1
|
Deng H, Han Y, Liu L, Zhang H, Liu D, Wen J, Huang M, Zhao L. Targeting Myeloid Leukemia-1 in Cancer Therapy: Advances and Directions. J Med Chem 2024; 67:5963-5998. [PMID: 38597264 DOI: 10.1021/acs.jmedchem.3c01998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
As a tripartite cell death switch, B-cell lymphoma protein 2 (Bcl-2) family members precisely regulate the endogenous apoptosis pathway in response to various cell signal stresses through protein-protein interactions. Myeloid leukemia-1 (Mcl-1), a key anti-apoptotic Bcl-2 family member, is positioned downstream in the endogenous apoptotic pathway and plays a central role in regulating mitochondrial function. Mcl-1 is highly expressed in a variety of hematological malignancies and solid tumors, contributing to tumorigenesis, poor prognosis, and chemoresistance, making it an attractive target for cancer treatment. This Perspective aims to discuss the mechanism by which Mcl-1 regulates apoptosis and non-apoptotic functions in cancer cells and to outline the discovery and optimization process of potent Mcl-1 modulators. In addition, we summarize the structural characteristics of potent inhibitors that bind to Mcl-1 through multiple co-crystal structures and analyze the cardiotoxicity caused by current Mcl-1 inhibitors, providing prospects for rational targeting of Mcl-1.
Collapse
Affiliation(s)
- Hongguang Deng
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Han
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Liang Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hong Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiachen Wen
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Min Huang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
2
|
Booth L, Roberts JL, West C, Dent P. GZ17-6.02 interacts with proteasome inhibitors to kill multiple myeloma cells. Oncotarget 2024; 15:159-174. [PMID: 38441437 PMCID: PMC10913917 DOI: 10.18632/oncotarget.28558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/23/2024] [Indexed: 03/07/2024] Open
Abstract
GZ17-6.02, a synthetically manufactured compound containing isovanillin, harmine and curcumin, has undergone phase I evaluation in patients with solid tumors (NCT03775525) with a recommended phase 2 dose (RP2D) of 375 mg PO BID. GZ17-6.02 was more efficacious as a single agent at killing multiple myeloma cells than had previously been observed in solid tumor cell types. GZ17-6.02 interacted with proteasome inhibitors in a greater than additive fashion to kill myeloma cells and alone it killed inhibitor-resistant cells to a similar extent. The drug combination of GZ17-6.02 and bortezomib activated ATM, the AMPK and PERK and inactivated ULK1, mTORC1, eIF2α, NFκB and the Hippo pathway. The combination increased ATG13 S318 phosphorylation and the expression of Beclin1, ATG5, BAK and BIM, and reduced the levels of BCL-XL and MCL1. GZ17-6.02 interacted with bortezomib to enhance autophagosome formation and autophagic flux, and knock down of ATM, AMPKα, ULK1, Beclin1 or ATG5 significantly reduced both autophagy and tumor cell killing. Knock down of BAK and BIM significantly reduced tumor cell killing. The expression of HDACs1/2/3 was significantly reduced beyond that previously observed in solid tumor cells and required autophagy. This was associated with increased acetylation and methylation of histone H3. Combined knock down of HDACs1/2/3 caused activation of ATM and the AMPK and caused inactivation of ULK1, mTORC1, NFκB and the Hippo pathway. HDAC knock down also enhanced ATG13 phosphorylation, increased BAK levels and reduced those of BCL-XL. Collectively, our present studies support performing additional in vivo studies with multiple myeloma cells.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jane L. Roberts
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Cameron West
- Genzada Pharmaceuticals, Hutchinson, KS 67502, USA
| | - Paul Dent
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
3
|
Schwarzer A, Oliveira M, Kleppa MJ, Slattery SD, Anantha A, Cooper A, Hannink M, Schambach A, Dörrie A, Kotlyarov A, Gaestel M, Hembrough T, Levine J, Luther M, Stocum M, Stiles L, Weinstock DM, Liesa M, Kostura MJ. Targeting Aggressive B-cell Lymphomas through Pharmacological Activation of the Mitochondrial Protease OMA1. Mol Cancer Ther 2023; 22:1290-1303. [PMID: 37643767 PMCID: PMC10723637 DOI: 10.1158/1535-7163.mct-22-0718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 07/02/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023]
