1
|
Gildea DT, Roswarski JL. Severe Lenalidomide-Associated Hyperbilirubinemia. Cureus 2023; 15:e34408. [PMID: 36874759 PMCID: PMC9977541 DOI: 10.7759/cureus.34408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
Immunomodulatory drugs (IMids), such as thalidomide and lenalidomide, are used to treat plasma cell neoplasms and B-cell malignancies. We present a case of severe direct hyperbilirubinemia in a patient taking lenalidomide-based therapy for plasmacytoma. Imaging was unrevealing, and liver biopsy showed only mild sinusoidal dilation. Roussel Uclaf Causality Assessment (RUCAM) score was 6, indicating lenalidomide was a probable cause of the injury. To our knowledge, this is the highest reported direct bilirubin regarding lenalidomide drug-induced liver injury (DILI), with a peak bilirubin of 41mg/dL. While a clear pathophysiology was not identified, this case provides important considerations regarding lenalidomide safety.
Collapse
Affiliation(s)
- Daniel T Gildea
- Internal Medicine, MedStar Georgetown University Hospital, Washington, D.C., USA
| | - Joseph L Roswarski
- Hematology and Oncology, MedStar Georgetown University Hospital, Washington, D.C., USA
| |
Collapse
|
2
|
Gong Y, Yang C, Wei Z, Liu J. SRPK1 promotes cell proliferation and tumor growth of osteosarcoma through activation of the NF-κB signaling pathway. Biol Chem 2021; 403:653-663. [PMID: 34964567 DOI: 10.1515/hsz-2020-0394] [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: 12/23/2020] [Accepted: 12/20/2021] [Indexed: 11/15/2022]
Abstract
To explore the expression and the functions of SRPK1 in osteosarcoma, we retrieved transcription profiling dataset by array of human bone specimens from patients with osteosarcoma from ArrayExpress (accession E-MEXP-3628) and from Gene Expression Omnibus (accession GSE16102) and analyzed expression level of SRPK1 and prognostic value in human osteosarcoma. Then we examined the effect of differential SRPK1 expression levels on the progression of osteosarcoma, including cell proliferation, cell cycle, apoptosis, and investigated its underlying molecular mechanism using in vitro osteosarcoma cell lines and in vivo nude mouse xenograft models. High expression level of SRPK1 was found in human osteosarcoma tissues and cell lines as compared to the normal bone tissues and osteoblast cells, and predicted poor prognosis of human osteosarcoma. Overexpression of SRPK1 in osteosarcoma U2OS cells led to cell proliferation but inhibition of apoptosis. In contrast, knockdown of SRPK1 in HOS cells impeded cell viability and induction of apoptosis. Moreover, silencing SRPK1 inhibited osteosarcoma tumor growth in nude mice. Mechanistic studies revealed that SRPK1 promoted cell cycle transition in osteosarcoma cells and activation of NF-κB is required for SRPK1 expression and its pro-survival signaling. SRPK1 promoted human osteosarcoma cell proliferation and tumor growth by regulating NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yubao Gong
- Department of Orthopedics, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
| | - Chen Yang
- Department of Orthopedics, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
| | - Zhengren Wei
- Department of Pharmacology, Basic Medical School, Jilin University, Changchun, China
| | - Jianguo Liu
- Department of Orthopedics, The First Hospital of Jilin University, No. 1 Xinmin Street, Changchun 130021, China
| |
Collapse
|
3
|
Li X, Liang Q, Gao S, Jiang Q, Zhang F, Zhang R, Ruan H, Li S, Luan J, Deng R, Zhou H, Huang H, Yang C. Lenalidomide attenuates post-inflammation pulmonary fibrosis through blocking NF-κB signaling pathway. Int Immunopharmacol 2021; 103:108470. [PMID: 34952465 DOI: 10.1016/j.intimp.2021.108470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a pathological consequence of interstitial pulmonary diseases, and is characterized by the persistence of fibroblasts and excessive deposition of extracellular matrix (ECM). The etiology of IPF is multifactorial. Although the role of inflammation in fibrogenesis is controversial, it is still recognized as an important component and epiphenomenon of IPF. Stimulus increase production of pro-inflammatory cytokines and activation of NF-κB, which will further promote inflammation response and myofibroblast transition. Lenalidomide is an immunomodulatory drug. Previous studies have revealed its anti-tumor effects through regulating immune response. Here we investigate the effect of lenalidomide on post-inflammation fibrosis. In vitro study revealed that lenalidomide inhibited NF-κB signaling in LPS-induced macrophage, and further attenuated macrophage-induced myofibroblast activation. Meanwhile, lenalidomide could inhibit TGF-β1-induced myofibroblast activation through suppressing TGF-β1 downstream MAPK signaling. In vivo study showed that lenalidomide inhibited pro-inflammatory cytokines TNF-α and IL-6 while enhanced anti-fibrotic cytokines IFN-γ and IL-10 in bleomycin-induced inflammation model, and attenuated pulmonary fibrosis and collagen deposition in the following fibrosis stage. In conclusion, our results demonstrate that lenalidomide possesses potential anti-fibrotic effects through suppressing NF-κB signaling.
Collapse
Affiliation(s)
- Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Qing Liang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Shaoyan Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Qiuyan Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Fangxia Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Ruiqin Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Hao Ruan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Shuangling Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Jiaoyan Luan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Ruxia Deng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China.
| | - Hui Huang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| |
Collapse
|
4
|
The Role of Chaperone-Mediated Autophagy in Bortezomib Resistant Multiple Myeloma. Cells 2021; 10:cells10123464. [PMID: 34943972 PMCID: PMC8700264 DOI: 10.3390/cells10123464] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/20/2021] [Accepted: 10/30/2021] [Indexed: 11/17/2022] Open
Abstract
Background: Multiple myeloma (MM) remains incurable despite high-dose chemotherapy, autologous stem cell transplants and novel agents. Even with the improved survival of MM patients treated with novel agents, including bortezomib (Bz), the therapeutic options in relapsed/refractory MM remain limited. The majority of MM patients eventually develop resistance to Bz, although the mechanisms of the resistance are poorly understood. Methods: Lysosomal associated membrane protein 2A (LAMP2A) mRNA and protein expression levels were assessed in ex vivo patient samples and a Bz-resistant MM cell line model by in real-rime PCR, western blotting and immunohistochemistry. In vitro modelling of chaperone-mediated autophagy (CMA) activity in response to ER stress were assessed by western blotting and confocal microscopy. The effects of CMA inhibition on MM cell viability and Bz sensitivity in MM cells were assessed by Annexin V/7AAD apoptosis assays using flow cytometry. Results: In this study, there is evidence that CMA, a chaperone-mediated protein degradation pathway, is upregulated in Bz-resistant MM and the inhibition of CMA sensitises resistant cells to Bz. The protein levels of LAMP2A, the rate-limiting factor of the CMA pathway, are significantly increased in MM patients resistant to Bz and within our Bz-resistant cell line model. Bz-resistant cell lines also possessed higher basal CMA activity than the Bz-sensitive parent cell line. In MM cell lines, CMA activity was upregulated in response to ER stress induced by Bz. The inhibition of CMA sensitises Bz-resistant cells to Bz and the combination of CMA inhibition and Bz in vitro had a more cytotoxic effect on myeloma cells than Bz alone. Conclusion: In summary, the upregulation of CMA is a potential mechanism of resistance to Bz and a novel target to overcome Bz-resistant MM.
Collapse
|
5
|
Chen M, Huang Z, Xia M, Ding Y, Shan T, Guan Z, Dai X, Xu X, Huang Y, Huang M, Zhao C. Glutathione-responsive copper-disulfiram nanoparticles for enhanced tumor chemotherapy. J Control Release 2021; 341:351-363. [PMID: 34856225 DOI: 10.1016/j.jconrel.2021.11.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/17/2022]
Abstract
Disulfiram (DSF), a familiar FDA-approved drug used for alcohol withdrawal, has recently been verified with potent antitumor therapeutic effect by generating Cu(DTC)2, which is the complex of its metabolite diethyldithiocarbamate (DTC) and copper. However, its poor tumor selectivity and insufficient endogenous Cu2+ concentration within tumor site largely hinders the application of DSF-based antitumor therapy. Therefore, a GSH-responsive coordination nanoparticles (Cu-IXZ@DSF) was established as a copper carrier to achieve synchronous but separate delivery of Cu2+ and DSF without antitumor ability, further to realize selectively triggered tumor in situ Cu(DTC)2 generation for antitumor therapy. A widely-used proteasome inhibitor ixazomib (IXZ) was chosen as ligands and Cu2+ was used as coordination nodes to form nanosized Cu-IXZ@DSF. The DSF encapsulated in Cu-IXZ@DSF could be reduced to DTC by intracellular GSH, which could contend for Cu2+ and realize in situ high toxic Cu(DTC)2 generation. Meanwhile, the chelation could lead to the disassembly of Cu-IXZ@DSF and release of IXZ to eventually achieve tumor specific "transformation from low toxicity to high toxicity" chemotherapy. The results of in vitro and in vivo experiments demonstrated that the as-prepared nanoplatform Cu-IXZ@DSF showed good biosafety and excellent antitumor effect via endoplasmic reticulum stress (ERS) as well as reactive oxygen species (ROS) generation pathway. Therefore, this nanocarrier provides an inspiring strategy with specific-triggered antitumor Cu(DTC)2 generation for DSF-based chemotherapy with high therapeutic effect and biosafety and showing great potential of treating cancer.
Collapse
Affiliation(s)
- Meixu Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Zeqian Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Meng Xia
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Yaqing Ding
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Ting Shan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Zilin Guan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Xiuling Dai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Xiaoyu Xu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Min Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China.
| |
Collapse
|
6
|
Spaan I, van de Stolpe A, Raymakers RA, Peperzak V. Multiple Myeloma Relapse Is Associated with Increased NFκB Pathway Activity and Upregulation of the Pro-Survival BCL-2 Protein BFL-1. Cancers (Basel) 2021; 13:cancers13184668. [PMID: 34572895 PMCID: PMC8467450 DOI: 10.3390/cancers13184668] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy that is still considered incurable due to the development of therapy resistance and subsequent relapse of disease. MM plasma cells (PC) use NFκB signaling to stimulate cell growth and disease progression, and for protection against therapy-induced apoptosis. Amongst its diverse array of target genes, NFκB regulates the expression of pro-survival BCL-2 proteins BCL-XL, BFL-1, and BCL-2. A possible role for BFL-1 in MM is controversial, since BFL-1, encoded by BCL2A1, is downregulated when mature B cells differentiate into antibody-secreting PC. NFκB signaling can be activated by many factors in the bone marrow microenvironment and/or induced by genetic lesions in MM PC. We used the novel signal transduction pathway activity (STA) computational model to quantify the functional NFκB pathway output in primary MM PC from diverse patient subsets at multiple stages of disease. We found that NFκB pathway activity is not altered during disease development, is irrespective of patient prognosis, and does not predict therapy outcome. However, disease relapse after treatment resulted in increased NFκB pathway activity in surviving MM PC, which correlated with increased BCL2A1 expression in a subset of patients. This suggests that BFL-1 upregulation, in addition to BCL-XL and BCL-2, may render MM PC resistant to therapy-induced apoptosis, and that BFL-1 targeting could provide a new approach to reduce therapy resistance in a subset of relapsed/refractory MM patients.
