|
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: 87] [Impact Index Per Article: 14.5] [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
|
34
|
Menotta M, Orazi S, Gioacchini AM, Spapperi C, Ricci A, Chessa L, Magnani M. Proteomics and transcriptomics analyses of ataxia telangiectasia cells treated with Dexamethasone. PLoS One 2018; 13:e0195388. [PMID: 29608596 PMCID: PMC5880408 DOI: 10.1371/journal.pone.0195388] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/21/2018] [Indexed: 12/21/2022] Open
Abstract
Ataxia telangiectasia (A-T) is an incurable and rare hereditary syndrome. In recent times, treatment with glucocorticoid analogues has been shown to improve the neurological symptoms that characterize this condition, but the molecular mechanism of action of these analogues remains unknown. Hence, the aim of this study was to gain insight into the molecular mechanism of action of glucocorticoid analogues in the treatment of A-T by investigating the role of Dexamethasone (Dexa) in A-T lymphoblastoid cell lines. We used 2DE and tandem MS to identify proteins that were influenced by the drug in A-T cells but not in healthy cells. Thirty-four proteins were defined out of a total of 746±63. Transcriptome analysis was performed by microarray and showed the differential expression of 599 A-T and 362 wild type (WT) genes and a healthy un-matching between protein abundance and the corresponding gene expression variation. The proteomic and transcriptomic profiles allowed the network pathway analysis to pinpoint the biological and molecular functions affected by Dexamethasone in Dexa-treated cells. The present integrated study provides evidence of the molecular mechanism of action of Dexamethasone in an A-T cellular model but also the broader effects of the drug in other tested cell lines.
Collapse
Affiliation(s)
- Michele Menotta
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Sara Orazi
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | | | - Chiara Spapperi
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Anastasia Ricci
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Luciana Chessa
- Department of Clinical and Molecular Medicine, Sapienza University, Rome, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| |
Collapse
|
35
|
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: 3.0] [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
|
36
|
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: 5] [Impact Index Per Article: 0.8] [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
|
37
|
A comparison of cell survival and heat shock protein expression after radiation in normal dermal fibroblasts, microvascular endothelial cells, and different head and neck squamous carcinoma cell lines. Clin Oral Investig 2018; 22:2251-2262. [PMID: 29307045 DOI: 10.1007/s00784-017-2323-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 12/20/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Head and neck squamous cell carcinoma (HNSCC) shows increased radioresistance due to the manipulation of homeostatic mechanisms like the heat shock response. This study intended to comparatively analyze effects of ionizing radiation on different HNSCC cell lines (PCI) and normal human dermal fibroblasts (NHFs) and human dermal microvascular endothelial cells (HDMECs) to uncover differences in radiation coping strategies. MATERIALS AND METHODS Proliferation (BrdU assay), apoptosis (caspase 3/7) and intracellular protein expression of heat shock protein (HSP)-70, and phosphorylated and total HSP27, determined by enzyme-linked immunosorbent assay (ELISA), were analyzed after exposure to increasing doses of ionizing radiation (2, 6, and 12 Gray, Gy). RESULTS Cell count decreased dose-dependently, but PCI cell lines consistently showed higher numbers compared to NHF and HDMEC. Likewise, high doses reduced cell proliferation, but low-dose radiation (2 Gy) instead increased proliferation in PCI 9 and 52. Apoptosis was not detectable in PCI cell lines. Basic HSP70 expression was high in PCI cells with little additional increase by irradiation. PCI cells yielded high basic total HSP27 concentrations but irradiation dose-dependently increased HSP27 in HDMEC, NHF, and PCI cells. Phosphorylated HSP27 concentrations were highest in NHF. CONCLUSION PCI cell lines showed higher resistance to dose-dependent reduction in cell number, proliferation, and protection from apoptosis compared to NHF and HDMEC. In parallel, we observed a high basic and radiation-induced expression of intracellular HSP70 leading to the assumption that the radioresistance of PCI cells is conferred by HSP70. CLINICAL RELEVANCE HNSCC use HSP to escape radiation-induced apoptosis and certain subtypes might increase proliferation after low-dose irradiation.