Abstract
DLBCL are aggressive, rapidly proliferating tumors that critically depend on the ATF4-mediated integrated stress response (ISR) to adapt to stress caused by uncontrolled growth, such as hypoxia, amino acid deprivation, and accumulation of misfolded proteins. Here, we show that ISR hyperactivation is a targetable liability in DLBCL. We describe a novel class of compounds represented by BTM-3528 and BTM-3566, which activate the ISR through the mitochondrial protease OMA1. Treatment of tumor cells with compound leads to OMA1-dependent cleavage of DELE1 and OPA1, mitochondrial fragmentation, activation of the eIF2α-kinase HRI, cell growth arrest, and apoptosis. Activation of OMA1 by BTM-3528 and BTM-3566 is mechanistically distinct from inhibitors of mitochondrial electron transport, as the compounds induce OMA1 activity in the absence of acute changes in respiration. We further identify the mitochondrial protein FAM210B as a negative regulator of BTM-3528 and BTM-3566 activity. Overexpression of FAM210B prevents both OMA1 activation and apoptosis. Notably, FAM210B expression is nearly absent in healthy germinal center B-lymphocytes and in derived B-cell malignancies, revealing a fundamental molecular vulnerability which is targeted by BTM compounds. Both compounds induce rapid apoptosis across diverse DLBCL lines derived from activated B-cell, germinal center B-cell, and MYC-rearranged lymphomas. Once-daily oral dosing of BTM-3566 resulted in complete regression of xenografted human DLBCL SU-DHL-10 cells and complete regression in 6 of 9 DLBCL patient-derived xenografts. BTM-3566 represents a first-of-its kind approach of selectively hyperactivating the mitochondrial ISR for treating DLBCL.
Collapse
Affiliation(s)
- Adrian Schwarzer
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostaseology, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Matheus Oliveira
- Department of Medicine, Endocrinology, UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Marc-Jens Kleppa
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | | | - Andy Anantha
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Alan Cooper
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Mark Hannink
- Biochemistry Department, Life Sciences Center and Ellis Fischel Cancer Center, University of Missouri, Columbia, Missouri
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Anneke Dörrie
- Department of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Alexey Kotlyarov
- Department of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Matthias Gaestel
- Department of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Todd Hembrough
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Jedd Levine
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Michael Luther
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Michael Stocum
- Bantam Pharmaceutical, Research Triangle Park, North Carolina
| | - Linsey Stiles
- Department of Medicine, Endocrinology, UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | - Marc Liesa
- Department of Medicine, Endocrinology, UCLA, David Geffen School of Medicine at UCLA, Los Angeles, California
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona, Catalonia, Spain
| | | |
Collapse
|
4
|
Liu Z, Xu X, Liu K, Zhang J, Ding D, Fu R. Immunogenic Cell Death in Hematological Malignancy Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207475. [PMID: 36815385 PMCID: PMC10161053 DOI: 10.1002/advs.202207475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/09/2023] [Indexed: 05/06/2023]
Abstract
Although the curative effect of hematological malignancies has been improved in recent years, relapse or drug resistance of hematological malignancies will eventually recur. Furthermore, the microenvironment disorder is an important mechanism in the pathogenesis of hematological malignancies. Immunogenic cell death (ICD) is a unique mechanism of regulated cell death (RCD) that triggers an intact antigen-specific adaptive immune response by firing a set of danger signals or damage-associated molecular patterns (DAMPs), which is an immunotherapeutic modality with the potential for the treatment of hematological malignancies. This review summarizes the existing knowledge about the induction of ICD in hematological malignancies and the current research on combining ICD inducers with other treatment strategies for hematological malignancies.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Xintong Xu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| | - Kaining Liu