Collapse
Affiliation(s)
- Ingrid Spaan
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Anja van de Stolpe
- Precision Diagnostics, Philips Research, 5656 AE Eindhoven, The Netherlands;
| | - Reinier A. Raymakers
- Department of Hematology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
| | - Victor Peperzak
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands;
- Correspondence: ; Tel.: +31-88-7567391
| |
Collapse
|
7
|
Sorrentino VG, Thota S, Gonzalez EA, Rameshwar P, Chang VT, Etchegaray JP. Hypomethylating Chemotherapeutic Agents as Therapy for Myelodysplastic Syndromes and Prevention of Acute Myeloid Leukemia. Pharmaceuticals (Basel) 2021; 14:641. [PMID: 34358067 PMCID: PMC8308509 DOI: 10.3390/ph14070641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Myelodysplastic Syndromes (MDSs) affect the elderly and can progress to Acute Myeloid Leukemia (AML). Epigenetic alterations including DNA methylation and chromatin modification may contribute to the initiation and progression of these malignancies. DNA hypomethylating agents such as decitabine and azacitidine are used as therapeutic treatments and have shown to promote expression of genes involved in tumor suppression, apoptosis, and immune response. Another anti-cancer drug, the proteasome inhibitor bortezomib, is used as a chemotherapeutic treatment for multiple myeloma (MM). Phase III clinical trials of decitabine and azacitidine used alone and in combination with other chemotherapeutics demonstrated their capacity to treat hematological malignancies and prolong the survival of MDS and AML patients. Although phase III clinical trials examining bortezomib's role in MDS and AML patients are limited, its underlying mechanisms in MM highlight its potential as a chemotherapeutic for such malignancies. Further research is needed to better understand how the epigenetic mechanisms mediated by these chemotherapeutic agents and their targeted gene networks are associated with the development and progression of MDS into AML. This review discusses the mechanisms by which decitabine, azacitidine, and bortezomib alter epigenetic programs and their results from phase III clinical trials.
Collapse
Affiliation(s)
- Vincent G. Sorrentino
- Department of Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA; (V.G.S.); (S.T.); (E.A.G.)
| | - Srijan Thota
- Department of Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA; (V.G.S.); (S.T.); (E.A.G.)
| | - Edward A. Gonzalez
- Department of Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA; (V.G.S.); (S.T.); (E.A.G.)
| | - Pranela Rameshwar
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA;
| | - Victor T. Chang
- Department of Medicine, Division of Hematology/Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA;
- Veteran Affairs New Jersey Health Care System, East Orange, NJ 07018, USA;
| | - Jean-Pierre Etchegaray
- Department of Biological Sciences, Rutgers University—Newark, Newark, NJ 07102, USA; (V.G.S.); (S.T.); (E.A.G.)
| |
Collapse
|
8
|
Yu X, Bao J, Cui X, DU F, Wang Y, Bi L, Sun J, Li L. Pyrrolidinedithiocarbamic Acid Ammonium Salt Inhibits Apoptosis and Phenotypic Transformation of Co-Culture of Myeloma Cells and Renal Tubular Epithelial Cells by Reducing the Secretion of Light Chain Protein. IRANIAN JOURNAL OF PUBLIC HEALTH 2021; 49:2078-2086. [PMID: 33708728 PMCID: PMC7917491 DOI: 10.18502/ijph.v49i11.4723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background: We investigate the effects of NFϰB inhibitor pyrrolidinedithiocarbamic acid ammonium salt (PDTC) on the viability, apoptosis and cell phenotype of HK-2 cells in the co-culture system of myeloma cells in renal tubular epithelial cells. Methods: This study was performed in Qiqihar Medical University, Qiqihar, China from Jun 2018 to Jan 2019. RPMI-8226 cells and HK-2 cells were inoculated in the co-culture chamber and cultured to establish the co-culture system. An immunoturbidimetric assay was performed to detect ϰ light chain and λ light chain in RPMI-8226 cells. The effect of PDTC on the secretion of ϰ light chain and λ light chain of RPMI-8226 cells was detected by immunoturbidimetry and the ratio was calculated. Results: PDTC significantly increased the viability of HK-2 cells. PDTC reduced the apoptosis of renal tubular epithelial cells. After PDTC treatment, the expression of cell surface marker E-cadherin decreased, and the expression of α-SMA increased, which induced the renal interstitial fibrosis. The secretion of ϰ light chain and λ light chain of RPMI-8226 cells was significantly decreased after the addition of PDTC, but the ratio was not changed. Conclusion: PDTC can inhibit the cell activity, promote apoptosis, and reduce the secretion of secretion of ϰ light chain and λ light chain through inhibiting the NF-ϰB pathway activation of myeloma cell RPMI-8226.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Department of Nephrology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar161000, P.R. China
| | - Jie Bao
- Department of Hematopathology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar161000, P.R. China
| | - Xinyu Cui
- Department of Hematopathology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar161000, P.R. China
| | - Fengxia DU
- Department of Pathogenic Biology, Qiqihar Medical University, Qiqihar161006, P.R. China
| | - Yuefei Wang
- Department of Physiology, Qiqihar Medical University, Qiqihar161006, P.R. China
| | - Lili Bi
- Department of Hematopathology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar161000, P.R. China
| | - Jun Sun
- Department of Hematopathology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar161000, P.R. China
| | - Ling Li
- Department of Hematopathology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar161000, P.R. China
| |
Collapse
|
9
|
Lee MS, Lim SH, Yu AR, Hwang CY, Kang I, Yeo EJ. Carfilzomib in Combination with Bortezomib Enhances Apoptotic Cell Death in B16-F1 Melanoma Cells. BIOLOGY 2021; 10:biology10020153. [PMID: 33671902 PMCID: PMC7918982 DOI: 10.3390/biology10020153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary The incidence rate of metastatic melanoma has been rapidly increasing worldwide and its 5-year survival rate is very low. Due to partial responses, various side effects, and resistance to any known cancer therapeutics, more potent and safer therapeutics are needed to increase the survival rate of patients with melanoma. Since proteasome inhibitors, such as bortezomib and carfilzomib, have been suggested as treatments for various cancers, we investigated their potential for the treatment of melanoma by studying their molecular mechanisms of action in B16-F1 melanoma cells. In this study, we found that both bortezomib and carfilzomib lead to apoptosis via ER stress as well as ROS accumulation and MMP loss in melanoma cells. Bortezomib and carfilzomib synergistically reduced B16-F1 tumor growth in vitro and in a C57BL/6 xenograft mouse model. Therefore, a combination therapy with carfilzomib and bortezomib at submaximal concentrations may reduce their side effects and be beneficial for melanoma treatment. Abstract Proteasome inhibitors, such as bortezomib (BZ) and carfilzomib (CFZ), have been suggested as treatments for various cancers. To utilize BZ and/or CFZ as effective therapeutics for treating melanoma, we studied their molecular mechanisms using B16-F1 melanoma cells. Flow cytometry of Annexin V-fluorescein isothiocyanate-labeled cells indicated apoptosis induction by treatment with BZ and CFZ. Apoptosis was evidenced by the activation of various caspases, including caspase 3, 8, 9, and 12. Treatment with BZ and CFZ induced endoplasmic reticulum (ER) stress, as indicated by an increase in eIF2α phosphorylation and the expression of ER stress-associated proteins, including GRP78, ATF6α, ATF4, XBP1, and CCAAT/enhancer-binding protein homologous protein. The effects of CFZ on ER stress and apoptosis were lower than that of BZ. Nevertheless, CFZ and BZ synergistically induced ER stress and apoptosis in B16-F1 cells. Furthermore, the combinational pharmacological interactions of BZ and CFZ against the growth of B16-F1 melanoma cells were assessed by calculating the combination index and dose-reduction index with the CompuSyn software. We found that the combination of CFZ and BZ at submaximal concentrations could obtain dose reduction by exerting synergistic inhibitory effects on cell growth. Moreover, this drug combination reduced tumor growth in C57BL/6 syngeneic mice. Taken together, these results suggest that CFZ in combination with BZ may be a beneficial and potential strategy for melanoma treatment.
Collapse
Affiliation(s)
- Min Seung Lee
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (M.S.L.); (S.H.L.)
| | - So Hyun Lim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (M.S.L.); (S.H.L.)
| | - Ah-Ran Yu
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; (A.-R.Y.); (C.Y.H.)
| | - Chi Yeon Hwang
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Korea; (A.-R.Y.); (C.Y.H.)
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (I.K.); (E.-J.Y.); Tel.: +82-29-610-922 (I.K.); +82-32-899-6050 (E.-J.Y.); Fax: +82-29-656-349 (I.K.); +82-32-899-6039 (E.-J.Y.)
| | - Eui-Ju Yeo
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Korea; (M.S.L.); (S.H.L.)
- Correspondence: (I.K.); (E.-J.Y.); Tel.: +82-29-610-922 (I.K.); +82-32-899-6050 (E.-J.Y.); Fax: +82-29-656-349 (I.K.); +82-32-899-6039 (E.-J.Y.)
| |
Collapse
|
10
|
Pancheri E, Guglielmi V, Wilczynski GM, Malatesta M, Tonin P, Tomelleri G, Nowis D, Vattemi G. Non-Hematologic Toxicity of Bortezomib in Multiple Myeloma: The Neuromuscular and Cardiovascular Adverse Effects. Cancers (Basel) 2020; 12:cancers12092540. [PMID: 32906684 PMCID: PMC7563977 DOI: 10.3390/cancers12092540] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is a still uncurable tumor of mainly elderly patients originating from the terminally differentiated B cells. Introduction to the treatment of MM patients of a new class of drugs called proteasome inhibitors (bortezomib followed by carfilzomib and ixazomib) significantly improved disease control. Proteasome inhibitors interfere with the major mechanism of protein degradation in a cell leading to the severe imbalance in the protein turnover that is deadly to MM cells. Currently, these drugs are the mainstream of MM therapy but are also associated with an increased rate of the injuries to multiple organs and tissues. In this review, we summarize the current knowledge on the molecular mechanisms of the first-in-class proteasome inhibitor bortezomib-induced disturbances in the function of peripheral nerves and cardiac and skeletal muscle. Abstract The overall approach to the treatment of multiple myeloma (MM) has undergone several changes during the past decade. and proteasome inhibitors (PIs) including bortezomib, carfilzomib, and ixazomib have considerably improved the outcomes in affected patients. The first-in-class selective PI bortezomib has been initially approved for the refractory forms of the disease but has now become, in combination with other drugs, the backbone of the frontline therapy for newly diagnosed MM patients, as well as in the maintenance therapy and relapsed/refractory setting. Despite being among the most widely used and highly effective agents for MM, bortezomib can induce adverse events that potentially lead to early discontinuation of the therapy with negative effects on the quality of life and outcome of the patients. Although peripheral neuropathy and myelosuppression have been recognized as the most relevant bortezomib-related adverse effects, cardiac and skeletal muscle toxicities are relatively common in MM treated patients, but they have received much less attention. Here we review the neuromuscular and cardiovascular side effects of bortezomib. focusing on the molecular mechanisms underlying its toxicity. We also discuss our preliminary data on the effects of bortezomib on skeletal muscle tissue in mice receiving the drug.
