1
|
Yamada M, Ikeda S, Kuroki W, Iwama S, Takahashi Y, Kitadate A, Tagawa H, Takahashi N. Comprehensive analysis of microRNAs modulated by histone deacetylase inhibitors identifies microRNA-7-5p with anti-myeloma effect. Int J Hematol 2024; 120:325-336. [PMID: 38954186 DOI: 10.1007/s12185-024-03812-1] [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: 02/08/2024] [Revised: 06/09/2024] [Accepted: 06/26/2024] [Indexed: 07/04/2024]
Abstract
Basic research to expand treatment options for multiple myeloma is greatly needed due to the refractory nature of the disease. Histone deacetylase (HDAC) inhibitors, which are epigenetic regulators, are attractive but have limited applications. MicroRNAs (miRNAs), which are also epigenetic regulators, are important molecules that may lead to future therapeutic breakthroughs. In this study, we comprehensively searched for miRNAs that are altered by HDAC inhibitors in myeloma cells. We identified miR-7-5p (miR-7) as a miRNA downregulated by HDAC inhibitors. Transfection of myeloma cell lines with miR-7 suppressed cell proliferation, induced apoptosis, and enhanced the effects of the HDAC inhibitor panobinostat. Expression of miR-7 was downregulated by c-Myc inhibition, but upregulated by bortezomib. Comprehensive examination of miR-7 targets revealed four candidates: SLC6A9, LRRC59, EXOSC2, and PSME3. Among these, we focused on PSME3, an oncogene involved in proteasome capacity in myeloma cells. PSME3 knockdown increases myeloma cell death and panobinostat sensitivity. In conclusion, miR-7, which is downregulated by HDAC inhibitors, is a tumor suppressor that targets PSME3. This miR-7 downregulation may be involved in HDAC inhibitor resistance. In addition, combinations of anti-myeloma drugs that complement changes in miRNA expression should be considered.
Collapse
Affiliation(s)
- Masahiro Yamada
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita, 0108543, Japan
| | - Sho Ikeda
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita, 0108543, Japan.
| | - Wataru Kuroki
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita, 0108543, Japan
| | - Sayaka Iwama
- Department of Life Science, Graduate School of Engineering Science, Akita University, Akita, Japan
| | - Yuto Takahashi
- Department of Life Science, Graduate School of Engineering Science, Akita University, Akita, Japan
| | - Akihiro Kitadate
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita, 0108543, Japan
| | - Hiroyuki Tagawa
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita, 0108543, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, Akita, 0108543, Japan
| |
Collapse
|
2
|
Maclachlan KH, Gitareja K, Kang J, Cuddihy A, Cao Y, Hein N, Cullinane C, Ang CS, Brajanovski N, Pearson RB, Khot A, Sanij E, Hannan RD, Poortinga G, Harrison SJ. Targeting the ribosome to treat multiple myeloma. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200771. [PMID: 38596309 PMCID: PMC10905045 DOI: 10.1016/j.omton.2024.200771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 04/11/2024]
Abstract
The high rates of protein synthesis and processing render multiple myeloma (MM) cells vulnerable to perturbations in protein homeostasis. The induction of proteotoxic stress by targeting protein degradation with proteasome inhibitors (PIs) has revolutionized the treatment of MM. However, resistance to PIs is inevitable and represents an ongoing clinical challenge. Our first-in-human study of the selective inhibitor of RNA polymerase I transcription of ribosomal RNA genes, CX-5461, has demonstrated a potential signal for anti-tumor activity in three of six heavily pre-treated MM patients. Here, we show that CX-5461 has potent anti-myeloma activity in PI-resistant MM preclinical models in vitro and in vivo. In addition to inhibiting ribosome biogenesis, CX-5461 causes topoisomerase II trapping and replication-dependent DNA damage, leading to G2/M cell-cycle arrest and apoptotic cell death. Combining CX-5461 with PI does not further enhance the anti-myeloma activity of CX-5461 in vivo. In contrast, CX-5461 shows synergistic interaction with the histone deacetylase inhibitor panobinostat in both the Vk∗MYC and the 5T33-KaLwRij mouse models of MM by targeting ribosome biogenesis and protein synthesis through distinct mechanisms. Our findings thus provide strong evidence to facilitate the clinical development of targeting the ribosome to treat relapsed and refractory MM.
Collapse
Affiliation(s)
- Kylee H. Maclachlan
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Clinical Hematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Kezia Gitareja
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine- St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Jian Kang
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine- St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Cuddihy
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Yuxi Cao
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Clinical Hematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Nadine Hein
- The ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Carleen Cullinane
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Ching-Seng Ang
- The Bio21 Institute of Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Natalie Brajanovski
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Richard B. Pearson
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Amit Khot
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Clinical Hematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Elaine Sanij
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- St Vincent’s Institute of Medical Research, Melbourne, VIC, Australia
- Department of Medicine- St Vincent’s Hospital, University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Ross D. Hannan
- The ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Gretchen Poortinga
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Simon J. Harrison
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Clinical Hematology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Ferro A, Pantazaka E, Athanassopoulos CM, Cuendet M. Histone deacetylase-based dual targeted inhibition in multiple myeloma. Med Res Rev 2023; 43:2177-2236. [PMID: 37191917 DOI: 10.1002/med.21972] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/08/2023] [Accepted: 04/30/2023] [Indexed: 05/17/2023]
Abstract
Despite enormous advances in terms of therapeutic strategies, multiple myeloma (MM) still remains an incurable disease with MM patients often becoming resistant to standard treatments. To date, multiple combined and targeted therapies have proven to be more beneficial compared to monotherapy approaches, leading to a decrease in drug resistance and an improvement in median overall survival in patients. Moreover, recent breakthroughs highlighted the relevant role of histone deacetylases (HDACs) in cancer treatment, including MM. Thus, the simultaneous use of HDAC inhibitors with other conventional regimens, such as proteasome inhibitors, is of interest in the field. In this review, we provide a general overview of HDAC-based combination treatments in MM, through a critical presentation of publications from the past few decades related to in vitro and in vivo studies, as well as clinical trials. Furthermore, we discuss the recent introduction of dual-inhibitor entities that could have the same beneficial effects as drug combinations with the advantage of having two or more pharmacophores in one molecular structure. These findings could represent a starting-point for both reducing therapeutic doses and lowering the risk of developing drug resistance.