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Jingtian Zhang
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical BiologyKey Laboratory of Bioactive, Materials, Ministry of Education and College of Life SciencesNankai UniversityTianjin300071P. R. China
| | - Rong Fu
- Department of HematologyTianjin Medical University General HospitalTianjin300052P. R. China
| |
Collapse
|
5
|
Negligible role of TRAIL death receptors in cell death upon endoplasmic reticulum stress in B-cell malignancies. Oncogenesis 2023; 12:6. [PMID: 36755015 PMCID: PMC9908905 DOI: 10.1038/s41389-023-00450-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
Impairments in protein folding in the endoplasmic reticulum (ER) lead to a condition called ER stress, which can trigger apoptosis via the mitochondrial or the death receptor (extrinsic) pathway. There is controversy concerning involvement of the death receptor (DR)4 and DR5-Caspase-8 -Bid pathway in ER stress-mediated cell death, and this axis has not been fully studied in B-cell malignancies. Using three B-cell lines from Mantle Cell Lymphoma, Waldenström's macroglobulinemia and Multiple Myeloma origins, we engineered a set of CRISPR KOs of key components of these cell death pathways to address this controversy. We demonstrate that DR4 and/or DR5 are essential for killing via TRAIL, however, they were dispensable for ER-stress induced-cell death, by Thapsigargin, Brefeldin A or Bortezomib, as were Caspase-8 and Bid. In contrast, the deficiency of Bax and Bak fully protected from ER stressors. Caspase-8 and Bid were cleaved upon ER-stress stimulation, but this was DR4/5 independent and rather a result of mitochondrial-induced feedback loop subsequent to Bax/Bak activation. Finally, combined activation of the ER-stress and TRAIL cell-death pathways was synergistic with putative clinical relevance for B-cell malignancies.
Collapse
|
6
|
Endoplasmic Reticulum Stress Signaling and Neuronal Cell Death. Int J Mol Sci 2022; 23:ijms232315186. [PMID: 36499512 PMCID: PMC9740965 DOI: 10.3390/ijms232315186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022] Open
Abstract
Besides protein processing, the endoplasmic reticulum (ER) has several other functions such as lipid synthesis, the transfer of molecules to other cellular compartments, and the regulation of Ca2+ homeostasis. Before leaving the organelle, proteins must be folded and post-translationally modified. Protein folding and revision require molecular chaperones and a favorable ER environment. When in stressful situations, ER luminal conditions or chaperone capacity are altered, and the cell activates signaling cascades to restore a favorable folding environment triggering the so-called unfolded protein response (UPR) that can lead to autophagy to preserve cell integrity. However, when the UPR is disrupted or insufficient, cell death occurs. This review examines the links between UPR signaling, cell-protective responses, and death following ER stress with a particular focus on those mechanisms that operate in neurons.
Collapse
|
7
|
Wang G, Fan F, Sun C, Hu Y. Looking into Endoplasmic Reticulum Stress: The Key to Drug-Resistance of Multiple Myeloma? Cancers (Basel) 2022; 14:5340. [PMID: 36358759 PMCID: PMC9654020 DOI: 10.3390/cancers14215340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/27/2022] [Indexed: 09/22/2023] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy, resulting from the clonal proliferation of malignant plasma cells within the bone marrow. Despite significant advances that have been made with novel drugs over the past two decades, MM patients often develop therapy resistance, especially to bortezomib, the first-in-class proteasome inhibitor that was approved for treatment of MM. As highly secretory monoclonal protein-producing cells, MM cells are characterized by uploaded endoplasmic reticulum stress (ERS), and rely heavily on the ERS response for survival. Great efforts have been made to illustrate how MM cells adapt to therapeutic stresses through modulating the ERS response. In this review, we summarize current knowledge on the mechanisms by which ERS response pathways influence MM cell fate and response to treatment. Moreover, based on promising results obtained in preclinical studies, we discuss the prospect of applying ERS modulators to overcome drug resistance in MM.