Collapse
Affiliation(s)
- Elia Pancheri
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, 37134 Verona, Italy; (E.P.); (V.G.); (P.T.); (G.T.)
| | - Valeria Guglielmi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, 37134 Verona, Italy; (E.P.); (V.G.); (P.T.); (G.T.)
| | - Grzegorz M. Wilczynski
- Laboratory of Molecular and Systemic Neuromorphology, Department of Neurophysiology Warsaw, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland;
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Anatomy and Histology, University of Verona, 37134 Verona, Italy;
| | - Paola Tonin
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, 37134 Verona, Italy; (E.P.); (V.G.); (P.T.); (G.T.)
| | - Giuliano Tomelleri
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, 37134 Verona, Italy; (E.P.); (V.G.); (P.T.); (G.T.)
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, 02-093 Warsaw, Poland;
- Laboratory of Experimental Medicine, Medical University of Warsaw, 02-093 Warsaw, Poland
| | - Gaetano Vattemi
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Clinical Neurology, University of Verona, 37134 Verona, Italy; (E.P.); (V.G.); (P.T.); (G.T.)
- Correspondence:
| |
Collapse
|
11
|
Akhtar S, Ali TA, Faiyaz A, Khan OS, Raza SS, Kulinski M, Omri HE, Bhat AA, Uddin S. Cytokine-Mediated Dysregulation of Signaling Pathways in the Pathogenesis of Multiple Myeloma. Int J Mol Sci 2020; 21:5002. [PMID: 32679860 PMCID: PMC7403981 DOI: 10.3390/ijms21145002] [Citation(s) in RCA: 25] [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: 01/21/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is a hematologic disorder of B lymphocytes characterized by the accumulation of malignant plasma cells (PCs) in the bone marrow. The altered plasma cells overproduce abnormal monoclonal immunoglobulins and also stimulate osteoclasts. The host's immune system and microenvironment are of paramount importance in the growth of PCs and, thus, in the pathogenesis of the disease. The interaction of MM cells with the bone marrow (BM) microenvironment through soluble factors and cell adhesion molecules causes pathogenesis of the disease through activation of multiple signaling pathways, including NF-κβ, PI3K/AKT and JAK/STAT. These activated pathways play a critical role in the inhibition of apoptosis, sustained proliferation, survival and migration of MM cells. Besides, these pathways also participate in developing resistance against the chemotherapeutic drugs in MM. The imbalance between inflammatory and anti-inflammatory cytokines in MM leads to an increased level of pro-inflammatory cytokines, which in turn play a significant role in dysregulation of signaling pathways and proliferation of MM cells; however, the association appears to be inadequate and needs more research. In this review, we are highlighting the recent findings on the roles of various cytokines and growth factors in the pathogenesis of MM and the potential therapeutic utility of aberrantly activated signaling pathways to manage the MM disease.
Collapse
Affiliation(s)
- Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
| | - Tayyiba A. Ali
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
| | - Ammara Faiyaz
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
| | - Omar S. Khan
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA;
| | - Syed Shadab Raza
- Department of Stem Cell Biology and Regenerative Medicine, Era University, Lucknow 226003, India;
| | - Michal Kulinski
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
| | - Halima El Omri
- National Cancer Care and Research, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Ajaz A. Bhat
- Translational Medicine, Research Branch, Sidra Medicine, Doha 26999, Qatar;
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; (S.A.); (T.A.A.); (A.F.); (M.K.)
- Dermatology Institute, Department of Dermatology and Venereology, Hamad Medical Corporation, Doha 3050, Qatar
| |
Collapse
|
12
|
Wu P, Oren O, Gertz MA, Yang EH. Proteasome Inhibitor-Related Cardiotoxicity: Mechanisms, Diagnosis, and Management. Curr Oncol Rep 2020; 22:66. [DOI: 10.1007/s11912-020-00931-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
13
|
Role of the Bone Marrow Milieu in Multiple Myeloma Progression and Therapeutic Resistance. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e752-e768. [PMID: 32651110 DOI: 10.1016/j.clml.2020.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/22/2020] [Accepted: 05/29/2020] [Indexed: 01/10/2023]
Abstract
Multiple myeloma (MM) is a cancer of the plasma cells within the bone marrow (BM). Studies have shown that the cellular and noncellular components of the BM milieu, such as cytokines and exosomes, play an integral role in MM pathogenesis and progression by mediating drug resistance and inducing MM proliferation. Moreover, the BM microenvironment of patients with MM facilitates cancer tolerance and immune evasion through the expansion of regulatory immune cells, inhibition of antitumor effector cells, and disruption of the antigen presentation machinery. These are of special relevance, especially in the current era of cancer immunotherapy. An improved understanding of the supportive role of the MM BM microenvironment will allow for the development of future therapies targeting MM in the context of the BM milieu to elicit deeper and more durable responses. In the present review, we have discussed our current understanding of the role of the BM microenvironment in MM progression and resistance to therapy and discuss novel potential approaches to alter its pro-MM function.
Collapse
|
14
|
Marino S, Petrusca DN, Roodman GD. Therapeutic targets in myeloma bone disease. Br J Pharmacol 2020; 178:1907-1922. [PMID: 31647573 DOI: 10.1111/bph.14889] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 09/09/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022] Open
Abstract
Multiple myeloma (MM) is the second most common haematological malignancy and is characterized by a clonal proliferation of neoplastic plasma cells within the bone marrow. MM is the most frequent cancer involving the skeleton, causing osteolytic lesions, bone pain and pathological fractures that dramatically decrease MM patients' quality of life and survival. MM bone disease (MBD) results from uncoupling of bone remodelling in which excessive bone resorption is not compensated by new bone formation, due to a persistent suppression of osteoblast activity. Current management of MBD includes antiresorptive agents, bisphosphonates and denosumab, that are only partially effective due to their inability to repair the existing lesions. Thus, research into agents that prevent bone destruction and more importantly repair existing lesions by inducing new bone formation is essential. This review discusses the mechanisms regulating the uncoupled bone remodelling in MM and summarizes current advances in the treatment of MBD. LINKED ARTICLES: This article is part of a themed issue on The molecular pharmacology of bone and cancer-related bone diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.9/issuetoc.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Medicine, Division Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Daniela N Petrusca
- Department of Medicine, Division Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - G David Roodman
- Department of Medicine, Division Hematology/Oncology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Roudebush VA Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
15
|
Ito S. Proteasome Inhibitors for the Treatment of Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12020265. [PMID: 31979059 PMCID: PMC7072336 DOI: 10.3390/cancers12020265] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/19/2020] [Accepted: 01/19/2020] [Indexed: 01/04/2023] Open
Abstract
Use of proteasome inhibitors (PIs) has been the therapeutic backbone of myeloma treatment over the past decade. Many PIs are being developed and evaluated in the preclinical and clinical setting. The first-in-class PI, bortezomib, was approved by the US food and drug administration in 2003. Carfilzomib is a next-generation PI, which selectively and irreversibly inhibits proteasome enzymatic activities in a dose-dependent manner. Ixazomib was the first oral PI to be developed and has a robust efficacy and favorable safety profile in patients with multiple myeloma. These PIs, together with other agents, including alkylators, immunomodulatory drugs, and monoclonal antibodies, have been incorporated into several regimens. This review summarizes the biological effects and the results of clinical trials investigating PI-based combination regimens and novel investigational inhibitors and discusses the future perspective in the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Shigeki Ito
- Hematology & Oncology, Department of Internal Medicine, Iwate Medical University School of Medicine, Yahaba-cho 028-3695, Japan
| |
Collapse
|
16
|
Mazzera L, Abeltino M, Lombardi G, Cantoni AM, Ria R, Ricca M, Saltarella I, Naponelli V, Rizzi FMA, Perris R, Corradi A, Vacca A, Bonati A, Lunghi P. Functional interplay between NF-κB-inducing kinase and c-Abl kinases limits response to Aurora inhibitors in multiple myeloma. Haematologica 2019; 104:2465-2481. [PMID: 30948493 PMCID: PMC6959191 DOI: 10.3324/haematol.2018.208280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/03/2019] [Indexed: 12/19/2022] Open
Abstract
Considering that Aurora kinase inhibitors are currently under clinical investigation in hematologic cancers, the identification of molecular events that limit the response to such agents is essential for enhancing clinical outcomes. Here, we discover a NF-κB-inducing kinase (NIK)-c-Abl-STAT3 signaling-centered feedback loop that restrains the efficacy of Aurora inhibitors in multiple myeloma. Mechanistically, we demonstrate that Aurora inhibition promotes NIK protein stabilization via downregulation of its negative regulator TRAF2. Accumulated NIK converts c-Abl tyrosine kinase from a nuclear proapoptotic into a cytoplasmic antiapoptotic effector by inducing its phosphorylation at Thr735, Tyr245 and Tyr412 residues, and, by entering into a trimeric complex formation with c-Abl and STAT3, increases both the transcriptional activity of STAT3 and expression of the antiapoptotic STAT3 target genes PIM1 and PIM2. This consequently promotes cell survival and limits the response to Aurora inhibition. The functional disruption of any of the components of the trimer NIK-c-Abl-STAT3 or the PIM survival kinases consistently enhances the responsiveness of myeloma cells to Aurora inhibitors. Importantly, concurrent inhibition of NIK or c-Abl disrupts Aurora inhibitor-induced feedback activation of STAT3 and sensitizes myeloma cells to Aurora inhibitors, implicating a combined inhibition of Aurora and NIK or c-Abl kinases as potential therapies for multiple myeloma. Accordingly, pharmacological inhibition of c-Abl together with Aurora resulted in substantial cell death and tumor regression in vivo The findings reveal an important functional interaction between NIK, Abl and Aurora kinases, and identify the NIK, c-Abl and PIM survival kinases as potential pharmacological targets for improving the efficacy of Aurora inhibitors in myeloma.