Collapse
Affiliation(s)
- Angelica Ferro
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Evangelia Pantazaka
- Synthetic Organic Chemistry Laboratory, Department of Chemistry, University of Patras, Patras, Greece
- Laboratory of Biochemistry/Metastatic Signaling, Section of Genetics, Cell Biology, and Development, Department of Biology, University of Patras, Patras, Greece
| | | | - Muriel Cuendet
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| |
Collapse
|
4
|
Tang S, Zhang F, Li J, Dong H, Yang Q, Liu J, Fu Y. The selective activator protein-1 inhibitor T-5224 regulates the IRF4/MYC axis and exerts cooperative antimyeloma activity with bortezomib. Chem Biol Interact 2023; 384:110687. [PMID: 37657595 DOI: 10.1016/j.cbi.2023.110687] [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: 04/19/2023] [Revised: 08/13/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023]
Abstract
The activating protein-1 (AP-1) transcription factors (TFs) have been associated with many different cancer types and are promising therapeutic targets in logical malignancies. However, the mechanisms of their role in multiple myeloma (MM) remain elusive. The present study determined and compared the mRNA and protein expression levels of the AP-1 family member JunB in CD138+ mononuclear cells from MM patients and healthy donors. Herein, we investigated the effect of T-5224, an inhibitor of JUN/AP-1, on MM. We found that the cytotoxicity of T-5224 toward myeloma is due to its ability to induce cell apoptosis, inhibit proliferation, and induce cell cycle arrest by increasing the levels of cleaved caspase3/7 and concomitantly inhibiting the IRF4/MYC axis. We also noticed that siJunB-mediated deletion of JunB/AP-1 enhanced MM cell apoptosis and affected cell proliferation. The software PROMO was used in the present study to predict the AP-1 TF that may bind the promoter region of IRF4. We confirmed the correlation between JunB/AP-1 and IRF4. Given that bortezomib (BTZ) facilitates IRF4 degradation in MM cells, we applied combination treatment of BTZ with T-5224. T-5224 and BTZ exerted synergistic effects, and T-5224 reversed the effect of BTZ on CD138+ primary resistance in MM cells, in part due to suppression of the IRF4/MYC axis. Our results suggest that targeting AP-1 TFs is a promising therapeutic strategy for MM. Additionally, targeting both AP-1 and IRF4 with T-5224 may be a synergistic therapeutic strategy for this clinically challenging subset of MM.
Collapse
Affiliation(s)
- Sishi Tang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Fangrong Zhang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Hang Dong
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, 410013, China
| | - Qin Yang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jing Liu
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Yunfeng Fu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| |
Collapse
|
5
|
Tang TF, Chan YT, Cheong HC, Cheok YY, Anuar NA, Looi CY, Gan GG, Wong WF. Regulatory network of BLIMP1, IRF4, and XBP1 triad in plasmacytic differentiation and multiple myeloma pathogenesis. Cell Immunol 2022; 380:104594. [PMID: 36081178 DOI: 10.1016/j.cellimm.2022.104594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/27/2022]
Abstract
Antibody secreting plasma cell plays an indispensable role in humoral immunity. As activated B cell undergoes germinal center reaction and develops into plasma cell, it gradually loses B cell characteristics and embraces functional changes associated with immunoglobulins production. Differentiation of B cell into plasma cell involves drastic changes in cell structure, granularity, metabolism, gene expression and epigenetic regulation that couple with the mounting capacity for synthesis of a large quantity of antigen-specific antibodies. The interplay between three hallmark transcriptional regulators IRF4, BLIMP1, and XBP1, is critical for supporting the cellular reprograming activities during B to plasma cell transition. IRF4 promotes plasma cell generation by directing immunoglobulin class switching, proliferation and survival; BLIMP1 serves as a transcriptional repressor that extinguishes B cell features; whereas XBP1 controls unfolded protein response that relieves endoplasmic reticulum stress and permits antibody release during terminal differentiation. Intriguingly, high expression of IRF4, BLIMP1, and XBP1 molecules have been reported in myeloma cells derived from multiple myeloma patients, which negatively impact treatment outcome, prognosis, and relapse frequency. Despite the introduction of immunomodulatory drugs in recent years, multiple myeloma is still an incurable disease with poor survival rate. An in-depth review of IRF4, BLIMP1, and XBP1 triad molecules in plasma cell generation and multiple myeloma tumorigenesis may provide clues to the possibility of targeting these molecules in disease management.