Collapse
Affiliation(s)
- Guangqi Wang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
| | - Fengjuan Fan
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
| | - Chunyan Sun
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yu Hu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Avenue 1277, Wuhan 430022, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
8
|
Benedetti R, Gilardini Montani MS, Romeo MA, Arena A, Santarelli R, D’Orazi G, Cirone M. Role of UPR Sensor Activation in Cell Death-Survival Decision of Colon Cancer Cells Stressed by DPE Treatment. Biomedicines 2021; 9:1262. [PMID: 34572447 PMCID: PMC8466673 DOI: 10.3390/biomedicines9091262] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 12/25/2022] Open
Abstract
Polyphenols have been shown to possess several beneficial properties, including properties involved in the prevention or treatment of cancer. Among these polyphenols, a leading role is played by dihydroxyphenylethanol (DPE), the most powerful antioxidant compound contained in the olive oil. DPE has been previously reported to induce endoplasmic reticulum (ER) stress and to reduce cell survival in colon cancer, one of the most common and aggressive cancers in developed countries. In this study, we further investigated the activation of UPR by DPE and explored the roles of the three UPR sensors, inositol-requiring enzyme (IRE) 1 alpha, protein kinase RNA-like endoplasmic reticulum kinase (PERK), and activating transcription factor (ATF6), in the cell death-survival decision of wt and mutp53 colon cancer cells and the underlying mechanisms involved. We also unveiled a new interplay between ATF6 and wt, as well as mutp53, which may have important implications in cancer therapy.
Collapse
Affiliation(s)
- Rossella Benedetti
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Maria Anele Romeo
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Andrea Arena
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Roberta Santarelli
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| | - Gabriella D’Orazi
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy;
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Mara Cirone
- Department of Experimental Medicine, La Sapienza University of Rome, Viale Regina Elena 324, 00185 Rome, Italy; (R.B.); (M.S.G.M.); (M.A.R.); (A.A.); (R.S.)
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, 00185 Rome, Italy
| |
Collapse
|
9
|
Li X, Wang Z, Zhang S, Yao Q, Chen W, Liu F. Ruxolitinib induces apoptosis of human colorectal cancer cells by downregulating the JAK1/2-STAT1-Mcl-1 axis. Oncol Lett 2021; 21:352. [PMID: 33747209 PMCID: PMC7967999 DOI: 10.3892/ol.2021.12613] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 12/14/2020] [Indexed: 01/09/2023] Open
Abstract
Under pathological conditions, the Janus kinase (JAK)/STAT signaling pathway can regulate the proliferation, differentiation and migration of tumor cells, including colorectal cancer (CRC). CRC is the third major types of cancer among males and the second among females worldwide. In China, CRC is the fifth common cancer among both males and females. Western blotting, flow cytometry, RNA interference, immunoprecipitation, xenografts models, and immunohistochemical staining were carried out to evaluate the possible mechanisms of acton of ruxolitinib. The present data suggested that ruxolitinib can suppress CRC cell proliferation by inducing apoptosis. Firstly, JAK1/2-STAT1 was identified as the target of ruxolitinib. Then, ruxolitinib downregulated myeloid cell leukemia-1 (Mcl-1) mRNA level and decreased its protein level, which enabled Bak to trigger CRC apoptosis. Furthermore, ruxolitinib exerted potent activity against CRC xenograft growth in vivo. High expression of phosphorylated STAT1 (S727) was also confirmed in 44 pairs of human colon carcinoma and adjacent normal tissues. Taken together, the results showed that ruxolitinib decreased JAK1/2-STAT1-Mcl-1 protein level and effectively suppressed CRC cell proliferation in vitro and in vivo. Therefore, ruxolitinib could be a promising anticancer agent for CRC treatment.
Collapse
Affiliation(s)
- Xia Li
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China.,Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China.,Zhejiang Cancer Research Institute, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, P.R. China
| | - Zhe Wang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Shengjie Zhang
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China.,Zhejiang Cancer Research Institute, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, P.R. China
| | - Qinghua Yao
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China.,Zhejiang Cancer Research Institute, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, P.R. China
| | - Wei Chen
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, P.R. China.,Zhejiang Cancer Research Institute, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, P.R. China
| | - Feiyan Liu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
10
|
Bai Y, Su X. Updates to the drug-resistant mechanism of proteasome inhibitors in multiple myeloma. Asia Pac J Clin Oncol 2020; 17:29-35. [PMID: 32920949 DOI: 10.1111/ajco.13459] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/21/2020] [Indexed: 12/11/2022]
Abstract
Proteasome inhibitors (PIs) have been a kind of backbone therapies for newly diagnosed as well as relapsed or refractory myeloma patients in the last two decades. Bortezomib, the first-in-class PI, was approved by the United States Food and Drug Administration in 2003. The key roles of this class of agents are targeting at the overstressed 26S proteasome, which are involved in the pathogenesis of the disease. Despite recent advancements in clinical antimyeloma treatment, the acquisition of resistance is a major limitation in PI therapy. This review aims at a better understanding of the pathways and biomarkers involved in MM drug resistance.
