1
|
Yang J, Liu HC, Zhang JQ, Zou JY, Zhang X, Chen WM, Gu Y, Hong H. The effect of metformin on senescence of T lymphocytes. Immun Ageing 2023; 20:73. [PMID: 38087369 PMCID: PMC10714529 DOI: 10.1186/s12979-023-00394-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/15/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Immunosenescence occurs as people age, leading to an increased incidence of age-related diseases. The number of senescent T cells also rises with age. T cell senescence and immune response dysfunction can result in a decline in immune function, especially in anti-tumor immune responses. Metformin has been shown to have various beneficial effects on health, such as lowering blood sugar levels, reducing the risk of cancer development, and slowing down the aging process. However, the immunomodulatory effects of metformin on senescent T cells still need to be investigated. METHODS PBMCs isolation from different age population (n = 88); Flow Cytometry is applied to determine the phenotypic characterization of senescent T lymphocytes; intracellular staining is applied to determine the function of senescent T cells; Enzyme-Linked Immunosorbent Assay (ELISA) is employed to test the telomerase concentration. The RNA-seq analysis of gene expression associated with T cell senescence. RESULTS The middle-aged group had the highest proportion of senescent T cells. We found that metformin could decrease the number of CD8 + senescent T cells. Metformin affects the secretion of SASP, inhibiting the secretion of IFN-γ in CD8 + senescent T cells. Furthermore, metformin treatment restrained the production of the proinflammatory cytokine IL-6 in lymphocytes. Metformin had minimal effects on Granzyme B secretion in senescent T cells, but it promoted the production of TNF-α in senescent T cells. Additionally, metformin increased the concentration of telomerase and the frequency of undifferentiated T cells. The results of RNA-seq showed that metformin promoted the expression of genes related to stemness and telomerase activity, while inhibiting the expression of DNA damage-associated genes. CONCLUSION Our findings reveal that metformin could inhibit T cell senescence in terms of cell number, effector function, telomerase content and gene expression in middle-aged individuals, which may serve as a promising approach for preventing age-related diseases in this population.
Collapse
Affiliation(s)
- Jia Yang
- The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhong Shan Two Road, Guang Zhou, 510000, Guang Dong, China
| | - Hai-Cheng Liu
- Key Laboratory of Tropical Disease Control of Sun Yat-Sen University, Ministry of Education, The Institute of Immunology of Zhong Shan Medical School, Sun Yat-Sen University, No.74 Zhong Shan Two Road, Guang Zhou, Guang Dong, 510000, China
| | - Jian-Qing Zhang
- The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhong Shan Two Road, Guang Zhou, 510000, Guang Dong, China
| | - Jian-Yong Zou
- The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhong Shan Two Road, Guang Zhou, 510000, Guang Dong, China
| | - Xin Zhang
- The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhong Shan Two Road, Guang Zhou, 510000, Guang Dong, China
| | - Wo-Ming Chen
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang West Road, Guang Zhou, 510120, China
| | - Yong Gu
- The First Affiliated Hospital of Sun Yat-Sen University, No.58 Zhong Shan Two Road, Guang Zhou, 510000, Guang Dong, China.
| | - Hai Hong
- Key Laboratory of Tropical Disease Control of Sun Yat-Sen University, Ministry of Education, The Institute of Immunology of Zhong Shan Medical School, Sun Yat-Sen University, No.74 Zhong Shan Two Road, Guang Zhou, Guang Dong, 510000, China.
| |
Collapse
|
2
|
Franza M, Albanesi J, Mancini B, Pennisi R, Leone S, Acconcia F, Bianchi F, di Masi A. The clinically relevant CHK1 inhibitor MK-8776 induces the degradation of the oncogenic protein PML-RARα and overcomes ATRA resistance in acute promyelocytic leukemia cells. Biochem Pharmacol 2023:115675. [PMID: 37406967 DOI: 10.1016/j.bcp.2023.115675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Acute promyelocytic leukemia (APL) is a hematological disease characterized by the expression of the oncogenic fusion protein PML-RARα. The current treatment approach for APL involves differentiation therapy using all-trans retinoic acid (ATRA) and arsenic trioxide (ATO). However, the development of resistance to therapy, occurrence of differentiation syndrome, and relapses necessitate the exploration of new treatment options that induce differentiation of leukemic blasts with low toxicity. In this study, we investigated the cellular and molecular effects of MK-8776, a specific inhibitor of CHK1, in ATRA-resistant APL cells. Treatment of APL cells with MK-8776 resulted in a decrease in PML-RARα levels, increased expression of CD11b, and increased granulocytic activity consistent with differentiation. Interestingly, we showed that the MK-8776-induced differentiating effect resulted synergic with ATO. We found that the reduction of PML-RARα by MK-8776 was dependent on both proteasome and caspases. Specifically, both caspase-1 and caspase-3 were activated by CHK1 inhibition, with caspase-3 acting upstream of caspase-1. Activation of caspase-3 was necessary to activate caspase-1 and promote PML-RARα degradation. Transcriptomic analysis revealed significant modulation of pathways and upstream regulators involved in the inflammatory response and cell cycle control upon MK-8776 treatment. Overall, the ability of MK-8776 to induce PML-RARα degradation and stimulate differentiation of immature APL cancer cells into more mature forms recapitulates the concept of differentiation therapy. Considering the in vivo tolerability of MK-8776, it will be relevant to evaluate its potential clinical benefit in APL patients resistant to standard ATRA/ATO therapy, as well as in patients with other forms of acute leukemias.
Collapse
Affiliation(s)
- Maria Franza
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Jacopo Albanesi
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Benedetta Mancini
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Rosa Pennisi
- Department of Oncology, University of Torino Medical School, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Torino, Italy
| | - Stefano Leone
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Filippo Acconcia
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarkers, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | - Alessandra di Masi
- Department of Sciences, Section of Biomedical Sciences and Technologies, Roma Tre University, Roma, Italy.
| |
Collapse
|
3
|
Shao J, Huang L, Lai W, Zou Y, Zhu Q. Design, Synthesis, and Biological Evaluation of Potent and Selective Inhibitors of Ataxia Telangiectasia Mutated and Rad3-Related (ATR) Kinase for the Efficient Treatment of Cancer. Molecules 2023; 28:molecules28114521. [PMID: 37298997 DOI: 10.3390/molecules28114521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Ataxia telangiectasia mutated and Rad3-related (ATR), a vital member of the phosphatidylinositol 3-kinase-related kinase (PIKK) family, plays a critical role in the DNA damage response (DDR). Tumor cells with a loss of DDR function or defects in the ataxia telangiectasia mutated (ATM) gene are generally more dependent on ATR for survival, suggesting that ATR is an attractive anticancer drug target based on its synthetic lethality. Herein, we present a potent and highly selective ATR inhibitor, ZH-12 (IC50 = 0.0068 μM). It showed potent antitumor activity as a single agent or in combination with cisplatin in the human colorectal adenocarcinoma LoVo tumor xenograft mouse model. Overall, ZH-12 may be a promising ATR inhibitor based on the principle of synthetic lethality and deserves further in-depth study.