Collapse
Affiliation(s)
- Laura Mazzera
- Department of Medicine and Surgery, University of Parma, Parma
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini," Brescia
| | | | - Guerino Lombardi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini," Brescia
| | | | - Roberto Ria
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari "Aldo Moro" Medical School, Bari
| | - Micaela Ricca
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini," Brescia
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari "Aldo Moro" Medical School, Bari
| | | | - Federica Maria Angela Rizzi
- Department of Medicine and Surgery, University of Parma, Parma
- Center for Molecular and Translational Oncology, University of Parma, Parma
| | - Roberto Perris
- Center for Molecular and Translational Oncology, University of Parma, Parma
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Attilio Corradi
- Department of Veterinary Science, University of Parma, Parma
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari "Aldo Moro" Medical School, Bari
| | - Antonio Bonati
- Department of Medicine and Surgery, University of Parma, Parma
- Center for Molecular and Translational Oncology, University of Parma, Parma
| | - Paolo Lunghi
- Center for Molecular and Translational Oncology, University of Parma, Parma
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| |
Collapse
|
17
|
Morita M, Nakanishi K, Masuda K, Yoshida K, Shimomura D, Ishida A, Shiga S, Ichiyama S. Acute non-heparin-induced thrombocytopenia during hemodiafiltration in a patient with multiple myeloma. Clin Case Rep 2019; 7:699-702. [PMID: 30997067 PMCID: PMC6452500 DOI: 10.1002/ccr3.1997] [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: 03/03/2018] [Revised: 11/19/2018] [Accepted: 12/04/2018] [Indexed: 11/13/2022] Open
Abstract
This report demonstrates that not only heparin-induced thrombocytopenia, but also hemodialysis conditions (platelet activation due to hemodiafiltration and heparin underdosing) may markedly reduce the platelet count and cause clotting in the hemodialysis circuit in patients in a hypercoagulable state. The clot prevention effects of bortezomib are therefore of great importance.
Collapse
Affiliation(s)
- Makiko Morita
- Department of Clinical LaboratoryKyoto University HospitalKyotoJapan
| | - Kayoko Nakanishi
- Department of Clinical LaboratoryKyoto University HospitalKyotoJapan
| | - Kenta Masuda
- Department of Clinical LaboratoryKyoto University HospitalKyotoJapan
| | | | | | - Atsumi Ishida
- Department of Clinical LaboratoryKyoto University HospitalKyotoJapan
| | - Shuichi Shiga
- Department of Clinical LaboratoryKyoto University HospitalKyotoJapan
| | - Satoshi Ichiyama
- Department of Clinical LaboratoryKyoto University HospitalKyotoJapan
| |
Collapse
|
18
|
The impact of NF-κB signaling on pathogenesis and current treatment strategies in multiple myeloma. Blood Rev 2019; 34:56-66. [DOI: 10.1016/j.blre.2018.11.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/14/2018] [Accepted: 11/22/2018] [Indexed: 12/13/2022]
|
19
|
Caffeic acid phenethyl ester exerts apoptotic and oxidative stress on human multiple myeloma cells. Invest New Drugs 2018; 37:837-848. [DOI: 10.1007/s10637-018-0701-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/14/2018] [Indexed: 01/02/2023]
|
20
|
Gu J, Song S, Han H, Xu H, Fan G, Qian C, Qiu Y, Zhou W, Zhuang W, Li B. The BET Bromodomain Inhibitor OTX015 Synergizes with Targeted Agents in Multiple Myeloma. Mol Pharm 2018; 15:5387-5396. [PMID: 30339013 DOI: 10.1021/acs.molpharmaceut.8b00880] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Treatment failure remains a main challenge in the management of high-risk multiple myeloma (MM) even with the expanding repertoire of new drugs. Combinatorial therapy is considered an encouraging strategy that can overcome the compensatory mechanisms and undesirable off-target effects that limit the benefits of many prospective agents. Preliminary results of a current phase I trial have indicated that the new BET bromodomain inhibitor OTX015 has favorable activity and tolerability. However, OTX015 is not efficacious enough as a monotherapy. Here, we provide evidence that synergistic drug combinations with OTX015 were generally more specific to particular cellular contexts than single agent activities. In addition, pairing OTX015 with three classes of drugs dramatically enhanced the antitumor activity in mouse models of disseminated human myeloma. Our studies further underscored that the BET inhibitor OTX015 sensitized MM cells by interrupting several pathways and genes critical for MM cell proliferation and drug response, which provided the rationale for multiple myeloma therapy with OTX015 combined with conventional chemotherapeutic drugs. Thus, the context specificity of synergistic combinations not only provide profound insights into therapeutically relevant selectivity but also improve control of complex biological systems.
Collapse
Affiliation(s)
- Jie Gu
- Department of Haematology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - Sha Song
- Department of Cell Biology, School of Biology & Basic Medical Sciences , Soochow University , Suzhou , China
| | - Huiying Han
- Department of Cell Biology, School of Biology & Basic Medical Sciences , Soochow University , Suzhou , China
| | - Hongxia Xu
- Department of Cell Biology, School of Biology & Basic Medical Sciences , Soochow University , Suzhou , China
| | - Gao Fan
- Department of Cell Biology, School of Biology & Basic Medical Sciences , Soochow University , Suzhou , China
| | - Chen'ao Qian
- Department of Bioinformatics, School of Biology & Basic Medical Sciences , Soochow University , Suzhou , China
| | - Yingchun Qiu
- Department of Cell Biology, School of Biology & Basic Medical Sciences , Soochow University , Suzhou , China
| | - Wenqi Zhou
- Department of Cell Biology, School of Biology & Basic Medical Sciences , Soochow University , Suzhou , China
| | - Wenzhuo Zhuang
- Department of Cell Biology, School of Biology & Basic Medical Sciences , Soochow University , Suzhou , China
| | - Bingzong Li
- Department of Haematology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| |
Collapse
|
21
|
|
22
|
Abstract
Multiple myeloma (MM) is the second-most-common hematologic malignancy and the most frequent cancer to involve bone. MM bone disease (MMBD) has devastating consequences for patients, including dramatic bone loss, severe bone pain, and pathological fractures that markedly decrease the quality of life and impact survival of MM patients. MMBD results from excessive osteoclastic bone resorption and persistent suppressed osteoblastic bone formation, causing lytic lesions that do not heal, even when patients are in complete and prolonged remission. This review discusses the cellular and molecular mechanisms that regulate the uncoupling of bone remodeling in MM, the effects of MMBD on tumor growth, and potential therapeutic approaches that may prevent severe bone loss and repair damaged bone in MM patients.
Collapse
Affiliation(s)
- Silvia Marino
- Department of Medicine, Division Hematology Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - G David Roodman
- Department of Medicine, Division Hematology Oncology, Indiana University School of Medicine, Indianapolis, Indiana 46202
- Roudebush VA Medical Center, Indianapolis, Indiana 46202
| |
Collapse
|
23
|
Targeting signaling pathways in multiple myeloma: Pathogenesis and implication for treatments. Cancer Lett 2018; 414:214-221. [DOI: 10.1016/j.canlet.2017.11.020] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/15/2022]
|
24
|
Abstract
Multiple myeloma (MM) is an incurable hematopoietic cancer that is characterized by malignant plasma cell infiltration of the bone marrow and/or extramedullary sites. Multi-modality approaches including "novel agents," traditional chemotherapy, and/or stem cell transplantation are used in MM therapy. Drug resistance, however, ultimately develops and the disease remains incurable for the vast majority of patients. In this chapter, we review both tumor cell-autonomous and non-autonomous (microenvironment-dependent) mechanisms of drug resistance. MM provides an attractive paradigm highlighting a number of current concepts and challenges in oncology. Firstly, identification of MM cancer stem cells and their unique drug resistance attributes may provide rational avenues towards MM eradication and cure. Secondly, the oligoclonal evolution of MM and alternation of "clonal tides" upon therapy challenge our current understanding of treatment responses. Thirdly, the success of MM "novel agents" provides exemplary evidence for the impact of therapies that target the immune and non-immune microenvironment. Fourthly, the rapid pace of drug approvals for MM creates an impetus for development of precision medicine strategies and biomarkers that promote efficacy and mitigate toxicity and cost. While routine cure of the disease remains the ultimate and yet unattainable prize, MM advances in the last 10-15 years have provided an astounding paradigm for the treatment of blood cancers in the modern era and have radically transformed patient outcomes.
Collapse
Affiliation(s)
- Athanasios Papadas
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- UW Carbone Cancer Center, Madison, WI, 53705, USA.
| | - Fotis Asimakopoulos
- Division of Hematology and Oncology, Department of Medicine, University of Wisconsin-Madison, Madison, WI, 53705, USA
- UW Carbone Cancer Center, Madison, WI, 53705, USA
| |
Collapse
|
25
|
Neuropilin-1 contributes to esophageal squamous cancer progression via promoting P65-dependent cell proliferation. Oncogene 2017; 37:935-943. [DOI: 10.1038/onc.2017.399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 09/11/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022]
|
26
|
Shvartsur A, Givechian KB, Garban H, Bonavida B. Overexpression of RKIP and its cross-talk with several regulatory gene products in multiple myeloma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:62. [PMID: 28476134 PMCID: PMC5420138 DOI: 10.1186/s13046-017-0535-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/26/2017] [Indexed: 12/27/2022]
Abstract
Multiple myeloma (MM) is a clonal plasma-cell neoplastic disorder arising from an indolent premalignant disease known as monoclonal gammopathy of undetermined significance (MGUS). MM is a biologically complex heterogeneous disease reflected by its variable clinical responses of patients receiving the same treatment. Therefore, a molecular identification of stage-specific biomarkers will support a more individualized precise diagnostic/prognostic approach, an effective therapeutic regime, and will assist in the identification of novel therapeutic molecular targets. The metastatic suppressor/anti-resistance factor Raf-1 kinase inhibitor protein (RKIP) is poorly expressed in the majority of cancers and is often almost absent in metastatic tumors. RKIP inhibits the Raf/MEK/ERK1/2 and the NF-κB pathways. Whereby all tumors examined exhibited low levels of RKIP, in contrast, our recent findings demonstrated that RKIP is overexpressed primarily in its inactive phosphorylated form in MM cell lines and patient-derived tumor tissues. The underlying mechanism of RKIP overexpression in MM, in contrast to other tumors, is not known. We examined transcriptomic datasets on Oncomine platform (Life Technologies) for the co-expression of RKIP and other gene products in both pre-MM and MM. The transcription of several gene products was found to be either commonly overexpressed (i.e., RKIP, Bcl-2, and DR5) or underexpressed (i.e., Bcl-6 and TNFR2) in both pre-MM and MM. Noteworthy, a significant inverse correlation of differentially expressed pro-apoptotic genes was observed in pre-MM: overexpression of Fas and TNF-α and underexpression of YY1 versus expression of anti-apoptotic genes in MM: overexpression of YY1 and underexpression of Fas and TNF-α. Based on the analysis on mRNA levels and reported studies on protein levels of the above various genes, we have constructed various schemes that illustrate the possible cross-talks between RKIP (active/inactive) and the identified gene products that underlie the mechanism of RKIP overexpression in MM. Clearly, such cross-talks would need to be experimentally validated in both MM cell lines and patient-derived tumor tissues. If validated, the differential molecular signatures between pre-MM and MM might lead to a more precise diagnosis/prognosis of the disease and disease stages and will also identify novel molecular therapeutic targets for pre-MM and MM.