Collapse
Affiliation(s)
- Yang Bai
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Xing Su
- The Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| |
Collapse
|
11
|
Cosialls AM, Sánchez-Vera I, Pomares H, Perramon-Andújar J, Sanchez-Esteban S, Palmeri CM, Iglesias-Serret D, Saura-Esteller J, Núñez-Vázquez S, Lavilla R, González-Barca EM, Pons G, Gil J. The BCL-2 family members NOXA and BIM mediate fluorizoline-induced apoptosis in multiple myeloma cells. Biochem Pharmacol 2020; 180:114198. [PMID: 32798467 DOI: 10.1016/j.bcp.2020.114198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022]
Abstract
Fluorizoline is a new synthetic molecule that induces apoptosis by selectively targeting prohibitins. In this study, we have assessed the pro-apoptotic effect of fluorizoline in 3 different multiple myeloma cell lines and 12 primary samples obtained from treatment-naïve multiple myeloma patients. Fluorizoline induced apoptosis in both multiple myeloma cell lines and primary samples at concentrations in the low micromolar range. All primary samples were sensitive to fluorizoline. Moreover, fluorizoline increased the mRNA and protein levels of the pro-apoptotic BCL-2 family member NOXA both in cell lines and primary samples analyzed. Finally, NOXA-depletion by CRISPR/Cas9 in cells that do not express BIM conferred resistance to fluorizoline-induced apoptosis in multiple myeloma cells. These results suggest that targeting prohibitins could be a new therapeutic strategy for myeloma multiple.
Collapse
Affiliation(s)
- Ana M Cosialls
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Ismael Sánchez-Vera
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Helena Pomares
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain; Servei d'Hematologia Clínica, Institut Català d'Oncologia, Oncobell-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Judit Perramon-Andújar
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sandra Sanchez-Esteban
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Claudia M Palmeri
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Daniel Iglesias-Serret
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain; Facultat de Medicina, Universitat de Vic - Universitat Central de Catalunya (UVic- UCC), Vic, Barcelona, Spain
| | - José Saura-Esteller
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sonia Núñez-Vázquez
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rodolfo Lavilla
- Laboratory of Medical Chemistry, Faculty of Pharmacy and Food Sciences and Institute of Medicine (IBUB), University of Barcelona, Barcelona, Spain
| | - Eva M González-Barca
- Servei d'Hematologia Clínica, Institut Català d'Oncologia, Oncobell-IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Gabriel Pons
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Joan Gil
- Departament de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Oncobell-IDIBELL (Institut d'Investigació Biomèdica de Bellvitge), L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
12
|
Florent R, Weiswald LB, Lambert B, Brotin E, Abeilard E, Louis MH, Babin G, Poulain L, N'Diaye M. Bim, Puma and Noxa upregulation by Naftopidil sensitizes ovarian cancer to the BH3-mimetic ABT-737 and the MEK inhibitor Trametinib. Cell Death Dis 2020; 11:380. [PMID: 32424251 PMCID: PMC7235085 DOI: 10.1038/s41419-020-2588-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022]
Abstract
Ovarian cancer represents the first cause of mortality from gynecologic malignancies due to frequent chemoresistance occurrence. Increasing the [BH3-only Bim, Puma, Noxa proapoptotic]/[Bcl-xL, Mcl-1 antiapoptotic] proteins ratio was proven to efficiently kill ovarian carcinoma cells and development of new molecules to imbalance Bcl-2 member equilibrium are strongly required. Drug repurposing constitutes an innovative approach to rapidly develop therapeutic strategies through exploitation of established drugs already approved for the treatment of noncancerous diseases. This strategy allowed a renewed interest for Naftopidil, an α1-adrenergic receptor antagonist commercialized in Japan for benign prostatic hyperplasia. Naftopidil was reported to decrease the incidence of prostate cancer and its derivative was described to increase BH3-only protein expression in some cancer models. Based on these arguments, we evaluated the effects of Naftopidil on ovarian carcinoma and showed that Naftopidil reduced cell growth and increased the expression of the BH3-only proteins Bim, Puma and Noxa. This effect was independent of α1-adrenergic receptors blocking and involved ATF4 or JNK pathway depending on cellular context. Finally, Naftopidil-induced BH3-only members sensitized our models to ABT-737 and Trametinib treatments, in vitro as well as ex vivo, in patient-derived organoid models.