Collapse
Affiliation(s)
- Jialu Shao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Huang
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology and Medicinal Chemistry, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Wenwen Lai
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Zou
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qihua Zhu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
4
|
Maiti S, Picard D. Cytosolic Hsp90 Isoform-Specific Functions and Clinical Significance. Biomolecules 2022; 12:1166. [PMID: 36139005 PMCID: PMC9496497 DOI: 10.3390/biom12091166] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 11/17/2022] Open
Abstract
The heat shock protein 90 (Hsp90) is a molecular chaperone and a key regulator of proteostasis under both physiological and stress conditions. In mammals, there are two cytosolic Hsp90 isoforms: Hsp90α and Hsp90β. These two isoforms are 85% identical and encoded by two different genes. Hsp90β is constitutively expressed and essential for early mouse development, while Hsp90α is stress-inducible and not necessary for survivability. These two isoforms are known to have largely overlapping functions and to interact with a large fraction of the proteome. To what extent there are isoform-specific functions at the protein level has only relatively recently begun to emerge. There are studies indicating that one isoform is more involved in the functionality of a specific tissue or cell type. Moreover, in many diseases, functionally altered cells appear to be more dependent on one particular isoform. This leaves space for designing therapeutic strategies in an isoform-specific way, which may overcome the unfavorable outcome of pan-Hsp90 inhibition encountered in previous clinical trials. For this to succeed, isoform-specific functions must be understood in more detail. In this review, we summarize the available information on isoform-specific functions of mammalian Hsp90 and connect it to possible clinical applications.
Collapse
Affiliation(s)
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH-1211 Geneve, Switzerland
| |
Collapse
|
5
|
The Role of Hsp90-R2TP in Macromolecular Complex Assembly and Stabilization. Biomolecules 2022; 12:biom12081045. [PMID: 36008939 PMCID: PMC9406135 DOI: 10.3390/biom12081045] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 01/27/2023] Open
Abstract
Hsp90 is a ubiquitous molecular chaperone involved in many cell signaling pathways, and its interactions with specific chaperones and cochaperones determines which client proteins to fold. Hsp90 has been shown to be involved in the promotion and maintenance of proper protein complex assembly either alone or in association with other chaperones such as the R2TP chaperone complex. Hsp90-R2TP acts through several mechanisms, such as by controlling the transcription of protein complex subunits, stabilizing protein subcomplexes before their incorporation into the entire complex, and by recruiting adaptors that facilitate complex assembly. Despite its many roles in protein complex assembly, detailed mechanisms of how Hsp90-R2TP assembles protein complexes have yet to be determined, with most findings restricted to proteomic analyses and in vitro interactions. This review will discuss our current understanding of the function of Hsp90-R2TP in the assembly, stabilization, and activity of the following seven classes of protein complexes: L7Ae snoRNPs, spliceosome snRNPs, RNA polymerases, PIKKs, MRN, TSC, and axonemal dynein arms.
Collapse
|
6
|
Emerging Link between Tsc1 and FNIP Co-Chaperones of Hsp90 and Cancer. Biomolecules 2022; 12:biom12070928. [PMID: 35883484 PMCID: PMC9312812 DOI: 10.3390/biom12070928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Heat shock protein-90 (Hsp90) is an ATP-dependent molecular chaperone that is tightly regulated by a group of proteins termed co-chaperones. This chaperone system is essential for the stabilization and activation of many key signaling proteins. Recent identification of the co-chaperones FNIP1, FNIP2, and Tsc1 has broadened the spectrum of Hsp90 regulators. These new co-chaperones mediate the stability of critical tumor suppressors FLCN and Tsc2 as well as the various classes of Hsp90 kinase and non-kinase clients. Many early observations of the roles of FNIP1, FNIP2, and Tsc1 suggested functions independent of FLCN and Tsc2 but have not been fully delineated. Given the broad cellular impact of Hsp90-dependent signaling, it is possible to explain the cellular activities of these new co-chaperones by their influence on Hsp90 function. Here, we review the literature on FNIP1, FNIP2, and Tsc1 as co-chaperones and discuss the potential downstream impact of this regulation on normal cellular function and in human diseases.
Collapse
|
7
|
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: 18] [Impact Index Per Article: 6.0] [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
|
8
|
Colangelo T, Carbone A, Mazzarelli F, Cuttano R, Dama E, Nittoli T, Albanesi J, Barisciano G, Forte N, Palumbo O, Graziano P, di Masi A, Colantuoni V, Sabatino L, Bianchi F, Mazzoccoli G. Loss of circadian gene Timeless induces EMT and tumor progression in colorectal cancer via Zeb1-dependent mechanism. Cell Death Differ 2022; 29:1552-1568. [PMID: 35034102 PMCID: PMC9345857 DOI: 10.1038/s41418-022-00935-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 01/05/2022] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
The circadian gene Timeless (TIM) provides a molecular bridge between circadian and cell cycle/DNA replication regulatory systems and has been recently involved in human cancer development and progression. However, its functional role in colorectal cancer (CRC), the third leading cause of cancer-related deaths worldwide, has not been fully clarified yet. Here, the analysis of two independent CRC patient cohorts (total 1159 samples) reveals that loss of TIM expression is an unfavorable prognostic factor significantly correlated with advanced tumor stage, metastatic spreading, and microsatellite stability status. Genome-wide expression profiling, in vitro and in vivo experiments, revealed that TIM knockdown induces the activation of the epithelial-to-mesenchymal transition (EMT) program. Accordingly, the analysis of a large set of human samples showed that TIM expression inversely correlated with a previously established gene signature of canonical EMT markers (EMT score), and its ectopic silencing promotes migration, invasion, and acquisition of stem-like phenotype in CRC cells. Mechanistically, we found that loss of TIM expression unleashes ZEB1 expression that in turn drives the EMT program and enhances the aggressive behavior of CRC cells. Besides, the deranged TIM-ZEB1 axis sets off the accumulation of DNA damage and delays DNA damage recovery. Furthermore, we show that the aggressive and genetically unstable 'CMS4 colorectal cancer molecular subtype' is characterized by a lower expression of TIM and that patients with the combination of low-TIM/high-ZEB1 expression have a poorer outcome. In conclusion, our results as a whole suggest the engagement of an unedited TIM-ZEB1 axis in key pathological processes driving malignant phenotype acquisition in colorectal carcinogenesis. Thus, TIM-ZEB1 expression profiling could provide a robust prognostic biomarker in CRC patients, supporting targeted therapeutic strategies with better treatment selection and patients' outcomes.
