1
|
Martins Rodrigues F, Jasielec J, Perpich M, Kim A, Moma L, Li Y, Storrs E, Wendl MC, Jayasinghe RG, Fiala M, Stefka A, Derman B, Jakubowiak AJ, DiPersio JF, Vij R, Godley LA, Ding L. Germline predisposition in multiple myeloma. iScience 2025; 28:111620. [PMID: 39845416 PMCID: PMC11750583 DOI: 10.1016/j.isci.2024.111620] [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: 09/08/2024] [Revised: 10/04/2024] [Accepted: 11/14/2024] [Indexed: 01/24/2025] Open
Abstract
We present a study of rare germline predisposition variants in 954 unrelated individuals with multiple myeloma (MM) and 82 MM families. Using a candidate gene approach, we identified such variants across all age groups in 9.1% of sporadic and 18% of familial cases. Implicated genes included genes suggested in other MM risk studies as potential risk genes (DIS3, EP300, KDM1A, and USP45); genes involved in predisposition to other cancers (ATM, BRCA1/2, CHEK2, PMS2, POT1, PRF1, and TP53); and BRIP1, EP300, and FANCM in individuals of African ancestry. Variants were characterized using loss of heterozygosity (LOH), biallelic events, and gene expression analyses, revealing 31 variants in 3.25% of sporadic cases for which pathogenicity was supported by multiple lines of evidence. Our results suggest that the disruption of DNA damage repair pathways may play a role in MM susceptibility. These results will inform improved surveillance in high-risk groups and potential therapeutic strategies.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jagoda Jasielec
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Melody Perpich
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Aelin Kim
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Luke Moma
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Yize Li
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erik Storrs
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael C. Wendl
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Reyka G. Jayasinghe
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark Fiala
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew Stefka
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Benjamin Derman
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Andrzej J. Jakubowiak
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - John F. DiPersio
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ravi Vij
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lucy A. Godley
- Division of Hematology/Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
2
|
Cancela MB, Winter U, Zugbi S, Dinardi M, Alves da Quinta D, Aschero R, Ganiewich D, Sampor C, Sgroi M, Lagomarsino E, Fandiño A, Llera AS, Chantada G, Carcaboso AM, Schaiquevich P. Mimicking Retinoblastoma Treatment With Repeated Topotecan or Melphalan Develops Cross-Resistance to Classic Agents But Not to Repurposed Drugs. Invest Ophthalmol Vis Sci 2024; 65:14. [PMID: 39636723 PMCID: PMC11622161 DOI: 10.1167/iovs.65.14.14] [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: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Purpose Refractory or recurrent retinoblastoma results from acquired chemoresistance and the management of these eyes often requires surgical removal. Our objective was to develop retinoblastoma models resistant to chemotherapy by exposing cancer cells to repeated chemotherapy mimicking the clinical scenario. These newly resistant cells were used to evaluate potential novel therapies. Methods Chemoresistant cells were obtained by exposing two primary retinoblastoma cell cultures to three weekly doses of melphalan or topotecan. The sensitivity of these resistant cells to each chemotherapy was evaluated, and cross-resistance to topotecan, melphalan, and carboplatin was assessed. Genomic alterations and differential expression of efflux/influx transporters between chemoresistant and parental cells were analyzed. Subsequently, sensitivity of both resistant and parental cells to the repurposed agents digoxin, methylene blue, and gemcitabine was assessed. Results Four chemoresistant models were successfully established, showing significantly higher half-maximal inhibitory concentration (IC50) values for melphalan and topotecan compared to their corresponding parental cells (P < 0.05). Cross-resistance between melphalan and topotecan was demonstrated, with a 3-fold increase in the IC50. Chemoresistant cells also showed reduced sensitivity to carboplatin (P < 0.05) compared to parental cells, whereas sensitivity to the evaluated repurposed agents remained unchanged. Genomic analysis revealed no selective alterations in the resistant cells, although differential expression of influx/efflux transporters was observed across all chemoresistant models. Conclusions In vitro simulation of patient treatment was useful to establish chemoresistant retinoblastomas and to identify strategies to overcome resistance to topotecan or melphalan through drug repurposed. Our results warrant further investigation to support the clinical translation.
Collapse
Affiliation(s)
- María Belen Cancela
- Unit of Innovative Treatments, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Ursula Winter
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Santiago Zugbi
- Unit of Innovative Treatments, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Milagros Dinardi
- Unit of Innovative Treatments, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Daniela Alves da Quinta
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir – Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Buenos Aires, Argentina
| | - Rosario Aschero
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Daiana Ganiewich
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir – Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Buenos Aires, Argentina
| | - Claudia Sampor
- Hematology-Oncology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Mariana Sgroi
- Ophthalmology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Eduardo Lagomarsino
- Pharmacy Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Adriana Fandiño
- Ophthalmology Service, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
| | - Andrea S. Llera
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
- Laboratory of Molecular and Cellular Therapy, Instituto Leloir – Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Buenos Aires, Argentina
| | - Guillermo Chantada
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Angel M. Carcaboso
- SJD Pediatric Cancer Center Barcelona, Hospital Sant Joan de Deu, Barcelona, Spain
- Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Paula Schaiquevich
- Unit of Innovative Treatments, Hospital de Pediatría JP Garrahan, Buenos Aires, Argentina
- National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| |
Collapse
|
3
|
Bellani MA, Shaik A, Majumdar I, Ling C, Seidman MM. Repair of genomic interstrand crosslinks. DNA Repair (Amst) 2024; 141:103739. [PMID: 39106540 PMCID: PMC11423799 DOI: 10.1016/j.dnarep.2024.103739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024]
Abstract
Genomic interstrand crosslinks (ICLs) are formed by reactive species generated during normal cellular metabolism, produced by the microbiome, and employed in cancer chemotherapy. While there are multiple options for replication dependent and independent ICL repair, the crucial step for each is unhooking one DNA strand from the other. Much of our insight into mechanisms of unhooking comes from powerful model systems based on plasmids with defined ICLs introduced into cells or cell free extracts. Here we describe the properties of exogenous and endogenous ICL forming compounds and provide an historical perspective on early work on ICL repair. We discuss the modes of unhooking elucidated in the model systems, the concordance or lack thereof in drug resistant tumors, and the evolving view of DNA adducts, including ICLs, formed by metabolic aldehydes.
Collapse
Affiliation(s)
- Marina A Bellani
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Althaf Shaik
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ishani Majumdar
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Chen Ling
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michael M Seidman
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
4
|
Li J, Jia Z, Wang R, Xiao B, Cai Y, Zhu T, Wang W, Zhang X, Fan S, Fan X, Han W, Lu X. Activated interferon response from DNA damage in multiple myeloma cells contributes to the chemotherapeutic effects of anthracyclines. Front Oncol 2024; 14:1357996. [PMID: 38800411 PMCID: PMC11116600 DOI: 10.3389/fonc.2024.1357996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Introduction Multiple myeloma (MM) is a malignant plasma cell disease caused by abnormal proliferation of clonal plasma cells in bone marrow. Upfront identification of tumor subgroups with specific biological markers has the potential to improve biologically-driven therapy. Previously, we established a molecular classification by stratifying multiple myeloma into two subtypes with a different prognosis based on a gene module co-expressed with MCL-1 (MCL1-M). Methods Gene Ontology (GO) analysis with differentially expressed genes was performed to identify signal pathway. Drug sensitivity was analyzed using the OncoPredict algorithm. Drug sensitivity of different myeloma cell lines was detected by CCK8 and flow cytometry. RNA-seq was performed on drug-sensitive cell lines before and after adriamycin treatment. RT-qPCR was used to further verify the sequencing results. The expression of γ-H2AX and dsDNA in sensitive and resistant cell lines was detected by immunofluorescence method. Results In our study, we demonstrated that MCL1-M low MM were more sensitive to anthracyclines. We treated different myeloma cell lines with doxorubicin in vitro and discovered the association of drug sensitivity with IFN signaling. Herein, we demonstrate that the doxorubicin-sensitive myeloma cell line showed significant DNA damage and up-regulated expression of genes related to the IFN response, which was not observed in drug-insensitive cell lines. Discussion Our results suggest that the active IFN signaling pathway may serve as a marker for predicting chemotherapy sensitivity in patients with myeloma. With our MCL1-M molecular classification system, we can screen patients with a potentially good response to the interferon signaling pathway and provide individualized treatment for MM. We propose IFN-a as adjuvant therapy for patients with myeloma sensitive to anthracyclines to further improve the therapeutic effect and prolong the survival of patients.
Collapse
Affiliation(s)
- Jin Li
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Zhuxia Jia
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Rongxuan Wang
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Bitao Xiao
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Yanan Cai
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Tianshu Zhu
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Weiya Wang
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Xinyue Zhang
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Shu Fan
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Xiaolong Fan
- Beijing Key Laboratory of Gene Resource and Molecular Development, Laboratory of Neuroscience and Brain Development, Beijing Normal University, Beijing, China
| | - Wenmin Han
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Xuzhang Lu
- Department of Hematology, Changzhou No. 2 People’s Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, China
| |
Collapse
|
5
|
Tonon G. Myeloma and DNA damage. Blood 2024; 143:488-495. [PMID: 37992215 DOI: 10.1182/blood.2023021384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/24/2023] Open
Abstract
ABSTRACT DNA-damaging agents have represented the first effective treatment for the blood cancer multiple myeloma, and after 65 years since their introduction to the clinic, they remain one of the mainstay therapies for this disease. Myeloma is a cancer of plasma cells. Despite exceedingly slow proliferation, myeloma cells present extended genomic rearrangements and intense genomic instability, starting at the premalignant stage of the disease. Where does such DNA damage stem from? A reliable model argues that the powerful oncogenes activated in myeloma as well the phenotypic peculiarities of cancer plasma cells, including the dependency on the proteasome for survival and the constant presence of oxidative stress, all converge on modulating DNA damage and repair. Beleaguered by these contraposing forces, myeloma cells survive in a precarious balance, in which the robust engagement of DNA repair mechanisms to guarantee cell survival is continuously challenged by rampant genomic instability, essential for cancer cells to withstand hostile selective pressures. Shattering this delicate equilibrium has been the goal of the extensive use of DNA-damaging agents since their introduction in the clinic, now enriched by novel approaches that leverage upon synthetic lethality paradigms. Exploiting the impairment of homologous recombination caused by myeloma genetic lesions or treatments, it is now possible to design therapeutic combinations that could target myeloma cells more effectively. Furthermore, DNA-damaging agents, as demonstrated in solid tumors, may sensitize cells to immune therapies. In all, targeting DNA damage and repair remains as central as ever in myeloma, even for the foreseeable future.
Collapse
Affiliation(s)
- Giovanni Tonon
- Università Vita-Salute San Raffaele, Milan, Italy
- Division of Experimental Oncology and Center for Omics Sciences, Functional Genomics of Cancer Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
6
|
Bruyer A, Dutrieux L, de Boussac H, Martin T, Chemlal D, Robert N, Requirand G, Cartron G, Vincent L, Herbaux C, Lutzmann M, Bret C, Pasero P, Moreaux J, Ovejero S. Combined inhibition of Wee1 and Chk1 as a therapeutic strategy in multiple myeloma. Front Oncol 2023; 13:1271847. [PMID: 38125947 PMCID: PMC10730928 DOI: 10.3389/fonc.2023.1271847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy characterized by an abnormal clonal proliferation of malignant plasma cells. Despite the introduction of novel agents that have significantly improved clinical outcome, most patients relapse and develop drug resistance. MM is characterized by genomic instability and a high level of replicative stress. In response to replicative and DNA damage stress, MM cells activate various DNA damage signaling pathways. In this study, we reported that high CHK1 and WEE1 expression is associated with poor outcome in independent cohorts of MM patients treated with high dose melphalan chemotherapy or anti-CD38 immunotherapy. Combined targeting of Chk1 and Wee1 demonstrates synergistic toxicities on MM cells and was associated with higher DNA double-strand break induction, as evidenced by an increased percentage of γH2AX positive cells subsequently leading to apoptosis. The therapeutic interest of Chk1/Wee1 inhibitors' combination was validated on primary MM cells of patients. The toxicity was specific of MM cells since normal bone marrow cells were not significantly affected. Using deconvolution approach, MM patients with high CHK1 expression exhibited a significant lower percentage of NK cells whereas patients with high WEE1 expression displayed a significant higher percentage of regulatory T cells in the bone marrow. These data emphasize that MM cell adaptation to replicative stress through Wee1 and Chk1 upregulation may decrease the activation of the cell-intrinsic innate immune response. Our study suggests that association of Chk1 and Wee1 inhibitors may represent a promising therapeutic approach in high-risk MM patients characterized by high CHK1 and WEE1 expression.