Collapse
Affiliation(s)
- Ting Fang Tang
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yee Teng Chan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Heng Choon Cheong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yi Ying Cheok
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Nur Adila Anuar
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- School of Bioscience, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Gin Gin Gan
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
6
|
Liu M, Liu N, Wang J, Fu S, Wang X, Chen D. Acetyl-CoA Synthetase 2 as a Therapeutic Target in Tumor Metabolism. Cancers (Basel) 2022; 14:cancers14122896. [PMID: 35740562 PMCID: PMC9221533 DOI: 10.3390/cancers14122896] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/04/2022] [Accepted: 06/08/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Acetyl-CoA Synthetase 2 (ACSS2) is highly expressed in a variety of tumors, which is very important for tumor growth, proliferation, invasion, and metastasis in the nutritional stress microenvironment. Studies have proven that ACSS2 inhibitors can be effective in halting cancer growth and can be combined with other antineoplastic drugs to reduce drug resistance. This article mainly reviews the mechanism of ACSS2-promoting tumor growth from many aspects and the prospect of clinical application of targeted inhibitors. Abstract Acetyl-CoA Synthetase 2 (ACSS2) belongs to a member of the acyl-CoA short-chain synthase family, which can convert acetate in the cytoplasm and nucleus into acetyl-CoA. It has been proven that ACSS2 is highly expressed in glioblastoma, breast cancer, liver cancer, prostate cancer, bladder cancer, renal cancer, and other tumors, and is closely related to tumor stage and the overall survival rate of patients. Accumulating studies show that hypoxia and a low serum level induce ACSS2 expression to help tumor cells cope with this nutrient-poor environment. The potential mechanisms are associated with the ability of ACSS2 to promote the synthesis of lipids in the cytoplasm, induce the acetylation of histones in the nucleus, and facilitate the expression of autophagy genes. Novel-specific inhibitors of ACSS2 are developed and confirmed to the effectiveness in pre-clinical tumor models. Targeting ACSS2 may provide novel approaches for tumor treatment. This review summarizes the biological function of ACSS2, its relation to survival and prognosis in different tumors, and how ACSS2 mediates different pathways to promote tumor metastasis, invasion, and drug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Xu Wang
- Correspondence: (X.W.); (D.C.)
| | | |
Collapse
|
7
|
Aksenova AY, Zhuk AS, Lada AG, Zotova IV, Stepchenkova EI, Kostroma II, Gritsaev SV, Pavlov YI. Genome Instability in Multiple Myeloma: Facts and Factors. Cancers (Basel) 2021; 13:5949. [PMID: 34885058 PMCID: PMC8656811 DOI: 10.3390/cancers13235949] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a malignant neoplasm of terminally differentiated immunoglobulin-producing B lymphocytes called plasma cells. MM is the second most common hematologic malignancy, and it poses a heavy economic and social burden because it remains incurable and confers a profound disability to patients. Despite current progress in MM treatment, the disease invariably recurs, even after the transplantation of autologous hematopoietic stem cells (ASCT). Biological processes leading to a pathological myeloma clone and the mechanisms of further evolution of the disease are far from complete understanding. Genetically, MM is a complex disease that demonstrates a high level of heterogeneity. Myeloma genomes carry numerous genetic changes, including structural genome variations and chromosomal gains and losses, and these changes occur in combinations with point mutations affecting various cellular pathways, including genome maintenance. MM genome instability in its extreme is manifested in mutation kataegis and complex genomic rearrangements: chromothripsis, templated insertions, and chromoplexy. Chemotherapeutic agents used to treat MM add another level of complexity because many of them exacerbate genome instability. Genome abnormalities are driver events and deciphering their mechanisms will help understand the causes of MM and play a pivotal role in developing new therapies.
Collapse
Affiliation(s)
- Anna Y. Aksenova
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anna S. Zhuk
- International Laboratory “Computer Technologies”, ITMO University, 197101 St. Petersburg, Russia;
| | - Artem G. Lada
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA;
| | - Irina V. Zotova
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (I.V.Z.); (E.I.S.)
- Vavilov Institute of General Genetics, St. Petersburg Branch, Russian Academy of Sciences, 199034 St. Petersburg, Russia
| | - Elena I. Stepchenkova
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (I.V.Z.); (E.I.S.)
- Vavilov Institute of General Genetics, St. Petersburg Branch, Russian Academy of Sciences, 199034 St. Petersburg, Russia
| | - Ivan I. Kostroma
- Russian Research Institute of Hematology and Transfusiology, 191024 St. Petersburg, Russia; (I.I.K.); (S.V.G.)
| | - Sergey V. Gritsaev
- Russian Research Institute of Hematology and Transfusiology, 191024 St. Petersburg, Russia; (I.I.K.); (S.V.G.)
| | - Youri I. Pavlov
- Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Departments of Biochemistry and Molecular Biology, Microbiology and Pathology, Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
8
|
Martínez-Martín S, Soucek L. MYC inhibitors in multiple myeloma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:842-865. [PMID: 35582389 PMCID: PMC8992455 DOI: 10.20517/cdr.2021.55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/30/2021] [Accepted: 08/12/2021] [Indexed: 11/25/2022]
Abstract
The importance of MYC function in cancer was discovered in the late 1970s when the sequence of the avian retrovirus that causes myelocytic leukemia was identified. Since then, over 40 years of unceasing research have highlighted the significance of this protein in malignant transformation, especially in hematologic diseases. Indeed, some of the earliest connections among the higher expression of proto-oncogenes (such as MYC), genetic rearrangements and their relation to cancer development were made in Burkitt lymphoma, chronic myeloid leukemia and mouse plasmacytomas. Multiple myeloma (MM), in particular, is a plasma cell malignancy strictly associated with MYC deregulation, suggesting that therapeutic strategies against it would be beneficial in treating this disease. However, targeting MYC was - and, somehow, still is - challenging due to its unique properties: lack of defined three-dimensional structure, nuclear localization and absence of a targetable enzymatic pocket. Despite these difficulties, however, many studies have shown the potential therapeutic impact of direct or indirect MYC inhibition. Different molecules have been tested, in fact, in the context of MM. In this review, we summarize the current status of the different compounds, including the results of their clinical testing, and propose to continue with the efforts to identify, repurpose, redesign or improve drug candidates to combine them with standard of care therapies to overcome resistance and enable better management of myeloma treatment.