Collapse
Affiliation(s)
- Tommaso Colangelo
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Biomarkers Unit, Viale Padre Pio 7, 71013, San Giovanni Rotondo, (FG), Italy
| | - Annalucia Carbone
- Fondazione IRCCS Casa Sollievo della Sofferenza, Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Viale Cappuccini snc, 71013, San Giovanni Rotondo, (FG), Italy
| | - Francesco Mazzarelli
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Biomarkers Unit, Viale Padre Pio 7, 71013, San Giovanni Rotondo, (FG), Italy
| | - Roberto Cuttano
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Biomarkers Unit, Viale Padre Pio 7, 71013, San Giovanni Rotondo, (FG), Italy
| | - Elisa Dama
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Biomarkers Unit, Viale Padre Pio 7, 71013, San Giovanni Rotondo, (FG), Italy
| | - Teresa Nittoli
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Biomarkers Unit, Viale Padre Pio 7, 71013, San Giovanni Rotondo, (FG), Italy
| | - Jacopo Albanesi
- Department of Sciences, Roma Tre University, Viale G. Marconi, 446, 00154, Rome, (RM), Italy
| | - Giovannina Barisciano
- Department of Sciences and Technologies, University of Sannio, Via Traiano, 3, 82100, Benevento, (BN), Italy
| | - Nicola Forte
- UOC- Patologia Clinica-Settore Anatomia Patologica, Ospedale Fatebenefratelli, Viale Principe di Napoli, 14/A, 82100, Benevento, (BN), Italy
| | - Orazio Palumbo
- Fondazione IRCCS Casa Sollievo della Sofferenza, Division of Medical Genetics, Viale Padre Pio, 7d, 71013, San Giovanni Rotondo, (FG), Italy
| | - Paolo Graziano
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini snc, 71013, San Giovanni Rotondo, (FG), Italy
| | - Alessandra di Masi
- Department of Sciences, Roma Tre University, Viale G. Marconi, 446, 00154, Rome, (RM), Italy
| | - Vittorio Colantuoni
- Department of Sciences and Technologies, University of Sannio, Via Traiano, 3, 82100, Benevento, (BN), Italy
| | - Lina Sabatino
- Department of Sciences and Technologies, University of Sannio, Via Traiano, 3, 82100, Benevento, (BN), Italy
| | - Fabrizio Bianchi
- Fondazione IRCCS Casa Sollievo della Sofferenza, Cancer Biomarkers Unit, Viale Padre Pio 7, 71013, San Giovanni Rotondo, (FG), Italy.
| | - Gianluigi Mazzoccoli
- Fondazione IRCCS Casa Sollievo della Sofferenza, Department of Medical Sciences, Division of Internal Medicine and Chronobiology Laboratory, Viale Cappuccini snc, 71013, San Giovanni Rotondo, (FG), Italy.
| |
Collapse
|
9
|
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
|
10
|
Cai H, Dai X, Guo X, Zhang L, Cao K, Yan F, Ji B, Liu Y. Ataxia telangiectasia mutated inhibitor-loaded copper sulfide nanoparticles for low-temperature photothermal therapy of hepatocellular carcinoma. Acta Biomater 2021; 127:276-286. [PMID: 33812073 DOI: 10.1016/j.actbio.2021.03.051] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer, and is ranked the sixth most common neoplasm and the third leading cause of cancer-related deaths. Photothermal therapy (PTT) for thermal ablation of local tumors has recently emerged as a therapeutic strategy. However, the relatively high temperature of over 50 °C may lead to unexpected heat-related damage to tumor-adjacent normal tissues. Herein, we designed and synthesized ataxia telangiectasia mutated (ATM) inhibitor loaded hollow-structured CuS NPs with surface modification with anti-TGF-β antibody (CuS-ATMi@TGF-β NPs). CuS-ATMi@TGF-β NPs are highly photo-stable, can release encapsulated drugs, and increase the temperature to an effective level in a near-infrared (NIR)-responsive manner. Moreover, CuS-ATMi@TGF-β NPs specifically target tumors and thereby significantly inhibit tumor growth on contribution to synergistic low-temperature PTT and chemotherapy. This system not only achieved low-temperature PTT but also resulted in reduced damage to normal tissues. Modification with anti-TGF-β antibody enhanced target specificity and immune activation. The combination of PTT and ATM inhibitor showed synergistic effects and significantly attenuated the growth of the HCC via down regulation of heat shock protein (HSP). CuS-ATMi@TGF-β NPs are a highly promising platform for targeted tumor ablation via hyperthermia-mediated tumor death with minimal damage to normal tissues at a low temperature. STATEMENT OF SIGNIFICANCE: We constructed ataxia telangiectasia mutated (ATM) inhibitor-loaded hollow-structured CuS NPs with surface modification with anti-TGF-β antibody (CuS-ATMi@TGF-β NPs). CuS-ATMi@TGF-β NPs not only achieved low-temperature photothermal therapy (PTT) but also resulted in reduced damage to normal tissues and sufficient biocompatibility. The modification with anti-TGF-β antibody enhanced targeted specificity, cell endocytosis, and immune activation. In addition, the combination of PTT and ATM inhibitor synergistically attenuated the growth of the HCC via downregulation of heat shock protein (HSP). This study provided proof-of-concept for the ATM inhibitor that mediated low-temperature PTT with a potential for future clinical applications.
Collapse
Affiliation(s)
- Hongqiao Cai
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Xinlun Dai
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Xingren Guo
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China
| | - Lingxiao Zhang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, International Joint Research Laboratory of Nano-Micro Architecture Chemistry (NMAC), International Research Center for Chemistry-Medicine Joint Innovation, College of Chemistry, Jilin University, 2699 Qianjin street, Changchun 130012, China
| | - Kunxia Cao
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, International Joint Research Laboratory of Nano-Micro Architecture Chemistry (NMAC), International Research Center for Chemistry-Medicine Joint Innovation, College of Chemistry, Jilin University, 2699 Qianjin street, Changchun 130012, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, International Joint Research Laboratory of Nano-Micro Architecture Chemistry (NMAC), International Research Center for Chemistry-Medicine Joint Innovation, College of Chemistry, Jilin University, 2699 Qianjin street, Changchun 130012, China
| | - Bai Ji
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China.
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, the First Hospital, Jilin University, 71 Xinmin Street, Changchun 130021, China.