Collapse
Affiliation(s)
| | - Laure Dutrieux
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
| | | | - Thibaut Martin
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
| | - Djamila Chemlal
- Diag2Tec, Montpellier, France
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
| | - Nicolas Robert
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Guilhem Requirand
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Guillaume Cartron
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
| | - Laure Vincent
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
| | - Charles Herbaux
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
| | - Malik Lutzmann
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
| | - Caroline Bret
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
| | - Philippe Pasero
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
| | - Jérôme Moreaux
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
- University of Montpellier, UFR Medicine, Montpellier, France
- Institut Universitaire de France (IUF), Paris, France
| | - Sara Ovejero
- Institute of Human Genetics, UMR CNRS-UM 9002, Montpellier, France
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| |
Collapse
|
7
|
Jiang N, Li W, Jiang S, Xie M, Liu R. Acetylation in pathogenesis: Revealing emerging mechanisms and therapeutic prospects. Biomed Pharmacother 2023; 167:115519. [PMID: 37729729 DOI: 10.1016/j.biopha.2023.115519] [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: 07/18/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 09/22/2023] Open
Abstract
Protein acetylation modifications play a central and pivotal role in a myriad of biological processes, spanning cellular metabolism, proliferation, differentiation, apoptosis, and beyond, by effectively reshaping protein structure and function. The metabolic state of cells is intricately connected to epigenetic modifications, which in turn influence chromatin status and gene expression patterns. Notably, pathological alterations in protein acetylation modifications are frequently observed in diseases such as metabolic syndrome, cardiovascular disorders, and cancer. Such abnormalities can result in altered protein properties and loss of function, which are closely associated with developing and progressing related diseases. In recent years, the advancement of precision medicine has highlighted the potential value of protein acetylation in disease diagnosis, treatment, and prevention. This review includes provocative and thought-provoking papers outlining recent breakthroughs in acetylation modifications as they relate to cardiovascular disease, mitochondrial metabolic regulation, liver health, neurological health, obesity, diabetes, and cancer. Additionally, it covers the molecular mechanisms and research challenges in understanding the role of acetylation in disease regulation. By summarizing novel targets and prognostic markers for the treatment of related diseases, we aim to contribute to the field. Furthermore, we discuss current hot topics in acetylation research related to health regulation, including N4-acetylcytidine and liquid-liquid phase separation. The primary objective of this review is to provide insights into the functional diversity and underlying mechanisms by which acetylation regulates proteins in disease contexts.
Collapse
Affiliation(s)
- Nan Jiang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Wenyong Li
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Shuanglin Jiang
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui 236037, China
| | - Ming Xie
- North China Petroleum Bureau General Hospital, Renqiu 062550, China.
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| |
Collapse
|
8
|
Deacetylation induced nuclear condensation of HP1γ promotes multiple myeloma drug resistance. Nat Commun 2023; 14:1290. [PMID: 36894562 PMCID: PMC9998874 DOI: 10.1038/s41467-023-37013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
Acquired chemoresistance to proteasome inhibitors is a major obstacle in managing multiple myeloma but key regulators and underlying mechanisms still remain to be explored. We find that high level of HP1γ is associated with low acetylation modification in the bortezomib-resistant myeloma cells using SILAC-based acetyl-proteomics assay, and higher HP1γ level is positively correlated with poorer outcomes in the clinic. Mechanistically, elevated HDAC1 in the bortezomib-resistant myeloma cells deacetylates HP1γ at lysine 5 and consequently alleviates the ubiquitin-mediated protein degradation, as well as the aberrant DNA repair capacity. HP1γ interacts with the MDC1 to induce DNA repair, and simultaneously the deacetylation modification and the interaction with MDC1 enhance the nuclear condensation of HP1γ protein and the chromatin accessibility of its target genes governing sensitivity to proteasome inhibitors, such as CD40, FOS and JUN. Thus, targeting HP1γ stability by using HDAC1 inhibitor re-sensitizes bortezomib-resistant myeloma cells to proteasome inhibitors treatment in vitro and in vivo. Our findings elucidate a previously unrecognized role of HP1γ in inducing drug resistance to proteasome inhibitors of myeloma cells and suggest that targeting HP1γ may be efficacious for overcoming drug resistance in refractory or relapsed multiple myeloma patients.
Collapse
|
9
|
Al-Odat OS, Guirguis DA, Schmalbach NK, Yao G, Budak-Alpdogan T, Jonnalagadda SC, Pandey MK. Autophagy and Apoptosis: Current Challenges of Treatment and Drug Resistance in Multiple Myeloma. Int J Mol Sci 2022; 24:ijms24010644. [PMID: 36614089 PMCID: PMC9820338 DOI: 10.3390/ijms24010644] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
Over the past two decades, the natural history of multiple myeloma (MM) has evolved dramatically, owing primarily to novel agents targeting MM in the bone marrow microenvironment (BMM) pathways. However, the mechanisms of resistance acquisition remain a mystery and are poorly understood. Autophagy and apoptosis are tightly controlled processes and play a critical role in the cell growth, development, and survival of MM. Genetic instability and abnormalities are two hallmarks of MM. During MM progression, plasma malignant cells become genetically unstable and activate various signaling pathways, resulting in the overexpression of abnormal proteins that disrupt autophagy and apoptosis biological processes. Thus, achieving a better understanding of the autophagy and apoptosis processes and the proteins that crosslinked both pathways, could provide new insights for the MM treatment and improve the development of novel therapeutic strategies to overcome resistance. This review presents a sufficient overview of the roles of autophagy and apoptosis and how they crosslink and control MM progression and drug resistance. Potential combination targeting of both pathways for improving outcomes in MM patients also has been addressed.
Collapse
Affiliation(s)
- Omar S. Al-Odat
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, USA
| | - Daniel A. Guirguis
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Nicole K. Schmalbach
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | - Gabriella Yao
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| | | | | | - Manoj K. Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
- Correspondence: ; Tel.: +1-856-956-2751
| |
Collapse
|
10
|
Ovejero S, Viziteu E, Dutrieux L, Devin J, Lin YL, Alaterre E, Jourdan M, Basbous J, Requirand G, Robert N, de Boussac H, Seckinger A, Hose D, Vincent L, Herbaux C, Constantinou A, Pasero P, Moreaux J. The BLM helicase is a new therapeutic target in multiple myeloma involved in replication stress survival and drug resistance. Front Immunol 2022; 13:983181. [PMID: 36569948 PMCID: PMC9780552 DOI: 10.3389/fimmu.2022.983181] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is a hematologic cancer characterized by accumulation of malignant plasma cells in the bone marrow. To date, no definitive cure exists for MM and resistance to current treatments is one of the major challenges of this disease. The DNA helicase BLM, whose depletion or mutation causes the cancer-prone Bloom's syndrome (BS), is a central factor of DNA damage repair by homologous recombination (HR) and genomic stability maintenance. Using independent cohorts of MM patients, we identified that high expression of BLM is associated with a poor outcome with a significant enrichment in replication stress signature. We provide evidence that chemical inhibition of BLM by the small molecule ML216 in HMCLs (human myeloma cell lines) leads to cell cycle arrest and increases apoptosis, likely by accumulation of DNA damage. BLM inhibition synergizes with the alkylating agent melphalan to efficiently inhibit growth and promote cell death in HMCLs. Moreover, ML216 treatment re-sensitizes melphalan-resistant cell lines to this conventional therapeutic agent. Altogether, these data suggest that inhibition of BLM in combination with DNA damaging agents could be of therapeutic interest in the treatment of MM, especially in those patients with high BLM expression and/or resistance to melphalan.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Elena Viziteu
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Laure Dutrieux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Julie Devin
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Yea-Lih Lin
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Elina Alaterre
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Michel Jourdan
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Jihane Basbous
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Guilhem Requirand
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | - Nicolas Robert
- Department of Biological Hematology, CHU Montpellier, Montpellier, France
| | | | | | - Dirk Hose
- Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Laure Vincent
- Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Charles Herbaux
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France,Department of Hematology and Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | | - Philippe Pasero
- Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France
| | - Jérôme Moreaux
- Department of Biological Hematology, CHU Montpellier, Montpellier, France,Institute of Human Genetics, UMR 9002 CNRS-UM, Montpellier, France,Department of Clinical Hematology, CHU Montpellier, Montpellier, France,*Correspondence: Jérôme Moreaux,
| |
Collapse
|
11
|
Solimando AG, Malerba E, Leone P, Prete M, Terragna C, Cavo M, Racanelli V. Drug resistance in multiple myeloma: Soldiers and weapons in the bone marrow niche. Front Oncol 2022; 12:973836. [PMID: 36212502 PMCID: PMC9533079 DOI: 10.3389/fonc.2022.973836] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is still an incurable disease, despite considerable improvements in treatment strategies, as resistance to most currently available agents is not uncommon. In this study, data on drug resistance in MM were analyzed and led to the following conclusions: resistance occurs via intrinsic and extrinsic mechanisms, including intraclonal heterogeneity, drug efflux pumps, alterations of drug targets, the inhibition of apoptosis, increased DNA repair and interactions with the bone marrow (BM) microenvironment, cell adhesion, and the release of soluble factors. Since MM involves the BM, interactions in the MM-BM microenvironment were examined as well, with a focus on the cross-talk between BM stromal cells (BMSCs), adipocytes, osteoclasts, osteoblasts, endothelial cells, and immune cells. Given the complex mechanisms that drive MM, next-generation treatment strategies that avoid drug resistance must target both the neoplastic clone and its non-malignant environment. Possible approaches based on recent evidence include: (i) proteasome and histone deacetylases inhibitors that not only target MM but also act on BMSCs and osteoclasts; (ii) novel peptide drug conjugates that target both the MM malignant clone and angiogenesis to unleash an effective anti-MM immune response. Finally, the role of cancer stem cells in MM is unknown but given their roles in the development of solid and hematological malignancies, cancer relapse, and drug resistance, their identification and description are of paramount importance for MM management.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
- Istituto di ricovero e cura a carattere scientifico (IRCCS) Istituto Tumori ‘Giovanni Paolo II’ of Bari, Bari, Italy
| | - Eleonora Malerba
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Marcella Prete
- Department of Interdisciplinary Medicine, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Carolina Terragna
- ’Seràgnoli’ Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Michele Cavo
- ’Seràgnoli’ Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Vito Racanelli
- Department of Interdisciplinary Medicine, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
- *Correspondence: Vito Racanelli,
| |
Collapse
|
12
|
Teoh PJ, An O, Chung TH, Vaiyapuri T, Raju A, Hoppe MM, Toh SHM, Wang W, Chan MC, Fullwood MJ, Jeyasekharan AD, Tergaonkar V, Chen L, Yang H, Chng WJ. p53-NEIL1 co-abnormalities induce genomic instability and promote synthetic lethality with Chk1 inhibition in multiple myeloma having concomitant 17p13(del) and 1q21(gain). Oncogene 2022; 41:2106-2121. [DOI: 10.1038/s41388-022-02227-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/21/2022] [Accepted: 02/01/2022] [Indexed: 11/09/2022]
|
13
|
Chen D, Yang X, Liu M, Zhang Z, Xing E. Roles of miRNA dysregulation in the pathogenesis of multiple myeloma. Cancer Gene Ther 2021; 28:1256-1268. [PMID: 33402729 PMCID: PMC8636266 DOI: 10.1038/s41417-020-00291-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/03/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023]
Abstract
Multiple myeloma (MM) is a malignant disease of plasma cells with complex pathology, causing significant morbidity due to its end-organ destruction. The outcomes of patients with myeloma have significantly improved in the past couple of decades with the introduction of novel agents, such as proteasome inhibitors, immunomodulators, and monoclonal antibodies. However, MM remains incurable and presents considerable individual heterogeneity. MicroRNAs (miRNAs) are short, endogenous noncoding RNAs of 19-22 nucleotides that regulate gene expression at the posttranscriptional level. Numerous studies have shown that miRNA deregulation is closely related to MM pathology, including tumor initiation, progression, metastasis, prognosis, and drug response, which make the complicated miRNA network an attractive and marvelous area of investigation for novel anti-MM therapeutic approaches. Herein, we mainly summarized the current knowledge on the roles of miRNAs, which are of great significance in regulating pathological factors involved in MM progressions, such as bone marrow microenvironment, methylation, immune regulation, genomic instability, and drug resistance. Meanwhile, their potential as novel prognostic biomarkers and therapeutic targets was also discussed.
Collapse
Affiliation(s)
- Dan Chen
- Department of Central Laboratory, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Xinhong Yang
- Department of Hematology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Min Liu
- Department of Hematology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China
| | - Zhihua Zhang
- Department of Hematology, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China.
| | - Enhong Xing
- Department of Central Laboratory, The Affiliated Hospital of Chengde Medical College, Chengde, Hebei, China.
| |
Collapse
|
14
|
Valikhani M, Rahimian E, Ahmadi SE, Chegeni R, Safa M. Involvement of classic and alternative non-homologous end joining pathways in hematologic malignancies: targeting strategies for treatment. Exp Hematol Oncol 2021; 10:51. [PMID: 34732266 PMCID: PMC8564991 DOI: 10.1186/s40164-021-00242-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/13/2021] [Indexed: 12/31/2022] Open
Abstract
Chromosomal translocations are the main etiological factor of hematologic malignancies. These translocations are generally the consequence of aberrant DNA double-strand break (DSB) repair. DSBs arise either exogenously or endogenously in cells and are repaired by major pathways, including non-homologous end-joining (NHEJ), homologous recombination (HR), and other minor pathways such as alternative end-joining (A-EJ). Therefore, defective NHEJ, HR, or A-EJ pathways force hematopoietic cells toward tumorigenesis. As some components of these repair pathways are overactivated in various tumor entities, targeting these pathways in cancer cells can sensitize them, especially resistant clones, to radiation or chemotherapy agents. However, targeted therapy-based studies are currently underway in this area, and furtherly there are some biological pitfalls, clinical issues, and limitations related to these targeted therapies, which need to be considered. This review aimed to investigate the alteration of DNA repair elements of C-NHEJ and A-EJ in hematologic malignancies and evaluate the potential targeted therapies against these pathways.