Collapse
Affiliation(s)
- Sandra Martínez-Martín
- Preclinical & Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona 08035, Spain
- Peptomyc S.L., Vall d’Hebron Barcelona Hospital Campus, Barcelona 08035, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Laura Soucek
- Preclinical & Translational Research Program, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona 08035, Spain
- Peptomyc S.L., Vall d’Hebron Barcelona Hospital Campus, Barcelona 08035, Spain
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona 08010, Spain
| |
Collapse
|
9
|
Nahi H, Kashif M, Klimkowska M, Karvouni M, Wallblom A, Gran C, Hauenstein J, Frengen N, Gustafsson C, Afram G, Uttervall K, Lund J, Månsson R, Wagner AK, Alici E. Low dose venetoclax as a single agent treatment of plasma cell malignancies harboring t(11;14). Am J Hematol 2021; 96:925-933. [PMID: 33901326 DOI: 10.1002/ajh.26207] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Approximately 20% of newly diagnosed multiple myeloma (NDMM) patients harbor t(11;14), a marker of inferior prognosis, resulting in up-regulation of CCND1. These patients respond to BCL2 inhibitor experimental drug venetoclax. Furthermore, t(11;14) is reported to be associated with increased BCL2/MCL1 ratio. We investigated the use of venetoclax (400 mg daily) in a cohort of 25 multiple myeloma (MM) and AL-amyloidosis patients harboring t(11;14) and assessed safety and efficacy. Efficacy was assessed by response rate (RR) and time on treatment. Furthermore, immunohistochemistry (IHC), for BCL2 family member expression was assessed at diagnosis and relapse in the venetoclax-treated group and analyzed for correlation with clinical RR. Additionally, patient material from venetoclax non-treated group including non-t(11;14) diagnosis (n = 27), t(11;14) diagnosis (n = 17), t(11;14) relapse (n = 7), hyperdiploidy (n = 6) and hyperdiploidy + t(11;14) (n = 6) was used for RNA sequencing (RNASeq) and validation by qPCR. Venetoclax treatment in t(11;14) patients demonstrated manageable safety and promising efficacy. Partial responses or better were observed in eleven patients (44%). Responding patients had significantly higher BCL2/MCL1 (p = 0.031) as well as BCL2/BCL-XL (p = 0.021) ratio, regardless of time of measurement before venetoclax treatment. Furthermore, an IRF5 motif was enriched (p < .001) in the downregulated genes in t(11;14) relapses vs diagnoses. The RR with single agent venetoclax was 71% in AL-amyloidosis and 33% in MM, and IHC proved useful in prediction of treatment outcome. We could also demonstrate possible resistance mechanisms of t(11;14), downregulation of IRF5 targeted genes, which can be exploited for therapeutic advantages.
Collapse
Affiliation(s)
- Hareth Nahi
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Muhammad Kashif
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
| | - Monika Klimkowska
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
- Department of Clinical Pathology and Cytology Karolinska University Hospital Stockholm Sweden
| | - Maria Karvouni
- Department of Medicine Karolinska Institutet Stockholm Sweden
| | - Ann Wallblom
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
| | - Charlotte Gran
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Clinical Chemistry Karolinska University Hospital Stockholm Sweden
| | - Julia Hauenstein
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Nicolai Frengen
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Charlotte Gustafsson
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | - Gabriel Afram
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Katarina Uttervall
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Johan Lund
- Department of Medicine Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
| | - Robert Månsson
- Center for Hematology and Regenerative Medicine (HERM) Karolinska Institutet Stockholm Sweden
- Department of Hematology Karolinska University Hospital Stockholm Sweden
- Department of Laboratory Medicine Karolinska Institutet Stockholm Sweden
| | | | - Evren Alici
- Department of Medicine Karolinska Institutet Stockholm Sweden
| |
Collapse
|
10
|
Jiang D, Zhang K, Zhu Y, Zhu Y, Zou L, Hu J, Cui Y, Zhou W, Chen F, He Y. Chidamide-Induced Accumulation of Reactive Oxygen Species Increases Lenalidomide Sensitivity Against Multiple Myeloma Cells. Onco Targets Ther 2021; 14:4061-4075. [PMID: 34262292 PMCID: PMC8274322 DOI: 10.2147/ott.s312249] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background Lenalidomide, an immunomodulatory drug (IMiD), is an effective therapy for the treatment of multiple myeloma (MM). However, prolonged treatment may be accompanied by toxicity, second primary malignancies, and drug resistance. There is an inherent vulnerability in MM cells that high rates of immunoglobulin synthesis resulting in the high level of reactive oxygen species (ROS). This provides a therapeutic potential for MM. Materials and Methods The intracellular ROS levels, H2O2 production and glutathione (GSH) levels were measured using detection kit. Cell viability was evaluated using cell-counting kit-8 (CCK-8) and soft agar colony formation assay. Apoptosis was determined in whole living cells using flow cytometry. Chidamide and its anti-myeloma efficacy in combination with lenalidomide were characterized in MM cell lines in vitro and in a mouse xenograft model. Moreover, Western blotting, immunofluorescence and immunohistochemical studies were performed. Results ROS levels increased in a time- and dose-dependent manner with chidamide treatment. Moreover, the GSH levels were decreased and the mRNA level of SLC7A11 downregulated after chidamide treatment. The co-treatment with chidamide and lenalidomide increased apoptosis and proliferation inhibition, with combination index (CI) in the synergistic range (0.2–0.5) using the Chou–Talalay method. The cooperative anti-myeloma efficacy was confirmed in the murine model, and immunohistochemical studies also supported this potentiation. Chidamide enhanced the effect of lenalidomide-induced degradation of IKZF1 and IKZF3 by elevating H2O2. In addition, co-treatment with chidamide and lenalidomide increased biomarkers of caspase and DNA damage. Conclusion Elevated ROS production may constitute a potential biochemical basis for anti-myeloma effects of chidamide plus lenalidomide. The results of this study confirm the synergistic effect of chidamide and lenalidomide against MM and provide a promising therapeutic strategy for MM.