Collapse
Affiliation(s)
- Anna Shvartsur
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kevin B Givechian
- Department of Biological Sciences, USC Dana and David Dornsife College of Letters, Arts and Sciences at the University of Southern California, Los Angeles, CA, 90089, USA
| | - Hermes Garban
- California NanoSystems Institute (CnSI), University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
27
|
Morotti A, Crivellaro S, Panuzzo C, Carrà G, Guerrasio A, Saglio G. IκB-α: At the crossroad between oncogenic and tumor-suppressive signals. Oncol Lett 2016; 13:531-534. [PMID: 28356925 PMCID: PMC5351326 DOI: 10.3892/ol.2016.5465] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 08/19/2016] [Indexed: 01/13/2023] Open
Abstract
Nuclear factor κB (NF-κB) is an essential component of tumorigenesis and resistance to cancer treatments. NFKB inhibitor α (IκB-α) acts as a negative regulator of the classical NF-κB pathway through its ability to maintain the presence of NF-κB in the cytoplasm. However, IκB-α is also able to form a complex with tumor protein p53, promoting its inactivation. Recently, we demonstrated that IκB-α is able to mediate p53 nuclear exclusion and inactivation in chronic myeloid leukemia, indicating that IκB-α can modulate either oncogenic or tumor-suppressive functions, with important implications for cancer treatment. The present review describes the role of IκB-α in cancer pathogenesis, with particular attention to hematological cancers, and highlights the involvement of IκB-α in the regulation of p53 tumor-suppressive functions.
Collapse
Affiliation(s)
- Alessandro Morotti
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Turin, Italy
| | - Sabrina Crivellaro
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Turin, Italy
| | - Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Turin, Italy
| | - Giovanna Carrà
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Turin, Italy
| | - Angelo Guerrasio
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Turin, Italy
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin, I-10043 Orbassano, Turin, Italy
| |
Collapse
|
28
|
Matthews GM, de Matos Simoes R, Dhimolea E, Sheffer M, Gandolfi S, Dashevsky O, Sorrell JD, Mitsiades CS. NF-κB dysregulation in multiple myeloma. Semin Cancer Biol 2016; 39:68-76. [PMID: 27544796 DOI: 10.1016/j.semcancer.2016.08.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 12/29/2022]
Abstract
The nuclear factor-κB (NF-κB) transcription factor family plays critical roles in the pathophysiology of hematologic neoplasias, including multiple myeloma. The current review examines the roles that this transcription factor system plays in multiple myeloma cells and the nonmalignant accessory cells of the local microenvironment; as well as the evidence indicating that a large proportion of myeloma patients harbor genomic lesions which perturb diverse genes regulating the activity of NF-κB. This article also discusses the therapeutic targeting of the NF-κB pathway using proteasome inhibitors, a pharmacological class that has become a cornerstone in the therapeutic management of myeloma; and reviews some of the future challenges and opportunities for NF-κB-related research in myeloma.
Collapse
Affiliation(s)
- Geoffrey M Matthews
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Ricardo de Matos Simoes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Eugen Dhimolea
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Michal Sheffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Sara Gandolfi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Olga Dashevsky
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Jeffrey D Sorrell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States
| | - Constantine S Mitsiades
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, United States.
| |
Collapse
|
29
|
Robinson AA, Wang J, Vardanyan S, Madden EK, Hebroni F, Udd KA, Spektor TM, Nosrati JD, Kitto AZ, Zahab M, Cheema S, Fors DH, Norberg A, Diehl J, Waterman GN, Swift RA, Crowley J, Berenson JR. Risk of skin cancer in multiple myeloma patients: a retrospective cohort study. Eur J Haematol 2016; 97:439-444. [PMID: 26872804 DOI: 10.1111/ejh.12748] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2016] [Indexed: 12/16/2022]
Affiliation(s)
| | - James Wang
- James R. Berenson, MD, Inc.; West Hollywood CA USA
| | - Suzie Vardanyan
- Institute for Myeloma and Bone Cancer Research; West Hollywood CA USA
| | - Erik K. Madden
- David Geffen School of Medicine; University of California; Los Angeles CA USA
| | - Frank Hebroni
- David Geffen School of Medicine; University of California; Los Angeles CA USA
| | - Kyle A. Udd
- James R. Berenson, MD, Inc.; West Hollywood CA USA
| | | | - Jason D. Nosrati
- Institute for Myeloma and Bone Cancer Research; West Hollywood CA USA
| | - Alex Z. Kitto
- Institute for Myeloma and Bone Cancer Research; West Hollywood CA USA
| | - Michael Zahab
- Institute for Myeloma and Bone Cancer Research; West Hollywood CA USA
| | - Simrin Cheema
- Fielding School of Public Health; University of California; Los Angeles CA USA
| | - Darron H. Fors
- David Geffen School of Medicine; University of California; Los Angeles CA USA
| | - Adam Norberg
- David Geffen School of Medicine; University of California; Los Angeles CA USA
| | - Joseph Diehl
- David Geffen School of Medicine; University of California; Los Angeles CA USA
| | - Gabriel N. Waterman
- Keck School of Medicine; University of Southern California; Los Angeles CA USA
| | | | - John Crowley
- Cancer Research and Biostatistics; Seattle WA USA
| | - James R. Berenson
- James R. Berenson, MD, Inc.; West Hollywood CA USA
- Institute for Myeloma and Bone Cancer Research; West Hollywood CA USA
- Oncotherapeutics; West Hollywood CA USA
| |
Collapse
|
30
|
Wang D, Chen J, Li R, Wu G, Sun Z, Wang Z, Zhai Z, Fang F, Guo Y, Zhong Y, Jiang M, Xu H, Chen M, Shen G, Sun J, Yan B, Yu C, Tian Z, Xiao W. PAX5 interacts with RIP2 to promote NF-κB activation and drug-resistance of B-lymphoproliferative disorders. J Cell Sci 2016; 129:2261-72. [DOI: 10.1242/jcs.183889] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 04/11/2016] [Indexed: 12/17/2022] Open
Abstract
Paired box protein 5 (PAX5) plays a lineage determination role in B-cell development. However, high expression of PAX5 has been also found in various malignant diseases including B-lymphoproliferative disorders (B-LPDs), but its functions and mechanisms in these diseases are still unclear. Here, we show that PAX5 induces drug-resistance through association and activation of receptor-interacting serine/threonine-protein kinase2 (RIP2) and subsequent activation of NF-κB signaling and anti-apoptosis genes expression in B-lymphoproliferative cells. Furthermore, PAX5 is able to interact with RIP1-3, modulating both RIP1- mediated TNFR and RIP2-mediated NOD1 and NOD2 pathways. Our findings describe a novel function of PAX5 in regulating RIP1 and RIP2 activation, which is at least involved in chemo drug-resistance in B-LPDs.
Collapse
Affiliation(s)
- Dong Wang
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Jingyu Chen
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Rui Li
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Guolin Wu
- Department of Hematology, Anhui Provincial Hospital, 17 Lujiang Road, Hefei, Anhui Province 230001, China
| | - Zimin Sun
- Department of Hematology, Anhui Provincial Hospital, 17 Lujiang Road, Hefei, Anhui Province 230001, China
| | - Zhitao Wang
- Department of Hematology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province 230601, China
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui Province 230601, China
| | - Fang Fang
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Yugang Guo
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Yongjun Zhong
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Ming Jiang
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Huan Xu
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Minhua Chen
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Guodong Shen
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Jie Sun
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Bailing Yan
- Emergency Department, the First Hospital of Jilin Univesity, Changchun 130021, China
| | - Chundong Yu
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Zhigang Tian
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| | - Weihua Xiao
- Key Laboratory of Innate Immunity and Chronic Disease of CAS, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Laboratory for Physical Sciences at Microscale, Hefei, Anhui 230027, China
- Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China
| |
Collapse
|
31
|
Inhibitory effects of parthenolide on the activity of NF-κB in multiple myeloma via targeting TRAF6. ACTA ACUST UNITED AC 2015; 35:343-349. [PMID: 26072071 DOI: 10.1007/s11596-015-1435-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/15/2015] [Indexed: 01/01/2023]
Abstract
This study examined the mechanism of the inhibitory effect of parthenolide (PTL) on the activity of NF-κB in multiple myeloma (MM). Human multiple myeloma cell line RPMI 8226 cells were treated with or without different concentrations of PTL for various time periods, and then MTT assay was used to detect cell proliferation. Cell cycle and apoptosis were flow cytometrically detected. The level of protein ubiquitination was determined by using immunoprecipitation. Western blotting was employed to measure the level of total protein ubiquitination, the expression of IκB-α in cell plasma and the content of p65 in nucleus. The content of p65 in nucleus before and after PTL treatment was also examined with immunofluorescence. Exposure of RPMI 8226 cells to PTL attenuated the level of ubiquitinated Nemo, increased the expression of IκB-α and reduced the level of p65 in nucleus, finally leading to the decrease of the activity of NF-κB. PTL inhibited cell proliferation, induced apoptosis and blocked cell cycle. Furthermore, the levels of ubiquitinated tumor necrosis factor receptor-associated factor 6 (TRAF6) and total proteins were decreased after PTL treatment. By using Autodock software package, we predicted that PTL could bind to TRAF6 directly and tightly. Taken together, our findings suggest that PTL inhibits the activation of NF-κB signaling pathway via directly binding with TRAF6, thereby suppressing MM cell proliferation and inducing apoptosis.