Collapse
Affiliation(s)
- Mohsen Valikhani
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Rahimian
- Department of Medical Translational Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Rouzbeh Chegeni
- Medical Laboratory Sciences, Program, College of Health and Human Sciences, Northern Illinois University, DeKalb, IL, USA
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Lyu L, Lin TC, McCarty N. TRIM44 mediated p62 deubiquitination enhances DNA damage repair by increasing nuclear FLNA and 53BP1 expression. Oncogene 2021; 40:5116-5130. [PMID: 34211088 PMCID: PMC9744239 DOI: 10.1038/s41388-021-01890-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/20/2021] [Accepted: 06/04/2021] [Indexed: 12/14/2022]
Abstract
Cancer cells show increases in protein degradation pathways, including autophagy, during progression to meet the increased protein degradation demand and support cell survival. On the other hand, reduced autophagy activity during aging is associated with a reduced DNA damage response and increased genomic instability. Therefore, it is a puzzling how DNA repair can be increased in cancer cells that are resistant to chemotherapies or during progression when autophagy activity is intact or increased. We discovered that tripartite motif containing 44 (TRIM44) is a pivotal element regulating the DNA damage response in cancer cells with intact autophagy. TRIM44 deubiquitinates p62, an autophagy substrate, which leads to its oligomerization. This prevents p62 localization to the nucleus upon irradiation. Increased cytoplasmic retention of p62 by TRIM44 prevents the degradation of FLNA and 53BP1, which increases DNA damage repair. Together, our data support TRIM44 a potential therapeutic target for therapy-resistant tumor cells with intact autophagy.
Collapse
Affiliation(s)
- Lin Lyu
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), the University of Texas-Health Science Center at Houston, Houston, Texas, 77030, USA
| | - Tsung-Chin Lin
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), the University of Texas-Health Science Center at Houston, Houston, Texas, 77030, USA
| | - Nami McCarty
- Center for Stem Cell and Regenerative Disease, Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM), the University of Texas-Health Science Center at Houston, Houston, Texas, 77030, USA.,Correspondence: Nami McCarty, Ph.D., University of Texas-Health Science Center at Houston, 1825 Pressler St., IMM-630A, Houston, TX 77030, USA, , Tel: 713-500-2495, Fax: 713-500-2424
| |
Collapse
|
16
|
Abstract
The concepts hybrid and hybridization are common in many scientific fields, as in the taxonomic parts of botany and zoology, in modern genetic, and in the quantum–mechanical theory of atomic–molecular orbitals, which are of foremost relevance in most aspects of modern chemistry. Years later, scientists applied the concept hybrid to colloids, if the particles’ domains are endowed with functionalities differing each from the other in nature and/or composition. For such denomination to be fully valid, the domains belonging to a given hybrid must be recognizable each from another in terms of some intrinsic features. Thus, the concept applies to particles where a given domain has its own physical state, functionality, or composition. Literature examples in this regard are many. Different domains that are present in hybrid colloids self-organize, self-sustain, and self-help, according to the constraints dictated by kinetic and/or thermodynamic stability rules. Covalent, or non-covalent, bonds ensure the formation of such entities, retaining the properties of a given family, in addition to those of the other, and, sometimes, new ones. The real meaning of this behavior is the same as in zoology; mules are pertinent examples, since they retain some features of their own parents (i.e., horses and donkeys) but also exhibit completely new ones, such as the loss of fertility. In colloid sciences, the concept hybrid refers to composites with cores of a given chemical type and surfaces covered by moieties differing in nature, or physical state. This is the result of a mimicry resembling the ones met in a lot of biological systems and foods, too. Many combinations may occur. Silica nanoparticles on which polymers/biopolymers are surface-bound (irrespective of whether binding is covalent or not) are pertinent examples. Here, efforts are made to render clear the concept, which is at the basis of many applications in the biomedical field, and not only. After a historical background and on some features of the species taking part to the formation of hybrids, we report on selected cases met in modern formulations of mixed, and sometimes multifunctional, colloid entities.
Collapse
|
17
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021. [DOI: 10.37349/etat.2020.00034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3University of Montpellier, UFR Medicine, 34093 Montpellier, France 4 Institut Universitaire de France (IUF), 75000 Paris France
| |
Collapse
|
18
|
Ovejero S, Moreaux J. Multi-omics tumor profiling technologies to develop precision medicine in multiple myeloma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:65-106. [PMID: 36046090 PMCID: PMC9400753 DOI: 10.37349/etat.2021.00034] [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: 10/17/2020] [Accepted: 01/06/2021] [Indexed: 11/19/2022] Open
Abstract
Multiple myeloma (MM), the second most common hematologic cancer, is caused by accumulation of aberrant plasma cells in the bone marrow. Its molecular causes are not fully understood and its great heterogeneity among patients complicates therapeutic decision-making. In the past decades, development of new therapies and drugs have significantly improved survival of MM patients. However, resistance to drugs and relapse remain the most common causes of mortality and are the major challenges to overcome. The advent of high throughput omics technologies capable of analyzing big amount of clinical and biological data has changed the way to diagnose and treat MM. Integration of omics data (gene mutations, gene expression, epigenetic information, and protein and metabolite levels) with clinical histories of thousands of patients allows to build scores to stratify the risk at diagnosis and predict the response to treatment, helping clinicians to make better educated decisions for each particular case. There is no doubt that the future of MM treatment relies on personalized therapies based on predictive models built from omics studies. This review summarizes the current treatments and the use of omics technologies in MM, and their importance in the implementation of personalized medicine.
Collapse
Affiliation(s)
- Sara Ovejero
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France
| | - Jerome Moreaux
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France 2Institute of Human Genetics, UMR 9002 CNRS-UM, 34000 Montpellier, France 3UFR Medicine, University of Montpellier, 34093 Montpellier, France 4Institut Universitaire de France (IUF), 75000 Paris, France
| |
Collapse
|
19
|
Saitoh T, Oda T. DNA Damage Response in Multiple Myeloma: The Role of the Tumor Microenvironment. Cancers (Basel) 2021; 13:504. [PMID: 33525741 PMCID: PMC7865954 DOI: 10.3390/cancers13030504] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/21/2021] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy characterized by genomic instability. MM cells present various forms of genetic instability, including chromosomal instability, microsatellite instability, and base-pair alterations, as well as changes in chromosome number. The tumor microenvironment and an abnormal DNA repair function affect genetic instability in this disease. In addition, states of the tumor microenvironment itself, such as inflammation and hypoxia, influence the DNA damage response, which includes DNA repair mechanisms, cell cycle checkpoints, and apoptotic pathways. Unrepaired DNA damage in tumor cells has been shown to exacerbate genomic instability and aberrant features that enable MM progression and drug resistance. This review provides an overview of the DNA repair pathways, with a special focus on their function in MM, and discusses the role of the tumor microenvironment in governing DNA repair mechanisms.
Collapse
Affiliation(s)
- Takayuki Saitoh
- Department of Laboratory Sciences, Graduate School of Health Sciences, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Tsukasa Oda
- Laboratory of Molecular Genetics, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma 371-8512, Japan;
| |
Collapse
|
20
|
Turner JG, Cui Y, Bauer AA, Dawson JL, Gomez JA, Kim J, Cubitt CL, Nishihori T, Dalton WS, Sullivan DM. Melphalan and Exportin 1 Inhibitors Exert Synergistic Antitumor Effects in Preclinical Models of Human Multiple Myeloma. Cancer Res 2020; 80:5344-5354. [PMID: 33023948 PMCID: PMC7718436 DOI: 10.1158/0008-5472.can-19-0677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 08/25/2020] [Accepted: 10/01/2020] [Indexed: 01/22/2023]
Abstract
High-dose chemotherapy with melphalan followed by autologous transplantation is a first-line treatment for multiple myeloma. Here, we present preclinical evidence that this treatment may be significantly improved by the addition of exportin 1 inhibitors (XPO1i). The XPO1i selinexor, eltanexor, and KOS-2464 sensitized human multiple myeloma cells to melphalan. Human 8226 and U266 multiple myeloma cell lines and melphalan-resistant cell lines (8226-LR5 and U266-LR6) were highly sensitized to melphalan by XPO1i. Multiple myeloma cells from newly diagnosed and relapsed/refractory multiple myeloma patients were also sensitized by XPO1i to melphalan. In NOD/SCIDγ mice challenged with either parental 8226 or U266 multiple myeloma and melphalan-resistant multiple myeloma tumors, XPO1i/melphalan combination treatments demonstrated stronger synergistic antitumor effects than single-agent melphalan with minimal toxicity. Synergistic cell death resulted from increased XPO1i/melphalan-induced DNA damage in a dose-dependent manner and decreased DNA repair. In addition, repair of melphalan-induced DNA damage was inhibited by selinexor, which decreased melphalan-induced monoubiquitination of FANCD2 in multiple myeloma cells. Knockdown of FANCD2 was found to replicate the effect of selinexor when used with melphalan, increasing DNA damage (γH2AX) by inhibiting DNA repair. Thus, combination therapies that include selinexor or eltanexor with melphalan may have the potential to improve treatment outcomes of multiple myeloma in melphalan-resistant and newly diagnosed patients. The combination of selinexor and melphalan is currently being investigated in the context of high-dose chemotherapy and autologous transplant (NCT02780609). SIGNIFICANCE: Inhibition of exportin 1 with selinexor synergistically sensitizes human multiple myeloma to melphalan by inhibiting Fanconi anemia pathway-mediated DNA repair.
Collapse
Affiliation(s)
- Joel G Turner
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Yan Cui
- Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Alexis A Bauer
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jana L Dawson
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Juan A Gomez
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Christopher L Cubitt
- Translational Research Core, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Taiga Nishihori
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - William S Dalton
- Department of Malignant Hematology, Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Daniel M Sullivan
- Chemical Biology and Molecular Medicine Program, Moffitt Cancer Center and Research Institute, Tampa, Florida.
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
21
|
Tang HL, Xu L, Chen XQ. [Bortezomib interferes with DNA repair and exerts synergistic anti-multiple myeloma activity with doxorubicin]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41:417-421. [PMID: 32536140 PMCID: PMC7342068 DOI: 10.3760/cma.j.issn.0253-2727.2020.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Indexed: 12/30/2022]
Affiliation(s)
- H L Tang
- Department of Hematology, Xijing Hospital, Air Force Medical University, Hematologic Diseases Center of Chinese People's Liberation Army, Xi'an 710032, China
| | - L Xu
- Department of Hematology, Xijing Hospital, Air Force Medical University, Hematologic Diseases Center of Chinese People's Liberation Army, Xi'an 710032, China
| | - X Q Chen
- Department of Hematology, Xijing Hospital, Air Force Medical University, Hematologic Diseases Center of Chinese People's Liberation Army, Xi'an 710032, China
| |
Collapse
|
22
|
[Bortezomib interferes with DNA repair and exerts synergistic anti-multiple myeloma activity with doxorubicin]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41. [PMID: 32536140 PMCID: PMC7342068 DOI: 10.3760/cma.j.issn.0253-2727.2020.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
23
|
Beksac M, Balli S, Akcora Yildiz D. Drug Targeting of Genomic Instability in Multiple Myeloma. Front Genet 2020; 11:228. [PMID: 32373151 PMCID: PMC7179656 DOI: 10.3389/fgene.2020.00228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/26/2020] [Indexed: 12/16/2022] Open
Abstract
Genomic instability can be observed at both chromosomal and chromatin levels. Instability at the macro level includes centrosome abnormalities (CA) resulting in numerical as well as structural chromosomal changes, whereas instability at the micro level is characterized by defects in DNA repair pathways resulting in microsatellite instability (MIN) or mutations. Genomic instability occurs during carcinogenesis without impairing survival and growth, though the precise mechanisms remain unclear. Solid tumors arising from most cells of epithelial origin are characterized by genomic instability which renders them resistant to chemotherapy and radiotherapy. This instability is also observed in 25% of myeloma patients and has been shown to be highly prognostic, independently of the international staging system (ISS). However, a biomarker of aberrant DNA repair and loss of heterozygosity (LOH), was only observed at a frequency of 5% in newly diagnosed patients. Several new molecules targeting the pathways involved in genomic instability are under development and some have already entered clinical trials. Poly(ADP-ribose) polymerase-1 (PARP) inhibitors have been FDA-approved for the treatment of breast cancer type 1 susceptibility protein (BRCA1)-mutated metastatic breast cancer, as well as ovarian and lung cancer. Topoisomerase inhibitors and epigenetic histone modification-targeting inhibitors, such as HDAC (Histone Deacetylase) inhibitors which are novel agents that can target genomic instability. Several of the small molecule inhibitors targeting chromosomal level instability such as PARP, Akt, Aurora kinase, cyclin dependent kinase or spindle kinase inhibitors have been tested in mouse models and early phase I/II trials. ATM, ATR kinase inhibitors and DNA helicase inhibitors are also promising novel agents. However, most of these drugs are not effective as single agents but appear to act synergistically with DNA damaging agents such as radiotherapy, platinum derivatives, immunomodulators, and proteasome inhibitors. In this review, new drugs targeting genomic instability and their mechanisms of action will be discussed.