Collapse
Affiliation(s)
- Duanfeng Jiang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, People's Republic of China
| | - Kaixuan Zhang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Yinghong Zhu
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, People's Republic of China
| | - Yan Zhu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Lang Zou
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Jian Hu
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Yajuan Cui
- Department of Hematology, the Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Wen Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Central South University, Changsha, 410078, People's Republic of China
| | - Fangping Chen
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha, 410013, People's Republic of China.,Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Yanjuan He
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| |
Collapse
|
11
|
Berdeja JG, Laubach JP, Richter J, Stricker S, Spencer A, Richardson PG, Chari A. Panobinostat From Bench to Bedside: Rethinking the Treatment Paradigm for Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:752-765. [PMID: 34340951 DOI: 10.1016/j.clml.2021.06.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/07/2021] [Accepted: 06/22/2021] [Indexed: 12/31/2022]
Abstract
Relapsed and refractory multiple myeloma (RRMM) presents a therapeutic challenge due to the development of drug resistance. Panobinostat is an oral histone deacetylase inhibitor (HDACi) that affects multiple cellular pathways and has demonstrated the ability to resensitize refractory-multiple myeloma cells in preclinical studies, as well as in patients with RRMM in clinical trials. Synergy of panobinostat with a number of different classes of antimyeloma drugs (proteasome inhibitors, immunomodulatory drugs and monoclonal antibodies) has also been shown. Panobinostat is a promising HDACi for the treatment of multiple myeloma. Here, we present a comprehensive review of preclinical and clinical studies of panobinostat.
Collapse
Affiliation(s)
- Jesus G Berdeja
- Sarah Cannon Research Institute, Nashville, TN; Tennessee Oncology PLLC, Nashville, TN
| | - Jacob P Laubach
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Joshua Richter
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY
| | | | - Andrew Spencer
- Alfred Hospital - Monash University, Melbourne, Australia
| | | | - Ajai Chari
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY.
| |
Collapse
|
12
|
Teramachi J, Tenshin H, Hiasa M, Oda A, Bat-Erdene A, Harada T, Nakamura S, Ashtar M, Shimizu S, Iwasa M, Sogabe K, Oura M, Fujii S, Kagawa K, Miki H, Endo I, Haneji T, Matsumoto T, Abe M. TAK1 is a pivotal therapeutic target for tumor progression and bone destruction in myeloma. Haematologica 2021; 106:1401-1413. [PMID: 32273474 PMCID: PMC8094086 DOI: 10.3324/haematol.2019.234476] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Indexed: 12/31/2022] Open
Abstract
Along with tumor progression, the bone marrow microenvironment is skewed in multiple myeloma (MM), which underlies the unique pathophysiology of MM and confers aggressiveness and drug resistance in MM cells. TGF-b-activated kinase-1 (TAK1) mediates a wide range of intracellular signaling pathways. We demonstrate here that TAK1 is constitutively overexpressed and phosphorylated in MM cells, and that TAK1 inhibition suppresses the activation of NF-κB, p38MAPK, ERK and STAT3 in order to decrease the expression of critical mediators for MM growth and survival, including PIM2, MYC, Mcl- 1, IRF4, and Sp1, along with a substantial reduction in the angiogenic factor VEGF in MM cells. Intriguingly, TAK1 phosphorylation was also induced along with upregulation of vascular cell adhesion molecule-1 (VCAM-1) in bone marrow stromal cells (BMSC) in cocultures with MM cells, which facilitated MM cell-BMSC adhesion while inducing IL-6 production and receptor activator of nuclear factor κ-B ligand (RANKL) expression by BMSC. TAK1 inhibition effectively impaired MM cell adhesion to BMSC to disrupt the support of MM cell growth and survival by BMSC. Furthermore, TAK1 inhibition suppressed osteoclastogenesis enhanced by RANKL in cocultures of bone marrow cells with MM cells, and restored osteoblastic differentiation suppressed by MM cells or inhibitory factors for osteoblastogenesis overproduced in MM. Finally, treatment with the TAK1 inhibitor LLZ1640-2 markedly suppressed MM tumor growth and prevented bone destruction and loss in mouse MM models. Therefore, TAK1 inhibition may be a promising therapeutic option targeting not only MM cells but also the skewed bone marrow microenvironment in MM.