| |
Collapse
|
11
|
George SL, Lorenzi F, King D, Hartlieb S, Campbell J, Pemberton H, Toprak UH, Barker K, Tall J, da Costa BM, van den Boogaard ML, Dolman MEM, Molenaar JJ, Bryant HE, Westermann F, Lord CJ, Chesler L. Therapeutic vulnerabilities in the DNA damage response for the treatment of ATRX mutant neuroblastoma. EBioMedicine 2020; 59:102971. [PMID: 32846370 PMCID: PMC7452577 DOI: 10.1016/j.ebiom.2020.102971] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In neuroblastoma, genetic alterations in ATRX, define a distinct poor outcome patient subgroup. Despite the need for new therapies, there is a lack of available models and a dearth of pre-clinical research. METHODS To evaluate the impact of ATRX loss of function (LoF) in neuroblastoma, we utilized CRISPR-Cas9 gene editing to generate neuroblastoma cell lines isogenic for ATRX. We used these and other models to identify therapeutically exploitable synthetic lethal vulnerabilities associated with ATRX LoF. FINDINGS In isogenic cell lines, we found that ATRX inactivation results in increased DNA damage, homologous recombination repair (HRR) defects and impaired replication fork processivity. In keeping with this, high-throughput compound screening showed selective sensitivity in ATRX mutant cells to multiple PARP inhibitors and the ATM inhibitor KU60019. ATRX mutant cells also showed selective sensitivity to the DNA damaging agents, sapacitabine and irinotecan. HRR deficiency was also seen in the ATRX deleted CHLA-90 cell line, and significant sensitivity demonstrated to olaparib/irinotecan combination therapy in all ATRX LoF models. In-vivo sensitivity to olaparib/irinotecan was seen in ATRX mutant but not wild-type xenografts. Finally, sustained responses to olaparib/irinotecan therapy were seen in an ATRX deleted neuroblastoma patient derived xenograft. INTERPRETATION ATRX LoF results in specific DNA damage repair defects that can be therapeutically exploited. In ATRX LoF models, preclinical sensitivity is demonstrated to olaparib and irinotecan, a combination that can be rapidly translated into the clinic. FUNDING This work was supported by Christopher's Smile, Neuroblastoma UK, Cancer Research UK, and the Royal Marsden Hospital NIHR BRC.
Collapse
Affiliation(s)
- Sally L George
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom; Children and Young People's Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT United Kingdom.
| | - Federica Lorenzi
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom
| | - David King
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, United Kingdom
| | - Sabine Hartlieb
- Neuroblastoma Genomics, Hopp Children`s Cancer Center Heidelberg (KiTZ) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - James Campbell
- Bioinformatics Core Facility, The Institute of Cancer Research, London, United Kingdom
| | - Helen Pemberton
- CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research London, SW3 6JB, United Kingdom
| | - Umut H Toprak
- Neuroblastoma Genomics, Hopp Children`s Cancer Center Heidelberg (KiTZ) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karen Barker
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom
| | - Jennifer Tall
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom
| | - Barbara Martins da Costa
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom
| | | | - M Emmy M Dolman
- Princess Maxima Center for Pediatric Cancer, Utrecht, The Netherlands
| | - Jan J Molenaar
- Princess Maxima Center for Pediatric Cancer, Utrecht, The Netherlands
| | - Helen E Bryant
- Academic Unit of Molecular Oncology, Sheffield Institute for Nucleic Acids (SInFoNiA), Department of Oncology and Metabolism, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, United Kingdom
| | - Frank Westermann
- Neuroblastoma Genomics, Hopp Children`s Cancer Center Heidelberg (KiTZ) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christopher J Lord
- CRUK Gene Function Laboratory and Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research London, SW3 6JB, United Kingdom
| | - Louis Chesler
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Sutton, Surrey SM2 5NG, United Kingdom; Children and Young People's Unit, Royal Marsden NHS Foundation Trust, Sutton, Surrey SM2 5PT United Kingdom
| |
Collapse
|
12
|
De Simone G, di Masi A, Vita GM, Polticelli F, Pesce A, Nardini M, Bolognesi M, Ciaccio C, Coletta M, Turilli ES, Fasano M, Tognaccini L, Smulevich G, Abbruzzetti S, Viappiani C, Bruno S, Ascenzi P. Mycobacterial and Human Nitrobindins: Structure and Function. Antioxid Redox Signal 2020; 33:229-246. [PMID: 32295384 DOI: 10.1089/ars.2019.7874] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Aims: Nitrobindins (Nbs) are evolutionary conserved all-β-barrel heme-proteins displaying a highly solvent-exposed heme-Fe(III) atom. The physiological role(s) of Nbs is almost unknown. Here, the structural and functional properties of ferric Mycobacterium tuberculosis Nb (Mt-Nb(III)) and ferric Homo sapiens Nb (Hs-Nb(III)) have been investigated and compared with those of ferric Arabidopsis thaliana Nb (At-Nb(III), Rhodnius prolixus nitrophorins (Rp-NP(III)s), and mammalian myoglobins. Results: Data here reported demonstrate that Mt-Nb(III), At-Nb(III), and Hs-Nb(III) share with Rp-NP(III)s the capability to bind selectively nitric oxide, but display a very low reactivity, if any, toward histamine. Data obtained overexpressing Hs-Nb in human embryonic kidney 293 cells indicate that Hs-Nb localizes mainly in the cytoplasm and partially in the nucleus, thanks to a nuclear localization sequence encompassing residues Glu124-Leu154. Human Hs-Nb corresponds to the C-terminal domain of the human nuclear protein THAP4 suggesting that Nb may act as a sensor possibly modulating the THAP4 transcriptional activity residing in the N-terminal region. Finally, we provide strong evidence that both Mt-Nb(III) and Hs-Nb(III) are able to scavenge peroxynitrite and to protect free l-tyrosine against peroxynitrite-mediated nitration. Innovation: Data here reported suggest an evolutionarily conserved function of Nbs related to their role as nitric oxide sensors and components of antioxidant systems. Conclusion: Human THAP4 may act as a sensing protein that couples the heme-based Nb(III) reactivity with gene transcription. Mt-Nb(III) seems to be part of the pool of proteins required to scavenge reactive nitrogen and oxygen species produced by the host during the immunity response.
Collapse
Affiliation(s)
| | | | | | - Fabio Polticelli
- Dipartimento di Scienze, Università Roma Tre, Roma, Italy.,Istituto Nazionale di Fisica Nucleare, Sezione di Roma Tre, Roma, Italy
| | | | - Marco Nardini
- Dipartimento di Bioscienze, Università di Milano, Milano, Italy
| | - Martino Bolognesi
- Dipartimento di Bioscienze, Università di Milano, Milano, Italy.,Centro di Ricerche Pediatriche R.E. Invernizzi, Università di Milano, Milano, Italy
| | - Chiara Ciaccio
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Roma, Italy
| | - Massimo Coletta
- Dipartimento di Scienze Cliniche e Medicina Traslazionale, Università di Roma Tor Vergata, Roma, Italy
| | - Emily Samuela Turilli
- Dipartimento di Scienza ed Alta Tecnologia, Università dell'Insubria, Busto Arsizio, Italy
| | - Mauro Fasano
- Dipartimento di Scienza ed Alta Tecnologia, Università dell'Insubria, Busto Arsizio, Italy
| | - Lorenzo Tognaccini
- Dipartimento di Chimica Ugo Schiff, Università di Firenze, Sesto Fiorentino, Italy
| | - Giulietta Smulevich
- Dipartimento di Chimica Ugo Schiff, Università di Firenze, Sesto Fiorentino, Italy
| | - Stefania Abbruzzetti
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università di Parma, Parma, Italy
| | - Cristiano Viappiani
- Dipartimento di Scienze Matematiche, Fisiche e Informatiche, Università di Parma, Parma, Italy
| | - Stefano Bruno
- Dipartimento di Scienze degli Alimenti e del Farmaco, Università di Parma, Parma, Italy
| | - Paolo Ascenzi
- Dipartimento di Scienze, Università Roma Tre, Roma, Italy
| |
Collapse
|
13
|
Chen YY, Jiang MJ, Tian L. Analysis of exosomal circRNAs upon irradiation in pancreatic cancer cell repopulation. BMC Med Genomics 2020; 13:107. [PMID: 32727565 PMCID: PMC7391519 DOI: 10.1186/s12920-020-00756-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/20/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most malignant tumors. However, radiotherapy can lead to tumor recurrence, which is caused by the residual surviving cells repopulation stimulated by some molecular released from dying cells. Exosomes may mediate cell-cell communication and transfer kinds of signals from the dying cells to the surviving cells for stimulating tumor repopulation. Circular RNAs (circRNAs) may be one vital kind of exosomal cargos involving in modulating cancer cell repopulation. METHODS Next generation sequencing (NGS) and bioinformatics were performed to analyze and annotate the expression and function of exosome-derived circRNAs in pancreatic cancer cells after radiation. Four circRNAs were chosen for qRT-PCR analysis to validate the sequencing results. RESULTS In this study, 3580 circRNAs were annotated in literatures and circBase among 12,572 identified circRNAs. There were 196 filtered differentially expressed circRNAs (the up-regulation and down-regulation respectively is 182 and 14, fold change > 2, p-value < 0.05). Regulation of metabolic process and lysine degradation were the main enriched biological processes and pathway according to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. CONCLUSIONS The hsa_circ_0002130-hsa_miR_4482-3p-NBN interaction network suggested potential sponging miRNA and target mRNA. Our results provided potential functions of circRNAs to explore molecular mechanisms and therapeutic targets in pancreatic cancer cell repopulation upon irradiation.