Collapse
Affiliation(s)
- Meral Beksac
- Department of Hematology, School of Medicine, Ankara University, Ankara, Turkey
| | - Sevinc Balli
- Kars Selim Public Hospital, Internal Medicine, Kars, Turkey
| | - Dilara Akcora Yildiz
- Department of Biology, Science & Art Faculty, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
24
|
Patel PR, Senyuk V, Sweiss K, Calip GS, Pan D, Rodriguez N, Oh A, Mahmud N, Rondelli D. PARP Inhibition Synergizes with Melphalan but Does not Reverse Resistance Completely. Biol Blood Marrow Transplant 2020; 26:1273-1279. [PMID: 32194286 DOI: 10.1016/j.bbmt.2020.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/21/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022]
Abstract
High-dose melphalan (MEL) and autologous stem cell transplantation (ASCT) is the standard of care in the treatment of multiple myeloma (MM). Resistance to MEL has been linked to increased DNA repair. Here we sought to identify whether inhibition of poly(ADP-ribose) polymerase (PARP) synergizes with MEL and can overcome resistance. We tested the synergistic cytotoxicity of 3 inhibitors of PARP (PARPi)-veliparib (VEL), olaparib (OLA), and niraparib (NIRA)-combined with MEL in RPMI8226 and U266 MM cell lines, as well as in their MEL resistance counterparts, RPMI8226-LR5 (LR5) and U266-LR6 (LR6). The addition of VEL, OLA, and NIRA to MEL reduced the half maximal inhibitory concentration (IC50) in RPMI8226 cells from 27.8 µM to 23.1 µM, 22.5 µM, and 18.0 µM, respectively. Similarly, the IC50 of MEL in U266 cells was decreased from 6.2 µM to 3.2 µM, 3.3 µM, and 3.0 µM, respectively. In LR5 and LR6 cells, PARPi did not reverse MEL resistance. We confirmed this in a NOD/SCID/gamma null xenograft mouse model with either MEL-sensitive (RPMI8226) or MEL-resistant (LR5) MM. Treatment with a MEL-VEL combination prolonged survival compared with MEL alone in RPMI8226 mice (107 days versus 67.5 days; P = .0009), but not in LR5 mice (41 versus 39 days; P = .09). We next tested whether 2 double-stranded DNA repair mechanisms, homologous recombination (HR) and nonhomologous end-joining (NHEJ), cause MEL resistance in LR5 and LR6 cells. In an HR assay, LR6 cells had a 4.5-fold greater HR capability than parent U226 cells (P = .05); however, LR5 cells had an equivalent HR ability as parent RPMI8226 cells. We hypothesized that NHEJ may be a mediator of MEL resistance in LR5 cells. Given that DNA-PK is integral to NHEJ and may be a therapeutic target, we treated LR5 cells with the DNA-PK inhibitor NU7026 in combination with MEL. Although NU7026 alone at 2.5 µM had no cytotoxicity, in combination it completely reversed resistance to MEL (MEL IC50, 46.4 µM versus 14.4 µM). We examined the clinical implications of our findings in a dataset of 414 patients treated with tandem ASCT. High PARP1 expressers had lower survival compared with patients with low expression (median 42.7 months versus median not reached; P = .003). We hypothesized that combined expression of the HR gene BRCA1, the NHEJ gene PRKDC (DNA-PK), and PARP1 may predict survival and found that overexpression of 0 (n = 101), 1 or 2 (n = 287), or all 3 (n = 26) genes had a negative impact on median survival (undefined versus 57.8 months versus 14.8 months; P < .0001). Here we demonstrate that PARPi synergized with MEL, but that resistance (which may be due to HR and NHEJ pathways) is not completely reversed by PARPi. In addition, we observed that a 3-gene analysis may be tested to identify patients resistant or sensitive to high-dose MEL.
Collapse
Affiliation(s)
- Pritesh R Patel
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois.
| | - Vitalyi Senyuk
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Karen Sweiss
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois
| | - Greg S Calip
- Department of Pharmacy Systems, Outcomes and Policy, University of Illinois at Chicago, Chicago, Illinois
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; Biomedical Research Center, Carle Foundation Hospital, Urbana, Illinois
| | - Natalie Rodriguez
- University of Illinois at Chicago, College of Medicine, Chicago, Illinois
| | - Annie Oh
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Nadim Mahmud
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Damiano Rondelli
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
25
|
Röpke EF, Theissig F, Ulrich G, Bäker K, Bochwitz C, Grundig A, Paasch C. Solitary plasmacytoma of thoracic vertebra in a woman with Lynch syndrome: A case report. Int J Surg Case Rep 2019; 65:44-47. [PMID: 31683141 PMCID: PMC6839014 DOI: 10.1016/j.ijscr.2019.10.052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/09/2019] [Accepted: 10/24/2019] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION One of the major disabling health conditions among elderly is back pain due to degenerative diseases. Less than 1% of spine disorders are caused by malignancies. Among the rare primary vertebral neoplasms the multiple myeloma and the plasmacytoma account for 26% of these cases. PRESENTATION OF CASE We are reporting a case of 64 year-old woman, who suffered from progressive upper back pain and intermittent neurological symptoms including lower limb weakness and voiding disorder under axial loading. Her medical history includes a Lynch syndrome (LS). Computed tomography (CT) and magnetic resonance imaging (MRI) detected a single malignant osteolytic process of the spine involving T5. Urgent surgery with laminectomy intralesional tumor removal and posterior stabilization (Th4-Th6) due to unstable pathologic fracture with spinal cord compression was conducted after interdisciplinary decision. Histopathological examination of the tumor revealed a solitary plasmacytoma. DISCUSSION To our knowledge this is the first case report of a solitary plasmacytoma of the bone (SPB) that arise in a patient who suffers from LS. Similar DNA mismatch repair malfunction is existent in LS and SPB. Hence, a hereditary correlation might be imaginable. CONCLUSION When detecting a lytic spinal tumor in a patient who suffers from LS a SPB should be taken under consideration.
Collapse
Affiliation(s)
- E F Röpke
- Department of Orthopaedics, Traumatology and Spine Surgery, Helios Klinik Jerichower Land, August - Bebel - Strasse 55a, 39288, Burg, Germany.
| | - F Theissig
- Institute of Pathology, Helios Klinikum Emil von Behring, Walterhöferstrasse 11, 14165, Berlin, Germany.
| | - G Ulrich
- Radiologie Sudenburg, Halberstädter Str. 125 - 127, 39112, Magdeburg, Germany.
| | - K Bäker
- Department of Orthopaedics, Traumatology and Spine Surgery, Helios Klinik Jerichower Land, August - Bebel - Strasse 55a, 39288, Burg, Germany.
| | - C Bochwitz
- Department of Orthopaedics, Traumatology and Spine Surgery, Helios Klinik Jerichower Land, August - Bebel - Strasse 55a, 39288, Burg, Germany.
| | - A Grundig
- Department of Orthopaedics, Traumatology and Spine Surgery, Helios Klinik Jerichower Land, August - Bebel - Strasse 55a, 39288, Burg, Germany.
| | - C Paasch
- Department of General, Visceral and Cancer Surgery, Helios Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125, Berlin, Germany.
| |
Collapse
|
26
|
Gourzones C, Bret C, Moreaux J. Treatment May Be Harmful: Mechanisms/Prediction/Prevention of Drug-Induced DNA Damage and Repair in Multiple Myeloma. Front Genet 2019; 10:861. [PMID: 31620167 PMCID: PMC6759943 DOI: 10.3389/fgene.2019.00861] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 08/19/2019] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma (MM) is a malignancy characterized by accumulation of malignant plasma cells within the bone marrow (BM). MM is considered mostly without definitive treatment because of the inability of standard of care therapies to overcome drug-resistant relapse. Genotoxic agents are used in the treatment of MM and exploit the fact that DNA double-strand breaks are highly cytotoxic for cancer cells. However, their mutagenic effects are well-established and described. According to these effects, chemotherapy could cause harmful DNA damage associated with new driver genomic abnormalities providing selective advantage, drug resistance, and higher relapse risk. Several mechanisms associated with MM cell (MMC) resistance to genotoxic agents have been described, underlining MM heterogeneity. The understanding of these mechanisms provides several therapeutic strategies to overcome drug resistance and limit mutagenic effects of treatment in MM. According to this heterogeneity, adopting precision medicine into clinical practice, with the development of biomarkers, has the potential to improve MM disease management and treatment.
Collapse
Affiliation(s)
| | - Caroline Bret
- IGH, CNRS, Univ Montpellier, France.,Department of Biological Hematology, CHU Montpellier, Montpellier, France.,Univ Montpellier, UFR de Médecine, Montpellier, France
| | - Jerome Moreaux
- IGH, CNRS, Univ Montpellier, France.,Department of Biological Hematology, CHU Montpellier, Montpellier, France.,Univ Montpellier, UFR de Médecine, Montpellier, France.,Institut Universitaire de France, Paris, France
| |
Collapse
|
27
|
Vlummens P, De Veirman K, Menu E, De Bruyne E, Offner F, Vanderkerken K, Maes K. The Use of Murine Models for Studying Mechanistic Insights of Genomic Instability in Multiple Myeloma. Front Genet 2019; 10:740. [PMID: 31475039 PMCID: PMC6704229 DOI: 10.3389/fgene.2019.00740] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell malignancy characterized by the accumulation of clonal plasma cells in the bone marrow. In normal plasma cell development, cells undergo programmed DNA breaks and translocations, a process necessary for generation of a wide repertoire of antigen-specific antibodies. This process also makes them vulnerable for the acquisition of chromosomal defects. Well-known examples of these aberrations, already seen at time of MM diagnosis, are hyperdiploidy or the translocations involving the immunoglobulin heavy chain. Over the recent years, however, novel aspects concerning genomic instability and its role in tumor development, disease progression and nascence of refractory disease were identified. As such, genomic instability is becoming a very relevant research topic with the potential identification of novel disease pathways. In this review, we aim to describe recent studies involving murine MM models focusing on the deregulation of processes implicated in genomic instability and their clinical impact. More specifically, we will discuss chromosomal instability, DNA damage and repair responses, development of drug resistance, and recent insights into the study of clonal hierarchy using different murine MM models. Lastly, we will discuss the importance and the use of murine MM models in the pre-clinical evaluation of promising novel therapeutic agents.
Collapse
Affiliation(s)
- Philip Vlummens
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium.,Department of Clinical Hematology, Ghent University Hospital, Gent, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fritz Offner
- Department of Clinical Hematology, Ghent University Hospital, Gent, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ken Maes
- Department of Hematology and Immunology-Myeloma Center Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
28
|
Yuan X, Ma R, Yang S, Jiang L, Wang Z, Zhu Z, Li H. miR-520g and miR-520h overcome bortezomib resistance in multiple myeloma via suppressing APE1. Cell Cycle 2019; 18:1660-1669. [PMID: 31204563 DOI: 10.1080/15384101.2019.1632138] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Background: Nowadays, microRNAs (miRNAs) attract much attention in regulating anticancer drug resistance in cancers including multiple myeloma (MM). Bortezomib is the first-line choice in MM treatment, and bortezomib resistance caused by aberrant DNA repair leads to the recurrence and therapeutic failure of MM. Objective: Our study aims to identify a miRNA that overcomes bortezomib resistance in MM. Methods: We established bortezomib-resistant MM cell lines, and screened several miRNAs that have aberrant expressions in MM cell lines. The expression of DNA-repair-related proteins were assessed by western blot, and cell viability was determined by the MTT assay in bortezomib-resistant cell lines. The binding between miRNAs and 3'-UTR of APE1 mRNA was confirmed by luciferase reporter assay. The mouse bortezomib-resistant xenograft was established to verify the therapeutic effect of miRNA overexpression. Results: miR-520g and miR-520h were significantly downregulated in bortezomib-resistant MM cell lines, and overexpression of miR-520g and miR-520h together inhibited expression of homologous recombination-related protein Rad51 and cell viability of bortezomib-resistant MM cells in vitro by binding with 3'-UTR of APE1 mRNA. Combined overexpression of miR-520g and miR-520h inhibited bortezomib-resistant MM tumor growth in vivo. Conclusion: Our findings demonstrated that combined overexpression of miR-520g and miR-520h overcomes bortezomib resistance in MM through inhibition of DNA repair, offering a promising therapeutic target for MM treatment.