Collapse
|
32
|
miRNA-202 in bone marrow stromal cells affects the growth and adhesion of multiple myeloma cells by regulating B cell-activating factor. Clin Exp Med 2015; 16:307-16. [PMID: 25971527 DOI: 10.1007/s10238-015-0355-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/24/2015] [Indexed: 10/23/2022]
Abstract
Bone marrow stromal cells (BMSCs) up-regulate B cell-activating factor (BAFF) in multiple myeloma. Increasing experimental evidence has shown that microRNAs play a causal role in hematology tumorigenesis. In this study, we characterized the role of miR-202 in regulating the expression of BAFF in BMSCs. It was found that expressions of BAFF mRNA and protein were increased in BMSCs treated with miR-202 inhibitor. The growth rate of miR-202 mimics transfection cells was significantly lower than that of non-transfected cells. The expression of Bcl-2 protein was down-regulated, and Bax protein was up-regulated after miR-202 mimics transfection. Over-expression of miR-202 in BMSCs rendered MM cells more sensitive to bortezomib. More significantly, the regulatory effect of miR-202 could inhibit the activation of NF-κB pathway in BMSCs. These results suggest that miR-202 functions as a modulator that can negatively regulate BAFF by inhibiting MM cell survival, growth, and adhesion in the bone marrow microenvironment.
Collapse
|
33
|
Multiple myeloma cells alter the senescence phenotype of bone marrow mesenchymal stromal cells under participation of the DLK1-DIO3 genomic region. BMC Cancer 2015; 15:68. [PMID: 25886144 PMCID: PMC4336751 DOI: 10.1186/s12885-015-1078-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/10/2015] [Indexed: 01/15/2023] Open
Abstract
Background Alterations and senescence in bone marrow mesenchymal stromal cells of multiple myeloma patients (MM-BMMSCs) have become an important research focus. However the role of senescence in the pathophysiology of MM is not clear. Methods Correlation between senescence, cell cycle and microRNA expression of MM-BMMSCs (n = 89) was analyzed. Gene expression analysis, copy number analysis and methylation specific PCR were performed by Real-Time PCR. Furthermore, cyclin E1, cyclin D1, p16 and p21 genes were analyzed at the protein level using ELISA. Cell cycle and senescence were analyzed by FACS. MiRNA transfection was performed with miR-485-5p inhibitor and mimic followed by downstream analysis of senescence and cell cycle characteristics of MM-BMMSCs. Results were analyzed by Mann–Whitney U test, Wilcoxon signed-rank test and paired t-test depending on the experimental set up. Results MM-BMMSCs displayed increased senescence associated β-galactosidase activity (SA-βGalA), cell cycle arrest in S phase and overexpression of microRNAs. The overexpressed microRNAs miR-485-5p and miR-519d are located on DLK1-DIO3 and C19MC, respectively. Analyses revealed copy number accumulation and hypomethylation of both clusters. KMS12-PE myeloma cells decreased SA-βGalA and influenced cell cycle characteristics of MM-BMMSCs. MiR-485-5p was significantly decreased in co-cultured MM-BMMSCs in connection with an increased methylation of DLK1-DIO3. Modification of miR-485-5p levels using microRNA mimic or inhibitor altered senescence and cell cycle characteristics of MM-BMMSCs. Conclusions Here, we show for the first time that MM-BMMSCs have aberrant methylation and copy number of the DLK1-DIO3 and C19MC genomic region. Furthermore, this is the first study pointing that multiple myeloma cells in vitro reduce both the senescence phenotype of MM-BMMSCs and the expression of miR-223 and miR-485-5p. Thus, it is questionable whether senescence of MM-BMMSCs plays a pathological role in active multiple myeloma or is more important when cell interaction with myeloma cells is inhibited. Furthermore, we found that MiR-485-5p, which is located on the DLK1-DIO3 cluster, seems to participate in the regulation of senescence status and cell cycle characteristics of MM-BMMSCs. Thus, further exploration of the microRNAs of DLK1-DIO3 could provide further insights into the origin of the senescence state and its reversal in MM-BMMSCs. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1078-3) contains supplementary material, which is available to authorized users.
Collapse
|
34
|
Siveen KS, Mustafa N, Li F, Kannaiyan R, Ahn KS, Kumar AP, Chng WJ, Sethi G. Thymoquinone overcomes chemoresistance and enhances the anticancer effects of bortezomib through abrogation of NF-κB regulated gene products in multiple myeloma xenograft mouse model. Oncotarget 2015; 5:634-48. [PMID: 24504138 PMCID: PMC3996662 DOI: 10.18632/oncotarget.1596] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) is a B cell malignancy characterized by clonal proliferation of plasma cells in the bone marrow. With the advent of novel targeted agents, the median survival rate has increased to 5−7 years. However, majority of patients with myeloma suffer relapse or develop chemoresistance to existing therapeutic agents. Thus, there is a need to develop novel alternative therapies for the treatment of MM. Thus in the present study, we investigated whether thymoquinone (TQ), a bioactive constituent of black seed oil, could suppress the proliferation and induce chemosensitization in human myeloma cells and xenograft mouse model. Our results show that TQ inhibited the proliferation of MM cells irrespective of their sensitivity to doxorubicin, melphalan or bortezomib. Interestingly, TQ treatment also resulted in a significant inhibition in the proliferation of CD138+ cells isolated from MM patient samples in a concentration dependent manner. TQ also potentiated the apoptotic effects of bortezomib in various MM cell lines through the activation of caspase-3, resulting in the cleavage of PARP. TQ treatment also inhibited chemotaxis and invasion induced by CXCL12 in MM cells. Furthermore, in a xenograft mouse model, TQ potentiated the antitumor effects of bortezomib (p < 0.05, vehicle versus bortezomib + TQ; p < 0.05, bortezomib versus bortezomib + TQ), and this correlated with modulation of various markers for survival and angiogenesis, such as Ki-67, vascular endothelial growth factor (VEGF), Bcl-2 and p65 expression. Overall, our results demonstrate that TQ can enhance the anticancer activity of bortezomib in vitro and in vivo and may have a substantial potential in the treatment of MM.
Collapse
|
35
|
Leoh LS, Morizono K, Kershaw KM, Chen ISY, Penichet ML, Daniels-Wells TR. Gene delivery in malignant B cells using the combination of lentiviruses conjugated to anti-transferrin receptor antibodies and an immunoglobulin promoter. J Gene Med 2014; 16:11-27. [PMID: 24436117 DOI: 10.1002/jgm.2754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 12/05/2013] [Accepted: 01/09/2014] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND We previously developed an antibody-avidin fusion protein (ch128.1Av) specific for the human transferrin receptor 1 (TfR1; CD71) to be used as a delivery vector for cancer therapy and showed that ch128.1Av delivers the biotinylated plant toxin saporin-6 into malignant B cells. However, as a result of widespread expression of TfR1, delivery of the toxin to normal cells is a concern. Therefore, we explored the potential of a dual targeted lentiviral-mediated gene therapy strategy to restrict gene expression to malignant B cells. Targeting occurs through the use of ch128.1Av or its parental antibody without avidin (ch128.1) and through transcriptional regulation using an immunoglobulin promoter. METHODS Flow cytometry was used to detect the expression of enhanced green fluorescent protein (EGFP) in a panel of cell lines. Cell viability after specific delivery of the therapeutic gene FCU1, a chimeric enzyme consisting of cytosine deaminase genetically fused to uracil phosphoribosyltransferse that converts the 5-fluorocytosine (5-FC) prodrug into toxic metabolites, was monitored using the MTS or WST-1 viability assay. RESULTS We found that EGFP was specifically expressed in a panel of human malignant B-cell lines, but not in human malignant T-cell lines. EGFP expression was observed in all cell lines when a ubiquitous promoter was used. Furthermore, we show the decrease of cell viability in malignant plasma cells in the presence of 5-FC and the FCU1 gene. CONCLUSIONS The present study demonstrates that gene expression can be restricted to malignant B cells and suggests that this dual targeted gene therapy strategy may help to circumvent the potential side effects of certain TfR1-targeted protein delivery approaches.
Collapse
Affiliation(s)
- Lai Sum Leoh
- Division of Surgical Oncology, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | | | | | | | | | |
Collapse
|
36
|
Offidani M, Corvatta L, Caraffa P, Gentili S, Maracci L, Leoni P. An evidence-based review of ixazomib citrate and its potential in the treatment of newly diagnosed multiple myeloma. Onco Targets Ther 2014; 7:1793-800. [PMID: 25302026 PMCID: PMC4189713 DOI: 10.2147/ott.s49187] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Proteasome inhibition represents one of the more important therapeutic targets in the treatment of multiple myeloma (MM), since by suppressing nuclear factor-κB activity, which promotes myelomagenesis, it makes plasma cells susceptible to proapoptotic signals. Bortezomib, the first proteasome inhibitor approved for MM therapy, has been shown to increase response rate and improve outcome in patients with relapsed/refractory disease and in the frontline setting, particularly when combined with immunomodulatory drugs and alkylating agents. Among second-generation proteasome inhibitors, ixazomib (MLN9708) is the first oral compound to be evaluated for the treatment of MM. Ixazomib has shown improved pharmacokinetic and pharmacodynamic parameters compared with bortezomib, in addition to similar efficacy in the control of myeloma growth and prevention of bone loss. Ixazomib was found to overcome bortezomib resistance and to trigger synergistic antimyeloma activity with dexamethasone, lenalidomide, and histone deacetylase inhibitors. Phase I/II studies using ixazomib weekly or twice weekly in relapsed/refractory MM patients suggested antitumor activity of the single agent, but more promising results have been obtained with the combination of ixazomib, lenalidomide, and dexamethasone in newly diagnosed MM. Ixazomib has also been used in systemic amyloidosis as a single agent, showing important activity in this difficult-to-treat plasma-cell dyscrasia. More frequent side effects observed during administration of ixazomib were thrombocytopenia, nausea, vomiting, diarrhea, fatigue, and rash, whereas severe peripheral neuropathy was rare. Here, we review the chemical characteristics of ixazomib, as well as its mechanism of action and results from preclinical and clinical trials.