Collapse
Affiliation(s)
- Yi-Yun Chen
- Shanghai Key Laboratory of Pancreatic Diseases & Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Science bldg. Rm 205, New Songjiang Rd No.650, Songjiang District, Shanghai, 201620, China
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Ming-Jie Jiang
- Shanghai Key Laboratory of Pancreatic Diseases & Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Science bldg. Rm 205, New Songjiang Rd No.650, Songjiang District, Shanghai, 201620, China
| | - Ling Tian
- Shanghai Key Laboratory of Pancreatic Diseases & Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Science bldg. Rm 205, New Songjiang Rd No.650, Songjiang District, Shanghai, 201620, China.
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
14
|
Backe SJ, Sager RA, Woodford MR, Makedon AM, Mollapour M. Post-translational modifications of Hsp90 and translating the chaperone code. J Biol Chem 2020; 295:11099-11117. [PMID: 32527727 DOI: 10.1074/jbc.rev120.011833] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
Cells have a remarkable ability to synthesize large amounts of protein in a very short period of time. Under these conditions, many hydrophobic surfaces on proteins may be transiently exposed, and the likelihood of deleterious interactions is quite high. To counter this threat to cell viability, molecular chaperones have evolved to help nascent polypeptides fold correctly and multimeric protein complexes assemble productively, while minimizing the danger of protein aggregation. Heat shock protein 90 (Hsp90) is an evolutionarily conserved molecular chaperone that is involved in the stability and activation of at least 300 proteins, also known as clients, under normal cellular conditions. The Hsp90 clients participate in the full breadth of cellular processes, including cell growth and cell cycle control, signal transduction, DNA repair, transcription, and many others. Hsp90 chaperone function is coupled to its ability to bind and hydrolyze ATP, which is tightly regulated both by co-chaperone proteins and post-translational modifications (PTMs). Many reported PTMs of Hsp90 alter chaperone function and consequently affect myriad cellular processes. Here, we review the contributions of PTMs, such as phosphorylation, acetylation, SUMOylation, methylation, O-GlcNAcylation, ubiquitination, and others, toward regulation of Hsp90 function. We also discuss how the Hsp90 modification state affects cellular sensitivity to Hsp90-targeted therapeutics that specifically bind and inhibit its chaperone activity. The ultimate challenge is to decipher the comprehensive and combinatorial array of PTMs that modulate Hsp90 chaperone function, a phenomenon termed the "chaperone code."
Collapse
Affiliation(s)
- Sarah J Backe
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Rebecca A Sager
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA.,College of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Mark R Woodford
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Alan M Makedon
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, New York, USA .,Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA.,Upstate Cancer Center, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
15
|
NBS1 interacts with HP1 to ensure genome integrity. Cell Death Dis 2019; 10:951. [PMID: 31836699 PMCID: PMC6911104 DOI: 10.1038/s41419-019-2185-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022]
Abstract
Heterochromatin Protein 1 (HP1) and the Mre11-Rad50-Nbs1 (MRN) complex are conserved factors that play crucial role in genome stability and integrity. Despite their involvement in overlapping cellular functions, ranging from chromatin organization, telomere maintenance to DNA replication and repair, a tight functional relationship between HP1 and the MRN complex has never been elucidated. Here we show that the Drosophila HP1a protein binds to the MRN complex through its chromoshadow domain (CSD). In addition, loss of any of the MRN members reduces HP1a levels indicating that the MRN complex acts as regulator of HP1a stability. Moreover, overexpression of HP1a in nbs (but not in rad50 or mre11) mutant cells drastically reduces DNA damage associated with the loss of Nbs suggesting that HP1a and Nbs work in concert to maintain chromosome integrity in flies. We have also found that human HP1α and NBS1 interact with each other and that, similarly to Drosophila, siRNA-mediated inhibition of NBS1 reduces HP1α levels in human cultured cells. Surprisingly, fibroblasts from Nijmegen Breakage Syndrome (NBS) patients, carrying the 657del5 hypomorphic mutation in NBS1 and expressing the p26 and p70 NBS1 fragments, accumulate HP1α indicating that, differently from NBS1 knockout cells, the presence of truncated NBS1 extends HP1α turnover and/or promotes its stability. Remarkably, an siRNA-mediated reduction of HP1α in NBS fibroblasts decreases the hypersensitivity to irradiation, a characteristic of the NBS syndrome. Overall, our data provide an unanticipated evidence of a close interaction between HP1 and NBS1 that is essential for genome stability and point up HP1α as a potential target to counteract chromosome instability in NBS patient cells.
Collapse
|
16
|
Gvozdenov Z, Kolhe J, Freeman BC. The Nuclear and DNA-Associated Molecular Chaperone Network. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a034009. [PMID: 30745291 PMCID: PMC6771373 DOI: 10.1101/cshperspect.a034009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Maintenance of a healthy and functional proteome in all cellular compartments is critical to cell and organismal homeostasis. Yet, our understanding of the proteostasis process within the nucleus is limited. Here, we discuss the identified roles of the major molecular chaperones Hsp90, Hsp70, and Hsp60 with client proteins working in diverse DNA-associated pathways. The unique challenges facing proteins in the nucleus are considered as well as the conserved features of the molecular chaperone system in facilitating DNA-linked processes. As nuclear protein inclusions are a common feature of protein-aggregation diseases (e.g., neurodegeneration), a better understanding of nuclear proteostasis is warranted.