Collapse
Affiliation(s)
- Jumpei Teramachi
- Dept. of Histology-Oral Histology and Dept. of Hematology, Tokushima University,Tokushima, Japan
| | - Hirofumi Tenshin
- Dept. of Hematology and Orthodontics and Dentofacial Orthopedics,Tokushima University, Japan
| | - Masahiro Hiasa
- Dept. of Hematology and Orthodontics and Dentofacial Orthopedics,Tokushima University, Japan
| | - Asuka Oda
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Ariunzaya Bat-Erdene
- Dept of Hematology, Tokushima University and University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Takeshi Harada
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Shingen Nakamura
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Mohannad Ashtar
- Dept. of Hematology and Orthodontics and Dentofacial Orthopedics,Tokushima University, Japan
| | - So Shimizu
- Dept. of Hematology and Orthodontics and Dentofacial Orthopedics,Tokushima University, Japan
| | - Masami Iwasa
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Kimiko Sogabe
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Masahiro Oura
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Shiro Fujii
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Kumiko Kagawa
- Department of Hematology, Tokushima University, Tokushima, Japan
| | - Hirokazu Miki
- Division of Transfusion Medicine and Cell Therapy, Tokushima University Hospital, Tokushima, Japan
| | - Itsuro Endo
- Department of Chronomedicine, Tokushima University, Tokushima, Japan
| | - Tatsuji Haneji
- Department of Histology and Oral Histology, Tokushima University, Tokushima, Japan
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Masahiro Abe
- Department of Hematology, Tokushima University, Tokushima, Japan
| |
Collapse
|
13
|
Dunphy K, Dowling P, Bazou D, O’Gorman P. Current Methods of Post-Translational Modification Analysis and Their Applications in Blood Cancers. Cancers (Basel) 2021; 13:1930. [PMID: 33923680 PMCID: PMC8072572 DOI: 10.3390/cancers13081930] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/04/2021] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Post-translational modifications (PTMs) add a layer of complexity to the proteome through the addition of biochemical moieties to specific residues of proteins, altering their structure, function and/or localization. Mass spectrometry (MS)-based techniques are at the forefront of PTM analysis due to their ability to detect large numbers of modified proteins with a high level of sensitivity and specificity. The low stoichiometry of modified peptides means fractionation and enrichment techniques are often performed prior to MS to improve detection yields. Immuno-based techniques remain popular, with improvements in the quality of commercially available modification-specific antibodies facilitating the detection of modified proteins with high affinity. PTM-focused studies on blood cancers have provided information on altered cellular processes, including cell signaling, apoptosis and transcriptional regulation, that contribute to the malignant phenotype. Furthermore, the mechanism of action of many blood cancer therapies, such as kinase inhibitors, involves inhibiting or modulating protein modifications. Continued optimization of protocols and techniques for PTM analysis in blood cancer will undoubtedly lead to novel insights into mechanisms of malignant transformation, proliferation, and survival, in addition to the identification of novel biomarkers and therapeutic targets. This review discusses techniques used for PTM analysis and their applications in blood cancer research.
Collapse
Affiliation(s)
- Katie Dunphy
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Paul Dowling
- Department of Biology, National University of Ireland, W23 F2K8 Maynooth, Ireland; (K.D.); (P.D.)
| | - Despina Bazou
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| | - Peter O’Gorman
- Department of Haematology, Mater Misericordiae University Hospital, D07 WKW8 Dublin, Ireland;
| |
Collapse
|
14
|
Laubach JP, Tuchman SA, Rosenblatt JM, Mitsiades CS, Colson K, Masone K, Warren D, Redd RA, Grayson D, Richardson PG. Phase 1 open-label study of panobinostat, lenalidomide, bortezomib + dexamethasone in relapsed and relapsed/refractory multiple myeloma. Blood Cancer J 2021; 11:20. [PMID: 33563894 PMCID: PMC7873303 DOI: 10.1038/s41408-021-00407-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/08/2020] [Accepted: 12/23/2020] [Indexed: 02/06/2023] Open
Abstract
Additional therapeutic options are needed for relapsed and refractory multiple myeloma (RRMM). We present data from a phase 1b, open-label, dose-escalation study (NCT01965353) of 20 patients with RRMM (median age: 63 years [range: 50–77]) and a median of four prior regimens (range: 2–14); 85% had refractory disease (lenalidomide [80%]; bortezomib [75%]; lenalidomide and bortezomib [50%]). Patients received a median of six cycles (range: 1–74) of panobinostat (10 or 15 mg), lenalidomide 15 mg, bortezomib 1 mg/m2, and dexamethasone 20 mg (pano-RVd). Median follow-up was ~14 months. Six dose-limiting toxicities were reported (mostly hematological); maximum tolerated dose of panobinostat (primary endpoint) was 10 mg. Most common adverse events (AEs) were diarrhea (60%) and peripheral neuropathy (60%); all grade 1/2. Grade 3/4 AEs occurred in 80% of patients and included decreased neutrophil (45%), platelet (25%) and white blood cell (25%) counts, anemia (25%) and hypophosphatemia (25%). No treatment-related discontinuations or mortality occurred. In evaluable patients (n = 18), overall response rate was 44%, and clinical benefit rate was 61%. Median duration of response was 9.2 months; progression-free survival was 7.4 months; overall survival was not reached. Pano-RVd proved generally well-tolerated and demonstrated potential to overcome lenalidomide and/or bortezomib resistance.