Collapse
Affiliation(s)
- Romane Florent
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Louis-Bastien Weiswald
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Bernard Lambert
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
- CNRS-Regional Delegation of Normandy, Caen, France
| | - Emilie Brotin
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Edwige Abeilard
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Marie-Hélène Louis
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Guillaume Babin
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
| | - Laurent Poulain
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France
- UNICANCER, Cancer Center François Baclesse, Caen, France
- Biological Ressources Center «OvaRessources», Cancer Center François Baclesse, Caen, France
| | - Monique N'Diaye
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Axis (Biology and Innovative Therapeutics for Ovarian Cancers), Caen, France.
- UNICANCER, Cancer Center François Baclesse, Caen, France.
| |
Collapse
|
13
|
Senichkin VV, Streletskaia AY, Zhivotovsky B, Kopeina GS. Molecular Comprehension of Mcl-1: From Gene Structure to Cancer Therapy. Trends Cell Biol 2019; 29:549-562. [DOI: 10.1016/j.tcb.2019.03.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 01/19/2023]
|
14
|
Serrano-Del Valle A, Anel A, Naval J, Marzo I. Immunogenic Cell Death and Immunotherapy of Multiple Myeloma. Front Cell Dev Biol 2019; 7:50. [PMID: 31041312 PMCID: PMC6476910 DOI: 10.3389/fcell.2019.00050] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/19/2019] [Indexed: 12/24/2022] Open
Abstract
Over the past decades, immunotherapy has demonstrated a prominent clinical efficacy in a wide variety of human tumors. For many years, apoptosis has been considered a non-immunogenic or tolerogenic process whereas necrosis or necroptosis has long been acknowledged to play a key role in inflammation and immune-related processes. However, the new concept of “immunogenic cell death” (ICD) has challenged this traditional view and has granted apoptosis with immunogenic abilities. This paradigm shift offers clear implications in designing novel anti-cancer therapeutic approaches. To date, several screening studies have been carried out to discover bona fide ICD inducers and reveal the inherent capacity of a wide variety of drugs to induce cell death-associated exposure of danger signals and to bring about in vivo anti-cancer immune responses. Recent shreds of evidence place ER stress at the core of all the scenarios where ICD occur. Furthermore, ER stress and the unfolded protein response (UPR) have emerged as important targets in different human cancers. Notably, in multiple myeloma (MM), a lethal plasma cell disorder, the elevated production of immunoglobulins leaves these cells heavily reliant on the survival arm of the UPR. For that reason, drugs that disrupt ER homeostasis and engage ER stress-associated cell death, such as proteasome inhibitors, which are currently used for the treatment of MM, as well as novel ER stressors are intended to be promising therapeutic agents in MM. This not only holds true for their capacity to induce cell death, but also to their potential ability to activate the immunogenic arm of the ER stress response, with the ensuing exposure of danger signals. We provide here an overview of the up-to-date knowledge regarding the cell death mechanisms involved in situations of ER stress with a special focus on the connections with the drug-induced ER stress pathways that evoke ICD. We will also discuss how this could assist in optimizing and developing better immunotherapeutic approaches, especially in MM treatment.