Collapse
Affiliation(s)
- Zlata Gvozdenov
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois 61801.,Department Chemie, Technische Universität München, Garching 85748, Germany
| | - Janhavi Kolhe
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois 61801
| | - Brian C Freeman
- Department of Cell and Developmental Biology, University of Illinois, Urbana-Champaign, Urbana, Illinois 61801
| |
Collapse
|
17
|
di Masi A, Leboffe L, Polticelli F, Tonon F, Zennaro C, Caterino M, Stano P, Fischer S, Hägele M, Müller M, Kleger A, Papatheodorou P, Nocca G, Arcovito A, Gori A, Ruoppolo M, Barth H, Petrosillo N, Ascenzi P, Di Bella S. Human Serum Albumin Is an Essential Component of the Host Defense Mechanism Against Clostridium difficile Intoxication. J Infect Dis 2019; 218:1424-1435. [PMID: 29868851 DOI: 10.1093/infdis/jiy338] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 06/01/2018] [Indexed: 01/05/2023] Open
Abstract
Background The pathogenic effects of Clostridium difficile are primarily attributable to the production of the large protein toxins (C difficile toxins [Tcd]) A (TcdA) and B (TcdB). These toxins monoglucosylate Rho GTPases in the cytosol of host cells, causing destruction of the actin cytoskeleton with cytotoxic effects. Low human serum albumin (HSA) levels indicate a higher risk of acquiring and developing a severe C difficile infection (CDI) and are associated with recurrent and fatal disease. Methods We used a combined approach based on docking simulation and biochemical analyses that were performed in vitro on purified proteins and in human epithelial colorectal adenocarcinoma cells (Caco-2), and in vivo on stem cell-derived human intestinal organoids and zebrafish embryos. Results Our results show that HSA specifically binds via its domain II to TcdA and TcdB and thereby induces their autoproteolytic cleavage at physiological concentrations. This process impairs toxin internalization into the host cells and reduces the toxin-dependent glucosylation of Rho proteins. Conclusions Our data provide evidence for a specific HSA-dependent self-defense mechanism against C difficile toxins and provide an explanation for the clinical correlation between CDI severity and hypoalbuminemia.
Collapse
Affiliation(s)
| | - Loris Leboffe
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Fabio Polticelli
- Department of Sciences, Roma Tre University, Roma, Italy.,National Institute of Nuclear Physics, Roma Tre Section, Roma, Italy
| | - Federica Tonon
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Cristina Zennaro
- Department of Medical, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Marianna Caterino
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli "Federico II", Napoli, Italy.,Associazione Culturale DiSciMuS RCF, Casoria, Napoli, Italy
| | - Pasquale Stano
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Stephan Fischer
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Marlen Hägele
- Department of Internal Medicine I, University of Ulm Medical Center, Germany
| | - Martin Müller
- Department of Internal Medicine I, University of Ulm Medical Center, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, University of Ulm Medical Center, Germany
| | - Panagiotis Papatheodorou
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg, Germany
| | - Giuseppina Nocca
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Sacred Heart, Roma, Italy.,Institute of Chemistry of Molecular Recognition, CNR, Roma, Italy
| | - Alessandro Arcovito
- Institute of Biochemistry and Clinical Biochemistry, Catholic University of Sacred Heart, Roma, Italy
| | - Andrea Gori
- Clinic of Infectious Diseases, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Napoli "Federico II", Napoli, Italy.,Associazione Culturale DiSciMuS RCF, Casoria, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Holger Barth
- Institute of Pharmacology and Toxicology, University of Ulm Medical Center, Ulm, Germany
| | - Nicola Petrosillo
- 2nd Infectious Diseases Division, National Institute for Infectious Diseases "L. Spallanzani", Roma, Italy
| | - Paolo Ascenzi
- Department of Sciences, Roma Tre University, Roma, Italy
| | - Stefano Di Bella
- 2nd Infectious Diseases Division, National Institute for Infectious Diseases "L. Spallanzani", Roma, Italy
| |
Collapse
|
18
|
Investigation of Precise Molecular Mechanistic Action of Tobacco-Associated Carcinogen `NNK´ Induced Carcinogenesis: A System Biology Approach. Genes (Basel) 2019; 10:genes10080564. [PMID: 31357510 PMCID: PMC6723528 DOI: 10.3390/genes10080564] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 12/21/2022] Open
Abstract
Cancer is the second deadliest disease listed by the WHO. One of the major causes of cancer disease is tobacco and consumption possibly due to its main component, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). A plethora of studies have been conducted in the past aiming to decipher the association of NNK with other diseases. However, it is strongly linked with cancer development. Despite these studies, a clear molecular mechanism and the impact of NNK on various system-level networks is not known. In the present study, system biology tools were employed to understand the key regulatory mechanisms and the perturbations that will happen in the cellular processes due to NNK. To investigate the system level influence of the carcinogen, NNK rewired protein–protein interaction network (PPIN) was generated from 544 reported proteins drawn out from 1317 articles retrieved from PubMed. The noise was removed from PPIN by the method of modulation. Gene ontology (GO) enrichment was performed on the seed proteins extracted from various modules to find the most affected pathways by the genes/proteins. For the modulation, Molecular COmplex DEtection (MCODE) was used to generate 19 modules containing 115 seed proteins. Further, scrutiny of the targeted biomolecules was done by the graph theory and molecular docking. GO enrichment analysis revealed that mostly cell cycle regulatory proteins were affected by NNK.
Collapse
|
19
|
Klimczak M, Biecek P, Zylicz A, Zylicz M. Heat shock proteins create a signature to predict the clinical outcome in breast cancer. Sci Rep 2019; 9:7507. [PMID: 31101846 PMCID: PMC6525249 DOI: 10.1038/s41598-019-43556-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 04/27/2019] [Indexed: 01/30/2023] Open
Abstract
Utilizing The Cancer Genome Atlas (TCGA) and KM plotter databases we identified six heat shock proteins associated with survival of breast cancer patients. The survival curves of samples with high and low expression of heat shock genes were compared by log-rank test (Mantel-Haenszel). Interestingly, patients overexpressing two identified HSPs – HSPA2 and DNAJC20 exhibited longer survival, whereas overexpression of other four HSPs – HSP90AA1, CCT1, CCT2, CCT6A resulted in unfavorable prognosis for breast cancer patients. We explored correlations between expression level of HSPs and clinicopathological features including tumor grade, tumor size, number of lymph nodes involved and hormone receptor status. Additionally, we identified a novel signature with the potential to serve as a prognostic model for breast cancer. Using univariate Cox regression analysis followed by multivariate Cox regression analysis, we built a risk score formula comprising prognostic HSPs (HSPA2, DNAJC20, HSP90AA1, CCT1, CCT2) and tumor stage to identify high-risk and low-risk cases. Finally, we analyzed the association of six prognostic HSP expression with survival of patients suffering from other types of cancer than breast cancer. We revealed that depending on cancer type, each of the six analyzed HSPs can act both as a positive, as well as a negative regulator of cancer development. Our study demonstrates a novel HSP signature for the outcome prediction of breast cancer patients and provides a new insight into ambiguous role of these proteins in cancer development.