Collapse
Affiliation(s)
- Xiaoli Yuan
- a Department of Hematology, Henan Key Laboratory for Hematology, Henan Provincial People's Hospital , People's Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Rongjun Ma
- a Department of Hematology, Henan Key Laboratory for Hematology, Henan Provincial People's Hospital , People's Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Shiwei Yang
- a Department of Hematology, Henan Key Laboratory for Hematology, Henan Provincial People's Hospital , People's Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Li Jiang
- a Department of Hematology, Henan Key Laboratory for Hematology, Henan Provincial People's Hospital , People's Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Zhen Wang
- a Department of Hematology, Henan Key Laboratory for Hematology, Henan Provincial People's Hospital , People's Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Zunmin Zhu
- a Department of Hematology, Henan Key Laboratory for Hematology, Henan Provincial People's Hospital , People's Hospital of Zhengzhou University , Zhengzhou , Henan , China
| | - Hongwei Li
- b Department of Neurosurgery , the First Affiliated Hospital of Zhengzhou University , Zhengzhou , Henan , China
| |
Collapse
|
29
|
Janz S, Zhan F, Sun F, Cheng Y, Pisano M, Yang Y, Goldschmidt H, Hari P. Germline Risk Contribution to Genomic Instability in Multiple Myeloma. Front Genet 2019; 10:424. [PMID: 31139207 PMCID: PMC6518313 DOI: 10.3389/fgene.2019.00424] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022] Open
Abstract
Genomic instability, a well-established hallmark of human cancer, is also a driving force in the natural history of multiple myeloma (MM) - a difficult to treat and in most cases fatal neoplasm of immunoglobulin producing plasma cells that reside in the hematopoietic bone marrow. Long recognized manifestations of genomic instability in myeloma at the cytogenetic level include abnormal chromosome numbers (aneuploidy) caused by trisomy of odd-numbered chromosomes; recurrent oncogene-activating chromosomal translocations that involve immunoglobulin loci; and large-scale amplifications, inversions, and insertions/deletions (indels) of genetic material. Catastrophic genetic rearrangements that either shatter and illegitimately reassemble a single chromosome (chromotripsis) or lead to disordered segmental rearrangements of multiple chromosomes (chromoplexy) also occur. Genomic instability at the nucleotide level results in base substitution mutations and small indels that affect both the coding and non-coding genome. Sometimes this generates a distinctive signature of somatic mutations that can be attributed to defects in DNA repair pathways, the DNA damage response (DDR) or aberrant activity of mutator genes including members of the APOBEC family. In addition to myeloma development and progression, genomic instability promotes acquisition of drug resistance in patients with myeloma. Here we review recent findings on the genetic predisposition to myeloma, including newly identified candidate genes suggesting linkage of germline risk and compromised genomic stability control. The role of ethnic and familial risk factors for myeloma is highlighted. We address current research gaps that concern the lack of studies on the mechanism by which germline risk alleles promote genomic instability in myeloma, including the open question whether genetic modifiers of myeloma development act in tumor cells, the tumor microenvironment (TME), or in both. We conclude with a brief proposition for future research directions, which concentrate on the biological function of myeloma risk and genetic instability alleles, the potential links between the germline genome and somatic changes in myeloma, and the need to elucidate genetic modifiers in the TME.
Collapse
Affiliation(s)
- Siegfried Janz
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Fenghuang Zhan
- Department of Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, United States.,Holden Comprehensive Cancer Center, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, United States
| | - Fumou Sun
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Yan Cheng
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michael Pisano
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States.,Interdisciplinary Graduate Program in Immunology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, United States
| | - Ye Yang
- The Third Affiliated Hospital, Nanjing University of Chinese Medicine, Nanjing, China.,Ministry of Education's Key Laboratory of Acupuncture and Medicine Research, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hartmut Goldschmidt
- Medizinische Klinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany.,Nationales Centrum für Tumorerkrankungen, Heidelberg, Germany
| | - Parameswaran Hari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
30
|
Gourzones C, Bellanger C, Lamure S, Gadacha OK, De Paco EG, Vincent L, Cartron G, Klein B, Moreaux J. Antioxidant Defenses Confer Resistance to High Dose Melphalan in Multiple Myeloma Cells. Cancers (Basel) 2019; 11:cancers11040439. [PMID: 30925767 PMCID: PMC6521290 DOI: 10.3390/cancers11040439] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 12/25/2022] Open
Abstract
Background: Multiple myeloma (MM) is the second most common hematological cancer after lymphoma. It is characterized by the accumulation of clonal malignant plasma cells within the bone marrow. The development of drug resistance remains a major problem for effective treatment of MM. Understand the mechanisms underlying drug resistance in MM is a focal point to improve MM treatment. Methods: In the current study, we analyzed further the role of redox imbalance induction in melphalan-induced toxicity both in human myeloma cell lines (HMCLs) and primary myeloma cells from patients. Results: We developed an in-vitro model of short-term resistance to high-dose melphalan and identified that pretreatment with physiological concentration of GSH protects HMCLs from melphalan-induced cell cycle arrest and cytotoxicity. We validated these results using primary MM cells from patients co-cultured with their bone marrow microenvironment. GSH did not affect the ability of melphalan to induce DNA damages in MM cells. Interestingly, melphalan induced reactive oxygen species, a significant decrease in GSH concentration, protein and lipd oxydation together with NRF2 (NF-E2-related factor 2) pathway activation. Conclusions: Our data demonstrate that antioxidant defenses confers resistance to high dose melphalan in MM cells, supporting that redox status in MM cells could be determinant for patients’ response to melphalan.
Collapse
Affiliation(s)
- Claire Gourzones
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
| | - Céline Bellanger
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
| | - Sylvain Lamure
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
| | | | | | - Laure Vincent
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
| | - Guillaume Cartron
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Univ Montpellier, UMR CNRS 5235, 34000 Montpellier, France.
| | - Bernard Klein
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France.
| | - Jérôme Moreaux
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France.
| |
Collapse
|
31
|
Cucè M, Gallo Cantafio ME, Siciliano MA, Riillo C, Caracciolo D, Scionti F, Staropoli N, Zuccalà V, Maltese L, Di Vito A, Grillone K, Barbieri V, Arbitrio M, Di Martino MT, Rossi M, Amodio N, Tagliaferri P, Tassone P, Botta C. Trabectedin triggers direct and NK-mediated cytotoxicity in multiple myeloma. J Hematol Oncol 2019; 12:32. [PMID: 30898137 PMCID: PMC6429746 DOI: 10.1186/s13045-019-0714-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/26/2019] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Genomic instability is a feature of multiple myeloma (MM), and impairment in DNA damaging response (DDR) has an established role in disease pathobiology. Indeed, a deregulation of DNA repair pathways may contribute to genomic instability, to the establishment of drug resistance to genotoxic agents, and to the escape from immune surveillance. On these bases, we evaluated the role of different DDR pathways in MM and investigated, for the first time, the direct and immune-mediated anti-MM activity of the nucleotide excision repair (NER)-dependent agent trabectedin. METHODS Gene-expression profiling (GEP) was carried out with HTA2.0 Affymetrix array. Evaluation of apoptosis, cell cycle, and changes in cytokine production and release have been performed in 2D and 3D Matrigel-spheroid models through flow cytometry on MM cell lines and patients-derived primary MM cells exposed to increasing nanomolar concentrations of trabectedin. DNA-damage response has been evaluated through Western blot, immunofluorescence, and DNA fragmentation assay. Trabectedin-induced activation of NK has been assessed by CD107a degranulation. miRNAs quantification has been done through RT-PCR. RESULTS By comparing GEP meta-analysis of normal and MM plasma cells (PCs), we observed an enrichment in DNA NER genes in poor prognosis MM. Trabectedin triggered apoptosis in primary MM cells and MM cell lines in both 2D and 3D in vitro assays. Moreover, trabectedin induced DDR activation, cellular stress with ROS production, and cell cycle arrest. Additionally, a significant reduction of MCP1 cytokine and VEGF-A in U266-monocytes co-cultures was observed, confirming the impairment of MM-promoting milieu. Drug-induced cell stress in MM cells led to upregulation of NK activating receptors ligands (i.e., NKG2D), which translated into increased NK activation and degranulation. Mechanistically, this effect was linked to trabectedin-induced inhibition of NKG2D-ligands negative regulators IRF4 and IKZF1, as well as to miR-17 family downregulation in MM cells. CONCLUSIONS Taken together, our findings indicate a pleiotropic activity of NER-targeting agent trabectedin, which appears a promising candidate for novel anti-MM therapeutic strategies.
Collapse
Affiliation(s)
- Maria Cucè
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Maria Eugenia Gallo Cantafio
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Maria Anna Siciliano
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Nicoletta Staropoli
- Medical and Translational Oncology Units, AOU Mater Domini, Catanzaro, Italy
| | | | | | - Anna Di Vito
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Vito Barbieri
- Medical and Translational Oncology Units, AOU Mater Domini, Catanzaro, Italy
| | - Mariamena Arbitrio
- Institute of Neurological Sciences, UOS of Pharmacology, Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
- Medical and Translational Oncology Units, AOU Mater Domini, Catanzaro, Italy
| | - Marco Rossi
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
- Medical and Translational Oncology Units, AOU Mater Domini, Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
- Medical and Translational Oncology Units, AOU Mater Domini, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy.
- Medical and Translational Oncology Units, AOU Mater Domini, Catanzaro, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| | - Cirino Botta
- Department of Experimental and Clinical Medicine, Magna Graecia University, Salvatore Venuta University Campus, Viale Europa, 88100, Catanzaro, Italy
| |
Collapse
|
32
|
Caracciolo D, Di Martino MT, Amodio N, Morelli E, Montesano M, Botta C, Scionti F, Talarico D, Altomare E, Gallo Cantafio ME, Zuccalà V, Maltese L, Todoerti K, Rossi M, Arbitrio M, Neri A, Tagliaferri P, Tassone P. miR-22 suppresses DNA ligase III addiction in multiple myeloma. Leukemia 2019; 33:487-498. [PMID: 30120376 PMCID: PMC6365379 DOI: 10.1038/s41375-018-0238-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/04/2018] [Accepted: 07/13/2018] [Indexed: 12/15/2022]
Abstract
Multiple myeloma (MM) is a hematologic malignancy characterized by high genomic instability. Here we provide evidence that hyper-activation of DNA ligase III (LIG3) is crucial for genomic instability and survival of MM cells. LIG3 mRNA expression in MM patients correlates with shorter survival and even increases with more advanced stage of disease. Knockdown of LIG3 impairs MM cells viability in vitro and in vivo, suggesting that neoplastic plasmacells are dependent on LIG3-driven repair. To investigate the mechanisms involved in LIG3 expression, we investigated the post-transcriptional regulation. We identified miR-22-3p as effective negative regulator of LIG3 in MM. Enforced expression of miR-22 in MM cells downregulated LIG3 protein, which in turn increased DNA damage inhibiting in vitro and in vivo cell growth. Taken together, our findings demonstrate that myeloma cells are addicted to LIG3, which can be effectively inhibited by miR-22, promoting a novel axis of genome stability regulation.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Eugenio Morelli
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Martina Montesano
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Cirino Botta
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | | | - Emanuela Altomare
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Maria Eugenia Gallo Cantafio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | | | | | - Katia Todoerti
- Department of Oncology and Hemato-oncology, University of Milan, and Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
| | - Marco Rossi
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Mariamena Arbitrio
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Antonino Neri
- Department of Oncology and Hemato-oncology, University of Milan, and Hematology, Fondazione Cà Granda IRCCS Policlinico, Milan, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Græcia University, Campus Salvatore Venuta, Catanzaro, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Vikova V, Jourdan M, Robert N, Requirand G, Boireau S, Bruyer A, Vincent L, Cartron G, Klein B, Elemento O, Kassambara A, Moreaux J. Comprehensive characterization of the mutational landscape in multiple myeloma cell lines reveals potential drivers and pathways associated with tumor progression and drug resistance. Theranostics 2019; 9:540-553. [PMID: 30809292 PMCID: PMC6376179 DOI: 10.7150/thno.28374] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 12/12/2018] [Indexed: 12/30/2022] Open
Abstract
Human multiple myeloma tumor cell lines (HMCLs) have been a cornerstone of research in multiple myeloma (MM) and have helped to shape our understanding of molecular processes that drive tumor progression. A comprehensive characterization of genomic mutations in HMCLs will provide a basis for choosing relevant cell line models to study a particular aspect of myeloma biology, or to screen for an antagonist of certain cancer pathways. Methods: We performed whole exome sequencing on a large cohort of 30 HMCLs, representative of a large molecular heterogeneity of MM, and 8 control samples (epstein-barr virus (EBV)-immortalized B-cells obtained from 8 different patients). We evaluated the sensitivity of HMCLs to ten drugs. Results: We identified a high confidence list of 236 protein-coding genes with mutations affecting the structure of the encoded protein. Among the most frequently mutated genes, there were known MM drivers, such as TP53, KRAS, NRAS, ATM and FAM46C, as well as novel mutated genes, including CNOT3, KMT2D, MSH3 and PMS1. We next generated a comprehensive map of altered key pathways in HMCLs. These include cell growth pathways (MAPK, JAK-STAT, PI(3)K-AKT and TP53 / cell cycle pathway), DNA repair pathway and chromatin modifiers. Importantly, our analysis highlighted a significant association between the mutation of several genes and the response to conventional drugs used in MM as well as targeted inhibitors. Conclusion: Taken together, this first comprehensive exome-wide analysis of the mutational landscape in HMCLs provides unique resources for further studies and identifies novel genes potentially associated with MM pathophysiology, some of which may be targets for future therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Nicolas Robert
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
| | - Guilhem Requirand
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
| | - Stéphanie Boireau
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
| | | | - Laure Vincent
- CHU Montpellier, Department of Clinical Hematology, Montpellier, France
| | - Guillaume Cartron
- Univ Montpellier, UFR de Médecine, Montpellier, France
- CHU Montpellier, Department of Clinical Hematology, Montpellier, France
- Univ Montpellier, UMR CNRS 5235, Montpellier, France
| | - Bernard Klein
- IGH, CNRS, Univ Montpellier, France
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
- Univ Montpellier, UFR de Médecine, Montpellier, France
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, 1305 York Avenue, New York, NY 10021, USA
| | - Alboukadel Kassambara
- IGH, CNRS, Univ Montpellier, France
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
| | - Jérôme Moreaux
- IGH, CNRS, Univ Montpellier, France
- CHU Montpellier, Department of Biological Hematology, Montpellier, France
- Univ Montpellier, UFR de Médecine, Montpellier, France
| |
Collapse
|
34
|
Patel K, Ahmed ZSO, Huang X, Yang Q, Ekinci E, Neslund-Dudas CM, Mitra B, Elnady FAEM, Ahn YH, Yang H, Liu J, Dou QP. Discovering proteasomal deubiquitinating enzyme inhibitors for cancer therapy: lessons from rational design, nature and old drug reposition. Future Med Chem 2018; 10:2087-2108. [PMID: 30066579 PMCID: PMC6123888 DOI: 10.4155/fmc-2018-0091] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 06/15/2018] [Indexed: 12/24/2022] Open
Abstract
The ubiquitin proteasome system has been validated as a target of cancer therapies evident by the US FDA approval of anticancer 20S proteasome inhibitors. Deubiquitinating enzymes (DUBs), an essential component of the ubiquitin proteasome system, regulate cellular processes through the removal of ubiquitin from ubiquitinated-tagged proteins. The deubiquitination process has been linked with cancer and other pathologies. As such, the study of proteasomal DUBs and their inhibitors has garnered interest as a novel strategy to improve current cancer therapies, especially for cancers resistant to 20S proteasome inhibitors. This article reviews proteasomal DUB inhibitors in the context of: discovery through rational design approach, discovery from searching natural products and discovery from repurposing old drugs, and offers a future perspective.