Collapse
Affiliation(s)
- Massimo Offidani
- Hematology Department, Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Italy
| | - Laura Corvatta
- Division of Medicine, Ospedale Stelluti Scala, Fabriano, Italy
| | - Patrizia Caraffa
- Hematology Department, Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Italy
| | - Silvia Gentili
- Hematology Department, Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Italy
| | - Laura Maracci
- Hematology Department, Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Italy
| | - Pietro Leoni
- Hematology Department, Azienda Ospedaliero Universitaria Ospedali Riuniti di Ancona, Italy
| |
Collapse
|
37
|
Hao M, Zang M, Wendlandt E, Xu Y, An G, Gong D, Li F, Qi F, Zhang Y, Yang Y, Zhan F, Qiu L. Low serum miR-19a expression as a novel poor prognostic indicator in multiple myeloma. Int J Cancer 2014; 136:1835-44. [PMID: 25220540 DOI: 10.1002/ijc.29199] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 07/18/2014] [Accepted: 08/15/2014] [Indexed: 01/03/2023]
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy characterized by the clonal expansion of plasma cells. Despite continuing advances, novel biomarkers are needed for diagnosis and prognosis of MM. In our study, we characterized the diagnostic and prognostic potential of circulating microRNAs (miRNAs) in MM. Serum miRNA levels were analyzed in 108 newly diagnosed symptomatic MM patients and 56 healthy donors (HDs). Our analysis identified 95 dysregulated miRNAs in newly diagnosed MM patients. Of the 95 dysregulated miRNAs, dysregulation of miR-19a, miR-92a, miR-214-3p, miR-135b-5p, miR-4254, miR-3658 and miR-33b was confirmed by quantitative reverse transcription PCR (RT-qPCR). Receiver operating characteristic analysis revealed that a combination of miR-19a and miR-4254 can distinguish MM from HD with a sensitivity of 91.7% and specificity of 90.5%. Decreased expression of miR-19a was positively correlated with international staging system advancement, del(13q14) and 1q21 amplification. Furthermore, downregulation of miR-19a resulted in significantly decreased progression-free survival (PFS) and overall survival (OS). Our analysis indicated that the poor prognostic correlation of miR-19a expression was independent of genetic abnormalities in MM. Multivariate analysis revealed that miR-19a was a significant predictor of shortened PFS and OS. Interestingly, although miR-19a levels portend a poor prognosis, patients with low miR-19a levels had an improved response to bortezomib compared to those with high miR-19a profile. Patients with downregulated miR-19a experienced a significantly extended survival upon bortezomib-based therapy. These data demonstrate that the expression patterns of serum microRNAs are altered in MM, and miR-19a levels are a valuable prognostic marker to identify high-risk MM.
Collapse
Affiliation(s)
- Mu Hao
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Celik S, Tangi F, Oktenli C. Increased frequency of Mediterranean fever gene variants in multiple myeloma. Oncol Lett 2014; 8:1735-1738. [PMID: 25202401 PMCID: PMC4156200 DOI: 10.3892/ol.2014.2407] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 07/01/2014] [Indexed: 11/06/2022] Open
Abstract
High frequencies of inherited variants in the Mediterranean fever (MEFV) gene have been identified in patients with multiple myeloma (MM). The sample size of the present pilot study was small, therefore, the actual frequency of inherited variants in the MEFV gene could be investigated in patients with MM. Twenty-eight patients with MM and 65 healthy controls were included in the study. Six heterozygous and one homozygous (E148Q/E148Q) variant was identified in patients with MM. None of the patients had a family history compatible with familial Mediterranean fever. In the healthy control group, 11 heterozygous variants were identified. The difference in the overall frequency of the inherited variants in the MEFV gene between the MM patients and the controls was statistically significant (χ2=4.905; P=0.027). In conclusion, a high frequency of inherited variants in the MEFV gene was identified in patients with MM. Based on the current data, it is hypothesized that the MEFV gene is a cancer susceptibility gene. Additional evidence, such as familial aggregation, monozygotic versus dizygotic twin concordance, and tumors in genetically engineered model organisms, is required in order to support this hypothesis.
Collapse
Affiliation(s)
- Serkan Celik
- Division of Oncology, GATA Haydarpasa Training Hospital, Istanbul, Turkey
| | - Fatih Tangi
- Division of Internal Medicine, GATA Haydarpasa Training Hospital, Istanbul, Turkey
| | - Cagatay Oktenli
- Department of Internal Medicine, Anadolu Medical Center, Kocaeli, Turkey
| |
Collapse
|
39
|
Bone marrow stromal cell-fueled multiple myeloma growth and osteoclastogenesis are sustained by protein kinase CK2. Leukemia 2014; 28:2094-7. [PMID: 24897506 DOI: 10.1038/leu.2014.178] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
40
|
Tian T, Wang M, Ma D. TNF-α, a good or bad factor in hematological diseases? Stem Cell Investig 2014; 1:12. [PMID: 27358858 PMCID: PMC4923506 DOI: 10.3978/j.issn.2306-9759.2014.04.02] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 04/20/2014] [Indexed: 01/22/2023]
Abstract
Tumor necrosis factor-alpha (TNF-α) is a highly pleiotropic cytokine involved in a spectrum of physiological processes that control inflammation, anti-tumor responses and homeostasis through two receptors, TNF-R1 and TNF-R2. In general, TNF-R1 mediates cytotoxicity, resistance to infection and stimulation of NF-κB. By contrast, TNF-R2 has been implicated in proliferation of T-cell line, thymocytes and human mononuclear cells. Hematological malignancies are the types of cancer that affect normal hematopoiesis, have a speedy development, high lethal rate and until now still have no effective treatment. Several studies have shown that inflammatory cytokines play an important role in the onset and progress of these diseases. In this review, we summarize the recent studies and evaluate the positive or negative role of TNF-α in some hematological malignancies or diseases with a malignant tendency.
Collapse
Affiliation(s)
- Tian Tian
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Min Wang
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital, Shandong University, Jinan 250012, China
| |
Collapse
|
41
|
Gasparini C, Celeghini C, Monasta L, Zauli G. NF-κB pathways in hematological malignancies. Cell Mol Life Sci 2014; 71:2083-102. [PMID: 24419302 PMCID: PMC11113378 DOI: 10.1007/s00018-013-1545-4] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/13/2013] [Accepted: 12/17/2013] [Indexed: 12/22/2022]
Abstract
The nuclear factor κB or NF-κB transcription factor family plays a key role in several cellular functions, i.e. inflammation, apoptosis, cell survival, proliferation, angiogenesis, and innate and acquired immunity. The constitutive activation of NF-κB is typical of most malignancies and plays a major role in tumorigenesis. In this review, we describe NF-κB and its two pathways: the canonical pathway (RelA/p50) and the non-canonical pathway (RelB/p50 or RelB/p52). We then consider the role of the NF-κB subunits in the development and functional activity of B cells, T cells, macrophages and dendritic cells, which are the targets of hematological malignancies. The relevance of the two pathways is described in normal B and T cells and in hematological malignancies, acute and chronic leukemias (ALL, AML, CLL, CML), B lymphomas (DLBCLs, Hodgkin's lymphoma), T lymphomas (ATLL, ALCL) and multiple myeloma. We describe the interaction of NF-κB with the apoptotic pathways induced by TRAIL and the transcription factor p53. Finally, we discuss therapeutic anti-tumoral approaches as mono-therapies or combination therapies aimed to block NF-κB activity and to induce apoptosis (PARAs and Nutlin-3).
Collapse
Affiliation(s)
- Chiara Gasparini
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Via dell'Istria 65/1, 34137, Trieste, Italy,
| | | | | | | |
Collapse
|
42
|
Nuclear factor-κB is involved in the protocadherin-10-mediated pro-apoptotic effect in multiple myeloma. Mol Med Rep 2014; 10:832-8. [PMID: 24888369 DOI: 10.3892/mmr.2014.2285] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 04/10/2014] [Indexed: 11/05/2022] Open
Abstract
The gene encoding protocadherin-10 (PCDH10), a member of the cadherin superfamily, has been recently identified as a tumor suppressor gene (TSG). PCDH10 plays important roles in the apoptosis of tumor cells in some cancer types. However, the exact role of PCDH10 in multiple myeloma (MM) is largely unknown. Increasing evidence has suggested that the activation of nuclear factor-κB (NF-κB) is crucial for apoptosis in myeloma cells. In this study, we investigated the pro-apoptotic effect of PCDH10 on myeloma cells and whether this effect may involve inhibition of the NF-κB pathway. We report here, for the first time to the best of our knowledge, that PCDH10 markedly induces apoptosis of myeloma cells, accompanied by an increase in activated caspase-3 and poly-ADP‑ribose polymerase (PARP) levels, and inhibited expression of anti‑apoptotic proteins. We also demonstrate that PCDH10 inhibits the activation of NF-κB, by inhibiting the expression of the inhibitor of nuclear factor-κB (IκB) kinase subunits (IKKs) and the phosphorylation of IκBα. Moreover, the constitutive NF-κB DNA-binding activity and the expression of the NF-κB‑regulated proteins cyclooxygenase-2 (COX-2), vascular endothelial growth factor (VEGF) and intercellular adhesion molecule 1 (ICAM-1) were inhibited by PCDH10 in MM cells. These results suggest that PCDH10 induces myeloma cell apoptosis, probably by inhibiting the NF-κB pathway.
Collapse
|
43
|
Mechanisms and consequences of constitutive NF-κB activation in B-cell lymphoid malignancies. Oncogene 2014; 33:5655-65. [DOI: 10.1038/onc.2013.565] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/09/2013] [Accepted: 12/09/2013] [Indexed: 12/13/2022]
|
44
|
Salazar L, Kashiwada T, Krejci P, Meyer AN, Casale M, Hallowell M, Wilcox WR, Donoghue DJ, Thompson LM. Fibroblast growth factor receptor 3 interacts with and activates TGFβ-activated kinase 1 tyrosine phosphorylation and NFκB signaling in multiple myeloma and bladder cancer. PLoS One 2014; 9:e86470. [PMID: 24466111 PMCID: PMC3900522 DOI: 10.1371/journal.pone.0086470] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 12/09/2013] [Indexed: 12/31/2022] Open
Abstract
Cancer is a major public health problem worldwide. In the United States alone, 1 in 4 deaths is due to cancer and for 2013 a total of 1,660,290 new cancer cases and 580,350 cancer-related deaths are projected. Comprehensive profiling of multiple cancer genomes has revealed a highly complex genetic landscape in which a large number of altered genes, varying from tumor to tumor, impact core biological pathways and processes. This has implications for therapeutic targeting of signaling networks in the development of treatments for specific cancers. The NFκB transcription factor is constitutively active in a number of hematologic and solid tumors, and many signaling pathways implicated in cancer are likely connected to NFκB activation. A critical mediator of NFκB activity is TGFβ-activated kinase 1 (TAK1). Here, we identify TAK1 as a novel interacting protein and target of fibroblast growth factor receptor 3 (FGFR3) tyrosine kinase activity. We further demonstrate that activating mutations in FGFR3 associated with both multiple myeloma and bladder cancer can modulate expression of genes that regulate NFκB signaling, and promote both NFκB transcriptional activity and cell adhesion in a manner dependent on TAK1 expression in both cancer cell types. Our findings suggest TAK1 as a potential therapeutic target for FGFR3-associated cancers, and other malignancies in which TAK1 contributes to constitutive NFκB activation.