Collapse
|
38
|
Kinzel L, Ernst A, Orth M, Albrecht V, Hennel R, Brix N, Frey B, Gaipl US, Zuchtriegel G, Reichel CA, Blutke A, Schilling D, Multhoff G, Li M, Niyazi M, Friedl AA, Winssinger N, Belka C, Lauber K. A novel HSP90 inhibitor with reduced hepatotoxicity synergizes with radiotherapy to induce apoptosis, abrogate clonogenic survival, and improve tumor control in models of colorectal cancer. Oncotarget 2017; 7:43199-43219. [PMID: 27259245 PMCID: PMC5190018 DOI: 10.18632/oncotarget.9774] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 05/24/2016] [Indexed: 12/20/2022] Open
Abstract
The chaperone heat shock protein 90 (HSP90) crucially supports the maturation, folding, and stability of a variety of client proteins which are of pivotal importance for the survival and proliferation of cancer cells. Consequently, targeting of HSP90 has emerged as an attractive strategy of anti-cancer therapy, and it appears to be particularly effective in the context of molecular sensitization towards radiotherapy as has been proven in preclinical models of different cancer entities. However, so far the clinical translation has largely been hampered by suboptimal pharmacological properties and serious hepatotoxicity of first- and second-generation HSP90 inhibitors. Here, we report on NW457, a novel radicicol-derived member of the pochoxime family with reduced hepatotoxicity, how it inhibits the DNA damage response and how it synergizes with ionizing irradiation to induce apoptosis, abrogate clonogenic survival, and improve tumor control in models of colorectal cancer in vitro and in vivo.
Collapse
Affiliation(s)
- Linda Kinzel
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Anne Ernst
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael Orth
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Valerie Albrecht
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Roman Hennel
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nikko Brix
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Gabriele Zuchtriegel
- Department of Otorhinolaryngology, Head and Neck Surgery, and Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christoph A Reichel
- Department of Otorhinolaryngology, Head and Neck Surgery, and Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology at the Center for Clinical Veterinary Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Daniela Schilling
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Minglun Li
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Anna A Friedl
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Nicolas Winssinger
- Department of Organic Chemistry, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Claus Belka
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
39
|
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: 2.7] [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
|
40
|
Seshacharyulu P, Baine MJ, Souchek JJ, Menning M, Kaur S, Yan Y, Ouellette MM, Jain M, Lin C, Batra SK. Biological determinants of radioresistance and their remediation in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2017; 1868:69-92. [PMID: 28249796 PMCID: PMC5548591 DOI: 10.1016/j.bbcan.2017.02.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/16/2017] [Accepted: 02/17/2017] [Indexed: 12/17/2022]
Abstract
Despite recent advances in radiotherapy, a majority of patients diagnosed with pancreatic cancer (PC) do not achieve objective responses due to the existence of intrinsic and acquired radioresistance. Identification of molecular mechanisms that compromise the efficacy of radiation therapy and targeting these pathways is paramount for improving radiation response in PC patients. In this review, we have summarized molecular mechanisms associated with the radio-resistant phenotype of PC. Briefly, we discuss the reversible and irreversible biological consequences of radiotherapy, such as DNA damage and DNA repair, mechanisms of cancer cell survival and radiation-induced apoptosis following radiotherapy. We further describe various small molecule inhibitors and molecular targeting agents currently being tested in preclinical and clinical studies as potential radiosensitizers for PC. Notably, we draw attention towards the confounding effects of cancer stem cells, immune system, and the tumor microenvironment in the context of PC radioresistance and radiosensitization. Finally, we discuss the need for examining selective radioprotectors in light of the emerging evidence on radiation toxicity to non-target tissue associated with PC radiotherapy.
Collapse
Affiliation(s)
- Parthasarathy Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michael J Baine
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Joshua J Souchek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Melanie Menning
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Ying Yan
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michel M. Ouellette
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Chi Lin
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
41
|
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: 3.9] [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
|
42
|
Cossar LH, Schache AG, Risk JM, Sacco JJ, Jones NJ, Lord R. Modulating the DNA Damage Response to Improve Treatment Response in Cervical Cancer. Clin Oncol (R Coll Radiol) 2017; 29:626-634. [PMID: 28336131 DOI: 10.1016/j.clon.2017.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 01/06/2023]
Abstract
Cervical cancer is the fourth most common cause of cancer-related death in women worldwide and new therapeutic approaches are needed to improve clinical outcomes for this group of patients. Current treatment protocols for locally advanced and metastatic disease consist of ionising radiation and chemotherapy. Chemoradiation induces cytotoxic levels of DNA double-strand breaks, which activates programmed cell death via the DNA damage response (DDR). Cervical cancers are unique given an almost exclusive association with human papillomavirus (HPV) infection; a potent manipulator of the DDR, with the potential to alter tumour sensitivity to DNA-damaging agents and influence treatment response. This review highlights the wide range of therapeutic strategies in development that have the potential to modulate DDR and sensitise cervical tumours to DNA-damaging agents in the context of HPV oncogenesis.