Collapse
Affiliation(s)
| | - Alberto Anel
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| | - Javier Naval
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| | - Isabel Marzo
- Department of Biochemistry and Molecular and Cell Biology, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
15
|
Song P, Yang S, Hua H, Zhang H, Kong Q, Wang J, Luo T, Jiang Y. The regulatory protein GADD34 inhibits TRAIL-induced apoptosis via TRAF6/ERK-dependent stabilization of myeloid cell leukemia 1 in liver cancer cells. J Biol Chem 2019; 294:5945-5955. [PMID: 30782845 DOI: 10.1074/jbc.ra118.006029] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 02/09/2019] [Indexed: 02/05/2023] Open
Abstract
GADD34 (growth arrest and DNA damage-inducible gene 34) plays a critical role in responses to DNA damage and endoplasmic reticulum stress. GADD34 has opposing effects on different stimuli-induced cell apoptosis events, but the reason for this is unclear. Here, using immunoblotting analyses and various molecular genetic approaches in HepG2 and SMMC-7721 cells, we demonstrate that GADD34 protects hepatocellular carcinoma (HCC) cells from tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by stabilizing a BCL-2 family member, myeloid cell leukemia 1 (MCL-1). We found that GADD34 knockdown decreased MCL-1 levels and that GADD34 overexpression up-regulated MCL-1 expression in HCC cells. GADD34 did not affect MCL-1 transcription but enhanced MCL-1 protein stability. The proteasome inhibitor MG132 abrogated GADD34 depletion-induced MCL-1 down-regulation, suggesting that GADD34 inhibits the proteasomal degradation of MCL-1. Furthermore, GADD34 overexpression promoted extracellular signal-regulated kinase (ERK) phosphorylation through a signaling axis that consists of the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor 6 (TRAF6) and transforming growth factor-β-activated kinase 1 (MAP3K7)-binding protein 1 (TAB1), which mediated the up-regulation of MCL-1 by GADD34. Of note, TRAIL up-regulated both GADD34 and MCL-1 levels, and knockdown of GADD34 and TRAF6 suppressed the induction of MCL-1 by TRAIL. Correspondingly, GADD34 knockdown potentiated TRAIL-induced apoptosis, and MCL-1 overexpression rescued TRAIL-treated and GADD34-depleted HCC cells from cell death. Taken together, these findings suggest that GADD34 inhibits TRAIL-induced HCC cell apoptosis through TRAF6- and ERK-mediated stabilization of MCL-1.
Collapse
Affiliation(s)
- Peiying Song
- From the State Key Laboratory of Biotherapy, Section of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041
| | - Songpeng Yang
- From the State Key Laboratory of Biotherapy, Section of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041
| | - Hui Hua
- the Laboratory of Stem Cell Biology, West China Hospital, Chengdu 610041
| | - Hongying Zhang
- From the State Key Laboratory of Biotherapy, Section of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041
| | - Qingbin Kong
- From the State Key Laboratory of Biotherapy, Section of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041
| | - Jiao Wang
- the School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 610075
| | - Ting Luo
- the Cancer Center, West China Hospital, Chengdu 610041, China
| | - Yangfu Jiang
- From the State Key Laboratory of Biotherapy, Section of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041.
| |
Collapse
|
16
|
Targeting KPNB1 overcomes TRAIL resistance by regulating DR5, Mcl-1 and FLIP in glioblastoma cells. Cell Death Dis 2019; 10:118. [PMID: 30742128 PMCID: PMC6370806 DOI: 10.1038/s41419-019-1383-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/16/2018] [Accepted: 01/09/2019] [Indexed: 01/08/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine with potential anticancer effect, but innate and adaptive TRAIL resistance in majority of cancers limit its clinical application. Karyopherin β1 (KPNB1) inhibition in cancer cells has been reported to abrogate the nuclear import of TRAIL receptor DR5 and facilitate its localization on the cell surface ready for TRAIL stimulation. However, our study reveals a more complicated mechanism. Genetic or pharmacological inhibition of KPNB1 potentiated TRAIL-induced apoptosis selectively in glioblastoma cells mainly by unfolded protein response (UPR). First, it augmented ATF4-mediated DR5 expression and promoted the assembly of death-inducing signaling complex (DISC). Second, it freed Bax and Bak from Mcl-1. Third, it downregulated FLIPL and FLIPS, inhibitors of caspase-8 cleavage, partly through upregulating ATF4–induced 4E-BP1 expression and disrupting the cap-dependent translation initiation. Meanwhile, KPNB1 inhibition-induced undesirable autophagy and accelerated cleaved caspase-8 clearance. Inhibition of autophagic flux maintained cleaved caspase-8 and aggravated apoptosis induced by KPNB1 inhibitor plus TRAIL, which were abolished by caspase-8 inhibitor. These results unveil new molecular mechanism for optimizing TRAIL-directed therapeutic efficacy against cancer.