Collapse
Affiliation(s)
- Marta Klimczak
- International Institute of Molecular and Cell Biology, Warsaw, Poland. .,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland.
| | - Przemyslaw Biecek
- Faculty of Mathematics, Informatics, and Mechanics, University of Warsaw, Warsaw, Poland.,Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
| | - Alicja Zylicz
- International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Maciej Zylicz
- International Institute of Molecular and Cell Biology, Warsaw, Poland.
| |
Collapse
|
20
|
Liu YH, Liu XM, Wang PC, Yu XX, Miao JK, Liu S, Wang YK, Du ZQ, Yang CX. Heat shock protein 90α couples with the MAPK-signaling pathway to determine meiotic maturation of porcine oocytes. J Anim Sci 2018; 96:3358-3369. [PMID: 29800308 DOI: 10.1093/jas/sky213] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022] Open
Abstract
Heat shock protein 90 (Hsp90) functions as a molecular chaperone in its interaction with clients to influence multiple cellular and physiological processes. However, our current understanding on Hsp90's relationship with mammalian oocyte maturation is still very limited. Here, we aimed to investigate Hsp90's effect on pig oocyte meiotic maturation. Endogenous Hsp90α was constantly expressed at both mRNA and protein levels in porcine maturing oocytes. Addition of 2 µM 17-allylamino-17-demethoxygeldanamycin (17-AAG), the Hsp90 inhibitor, to in vitro mature cumulus-oocyte complexes (COC) significantly decreased Hsp90α protein level (P < 0.05), delayed germinal vesicle breakdown (GVBD) (P < 0.05), and impeded the first polar body (PB1) extrusion (P < 0.01) of porcine oocytes. 2 µM 17-AAG treatment during in vitro maturation also decreased the subsequent development competence as indicated by the lower cleavage (P < 0.001) and higher fragmentation (P < 0.001) rates of parthenotes, whereas no effects on the percentage and average cell number of blastocysts were found. Immunodepletion of Hsp90α by antibody microinjection into porcine oocytes at germinal vesicle and metaphase II stages induced similar defects of meiotic maturation and parthenote development, to that resulted from 2 µM inhibitor 17-AAG. For oocytes treated by 2 µM 17-AAG, the cytoplasm and membrane actin levels were weakened (P < 0.01), and the spindle assembly was disturbed (P < 0.05), due to decreased p-ERK1/2 level (P < 0.05). However, the mitochondrial function and early apoptosis were not affected, as demonstrated by rhodamine 123 staining and Annexin V assays. Our findings indicate that Hsp90α can couple with mitogen-activated protein kinase to regulate cytoskeletal structure and orchestrate meiotic maturation of porcine oocytes.
Collapse
Affiliation(s)
- Yun-Hua Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiao-Man Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Pei-Chao Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Xiao-Xia Yu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Jia-Kun Miao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shuai Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yan-Kui Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Zhi-Qiang Du
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Cai-Xia Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| |
Collapse
|
21
|
Wang W, Zhan L, Guo D, Xiang Y, Zhang Y, Tian M, Han Z. Transcriptome analysis of pancreatic cancer cell response to treatment with grape seed proanthocyanidins. Oncol Lett 2018; 17:1741-1749. [PMID: 30675233 PMCID: PMC6341838 DOI: 10.3892/ol.2018.9807] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 10/25/2018] [Indexed: 01/09/2023] Open
Abstract
Grape seed proanthocyanidins (GSPs) have been demonstrated to exhibit potential chemotherapeutic efficacy against various cancer types. To determine the underlying molecular mechanisms involved in GSP-induced apoptosis, the present study prepared pancreatic cancer (PC) cells samples, S3, S12 and S24, which were treated with 20 µg/ml GSPs for 3, 12 and 24 h, respectively. Control cell samples, C3, C12 and C24, were also prepared. Using RNA-sequencing, transcriptome comparisons were performed, which identified 966, 3,543 and 4,944 differentially-expressed genes (DEGs) in S3 vs. C3, S12 vs. C12 and S24 vs. C24, respectively. Gene Ontology analysis of the DEGs, revealed that treatment with GSPs is associated with disruption of the cell cycle (CC) in PC cells. Additionally, disruption of transcription, DNA replication and DNA repair were associated with GSP-treatment in PC cells. Network analysis demonstrated that the common DEGs involved in the CC, transcription, DNA replication and DNA repair were integrated, and served essential roles in the control of CC progression in cancer cells. In summary, GSPs may exhibit a potential chemotherapeutic effect on PC cell proliferation.
Collapse
Affiliation(s)
- Weihua Wang
- Department of Food Science, College of Life Sciences, Tarim University, Alar, Xinjiang 843300, P.R. China.,Xinjiang Production and Construction Corps Key Laboratory of Deep Processing of Agricultural Products in South Xinjiang, Alar, Xinjiang 843300, P.R. China
| | - Leilei Zhan
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei 430075, P.R. China
| | - Dongqi Guo
- Department of Food Science, College of Life Sciences, Tarim University, Alar, Xinjiang 843300, P.R. China.,Xinjiang Production and Construction Corps Key Laboratory of Deep Processing of Agricultural Products in South Xinjiang, Alar, Xinjiang 843300, P.R. China
| | - Yanju Xiang
- Department of Food Science, College of Life Sciences, Tarim University, Alar, Xinjiang 843300, P.R. China.,Xinjiang Production and Construction Corps Key Laboratory of Deep Processing of Agricultural Products in South Xinjiang, Alar, Xinjiang 843300, P.R. China
| | - Yu Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, Hubei 430075, P.R. China
| | - Muxing Tian
- Department of Food Science, College of Life Sciences, Tarim University, Alar, Xinjiang 843300, P.R. China.,Xinjiang Production and Construction Corps Key Laboratory of Deep Processing of Agricultural Products in South Xinjiang, Alar, Xinjiang 843300, P.R. China
| | - Zhanjiang Han
- Department of Food Science, College of Life Sciences, Tarim University, Alar, Xinjiang 843300, P.R. China.,Xinjiang Production and Construction Corps Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Alar, Xinjiang 843300, P.R. China
| |
Collapse
|
22
|
Morishima Y, Mehta RK, Yoshimura M, Lau M, Southworth DR, Lawrence TS, Pratt WB, Nyati MK, Osawa Y. Chaperone Activity and Dimerization Properties of Hsp90 α and Hsp90 β in Glucocorticoid Receptor Activation by the Multiprotein Hsp90/Hsp70-Dependent Chaperone Machinery. Mol Pharmacol 2018; 94:984-991. [PMID: 29941666 PMCID: PMC6064783 DOI: 10.1124/mol.118.112516] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/20/2018] [Indexed: 11/22/2022] Open
Abstract
Several hundred proteins cycle into heterocomplexes with a dimer of the chaperone heat shock protein 90 (Hsp90), regulating their activity and turnover. There are two isoforms of Hsp90, Hsp90α and Hsp90β, and their relative chaperone activities and composition in these client protein•Hsp90 heterocomplexes has not been determined. Here, we examined the activity of human Hsp90α and Hsp90β in a purified five-protein chaperone machinery that assembles glucocorticoid receptor (GR)•Hsp90 heterocomplexes to generate high-affinity steroid-binding activity. We found that human Hsp90α and Hsp90β have equivalent chaperone activities, and when mixed together in this assay, they formed only GR•Hsp90αα and GR•Hsp90ββ homodimers and no GR•Hsp90αβ heterodimers. In contrast, GR•Hsp90 heterocomplexes formed in human embryonic kidney (HEK) cells also contain GR•Hsp90αβ heterodimers. The formation of GR•Hsp90αβ heterodimers in HEK cells probably reflects the longer time permitted for exchange to form Hsp90αβ heterodimers in the cell versus in the cell-free assembly conditions. This purified GR-activating chaperone machinery can be used to determine how modifications of Hsp90 affect its chaperone activity. To that effect, we have tested whether the unique phosphorylation of Hsp90α at threonines 5 and 7 that occurs during DNA damage repair affects its chaperone activity. We showed that the phosphomimetic mutant Hsp90α T5/7D has the same intrinsic chaperone activity as wild-type human Hsp90α in activation of GR steroid-binding activity by the five-protein machinery, supporting the conclusion that T5/7 phosphorylation does not affect Hsp90α chaperone activity.