Collapse
Affiliation(s)
- L H Cossar
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Clatterbridge Cancer Centre, Wirral, UK.
| | - A G Schache
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - J M Risk
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - J J Sacco
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Clatterbridge Cancer Centre, Wirral, UK
| | - N J Jones
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - R Lord
- Clatterbridge Cancer Centre, Wirral, UK
| |
Collapse
|
43
|
Kudryavtsev VA, Khokhlova AV, Mosina VA, Selivanova EI, Kabakov AE. Induction of Hsp70 in tumor cells treated with inhibitors of the Hsp90 activity: A predictive marker and promising target for radiosensitization. PLoS One 2017; 12:e0173640. [PMID: 28291803 PMCID: PMC5349677 DOI: 10.1371/journal.pone.0173640] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/23/2017] [Indexed: 01/03/2023] Open
Abstract
We studied a role of the inducible heat shock protein 70 (Hsp70) in cellular response to radiosensitizing treatments with inhibitors of the heat shock protein 90 (Hsp90) chaperone activity. Cell lines derived from solid tumors of different origin were treated with the Hsp90 inhibitors (17AAG, geldanamycin, radicicol, NVP-AUY922) or/and γ-photon radiation. For comparison, human cells of the non-cancerous origin were subjected to the same treatments. We found that the Hsp90 inhibitors yielded considerable radiosensitization only when they cause early and pronounced Hsp70 induction; moreover, a magnitude of radiosensitization was positively correlated with the level of Hsp70 induction. The quantification of Hsp70 levels in Hsp90 inhibitor-treated normal and cancer cells enabled to predict which of them will be susceptible to any Hsp90-inhibiting radiosensitizer as well as what concentrations of the inhibitors ensure the preferential cytotoxicity in the irradiated tumors without aggravating radiation damage to adjacent normal tissues. Importantly, the Hsp70 induction in the Hsp90 inhibitor-treated cancer cells appears to be their protective response that alleviates the tumor-sensitizing effects of the Hsp90 inactivation. Combination of the Hsp70-inducing inhibitors of Hsp90 with known inhibitors of the Hsp induction such as quercetin, triptolide, KNK437, NZ28 prevented up-regulation of Hsp70 in the cancer cells thereby increasing their post-radiation apoptotic/necrotic death and decreasing their post-radiation viability/clonogenicity. Similarly, co-treatment with the two inhibitors conferred the enhanced radiosensitization of proliferating rather than quiescent human vascular endothelial cells which may be used for suppressing the tumor-stimulated angiogenesis. Thus, the easily immunodetectable Hsp70 induction can be a useful marker for predicting effects of Hsp90-inhibiting radiosensitizers on tumors and normal tissues exposed to ionizing radiation. Moreover, targeting the Hsp70 induction in Hsp90 inhibitor-treated cancer cells and tumor vasculature cells may beneficially enhance the radiosensitizing effect.