Collapse
Affiliation(s)
- Kush Patel
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Zainab SO Ahmed
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Giza 12613, Egypt
| | - Xuemei Huang
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- School of Life Science & Technology, Harbin Institute of Technology, Harbin 150001, PR China
| | - Qianqian Yang
- Protein Modification & Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou 510000, PR China
| | - Elmira Ekinci
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Christine M Neslund-Dudas
- Department of Public Health Sciences & Henry Ford Cancer Institute, Henry Ford Health System, One Ford Place, Suite 5C, Detroit, MI 48202, USA
| | - Bharati Mitra
- Department of Biochemistry, Microbiology & Immunology, Wayne State University School of Medicine, 540 E. Canfield Avenue, Detroit, MI 48201, USA
| | - Fawzy AEM Elnady
- Department of Anatomy & Embryology, Faculty of Veterinary Medicine, Cairo University, Giza, Giza 12613, Egypt
| | - Young-Hoon Ahn
- Department of Chemistry, Wayne State University, Detroit, MI 48202, USA
| | - Huanjie Yang
- School of Life Science & Technology, Harbin Institute of Technology, Harbin 150001, PR China
| | - Jinbao Liu
- Protein Modification & Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou 510000, PR China
| | - Qing Ping Dou
- Departments of Oncology, Pharmacology & Pathology, School of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
- Protein Modification & Degradation Lab, School of Basic Medical Sciences, Affiliated Tumor Hospital of Guangzhou Medical University, Guangzhou 510000, PR China
| |
Collapse
|
35
|
Guo X, He D, Zhang E, Chen J, Chen Q, Li Y, Yang L, Yang Y, Zhao Y, Wang G, He J, Cai Z. HMGB1 knockdown increases MM cell vulnerability by regulating autophagy and DNA damage repair. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:205. [PMID: 30157958 PMCID: PMC6114506 DOI: 10.1186/s13046-018-0883-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 08/16/2018] [Indexed: 02/06/2023]
Abstract
Background With the development of novel therapeutic agents, the survival of multiple myeloma (MM) patients has much improved. However, the disease is incurable due to drug resistance. Previous studies have found that high-mobility group box 1 (HMGB1) is involved in inflammation, angiogenesis, DNA damage repair, and cancer invasion, progression, metastasis and drug resistance and that high HMGB1 expression is associated with poor MM prognosis, yet the role and mechanism of HMGB1 in MM remains unclear. Methods Through gene expression and Oncomine database analyses, we found that HMGB1 is associated with a poor prognosis in MM patients. RNA interference together with gene array analysis, cell proliferation and apoptosis assays, autophagy detection assays, western blotting, and in vivo xenograft models were employed to evaluate the effect of HMGB1 and the mechanism involved in MM drug resistance. Results MM cell lines and primary MM samples were found to express high levels of HMGB1, which was negatively associated with the 3-year survival of MM patients. HMGB1 knockdown in MM cells enhanced the inhibitory effect of chemotherapy with dexamethasone (Dex) via apoptosis induction. Furthermore, downregulation of HMGB1 activated the mTOR pathway, inhibited autophagy and increased DNA damage induced by Dex by modulating expression of related genes. In vivo, xenograft models showed that after Dex treatment, the tumor burden of HMGB1-knockdown mice was decreased compared with that of control mice. Conclusions Our research shows that HMGB1 participates in autophagy and DNA damage repair and that downregulation of HMGB1 enhances the sensitivity of MM cells to Dex, suggesting that HMGB1 may serve as a target for MM treatment.
Collapse
Affiliation(s)
- Xing Guo
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Donghua He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Jing Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Qingxiao Chen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Yi Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Li Yang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Yang Yang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Yi Zhao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Gang Wang
- Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China
| | - Jingsong He
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
36
|
Yang X, Ye H, He M, Zhou X, Sun N, Guo W, Lin X, Huang H, Lin Y, Yao R, Wang H. LncRNA PDIA3P interacts with c-Myc to regulate cell proliferation via induction of pentose phosphate pathway in multiple myeloma. Biochem Biophys Res Commun 2018; 498:207-213. [PMID: 29501744 DOI: 10.1016/j.bbrc.2018.02.211] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 02/28/2018] [Indexed: 12/24/2022]
Abstract
Multiple myeloma (MM), the second most common hematologic malignancy, is an incurable disease characterized by the accumulation of malignant plasma cells within the bone marrow. Though great progresses have been made in understanding the mechanisms of MM, metabolic plasticity and drug resistance remain largely unknown. In this study, we found lncRNA Protein disulfide isomerase family A member 3 pseudogene 1 (PDIA3P) is highly expressed in MM and is associated with the survival rate of MM patients. PDIA3P regulates MM growth and drug resistance through Glucose 6-phosphate dehydrogenase (G6PD) and the pentose phosphate pathway (PPP). Mechanistically, we revealed that PDIA3P interacts with c-Myc to enhance its transactivation activity and binding to G6PD promoter, stimulating G6PD expression and PPP flux. Our study identified PDIA3P as a novel c-Myc interacting lncRNA and elucidated crucial roles for PDIA3P in metabolic regulation of MM, providing a potential therapeutic target for MM patients.
Collapse
Affiliation(s)
- Xiangchou Yang
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Haihao Ye
- Department of Cardiology, Wenzhou TCM Hospital, Wenzhou, 325000, China
| | - Muqing He
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaohai Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ni Sun
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Wenjian Guo
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaoji Lin
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - He Huang
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ying Lin
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Rongxin Yao
- Department of Hematology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hong Wang
- Department of Rheumatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
37
|
Tibullo D, Barbagallo I, Giallongo C, Vanella L, Conticello C, Romano A, Saccone S, Godos J, Di Raimondo F, Li Volti G. Heme oxygenase-1 nuclear translocation regulates bortezomibinduced cytotoxicity and mediates genomic instability in myeloma cells. Oncotarget 2018; 7:28868-80. [PMID: 26930712 PMCID: PMC5045362 DOI: 10.18632/oncotarget.7563] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/20/2016] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is a clonal B-cell malignancy characterized by an accumulation of clonal plasma cells in the bone marrow leading to bone destruction and bone marrow failure. Several molecular mechanisms underlie chemoresistance among which heme oxygenase-1 (HO-1) could play a major role. The aim of the present research was to evaluate the impact of HO-1 in MM following bortezomib (BTZ) treatment and how HO-1 is implicated in the mechanisms of chemoresistance. MM cells were treated for 24h with BTZ (15 nM), a boronic acid dipeptide inhibitor of the 26S proteasome used in the treatment of patients with MM as first-line therapy. We evaluated cell viability, reactive oxygen species (ROS) formation, endoplasmic reticulum (ER) stress, HO-1 expression and compartmentalization and cellular genetic instability. Results showed that BTZ significantly reduced cell viability in different MM cell lines and induced ER-stress and ROS formation. Concomitantly, we observed a significant overexpression of both HO-1 gene and protein levels. This effect was abolished by concomitant treatment with 4-phenybutirric acid, a molecular chaperone, which is known to reduce ER-stress. Surprisingly, inhibition of HO activity with SnMP (10μM) failed to increase BTZ sensitivity in MM cells whereas inhibition of HO-1 nuclear translocation by E64d, a cysteine protease inhibitor, increased sensitivity to BTZ and decreased genetic instability as measured by cytokinesis-block micronucleus assay. In conclusion, our data suggest that BTZ sensitivity depends on HO-1 nuclear compartmentalization and not on its enzymatic activity and this finding may represent an important tool to overcome BTZ chemoresistance in MM patients.
Collapse
Affiliation(s)
- Daniele Tibullo
- Division of Haematology, AOU "Policlinico - Vittorio Emanuele", University of Catania, Catania, Italy.,Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | | | - Cesarina Giallongo
- Division of Haematology, AOU "Policlinico - Vittorio Emanuele", University of Catania, Catania, Italy
| | - Luca Vanella
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Concetta Conticello
- Division of Haematology, AOU "Policlinico - Vittorio Emanuele", University of Catania, Catania, Italy
| | - Alessandra Romano
- Division of Haematology, AOU "Policlinico - Vittorio Emanuele", University of Catania, Catania, Italy
| | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Justyna Godos
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Francesco Di Raimondo
- Division of Haematology, AOU "Policlinico - Vittorio Emanuele", University of Catania, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,EuroMediterranean Institute of Science and Technology, Palermo, Italy
| |
Collapse
|
38
|
Handa H, Sasaki Y, Hattori H, Alkebsi L, Kasamatsu T, Saitoh T, Mitsui T, Yokohama A, Tsukamoto N, Matsumoto M, Murakami H. Recurrent alterations of the WW domain containing oxidoreductase gene spanning the common fragile site FRA16D in multiple myeloma and monoclonal gammopathy of undetermined significance. Oncol Lett 2017; 14:4372-4378. [PMID: 28943951 DOI: 10.3892/ol.2017.6672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 03/03/2017] [Indexed: 11/05/2022] Open
Abstract
The putative tumor suppressor gene WW domain containing oxidoreductase (WWOX) spans a common fragile site (CFS) on chromosome 16q23.3. CFSs are regions of profound genomic instability and sites for genomic deletions in cancer cells. Therefore, WWOX is structurally altered in diverse nonhematological cancer types. However, the function of WWOX in hematological tumor types, including multiple myeloma (MM) and monoclonal gammopathy of undetermined significance (MGUS) remains unclear. WWOX expression and methylation in patients with MM, MGUS, or noninvasive lymphoma (control) were analyzed using reverse transcription- and methylation specific-polymerase chain reaction analysis. Variant WWOX transcripts were detected in 65 and 50% of patients with MM and MGUS, respectively, compared with 10% of controls. WWOX expression was higher in patients with MM, and WWOX promoter methylation was detected in 35% of patients with MM compared with 5% of patients with MGUS and 4% of controls. WWOX promoter methylation was significantly associated with shorter overall survival time of patients, in particular those with MM who were never treated with novel agents. Genomic alterations, including deletions and promoter methylation that affect WWOX expression occur early and may be involved in the pathogenesis, progression, and prognosis of MM.
Collapse
Affiliation(s)
- Hiroshi Handa
- Department of Medicine and Clinical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Yoshiko Sasaki
- Department of Laboratory Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Hikaru Hattori
- Department of Laboratory Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Lobna Alkebsi
- Department of Laboratory Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Tetsuhiro Kasamatsu
- Department of Laboratory Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Takayuki Saitoh
- Department of Laboratory Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Takeki Mitsui
- Department of Medicine and Clinical Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Akihiko Yokohama
- Blood Transfusion Service, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Norifumi Tsukamoto
- Oncology Center, Gunma University Hospital, Maebashi, Gunma 371-8511, Japan
| | - Morio Matsumoto
- Department of Hematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, Gunma 377-0280, Japan
| | - Hirokazu Murakami
- Department of Laboratory Science, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
39
|
Nucleotide excision repair is a potential therapeutic target in multiple myeloma. Leukemia 2017; 32:111-119. [PMID: 28588253 PMCID: PMC5720937 DOI: 10.1038/leu.2017.182] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/05/2017] [Accepted: 05/24/2017] [Indexed: 12/20/2022]
Abstract
Despite the development of novel drugs, alkylating agents remain an important component of therapy in multiple myeloma (MM). DNA repair processes contribute towards sensitivity to alkylating agents and therefore we here evaluate the role of nucleotide excision repair (NER), which is involved in the removal of bulky adducts and DNA crosslinks in MM. We first evaluated NER activity using a novel functional assay and observed a heterogeneous NER efficiency in MM cell lines and patient samples. Using next-generation sequencing data, we identified that expression of the canonical NER gene, excision repair cross-complementation group 3 (ERCC3), significantly impacted the outcome in newly diagnosed MM patients treated with alkylating agents. Next, using small RNA interference, stable knockdown and overexpression, and small-molecule inhibitors targeting xeroderma pigmentosum complementation group B (XPB), the DNA helicase encoded by ERCC3, we demonstrate that NER inhibition significantly increases sensitivity and overcomes resistance to alkylating agents in MM. Moreover, inhibiting XPB leads to the dual inhibition of NER and transcription and is particularly efficient in myeloma cells. Altogether, we show that NER impacts alkylating agents sensitivity in myeloma cells and identify ERCC3 as a potential therapeutic target in MM.