Collapse
Affiliation(s)
- Yoshihiro Morishima
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Ranjit K Mehta
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Miyako Yoshimura
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Miranda Lau
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Daniel R Southworth
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Theodore S Lawrence
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - William B Pratt
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Mukesh K Nyati
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| | - Yoichi Osawa
- Departments of Pharmacology (Y.M., M.Y., M.L., W.B.P., Y.O.) and Radiation Oncology (R.K.M., T.S.L., M.K.N.), University of Michigan Medical School, Ann Arbor, Michigan; and Life Sciences Institute and Departments of Biochemistry and Biophysics, University of Michigan, Ann Arbor, Michigan (D.R.S.)
| |
Collapse
|
23
|
Hartlerode AJ, Regal JA, Ferguson DO. Reversible mislocalization of a disease-associated MRE11 splice variant product. Sci Rep 2018; 8:10121. [PMID: 29973640 PMCID: PMC6031676 DOI: 10.1038/s41598-018-28370-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/20/2018] [Indexed: 01/13/2023] Open
Abstract
Ataxia-telangiectasia (AT) and related disorders feature cancer predisposition, neurodegeneration, and immunodeficiency resulting from failure to respond to DNA damage. Hypomorphic mutations in MRE11 cause an AT-like disorder (ATLD) with variable clinical presentation. We have sought to understand how diverse MRE11 mutations may provide unique therapeutic opportunities, and potentially correlate with clinical variability. Here we have undertaken studies of an MRE11 splice site mutation that was found in two ATLD siblings that died of pulmonary adenocarcinoma at the young ages of 9 and 16. The mutation, termed MRE11 alternative splice mutation (MRE11ASM), causes skipping of a highly conserved exon while preserving the protein's open reading frame. A new mouse model expressing Mre11ASM from the endogenous locus demonstrates that the protein is present at very low levels, a feature in common with the MRE11ATLD1 mutant found in other patients. However, the mechanisms causing low protein levels are distinct. MRE11ASM is mislocalized to the cytoplasm, in contrast to MRE11ATLD1, which remains nuclear. Strikingly, MRE11ASM mislocalization is corrected by inhibition of the proteasome, implying that the protein undergoes strict protein quality control in the nucleus. These findings raise the prospect that inhibition of poorly understood nuclear protein quality control mechanisms might have therapeutic benefit in genetic disorders causing cytoplasmic mislocalization.
Collapse
Affiliation(s)
- Andrea J Hartlerode
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI, 48109-2200, USA
| | - Joshua A Regal
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI, 48109-2200, USA
- Molecular and Cellular Pathology Graduate Program, The University of Michigan Medical School, Ann Arbor, MI, 48109-2200, USA
| | - David O Ferguson
- Department of Pathology, The University of Michigan Medical School, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
24
|
Syed A, Tainer JA. The MRE11-RAD50-NBS1 Complex Conducts the Orchestration of Damage Signaling and Outcomes to Stress in DNA Replication and Repair. Annu Rev Biochem 2018; 87:263-294. [PMID: 29709199 PMCID: PMC6076887 DOI: 10.1146/annurev-biochem-062917-012415] [Citation(s) in RCA: 287] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Genomic instability in disease and its fidelity in health depend on the DNA damage response (DDR), regulated in part from the complex of meiotic recombination 11 homolog 1 (MRE11), ATP-binding cassette-ATPase (RAD50), and phosphopeptide-binding Nijmegen breakage syndrome protein 1 (NBS1). The MRE11-RAD50-NBS1 (MRN) complex forms a multifunctional DDR machine. Within its network assemblies, MRN is the core conductor for the initial and sustained responses to DNA double-strand breaks, stalled replication forks, dysfunctional telomeres, and viral DNA infection. MRN can interfere with cancer therapy and is an attractive target for precision medicine. Its conformations change the paradigm whereby kinases initiate damage sensing. Delineated results reveal kinase activation, posttranslational targeting, functional scaffolding, conformations storing binding energy and enabling access, interactions with hub proteins such as replication protein A (RPA), and distinct networks at DNA breaks and forks. MRN biochemistry provides prototypic insights into how it initiates, implements, and regulates multifunctional responses to genomic stress.
Collapse
Affiliation(s)
- Aleem Syed
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA; ,
| | - John A Tainer
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA; ,
- Molecular Biophysics and Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| |
Collapse
|
25
|
Chenette EJ, Martin SJ. 50 years of The FEBS Journal: looking back as well as ahead. FEBS J 2018; 284:4162-4171. [PMID: 29251437 DOI: 10.1111/febs.14328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In this last issue of 2017, we're celebrating the 50th anniversary of The FEBS Journal. This Editorial considers how the journal has grown and changed from volume 1, issue 1 and outlines our exciting plans for the future.
Collapse
Affiliation(s)
| | - Seamus J Martin
- The FEBS Journal Editorial Office, Cambridge, UK.,Department of Genetics, The Smurfit Institute, Trinity College, Dublin, Ireland
| |
Collapse
|
26
|
Corrigendum. FEBS J 2017; 284:3968. [DOI: 10.1111/febs.14298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
27
|
Abstract
The NBN component of the MRE11-RAD50-NBN (MRN) complex and the ATM kinase have been identified as clients of the HSP90α chaperone. Inhibition of HSP90 leads to reduced stability of NBN and ATM and an impaired DNA damage response. These results identify new regulatory details of the DNA damage response and further explain the chemosensitizing effects of HSP90 inhibitors.
Collapse
Affiliation(s)
- Travis H Stracker
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Spain
| |
Collapse
|