Collapse
Affiliation(s)
- Vladimir A. Kudryavtsev
- Department of Radiation Biochemistry, A. Tsyb Medical Radiological Research Center, Obninsk, Russia
| | - Anna V. Khokhlova
- Department of Radiation Biochemistry, A. Tsyb Medical Radiological Research Center, Obninsk, Russia
| | - Vera A. Mosina
- Department of Radiation Biochemistry, A. Tsyb Medical Radiological Research Center, Obninsk, Russia
| | - Elena I. Selivanova
- Department of Radiation Biochemistry, A. Tsyb Medical Radiological Research Center, Obninsk, Russia
| | - Alexander E. Kabakov
- Department of Radiation Biochemistry, A. Tsyb Medical Radiological Research Center, Obninsk, Russia
- * E-mail:
| |
Collapse
|
44
|
HSP90 inhibition sensitizes head and neck cancer to platin-based chemoradiotherapy by modulation of the DNA damage response resulting in chromosomal fragmentation. BMC Cancer 2017; 17:86. [PMID: 28143445 PMCID: PMC5282703 DOI: 10.1186/s12885-017-3084-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 01/23/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Concurrent cisplatin radiotherapy (CCRT) is a current standard-of-care for locally advanced head and neck squamous cell carcinoma (HNSCC). However, CCRT is frequently ineffective in patients with advanced disease. It has previously been shown that HSP90 inhibitors act as radiosensitizers, but these studies have not focused on CCRT in HNSCC. Here, we evaluated the HSP90 inhibitor, AUY922, combined with CCRT. METHODS The ability of AUY922 to sensitize to CCRT was assessed in p53 mutant head and neck cell lines by clonogenic assay. Modulation of the CCRT induced DNA damage response (DDR) by AUY922 was characterized by confocal image analysis of RAD51, BRCA1, 53BP1, ATM and mutant p53 signaling. The role of FANCA depletion by AUY922 was examined using shRNA. Cell cycle checkpoint abrogation and chromosomal fragmentation was assessed by western blot, FACS and confocal. The role of ATM was also assessed by shRNA. AUY922 in combination with CCRT was assessed in vivo. RESULTS The combination of AUY922 with cisplatin, radiation and CCRT was found to be synergistic in p53 mutant HNSCC. AUY922 leads to significant alterations to the DDR induced by CCRT. This comprises inhibition of homologous recombination through decreased RAD51 and pS1524 BRCA1 with a corresponding increase in 53BP1 foci, activation of ATM and signaling into mutant p53. A shift to more error prone repair combined with a loss of checkpoint function leads to fragmentation of chromosomal material. The degree of disruption to DDR signalling correlated to chromosomal fragmentation and loss of clonogenicity. ATM shRNA indicated a possible rationale for the combination of AUY922 and CCRT in cells lacking ATM function. CONCLUSIONS This study supports future clinical studies combining AUY922 and CCRT in p53 mutant HNSCC. Modulation of the DDR and chromosomal fragmentation are likely to be analytical points of interest in such trials.
Collapse
|
45
|
Jiang J, Lu Y, Li Z, Li L, Niu D, Xu W, Liu J, Fu L, Zhou Z, Gu Y, Xia F. Ganetespib overcomes resistance to PARP inhibitors in breast cancer by targeting core proteins in the DNA repair machinery. Invest New Drugs 2017; 35:251-259. [PMID: 28111726 DOI: 10.1007/s10637-016-0424-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 12/30/2016] [Indexed: 11/24/2022]
Abstract
DNA damage repair plays essential roles in drug resistance, especially resistance to Poly (ADP-ribose) polymerase (PARP) inhibitors in the clinic. A subset of DNA repair proteins such as Breast cancer gene 1 (BRCA1), BRCA2 and RecA homolog (RAD51) are client proteins of heat shock protein 90 (Hsp90). Clearance of these DNA repair proteins by inhibition of Hsp90 is a promising strategy for overcoming resistance to PARP inhibitors. Here we report the pharmacological analysis of the highly potent second-generation Hsp90 inhibitor, ganetespib. Methods Nuclear BRCA1, BRCA2, and RAD51 expression in breast cancer cells were detected by subcellular fractionation and western blot analysis. Formation of nuclear RAD51 and γ-H2AX foci was analyzed by immunofluorescent staining. The cytotoxicity of ganetespib and ABT-888 in breast cancer cells were evaluated by cell proliferation, colony survival, and apoptosis assay. To investigate the efficacy of this therapy in vivo, SCID mice bearing MCF7 xenografts were treated with ganetespib and ABT-888, both as single agents and in combination. Results Ganetespib significantly destabilized nuclear BRCA1, BRCA2, and RAD51, and efficiently disrupted homologous recombination-mediated DNA double-strand break repair in breast cancer cells. The synergistic antitumor effects of ganetespib and the PARP inhibitor, ABT-888 were observed, and concurrent treatment with both inhibitors synergistically inhibited xenograft tumor growth. Importantly, the combined treatment was well tolerated, without significant loss of body weight or major histological changes in the breast cancer xenograft model. Conclusion These data provide a novel strategy for the treatment of breast cancer with wild type BRCA1 using combination therapy targeting Hsp90 to overcome resistance to PARP inhibitors.