Collapse
MESH Headings
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Cell Adhesion
- Cell Proliferation
- Gene Expression Profiling
- Humans
- Immunoprecipitation
- MAP Kinase Kinase Kinases/genetics
- MAP Kinase Kinase Kinases/metabolism
- Multiple Myeloma/genetics
- Multiple Myeloma/metabolism
- Multiple Myeloma/pathology
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Oligonucleotide Array Sequence Analysis
- Peptide Fragments
- Phosphorylation
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Tumor Cells, Cultured
- Two-Hybrid System Techniques
- Tyrosine/metabolism
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/pathology
Collapse
Affiliation(s)
- Lisa Salazar
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California, United States of America
| | - Tamara Kashiwada
- Department of Biological Chemistry, University of California Irvine, Irvine, California, United States of America
| | - Pavel Krejci
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Institute of Experimental Biology, Masaryk University and Department of Cytokinetics, Institute of Biophysics AS CR, v.v.i., Brno, Czech Republic
- Department of Pediatrics, UCLA School of Medicine, Los Angeles, California, United States of America
| | - April N. Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Malcolm Casale
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
| | - Matthew Hallowell
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California, United States of America
| | - William R. Wilcox
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Pediatrics, UCLA School of Medicine, Los Angeles, California, United States of America
| | - Daniel J. Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
- Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Leslie Michels Thompson
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, California, United States of America
- Department of Biological Chemistry, University of California Irvine, Irvine, California, United States of America
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, California, United States of America
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
45
|
Khan RZ, Badros A. Role of carfilzomib in the treatment of multiple myeloma. Expert Rev Hematol 2014; 5:361-72. [DOI: 10.1586/ehm.12.26] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
46
|
Meyer AN, Drafahl KA, McAndrew CW, Gilda JE, Gallo LH, Haas M, Brill LM, Donoghue DJ. Tyrosine phosphorylation allows integration of multiple signaling inputs by IKKβ. PLoS One 2014; 8:e84497. [PMID: 24386391 PMCID: PMC3873999 DOI: 10.1371/journal.pone.0084497] [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: 09/26/2013] [Accepted: 11/14/2013] [Indexed: 02/06/2023] Open
Abstract
Signaling regulated by NFκB and related transcription factors is centrally important to many inflammatory and autoimmune diseases, cancer, and stress responses. The kinase that directly regulates the canonical NFκB transcriptional pathway, Inhibitor of κB kinase β (IKKβ), undergoes activation by Ser phosphorylation mediated by NIK or TAK1 in response to inflammatory signals. Using titanium dioxide-based phosphopeptide enrichment (TiO2)-liquid chromatography (LC)-high mass accuracy tandem mass spectrometry (MS/MS), we analyzed IKKβ phosphorylation in human HEK293 cells expressing IKKβ and FGFR2, a Receptor tyrosine kinase (RTK) essential for embryonic differentiation and dysregulated in several cancers. We attained unusually high coverage of IKKβ, identifying an abundant site of Tyr phosphorylation at Tyr169 within the Activation Loop. The phosphomimic at this site confers a level of kinase activation and NFκB nuclear localization exceeding the iconic mutant S177E/S181E, demonstrating that RTK-mediated Tyr phosphorylation of IKKβ has the potential to directly regulate NFκB transcriptional activation.
Collapse
Affiliation(s)
- April N. Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Kristine A. Drafahl
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Christopher W. McAndrew
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Jennifer E. Gilda
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Leandro H. Gallo
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Martin Haas
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Laurence M. Brill
- Sanford-Burnham Medical Research Institute, La Jolla, California, United States of America
| | - Daniel J. Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
- Moores UCSD Cancer Center, University of California San Diego, La Jolla, California, United States of America
- *
| |
Collapse
|
47
|
Morelli MB, Offidani M, Alesiani F, Discepoli G, Liberati S, Olivieri A, Santoni M, Santoni G, Leoni P, Nabissi M. The effects of cannabidiol and its synergism with bortezomib in multiple myeloma cell lines. A role for transient receptor potential vanilloid type-2. Int J Cancer 2013; 134:2534-46. [DOI: 10.1002/ijc.28591] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 10/22/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Maria Beatrice Morelli
- Section of Experimental Medicine, School of Pharmacy; University of Camerino; Camerino Italy
| | - Massimo Offidani
- Clinica di Ematologia, Azienda Ospedaliero - Universitaria Ospedali Riuniti di Ancona; Ancona Italy
| | - Francesco Alesiani
- Unità di Oncoematologia, Ospedale di San Severino; San Severino Marche Italy
| | - Giancarlo Discepoli
- Laboratorio di Genetica Medica, Clinica di Pediatria, Ospedali Riuniti di Ancona; Ancona Italy
| | - Sonia Liberati
- Department of Molecular Medicine; Sapienza University; Rome Italy
| | - Attilio Olivieri
- Clinica di Ematologia, Azienda Ospedaliero - Universitaria Ospedali Riuniti di Ancona; Ancona Italy
| | - Matteo Santoni
- Section of Experimental Medicine, School of Pharmacy; University of Camerino; Camerino Italy
| | - Giorgio Santoni
- Section of Experimental Medicine, School of Pharmacy; University of Camerino; Camerino Italy
| | - Pietro Leoni
- Clinica di Ematologia, Azienda Ospedaliero - Universitaria Ospedali Riuniti di Ancona; Ancona Italy
| | - Massimo Nabissi
- Section of Experimental Medicine, School of Pharmacy; University of Camerino; Camerino Italy
| |
Collapse
|
48
|
Yarde DN, Nace RA, Russell SJ. Oncolytic vesicular stomatitis virus and bortezomib are antagonistic against myeloma cells in vitro but have additive anti-myeloma activity in vivo. Exp Hematol 2013; 41:1038-49. [PMID: 24067362 DOI: 10.1016/j.exphem.2013.09.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 08/18/2013] [Accepted: 09/09/2013] [Indexed: 11/26/2022]
Abstract
Multiple myeloma cells are highly sensitive to the oncolytic effects of vesicular stomatitis virus (VSV), which specifically targets and kills cancer cells. Myeloma cells are also exquisitely sensitive to the cytotoxic effects of the clinically approved proteasome inhibitor bortezomib. Therefore, we sought to determine whether the combination of VSV and bortezomib would enhance tumor cell killing. However, as shown here, combining these two agents in vitro results in antagonism. We show that bortezomib inhibits VSV replication and spread. We found that bortezomib inhibits VSV-induced NF-κB activation and, using the NF-κB-specific inhibitor BMS-345541, that VSV requires NF-κB activity to spread efficiently in myeloma cells. In contrast to other cancer cell lines, viral titer is not recovered by BMS-345541 when myeloma cells are pretreated with interferon β. Thus, inhibiting NF-κB activity, either with bortezomib or BMS-345541, results in reduced VSV titers in myeloma cells in vitro. However, when VSV and bortezomib are combined in vivo in two syngeneic, immunocompetent myeloma models, the combination reduces tumor burden to a greater degree than VSV does as a single agent. Intratumoral VSV viral load is unchanged when mice are treated concomitantly with bortezomib compared to VSV treatment alone. To our knowledge, this report is the first to analyze the combination of VSV and bortezomib in vivo. Although antagonism between VSV and bortezomib is seen in vitro, analyzing these cells in the context of their host environment shows that bortezomib enhances VSV response, suggesting that this combination will also enhance response in myeloma patients.
Collapse
Affiliation(s)
- Danielle N Yarde
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | |
Collapse
|
49
|
Aurora and IKK kinases cooperatively interact to protect multiple myeloma cells from Apo2L/TRAIL. Blood 2013; 122:2641-53. [PMID: 23974204 DOI: 10.1182/blood-2013-02-482356] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Constitutive activation of the canonical and noncanonical nuclear factor-κB (NF-κB) pathways is frequent in multiple myeloma (MM) and can compromise sensitivity to TRAIL. In this study, we demonstrate that Aurora kinases physically and functionally interact with the key regulators of canonical and noncanonical NF-κB pathways IκB kinase β (IKKβ) and IKKα to activate NF-κB in MM, and the pharmacological blockade of Aurora kinase activity induces TRAIL sensitization in MM because it abrogates TRAIL-induced activation of NF-κB. We specifically found that TRAIL induces prosurvival signaling by increasing the phosphorylation state of both Aurora and IKK kinases and their physical interactions, and the blockade of Aurora kinase activity by pan-Aurora kinase inhibitors (pan-AKIs) disrupts TRAIL-induced survival signaling by effectively reducing Aurora-IKK kinase interactions and NF-κB activation. Pan-AKIs consistently blocked TRAIL induction of the antiapoptotic NF-κB target genes A1/Bfl-1 and/or Mcl-1, both important targets for TRAIL sensitization in MM cells. In summary, these results identify a novel interaction between Aurora and IKK kinases and show that these pathways can cooperate to promote TRAIL resistance. Finally, combining pan-AKIs with TRAIL in vivo showed dramatic efficacy in a multidrug-resistant human myeloma xenograft model. These findings suggest that combining Aurora kinase inhibitors with TRAIL may have therapeutic benefit in MM.
Collapse
|
50
|
Cormier F, Monjanel H, Fabre C, Billot K, Sapharikas E, Chereau F, Bordereaux D, Molina TJ, Avet-Loiseau H, Baud V. Frequent engagement of RelB activation is critical for cell survival in multiple myeloma. PLoS One 2013; 8:e59127. [PMID: 23555623 PMCID: PMC3610937 DOI: 10.1371/journal.pone.0059127] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 02/11/2013] [Indexed: 12/17/2022] Open
Abstract
The NF-κB family of transcription factors has emerged as a key player in the pathogenesis of multiple myeloma (MM). NF-κB is activated by at least two major signaling pathways. The classical pathway results in the activation of mainly RelA containing dimers, whereas the alternative pathway leads to the activation of RelB/p52 and RelB/p50 heterodimers. Activating mutations in regulators of the alternative pathway have been identified in 17% of MM patients. However, the status of RelB activation per se and its role in the regulation of cell survival in MM has not been investigated. Here, we reveal that 40% of newly diagnosed MM patients have a constitutive RelB DNA-binding activity in CD138(+) tumor cells, and we show an association with increased expression of a subset of anti-apoptotic NF-κB target genes, such as cIAP2. Furthermore, we demonstrate that RelB exerts a crucial anti-apoptotic activity in MM cells. Our findings indicate that RelB activation is key for promoting MM cell survival through the upregulation of anti-apoptotic proteins. Altogether, our study provides the framework for the development of new molecules targeting RelB in the treatment of MM.
Collapse
|