Collapse
Affiliation(s)
- Jacob P Laubach
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | | | | | | | - Kathleen Colson
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kelly Masone
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Diane Warren
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Robert A Redd
- Dana-Farber Cancer Institute, Department of Data Sciences, Boston, MA, USA
| | | | - Paul G Richardson
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Zhou Z, Ma D, Liu P, Wang P, Wei D, Yu K, Li P, Fang Q, Wang J. Deletion of HO-1 blocks development of B lymphocytes in mice. Cell Signal 2019; 63:109378. [PMID: 31369826 DOI: 10.1016/j.cellsig.2019.109378] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/25/2019] [Accepted: 07/27/2019] [Indexed: 12/29/2022]
Abstract
B lymphocytes, a key cluster of cells composing the immune system, can protect against abnormal biological factors. Heme oxygenase-1 (HO-1) plays important roles in cell proliferation and immune regulation, but its effects on the development and growth of B lymphocytes are still unknown. Herein, the count of B lymphocytes in HO-1 gene knockout (HO-1+/-) mice was significantly lower than that of the HO-1 gene wild-type (HO-1WT) mice. Meanwhile, the cell count of HO-1+/- mice did not recover after irradiation for one week, due to the G0/G1 phase arrest of Pro-B cells and the augmented apoptosis of Pre-B cells. Up-regulation of HO-1 by lentivirus attenuated the Pro-B cell cycle arrest and Pre-B cell apoptosis. To understand the molecular mechanism by which HO-1 knockout blocked B lymphocyte development, protein-to-protein interaction network and Western blot were used. The PI3K/AKT signaling pathway mediated the regulatory effects of HO-1 on B lymphocytes. In conclusion, HO-1 is a crucial transcriptional repressor for B cell development.
Collapse
Affiliation(s)
- Zhen Zhou
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Ping Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Ping Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Danna Wei
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Kunling Yu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Peifan Li
- Department of Psychiatry, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China.
| |
Collapse
|
16
|
Zhou Z, Fang Q, Li P, Ma D, Zhe N, Ren M, Chen B, He Z, Wang J, Zhong Q, Wang J. Entinostat combined with Fludarabine synergistically enhances the induction of apoptosis in TP53 mutated CLL cells via the HDAC1/HO-1 pathway. Life Sci 2019; 232:116583. [PMID: 31226417 DOI: 10.1016/j.lfs.2019.116583] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/28/2019] [Accepted: 06/17/2019] [Indexed: 02/02/2023]
Abstract
TP53 mutation is an indicator of poor prognostic in chronic lymphocytic leukemia (CLL). Worse still, CLL patients with TP53 mutation are associated with poor efficacy to current chemotherapeutic, such as Fludarabine. Here, we confirmed that high expression of HDAC1 in CLL patients with TP53 mutation, which is closely related to poor prognosis and drug-resistance. Subsequently, we demonstrated Entinostat (HDAC1 inhibitor) combination with Fludarabine significantly induced apoptosis in TP53 mutations CLL cells. Its mechanism was associated with up-regulation of the pro-apoptotic protein Bax and the down-regulation of HDAC1, HO-1 and BCL-2 proteins. More importantly, we also confirmed that upregulation of HDAC1 could resistant Entinostat-induced apoptosis in TP53 mutations CLL cells by activating the HDAC1/P38/HO-1 pathway. In vivo, we found that Entinostat combination with Fludarabine significantly induced tumor cells apoptosis and prolong survival time in xenograft mouse model. Finally, combining vitro and vivo experiments, we presented the first demonstration that Entinostat combination with Fludarabine had a synergistic effect on the induction of apoptosis in TP53 mutations CLL cells. In conclusion, we provide valuable pre-clinical experimental evidence for the treatment of CLL patients with poor prognosis, especially for TP53 mutations.
Collapse
Affiliation(s)
- Zhen Zhou
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China
| | - Qin Fang
- Department of Pharmacy, Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Pharmacy, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Peifan Li
- Clinical Research Centre, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Nana Zhe
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Mei Ren
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Bingqing Chen
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China
| | - Zhengchang He
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China
| | - Jun Wang
- Clinical Research Centre, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Qin Zhong
- Clinical Research Centre, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Key Laboratory of Hematological Disease Diagnostic and Treat Centre of Guizhou Province, Guiyang 550004, China; Department of Hematology, Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Center, Guiyang 550004, China.
| |
Collapse
|
17
|
He ZC, Li XY, Guo YL, Ma D, Fang Q, Ren LL, Zhang ZY, Wang W, Yu ZY, Zhao P, Wang JS. Heme oxygenase-1 attenuates the inhibitory effect of bortezomib against the APRIL-NF-κB-CCL3 signaling pathways in multiple myeloma cells: Corelated with bortezomib tolerance in multiple myeloma. J Cell Biochem 2019; 120:6972-6987. [PMID: 30368867 DOI: 10.1002/jcb.27879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/21/2018] [Indexed: 01/24/2023]
Abstract
Osteoclasts (OCs) play an essential role in bone destruction in patients with multiple myeloma (MM). Bortezomib can ameliorate bone destruction in patients with MM, but advanced MM often resists bortezomib. We studied the molecular mechanisms of bortezomib tolerance in MM. The expression of the MM-related genes in newly diagnosed patients with MM and normal donors were studied. C-C motif chemokine ligand 3 (CCL3) is a cytokine involved in the differentiation of OCs, and its expression is closely related to APRIL (a proliferation-inducing ligand). We found that bortezomib treatment inhibited APRIL and CCL3. But the heme oxygenase-1 (HO-1) activator hemin attenuated the inhibitory effects of bortezomib on APRIL and CCL3. We induced mononuclear cells to differentiate into OCs, and the enzyme-linked immunosorbent assay showed that the more OCs differentiated, the higher the levels CCL3 secretions detected. Animal experiments showed that hemin promoted MM cell infiltration in mice. The weight and survival rate of tumor mice were associated with HO-1 expression. Immunohistochemical staining showed that HO-1, APRIL, and CCL3 staining were positively stained in the tumor infiltrating sites. Then, MM cells were transfected with L-HO-1/si-HO-1 expression vectors and cultured with an nuclear factor (NF)-kappa B (κB) pathway inhibitor, QNZ. The results showed that HO-1 was the upstream gene of APRIL, NF-κB, and CCL3. We showed that HO-1 could attenuate the inhibitory effect of bortezomib against the APRIL-NF-κB-CCL3 signaling pathways in MM cells, and the tolerance of MM cells to bortezomib and the promotion of bone destruction are related to HO-1.