Collapse
Affiliation(s)
- Juhong Jiang
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yuanzhi Lu
- Department of Pathology, the First Affiliated Hospital, Jinan University, #601 West Huangpu Avenue, Guangzhou, Guangdong, 510632, China
| | - Zhi Li
- Department of Pathology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Liping Li
- Department of Biochemistry, Guangdong Medical University, Dongguang, Guangdong, 523808, China
| | - Daoli Niu
- Department of Radiation Oncology, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Wenwei Xu
- Department of Radiation Oncology, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510120, China
| | - Jing Liu
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Lin Fu
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Ziqing Zhou
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yingying Gu
- The State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
| | - Fen Xia
- Arkansas College of Medicine, 4301 W. Markham, Slot 771, Little Rock, AR, 72205, USA.
| |
Collapse
|
46
|
Kramer D, Stark N, Schulz-Heddergott R, Erytch N, Edmunds S, Roßmann L, Bastians H, Concin N, Moll UM, Dobbelstein M. Strong antitumor synergy between DNA crosslinking and HSP90 inhibition causes massive premitotic DNA fragmentation in ovarian cancer cells. Cell Death Differ 2016; 24:300-316. [PMID: 27834954 DOI: 10.1038/cdd.2016.124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 08/13/2016] [Accepted: 09/26/2016] [Indexed: 12/29/2022] Open
Abstract
All current regimens for treating ovarian cancer center around carboplatin as standard first line. The HSP90 inhibitor ganetespib is currently being assessed in advanced clinical oncology trials. Thus, we tested the combined effects of ganetespib and carboplatin on a panel of 15 human ovarian cancer lines. Strikingly, the two drugs strongly synergized in cytotoxicity in tumor cells lacking wild-type p53. Mechanistically, ganetespib and carboplatin in combination, but not individually, induced persistent DNA damage causing massive global chromosome fragmentation. Live-cell microscopy revealed chromosome fragmentation occurring to a dramatic degree when cells condensed their chromatin in preparation for mitosis, followed by cell death in mitosis or upon aberrant exit from mitosis. HSP90 inhibition caused the rapid decay of key components of the Fanconi anemia pathway required for repair of carboplatin-induced interstrand crosslinks (ICLs), as well as of cell cycle checkpoint mediators. Overexpressing FancA rescued the DNA damage induced by the drug combination, indicating that FancA is indeed a key client of Hsp90 that enables cancer cell survival in the presence of ICLs. Conversely, depletion of nuclease DNA2 prevented chromosomal fragmentation, pointing to an imbalance of defective repair in the face of uncontrolled nuclease activity as mechanistic basis for the observed premitotic DNA fragmentation. Importantly, the drug combination induced robust antitumor activity in xenograft models, again accompanied with depletion of FancA. In sum, our findings indicate that ganetespib strongly potentiates the antitumor efficacy of carboplatin by causing combined inhibition of DNA repair and cell cycle control mechanisms, thus triggering global chromosome disruption, aberrant mitosis and cell death.
Collapse
Affiliation(s)
- Daniela Kramer
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany
| | - Nadine Stark
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany
| | - Ramona Schulz-Heddergott
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany
| | - Norman Erytch
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany
| | - Shelley Edmunds
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany
| | - Laura Roßmann
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany
| | - Holger Bastians
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany
| | - Nicole Concin
- Department of Obstetrics and Gynaecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ute M Moll
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany.,Department of Pathology, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Matthias Dobbelstein
- Institute of Molecular Oncology, Göttingen Center of Molecular Biosciences (GZMB), University Medical Center Göttingen, Göttingen D-37077, Germany
| |
Collapse
|
47
|
Lee Y, Sunada S, Hirakawa H, Fujimori A, Nickoloff JA, Okayasu R. TAS-116, a Novel Hsp90 Inhibitor, Selectively Enhances Radiosensitivity of Human Cancer Cells to X-rays and Carbon Ion Radiation. Mol Cancer Ther 2016; 16:16-24. [PMID: 28062703 DOI: 10.1158/1535-7163.mct-16-0573] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 01/09/2023]
Abstract
Hsp90 inhibitors have been investigated as cancer therapeutics in monotherapy and to augment radiotherapy; however, serious adverse effects of early-generation Hsp90 inhibitors limited their development. TAS-116 is a novel Hsp90 inhibitor with lower adverse effects than other Hsp90 inhibitors, and here, we investigated the radiosensitizing effects of TAS-116 in low linear energy transfer (LET) X-ray and high LET carbon ion-irradiated human cancer cells and mouse tumor xenografts. TAS-116 decreased cell survival of both X-ray and carbon ion-irradiated human cancer cell lines (HeLa and H1299 cells), and similar to other Hsp90 inhibitors, it did not affect radiosensitivity of noncancerous human fibroblasts. TAS-116 increased the number of radiation-induced γ-H2AX foci and delayed the repair of DNA double-strand breaks (DSB). TAS-116 reduced the expression of proteins that mediate repair of DSBs by homologous recombination (RAD51) and nonhomologous end joining (Ku, DNA-PKcs), and suppressed formation of RAD51 foci and phosphorylation/activation of DNA-PKcs. TAS-116 also decreased expression of the cdc25 cell-cycle progression marker, markedly increasing G2-M arrest. Combined treatment of mouse tumor xenografts with carbon ions and TAS-116 showed promising delay in tumor growth compared with either individual treatment. These results demonstrate that TAS-116 radiosensitizes human cancer cells to both X-rays and carbon ions by inhibiting the two major DSB repair pathways, and these effects were accompanied by marked cell-cycle arrest. The promising results of combination TAS-116 + carbon ion radiotherapy of tumor xenografts justify further exploration of TAS-116 as an adjunct to radiotherapy using low or high LET radiation. Mol Cancer Ther; 16(1); 16-24. ©2016 AACR.