Collapse
|
40
|
Kim J, Choi S, Saxena N, Singh AK, Singh I, Won JS. Regulation of STAT3 and NF-κB activations by S-nitrosylation in multiple myeloma. Free Radic Biol Med 2017; 106:245-253. [PMID: 28232202 PMCID: PMC5826580 DOI: 10.1016/j.freeradbiomed.2017.02.039] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 02/09/2017] [Accepted: 02/20/2017] [Indexed: 10/20/2022]
Abstract
Numerous reports suggest that aberrant activations of STAT3 and NF-κB promote survival and proliferation of multiple myeloma (MM) cells. In the present report, we demonstrate that a synthetic S-nitrosothiol compound, S-nitroso-N-acetylcysteine (SNAC), inhibits proliferation and survival of multiple MM cells via S-nitrosylation-dependent inhibition of STAT3 and NF-κB. In human MM cells (e.g. U266, H929, and IM-9 cells), SNAC treatment increased S-nitrosylation of STAT3 and NF-κB and inhibited their activities. Consequently, SNAC treatment resulted in MM cell cycle arrest at G1/S check point and inhibited their proliferation. SNAC also decreased the expression of cell survival factors and increased the activities of caspases, thus increased sensitivity of MM cells to melphalan, a chemotherapeutic agent for MM. In U266 xenografted mice, SNAC treatment decreased the activity of STAT3 and reduced the growth of human CD138 positive cells (U266 cells) in the bone marrow and also reduced their production of human IgE into the serum. Taken together, these data document the S-nitrosylation mediated inhibition of MM cell proliferation and cell survival via inhibition of STAT3 and NF-κB pathways and its efficacy in animal model of MM.
Collapse
Affiliation(s)
- Jinsu Kim
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Seungho Choi
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Nishant Saxena
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, United States; Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, United States
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, United States.
| | - Je-Seong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, United States.
| |
Collapse
|
41
|
RECQ1 helicase is involved in replication stress survival and drug resistance in multiple myeloma. Leukemia 2017; 31:2104-2113. [PMID: 28186131 PMCID: PMC5629372 DOI: 10.1038/leu.2017.54] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 12/29/2016] [Accepted: 01/27/2017] [Indexed: 12/19/2022]
Abstract
Multiple myeloma (MM) is a plasma cell cancer with poor survival, characterized by the expansion of multiple myeloma cells (MMCs) in the bone marrow. Using a microarray-based genome-wide screen for genes responding to DNA methyltransferases (DNMT) inhibition in MM cells, we identified RECQ1 among the most downregulated genes. RecQ helicases are DNA unwinding enzymes involved in the maintenance of chromosome stability. Here we show that RECQ1 is significantly overexpressed in MMCs compared to normal plasma cells and that increased RECQ1 expression is associated with poor prognosis in three independent cohorts of patients. Interestingly, RECQ1 knockdown inhibits cells growth and induces apoptosis in MMCs. Moreover, RECQ1 depletion promotes the development of DNA double-strand breaks, as evidenced by the formation of 53BP1 foci and the phosphorylation of ataxia-telangiectasia mutated (ATM) and histone variant H2A.X (H2AX). In contrast, RECQ1 overexpression protects MMCs from melphalan and bortezomib cytotoxicity. RECQ1 interacts with PARP1 in MMCs exposed to treatment and RECQ1 depletion sensitizes MMCs to poly(ADP-ribose) polymerase (PARP) inhibitor. DNMT inhibitor treatment results in RECQ1 downregulation through miR-203 deregulation in MMC. Altogether, these data suggest that association of DNA damaging agents and/or PARP inhibitors with DNMT inhibitors may represent a therapeutic approach in patients with high RECQ1 expression associated with a poor prognosis.
Collapse
|
42
|
Nadiminti K, Singh Abbi KK, Mott SL, Dozeman L, Tricot A, Schultz A, Behrends S, Zhan F, Tricot G. VTD-melphalan is well tolerated and results in very high rates of stringent CR and MRD-negative status in multiple myeloma. Onco Targets Ther 2017; 10:217-226. [PMID: 28123303 PMCID: PMC5229169 DOI: 10.2147/ott.s112423] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The addition of cytotoxic drugs to high-dose melphalan as a preparative regimen for autologous stem cell transplantation in multiple myeloma has not resulted in superior activity. Although novel agents have significantly improved outcome in multiple myeloma, their role in preparative regimens remains largely unknown. We have evaluated the toxicity and efficacy of combining bortezomib, thalidomide, and dexamethasone with high-dose melphalan. An institutional review board-approved retrospective analysis was performed on 100 consecutive patients receiving 153 transplants; 53 had tandem transplants; 64 patients received early transplants; and 36 had salvage transplantation. Endpoints were treatment-related toxicity and mortality, and quality of response post-transplantation with assessment of stringent complete remission (sCR) and minimal residual disease (MRD) status. Median age was 61 years, and median follow-up was 16.2 months. At 6 months, sCR was attained in 56% of patients and CR in 20%. An MRD status, assessed by sensitive (10−4) multiparameter flow cytometry, was achieved in 85%. The 100-day mortality rate was 2.6% (4/153); 1.8% for early transplants and 4.5% for salvage transplants. Grade 3–5 non-hematologic toxicities were mainly related to metabolism/nutrition; gastrointestinal and infectious problems. Median time to absolute neutrophil count of >500/µL was 12 days for both early and salvage transplantations. No significant differences in quality of response were observed between early and salvage transplantation or between single and tandem autologous stem cell transplantation. Since both sCR and MRD are excellent early surrogate markers for progression-free and overall survival, this regimen will likely be superior to melphalan alone, but it needs to be formally assessed in a randomized study.
Collapse
Affiliation(s)
- Kalyan Nadiminti
- Division of Hematology/Oncology; Holden Comprehensive Cancer Center
| | - Kamal Kant Singh Abbi
- Holden Comprehensive Cancer Center; Department of Internal Medicine, Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | | | | | | | | - Sonya Behrends
- Holden Comprehensive Cancer Center; Department of Internal Medicine, Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Fenghuang Zhan
- Holden Comprehensive Cancer Center; Department of Internal Medicine, Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Guido Tricot
- Holden Comprehensive Cancer Center; Department of Internal Medicine, Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
43
|
Coudre C, Alani J, Ritchie W, Marsaud V, Sola B, Cahu J. HIF-1α and rapamycin act as gerosuppressant in multiple myeloma cells upon genotoxic stress. Cell Cycle 2016; 15:2174-2182. [PMID: 27340936 PMCID: PMC4993538 DOI: 10.1080/15384101.2016.1196302] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (MM) is still an incurable hematological malignancy. Despite recent progress due to new anti-myeloma agents, the pathology is characterized by a high frequency of de novo or acquired resistance. Delineating the mechanisms of MM resistance is essential for therapeutic advances. We previously showed that long-term genotoxic stress induces the establishment of a senescence-associated secretory phenotype, a pro-inflammatory response that favors the emergence of cells with cancer stem-like properties. Here, we studied the short-term response of MM cells following treatment with various DNA damaging agents such as the energetic C-ion irradiation. MM cells are highly resistant to all treatments and do not enter apoptosis after they arrest cycling at the G2 phase. Although the DNA damage response pathway was activated, DNA breaks remained chronically in damaged MM cells. We found, using a transcriptomic approach that RAD50, a major DNA repair gene was downregulated early after genotoxic stress. In two gerosuppression situations: induction of hypoxia and inhibition of the mammalian target of rapamycin (mTOR) pathway, we observed, after the treatment with a DNA damaging agent, a normalization of RAD50 expression concomitant with the absence of cell cycle arrest. We propose that combining inhibitors of mTOR with genotoxic agents could avoid MM cells to senesce and secrete pro-inflammatory factors responsible for cancer stem-like cell emergence and, in turn, relapse of MM patients.
Collapse
Affiliation(s)
| | - Julien Alani
- Normandie Univ, UNICAEN, EA4652, MICAH team, Caen, France
| | - William Ritchie
- Centenary Institute, University of Sydney, Sydney, Australia
| | | | - Brigitte Sola
- Normandie Univ, UNICAEN, EA4652, MICAH team, Caen, France
| | - Julie Cahu
- Normandie Univ, UNICAEN, EA4652, MICAH team, Caen, France
| |
Collapse
|
44
|
MMSET/WHSC1 enhances DNA damage repair leading to an increase in resistance to chemotherapeutic agents. Oncogene 2016; 35:5905-5915. [PMID: 27109101 PMCID: PMC6071667 DOI: 10.1038/onc.2016.116] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 02/22/2016] [Accepted: 02/29/2016] [Indexed: 12/21/2022]
Abstract
MMSET/WHSC1 is a histone methyltransferase (HMT) overexpressed in t(4;14)+ multiple myeloma (MM) patients, believed to be the driving factor in the pathogenesis of this MM subtype. MMSET overexpression in MM leads to an increase in histone 3 lysine 36 dimethylation (H3K36me2), and a decrease in histone 3 lysine 27 trimethylation (H3K27me3), as well as changes in proliferation, gene expression, and chromatin accessibility. Prior work linked methylation of histones to the ability of cells to undergo DNA damage repair. In addition, t(4;14)+ patients frequently relapse after regimens that include DNA damage-inducing agents, suggesting that MMSET may play a role in DNA damage repair and response. In U2OS cells, we found that MMSET is required for efficient non-homologous end joining as well as homologous recombination. Loss of MMSET led to loss of expression of several DNA repair proteins, as well as decreased recruitment of DNA repair proteins to sites of DNA double strand breaks (DSBs). Using genetically matched MM cell lines that had either high (pathological) or low (physiological) expression of MMSET, we found that MMSET high cells had increased damage at baseline. Upon addition of a DNA damaging agent, MMSET high cells repaired DNA damage at an enhanced rate and continued to proliferate, whereas MMSET low cells accumulated DNA damage and entered cell cycle arrest. In a murine xenograft model using t(4;14)+ KMS11 MM cells harboring an inducible MMSET shRNA, depletion of MMSET enhanced the efficacy of chemotherapy, inhibiting tumor growth and extending survival. These findings help explain the poorer prognosis of t(4;14) MM and further validate MMSET as a potential therapeutic target in MM and other cancers.
Collapse
|
45
|
Lun X, Wells JC, Grinshtein N, King JC, Hao X, Dang NH, Wang X, Aman A, Uehling D, Datti A, Wrana JL, Easaw JC, Luchman A, Weiss S, Cairncross JG, Kaplan DR, Robbins SM, Senger DL. Disulfiram when Combined with Copper Enhances the Therapeutic Effects of Temozolomide for the Treatment of Glioblastoma. Clin Cancer Res 2016; 22:3860-75. [PMID: 27006494 DOI: 10.1158/1078-0432.ccr-15-1798] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 03/11/2016] [Indexed: 12/16/2022]
Abstract
PURPOSE Glioblastoma is one of the most lethal cancers in humans, and with existing therapy, survival remains at 14.6 months. Current barriers to successful treatment include their infiltrative behavior, extensive tumor heterogeneity, and the presence of a stem-like population of cells, termed brain tumor-initiating cells (BTIC) that confer resistance to conventional therapies. EXPERIMENTAL DESIGN To develop therapeutic strategies that target BTICs, we focused on a repurposing approach that explored already-marketed (clinically approved) drugs for therapeutic potential against patient-derived BTICs that encompass the genetic and phenotypic heterogeneity of glioblastoma observed clinically. RESULTS Using a high-throughput in vitro drug screen, we found that montelukast, clioquinol, and disulfiram (DSF) were cytotoxic against a large panel of patient-derived BTICs. Of these compounds, disulfiram, an off-patent drug previously used to treat alcoholism, in the presence of a copper supplement, showed low nanomolar efficacy in BTICs including those resistant to temozolomide and the highly infiltrative quiescent stem-like population. Low dose DSF-Cu significantly augmented temozolomide activity in vitro, and importantly, prolonged in vivo survival in patient-derived BTIC models established from both newly diagnosed and recurrent tumors. Moreover, we found that in addition to acting as a potent proteasome inhibitor, DSF-Cu functionally impairs DNA repair pathways and enhances the effects of DNA alkylating agents and radiation. These observations suggest that DSF-Cu inhibits proteasome activity and augments the therapeutic effects of DNA-damaging agents (temozolomide and radiation). CONCLUSIONS DSF-Cu should be considered as an adjuvant therapy for the treatment of patients with glioblastoma in both newly diagnosed and recurrent settings. Clin Cancer Res; 22(15); 3860-75. ©2016 AACR.