Collapse
Affiliation(s)
- Zheng C He
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xin Y Li
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yong L Guo
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Dan Ma
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Qin Fang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ling L Ren
- Department of Hematology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhao Y Zhang
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Weili Wang
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zheng Y Yu
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Peng Zhao
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ji S Wang
- Department of Hematology, Affiliated Hospital of Medical University, Guiyang, China.,Hematological Institute of Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Guizhou Provincial Laboratory of Hematopoietic Stem Cell Transplantation Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
18
|
Imai Y, Hirano M, Kobayashi M, Futami M, Tojo A. HDAC Inhibitors Exert Anti-Myeloma Effects through Multiple Modes of Action. Cancers (Basel) 2019; 11:cancers11040475. [PMID: 30987296 PMCID: PMC6520917 DOI: 10.3390/cancers11040475] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 11/29/2022] Open
Abstract
HDACs are critical regulators of gene expression that function through histone modification. Non-histone proteins and histones are targeted by these proteins and the inhibition of HDACs results in various biological effects. Moreover, the aberrant expression and function of these proteins is thought to be related to the pathogenesis of multiple myeloma (MM) and several inhibitors have been introduced or clinically tested. Panobinostat, a pan-HDAC inhibitor, in combination with a proteasome inhibitor and dexamethasone has improved survival in relapsing/refractory MM patients. We revealed that panobinostat inhibits MM cell growth by degrading the protein PPP3CA, a catalytic subunit of calcineurin. This degradation was suggested to be mediated by suppression of the chaperone function of HSP90 due to HDAC6 inhibition. Cytotoxicity due to the epigenetic regulation of tumor-associated genes by HDAC inhibitors has also been reported. In addition, HDAC6 inhibition enhances tumor immunity and has been suggested to strengthen the cytotoxic effects of therapeutic antibodies against myeloma. Furthermore, therapeutic strategies to enhance the anti-myeloma effects of HDAC inhibitors through the addition of other agents has been intensely evaluated. Thus, the treatment of patients with MM using HDAC inhibitors is promising as these drugs exert their effects through multiple modes of action.
Collapse
Affiliation(s)
- Yoichi Imai
- Department of Hematology/Oncology, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan.
| | - Mitsuhito Hirano
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan.
| | - Masayuki Kobayashi
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan.
| | - Muneyoshi Futami
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan.
| | - Arinobu Tojo
- Department of Hematology/Oncology, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan.
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan.
| |
Collapse
|
19
|
Meng X, Yuan Y, Shen F, Li C. Heme oxygenase-1 ameliorates hypoxia/reoxygenation via suppressing apoptosis and enhancing autophagy and cell proliferation though Sirt3 signaling pathway in H9c2 cells. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:189-198. [PMID: 30415272 DOI: 10.1007/s00210-018-1575-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022]
Abstract
Cardiomyocyte infarction could lead to high morbidity and mortality worldwide. Recent studies demonstrated that Heme oxygenase-1 (HO-1) could exert cardiac protective effect and arouse attention. However, the detailed mechanism is still unclear. Our study provided evidences of the protective effect of HO-1 overexpression on cardiomyocytes against hypoxia/reoxygenation (H/R). We divided the treatment into four groups: the control group, H/R group, H/R+HO-1 group, and H/R+Null group. Immunofluorescent study was utilized to label the BrdU-positive and LC3-positive cells. Flow cytometry and TUNEL assay were used to examine the cell apoptosis. Protein levels of Bax, Bcl-2, Sirt3, beclin-1, LC3-I, and LC3-II were both measured using western blotting. The results indicated that HO-1 overexpression decreased the cell apoptosis and enhanced the cell proliferation. The level of Sirt3 and autophagy were also increased in H/R+HO-1 group compared with H/R group. However, ZnPP, a HO-1 inhibitor, and SiRNA of Sirt3 are both reversed the decrease of cell apoptosis of HO-1 overexpression. Moreover, ZnPP also decreased the expression of Sirt3 in HO-1 overexpression treatment group. In summary, HO-1 overexpression protects cardiomyocytes against H/R injury via ameliorating cell apoptosis and enhancing cell proliferation and autophagy through Sirt3 signaling pathway.
Collapse
Affiliation(s)
- Xiangli Meng
- Emergency department, The Zoucheng People's Hospital, 59 Qianquan Road, Jining, Shandong, 273500, People's Republic of China
| | - Yuxiang Yuan
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, Jining, Shandong, 273500, People's Republic of China
| | - Fengjuan Shen
- Emergency department, The Zoucheng People's Hospital, 59 Qianquan Road, Jining, Shandong, 273500, People's Republic of China.
| | - Chengqiu Li
- Department of Cardiovascular Medicine, The Zoucheng People's Hospital, 59 Qianquan Road, Jining, Shandong, 273500, People's Republic of China.
| |
Collapse
|
20
|
Agnarelli A, Chevassut T, Mancini EJ. IRF4 in multiple myeloma—Biology, disease and therapeutic target. Leuk Res 2018; 72:52-58. [DOI: 10.1016/j.leukres.2018.07.025] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 01/29/2023]
|