Collapse
Affiliation(s)
- Younghyun Lee
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Inage-ku, Chiba, Japan
| | - Shigeaki Sunada
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Inage-ku, Chiba, Japan.,Department of Nuclear Engineering and Management, School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Hirokazu Hirakawa
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Inage-ku, Chiba, Japan
| | - Akira Fujimori
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Inage-ku, Chiba, Japan
| | - Jac A Nickoloff
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Ryuichi Okayasu
- Department of Basic Medical Sciences for Radiation Damages, National Institute of Radiological Sciences, Inage-ku, Chiba, Japan.
| |
Collapse
|
48
|
Dallas LJ, Bean TP, Turner A, Lyons BP, Jha AN. Exposure to tritiated water at an elevated temperature: Genotoxic and transcriptomic effects in marine mussels (M. galloprovincialis). JOURNAL OF ENVIRONMENTAL RADIOACTIVITY 2016; 164:325-336. [PMID: 27552656 DOI: 10.1016/j.jenvrad.2016.07.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/27/2016] [Indexed: 06/06/2023]
Abstract
Temperature is an abiotic factor of particular concern for assessing the potential impacts of radionuclides on marine species. This is particularly true for tritium, which is discharged as tritiated water (HTO) in the process of cooling nuclear institutions. Additionally, with sea surface temperatures forecast to rise 0.5-3.5 °C in the next 30-100 years, determining the interaction of elevated temperature with radiological exposure has never been more relevant. We assessed the tissue-specific accumulation, transcriptional expression of key genes, and genotoxicity of tritiated water to marine mussels at either 15 or 25 °C, over a 7 day time course with sampling after 1 h, 12 h, 3 d and 7d. The activity concentration used (15 MBq L-1) resulted in tritium accumulation that varied with both time and temperature, but consistently produced dose rates (calculated using the ERICA tool) of <20 Gy h-1, i.e. considerably below the recommended guidelines of the IAEA and EURATOM. Despite this, there was significant induction of DNA strand breaks (as measured by the comet assay), which also showed a temperature-dependent time shift. At 15 °C, DNA damage was only significantly elevated after 7 d, in contrast to 25 °C where a similar response was observed after only 3 d. The transcription profiles of two isoforms of hsp70, hsp90, mt20, p53 and rad51 indicated potential mechanisms behind this temperature-induced acceleration of genotoxicity, which may be the result of compromised defence. Specifically, genes involved in protein folding, DNA double strand break repair and cell cycle checkpoint control were upregulated after 3 d HTO exposure at 15 °C, but significantly downregulated when the same exposure occurred at 25 °C. This study is the first to investigate temperature effects on radiation-induced genotoxicity in an ecologically relevant marine invertebrate, Mytilus galloprovincialis. From an ecological perspective, our study suggests that mussels (or similar marine species) exposed to increased temperature and HTO may have a compromised ability to defend against genotoxic stress.