Collapse
Affiliation(s)
- Xueqing Lun
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada. Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - J Connor Wells
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada
| | - Natalie Grinshtein
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jennifer C King
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada. Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - Xiaoguang Hao
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada. Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - Ngoc-Ha Dang
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada. Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - Xiuling Wang
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada. Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada
| | - Ahmed Aman
- Drug Discovery Platform, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - David Uehling
- Drug Discovery Platform, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Alessandro Datti
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital Toronto, Ontario, Canada. Department of Agricultural, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Jeffrey L Wrana
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital Toronto, Ontario, Canada. Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Jacob C Easaw
- Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada. Department of Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Artee Luchman
- Department of Cell Biology & Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - Samuel Weiss
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada. Department of Cell Biology & Anatomy, University of Calgary, Calgary, Alberta, Canada
| | - J Gregory Cairncross
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada. Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada. Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - David R Kaplan
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, Ontario, Canada. Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Stephen M Robbins
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada. Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada. Department of Oncology, University of Calgary, Calgary, Alberta, Canada.
| | - Donna L Senger
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada. Clark H. Smith Brain Tumour Centre, University of Calgary, Calgary, Alberta, Canada. Department of Oncology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
46
|
Cea M, Cagnetta A, Adamia S, Acharya C, Tai YT, Fulciniti M, Ohguchi H, Munshi A, Acharya P, Bhasin MK, Zhong L, Carrasco R, Monacelli F, Ballestrero A, Richardson P, Gobbi M, Lemoli RM, Munshi N, Hideshima T, Nencioni A, Chauhan D, Anderson KC. Evidence for a role of the histone deacetylase SIRT6 in DNA damage response of multiple myeloma cells. Blood 2016; 127:1138-50. [PMID: 26675349 PMCID: PMC4778164 DOI: 10.1182/blood-2015-06-649970] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 12/02/2015] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is characterized by a highly unstable genome, with aneuploidy observed in nearly all patients. The mechanism causing this karyotypic instability is largely unknown, but recent observations have correlated these abnormalities with dysfunctional DNA damage response. Here, we show that the NAD(+)-dependent deacetylase SIRT6 is highly expressed in MM cells, as an adaptive response to genomic stability, and that high SIRT6 levels are associated with adverse prognosis. Mechanistically, SIRT6 interacts with the transcription factor ELK1 and with the ERK signaling-related gene. By binding to their promoters and deacetylating H3K9 at these sites, SIRT6 downregulates the expression of mitogen-activated protein kinase (MAPK) pathway genes, MAPK signaling, and proliferation. In addition, inactivation of ERK2/p90RSK signaling triggered by high SIRT6 levels increases DNA repair via Chk1 and confers resistance to DNA damage. Using genetic and biochemical studies in vitro and in human MM xenograft models, we show that SIRT6 depletion both enhances proliferation and confers sensitization to DNA-damaging agents. Our findings therefore provide insights into the functional interplay between SIRT6 and DNA repair mechanisms, with implications for both tumorigenesis and the treatment of MM.
Collapse
Affiliation(s)
- Michele Cea
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Clinic of Hematology, Department of Internal Medicine, University of Genoa, Istituto di Ricovero e Cura a Carattere Scientifico Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Genoa, Italy
| | - Antonia Cagnetta
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Clinic of Hematology, Department of Internal Medicine, University of Genoa, Istituto di Ricovero e Cura a Carattere Scientifico Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Genoa, Italy
| | - Sophia Adamia
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Chirag Acharya
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Mariateresa Fulciniti
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Hiroto Ohguchi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Aditya Munshi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Prakrati Acharya
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Manoj K Bhasin
- Beth Israel Deaconess Medical Center Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA; and
| | - Lei Zhong
- The Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Ruben Carrasco
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Fiammetta Monacelli
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Istituto di Ricovero e Cura a Carattere Scientifico Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Genoa, Italy
| | - Alberto Ballestrero
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Istituto di Ricovero e Cura a Carattere Scientifico Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Genoa, Italy
| | - Paul Richardson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Marco Gobbi
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Istituto di Ricovero e Cura a Carattere Scientifico Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Genoa, Italy
| | - Roberto M Lemoli
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Istituto di Ricovero e Cura a Carattere Scientifico Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Genoa, Italy
| | - Nikhil Munshi
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Teru Hideshima
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Alessio Nencioni
- Clinic of Hematology, Department of Internal Medicine, University of Genoa, Istituto di Ricovero e Cura a Carattere Scientifico Scientifico Azienda Ospedaliera Universitaria San Martino-Istituto Scientifico Tumori, Genoa, Italy
| | - Dharminder Chauhan
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Kenneth C Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Center for Multiple Myeloma Research, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
47
|
Galgano A, Barinov A, Vasseur F, de Villartay JP, Rocha B. CD8 Memory Cells Develop Unique DNA Repair Mechanisms Favoring Productive Division. PLoS One 2015; 10:e0140849. [PMID: 26485718 PMCID: PMC4613136 DOI: 10.1371/journal.pone.0140849] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/01/2015] [Indexed: 11/19/2022] Open
Abstract
Immune responses are efficient because the rare antigen-specific naïve cells are able to proliferate extensively and accumulate upon antigen stimulation. Moreover, differentiation into memory cells actually increases T cell accumulation, indicating improved productive division in secondary immune responses. These properties raise an important paradox: how T cells may survive the DNA lesions necessarily induced during their extensive division without undergoing transformation. We here present the first data addressing the DNA damage responses (DDRs) of CD8 T cells in vivo during exponential expansion in primary and secondary responses in mice. We show that during exponential division CD8 T cells engage unique DDRs, which are not present in other exponentially dividing cells, in T lymphocytes after UV or X irradiation or in non-metastatic tumor cells. While in other cell types a single DDR pathway is affected, all DDR pathways and cell cycle checkpoints are affected in dividing CD8 T cells. All DDR pathways collapse in secondary responses in the absence of CD4 help. CD8 T cells are driven to compulsive suicidal divisions preventing the propagation of DNA lesions. In contrast, in the presence of CD4 help all the DDR pathways are up regulated, resembling those present in metastatic tumors. However, this up regulation is present only during the expansion phase; i.e., their dependence on antigen stimulation prevents CD8 transformation. These results explain how CD8 T cells maintain genome integrity in spite of their extensive division, and highlight the fundamental role of DDRs in the efficiency of CD8 immune responses.
Collapse
Affiliation(s)
- Alessia Galgano
- INSERM, U1020, CNRS, UMR 8253, Medical Faculty Paris Descartes, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Aleksandr Barinov
- INSERM, U1020, CNRS, UMR 8253, Medical Faculty Paris Descartes, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Florence Vasseur
- INSERM, U1020, CNRS, UMR 8253, Medical Faculty Paris Descartes, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | | | - Benedita Rocha
- INSERM, U1020, CNRS, UMR 8253, Medical Faculty Paris Descartes, Université Paris Descartes Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
48
|
Liang BY, Xiong M, Ji GB, Zhang EL, Zhang ZY, Dong KS, Chen XP, Huang ZY. Synergistic suppressive effect of PARP-1 inhibitor PJ34 and HDAC inhibitor SAHA on proliferation of liver cancer cells. JOURNAL OF HUAZHONG UNIVERSITY OF SCIENCE AND TECHNOLOGY. MEDICAL SCIENCES = HUA ZHONG KE JI DA XUE XUE BAO. YI XUE YING DE WEN BAN = HUAZHONG KEJI DAXUE XUEBAO. YIXUE YINGDEWEN BAN 2015; 35:535-540. [PMID: 26223923 DOI: 10.1007/s11596-015-1466-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/26/2015] [Indexed: 10/23/2022]
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors and histone deacetylase (HDAC) inhibitors have recently emerged as promising anticancer drugs. The aim of this study was to investigate the effect of combination treatment with the PARP inhibitor PJ34 and HDAC inhibitor SAHA on the proliferation of liver cancer cells. Cell proliferation and apoptosis were assessed in three human liver cancer cell lines (HepG2, Hep3B and HCC-LM3) treated with PJ34 (8 μmol/L) and SAHA (1 μmol/L), alone or combined, by Cell Counting Kit-8 assay and flow cytometry, respectively. The nude mice bearing subcutaneous HepG2 tumors were administered different groups of drugs (10 mg/kg PJ34, 25 mg/kg SAHA, 10 mg/kg PJ34+25 mg/kg SAHA), and the inhibition rates of tumor growth were compared between groups. The results showed that combined use of PJ34 and SAHA could synergistically inhibit the proliferation of liver cancer cell lines HepG2, Hep3B and HCC-LM3. The apoptosis rate of HepG2 cells treated with PJ34+SAHA was significantly higher than that of HepG2 cells treated with PJ34 or SAHA alone (P<0.05). In vivo, the tumor inhibition rates were 53.5%, 61.4% and 82.6% in PJ34, SAHA and PJ34+SAHA groups, respectively. The combined use of PJ34 and SAHA could significantly inhibit the xenograft tumor growth when compared with use of PJ34 or SAHA alone (P<0.05). It was led to conclude that PJ34 and SAHA can synergistically suppress the proliferation of liver cancer cells.
Collapse
Affiliation(s)
- Bin-Yong Liang
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Min Xiong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Gui-Bao Ji
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of General Surgery, Pu'ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Er-Lei Zhang
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zun-Yi Zhang
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ke-Shuai Dong
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Ping Chen
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhi-Yong Huang
- Research Laboratory and Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
49
|
NKG2D and DNAM-1 Ligands: Molecular Targets for NK Cell-Mediated Immunotherapeutic Intervention in Multiple Myeloma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:178698. [PMID: 26161387 PMCID: PMC4486747 DOI: 10.1155/2015/178698] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 03/26/2015] [Indexed: 01/23/2023]
Abstract
A pivotal strategy to improve NK cell-mediated antitumor activity involves the upregulation of activating ligands on tumor cells. Enhancement of NK cell-mediated recognition of multiple myeloma cells was reported by us and others showing increased surface expression of NKG2D and DNAM-1 ligands on tumor cells following treatment with a number of chemotherapeutic agents, such as genotoxic drugs or inhibitors of proteasome, histone deacetylases, GSK3, and HSP-90. These compounds have the capability to affect tumor survival but also to activate specific transduction pathways associated with the upregulation of different NK cell activating ligands on the tumor cells. Here, we will summarize and discuss the molecular pathways whereby these drugs can regulate the expression of NK cell activating ligands in multiple myeloma cells.
Collapse
|
50
|
Ahmed-Seghir S, Pouvelle C, Despras E, Cordonnier A, Sarasin A, Kannouche PL. Aberrant C-terminal domain of polymerase η targets the functional enzyme to the proteosomal degradation pathway. DNA Repair (Amst) 2015; 29:154-65. [PMID: 25766642 DOI: 10.1016/j.dnarep.2015.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 02/14/2015] [Accepted: 02/16/2015] [Indexed: 12/22/2022]
Abstract
Xeroderma pigmentosum variant (XP-V) is a rare genetic disease, characterized by sunlight sensitivity and predisposition to cutaneous malignancies. XP-V is caused by a deficiency in DNA polymerase eta (Polη) that plays a pivotal role in translesion synthesis by bypassing UV-induced pyrimidine dimers. Previously we identified a new Polη variant containing two missense mutations, one mutation within the bipartite NLS (T692A) and a second mutation on the stop codon (X714W) leading to a longer protein with an extra 8 amino acids (721 instead of 713 AA). First biochemical analysis revealed that this Polη missense variant was barely detectable by western blot. As this mutant is extremely unstable and is nearly undetectable, a definitive measure of its functional deficit in cells has not been explored. Here we report the molecular and cellular characterization of this missense variant. In cell free extracts, the extra 8 amino acids in the C-terminal of Polη(721) only slightly reduce the bypass efficiency through CPD lesions. In vivo, Polη(721) accumulates in replication factories and interacts with mUb-PCNA albeit at lower level than Polη(wt). XP-V cells overexpressing Polη(721) were only slightly UV-sensitive. Altogether, our data strongly suggest that Polη(721) is functional and that the patient displays a XP-V phenotype because the mutant protein is excessively unstable. We then investigated the molecular mechanisms involved in this excessive proteolysis. We showed that Polη(721) is degraded by the proteasome in an ubiquitin-dependent manner and that this proteolysis is independent of the E3 ligases, CRL4(cdt2) and Pirh2, reported to promote Polη degradation. We then demonstrated that the extra 8 amino acids of Polη(721) do not act as a degron but rather induce a conformational change of the Polη C-terminus exposing its bipartite NLS as well as a sequence close to its UBZ to the ubiquitin/proteasome system. Interestingly we showed that the clinically approved proteasome inhibitor, Bortezomib restores the levels of Polη(721) suggesting that this might be a therapeutic approach to preventing tumor development in certain XP-V patients harboring missense mutations.
Collapse
Affiliation(s)
- Sana Ahmed-Seghir
- Université Paris-Sud, CNRS-UMR8200, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Caroline Pouvelle
- Université Paris-Sud, CNRS-UMR8200, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Emmanuelle Despras
- Université Paris-Sud, CNRS-UMR8200, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | | | - Alain Sarasin
- Université Paris-Sud, CNRS-UMR8200, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France
| | - Patricia L Kannouche
- Université Paris-Sud, CNRS-UMR8200, Equipe labellisée Ligue Contre le Cancer, Gustave Roussy, Villejuif, France.
| |
Collapse
|