Collapse
Affiliation(s)
- Lorna J Dallas
- School of Biological Sciences, Plymouth University, Drake Circus, Plymouth, PL4 8AA, UK
| | - Tim P Bean
- Centre for Environment, Fisheries and Aquaculture Science, The Nothe, Barrack Road, Weymouth, DT4 8UB, UK
| | - Andrew Turner
- School of Geography, Earth & Environmental Sciences, Plymouth University, Drake Circus, Plymouth, PL4 8AA, UK
| | - Brett P Lyons
- Centre for Environment, Fisheries and Aquaculture Science, The Nothe, Barrack Road, Weymouth, DT4 8UB, UK
| | - Awadhesh N Jha
- School of Biological Sciences, Plymouth University, Drake Circus, Plymouth, PL4 8AA, UK.
| |
Collapse
|
49
|
Rae C, Mairs RJ. Evaluation of the radiosensitizing potency of chemotherapeutic agents in prostate cancer cells. Int J Radiat Biol 2016; 93:194-203. [PMID: 27600766 DOI: 10.1080/09553002.2017.1231946] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Despite recent advances in the treatment of metastatic prostate cancer, survival rates are low and treatment options are limited to chemotherapy and hormonal therapy. Although ionizing radiation is used to treat localized and metastatic prostate cancer, the most efficient use of radiotherapy is yet to be defined. Our purpose was to determine in vitro the potential benefit to be gained by combining radiation treatment with cytotoxic drugs. MATERIALS AND METHODS Inhibitors of DNA repair and heat shock protein 90 and an inducer of oxidative stress were evaluated in combination with X-radiation for their capacity to reduce clonogenic survival and delay the growth of multicellular tumor spheroids. RESULTS Inhibitors of the PARP DNA repair pathway, olaparib and rucaparib, and the HSP90 inhibitor 17-DMAG, enhanced the clonogenic cell kill and spheroid growth delay induced by X-radiation. However, the oxidative stress-inducing drug elesclomol failed to potentiate the effects of X-radiation. PARP inhibitors arrested cells in the G2/M phase when administered as single agents or in combination with radiation, whereas elesclomol and 17-DMAG did not affect radiation-induced cell cycle modulation. CONCLUSION These results indicate that radiotherapy of prostate cancer may be optimized by combination with inhibitors of PARP or HSP90, but not elesclomol.
Collapse
Affiliation(s)
- Colin Rae
- a Radiation Oncology , Institute of Cancer Sciences, University of Glasgow , Glasgow , UK
| | - Robert J Mairs
- a Radiation Oncology , Institute of Cancer Sciences, University of Glasgow , Glasgow , UK
| |
Collapse
|
50
|
Yan S, Li X, Jin Q, Yuan J. MicroRNA-145 sensitizes cervical cancer cells to low-dose irradiation by downregulating OCT4 expression. Exp Ther Med 2016; 12:3130-3136. [PMID: 27882128 DOI: 10.3892/etm.2016.3731] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/23/2016] [Indexed: 12/13/2022] Open
Abstract
Poor elucidation of the mechanisms involved in regulating the radiosensitivity of cancers prevents the extensive application of low-dose radiotherapy in clinical settings. The present study was conducted to investigate the role of microRNA-145 (miR-145) in the modulation of cervical cancer cell radiosensitivity, as well as to identify the underlying target of miR-145 during this process. Cervical cancer tera cells were initially exposed to doses of radiation between 1 and 6 Gy before the assessments of the cell viability and apoptosis rate. Irradiation at dose of 1 Gy was screened as optimum dose and used in subsequent experiments. A dual luciferase reporter assay was performed to demonstrate that octamer-binding transcription factor 4 (OCT4) is a target of miR-145 in cervical cancer. Consequently, OCT4 was suggested to be a target of miR-145, as a dual luciferase vector that was ligated to a fragment corresponding to the predicted target site of miR-145 in OCT4 3'-UTR showed an 83% reduction in fluorescence. Following exposure to 1 Gy irradiation, tera cells transfected with miR-145 mimics, which showed downregulation of OCT4 and cyclin D1, had lower cell viability and cell migration rate and higher apoptosis rate compared to non-transfected cells. However, the co-transfection of miR-145 mimics and OCT4 expression vector restored OCT4 and cyclin D1 expression levels and made no significant difference in terms of cell viability, cell migration rate and apoptosis rate. The present results indicate that miR-145 increases the radiosensitivity of cervical cancer cells by silencing OCT4, that cyclin D1 is putatively under the positive regulation of OCT4 and mediates miR-145 function.
Collapse
Affiliation(s)
- Siqi Yan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiangjun Li
- Department of Oncology, The Second People's Hospital, Changsha, Hunan 410000, P.R. China
| | - Qiao Jin
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Jun Yuan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|