Collapse
|
17
|
Almanza A, Carlesso A, Chintha C, Creedican S, Doultsinos D, Leuzzi B, Luís A, McCarthy N, Montibeller L, More S, Papaioannou A, Püschel F, Sassano ML, Skoko J, Agostinis P, de Belleroche J, Eriksson LA, Fulda S, Gorman AM, Healy S, Kozlov A, Muñoz‐Pinedo C, Rehm M, Chevet E, Samali A. Endoplasmic reticulum stress signalling - from basic mechanisms to clinical applications. FEBS J 2019; 286:241-278. [PMID: 30027602 PMCID: PMC7379631 DOI: 10.1111/febs.14608] [Citation(s) in RCA: 566] [Impact Index Per Article: 113.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 06/24/2018] [Accepted: 07/18/2018] [Indexed: 02/06/2023]
Abstract
The endoplasmic reticulum (ER) is a membranous intracellular organelle and the first compartment of the secretory pathway. As such, the ER contributes to the production and folding of approximately one-third of cellular proteins, and is thus inextricably linked to the maintenance of cellular homeostasis and the fine balance between health and disease. Specific ER stress signalling pathways, collectively known as the unfolded protein response (UPR), are required for maintaining ER homeostasis. The UPR is triggered when ER protein folding capacity is overwhelmed by cellular demand and the UPR initially aims to restore ER homeostasis and normal cellular functions. However, if this fails, then the UPR triggers cell death. In this review, we provide a UPR signalling-centric view of ER functions, from the ER's discovery to the latest advancements in the understanding of ER and UPR biology. Our review provides a synthesis of intracellular ER signalling revolving around proteostasis and the UPR, its impact on other organelles and cellular behaviour, its multifaceted and dynamic response to stress and its role in physiology, before finally exploring the potential exploitation of this knowledge to tackle unresolved biological questions and address unmet biomedical needs. Thus, we provide an integrated and global view of existing literature on ER signalling pathways and their use for therapeutic purposes.
Collapse
Affiliation(s)
- Aitor Almanza
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| | - Antonio Carlesso
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGöteborgSweden
| | - Chetan Chintha
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| | | | - Dimitrios Doultsinos
- INSERM U1242University of RennesFrance
- Centre de Lutte Contre le Cancer Eugène MarquisRennesFrance
| | - Brian Leuzzi
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| | - Andreia Luís
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CentreViennaAustria
| | - Nicole McCarthy
- Institute for Experimental Cancer Research in PaediatricsGoethe‐UniversityFrankfurtGermany
| | - Luigi Montibeller
- Neurogenetics GroupDivision of Brain SciencesFaculty of MedicineImperial College LondonUK
| | - Sanket More
- Department Cellular and Molecular MedicineLaboratory of Cell Death and TherapyKU LeuvenBelgium
| | - Alexandra Papaioannou
- INSERM U1242University of RennesFrance
- Centre de Lutte Contre le Cancer Eugène MarquisRennesFrance
| | - Franziska Püschel
- Cell Death Regulation GroupOncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Maria Livia Sassano
- Department Cellular and Molecular MedicineLaboratory of Cell Death and TherapyKU LeuvenBelgium
| | - Josip Skoko
- Institute of Cell Biology and ImmunologyUniversity of StuttgartGermany
| | - Patrizia Agostinis
- Department Cellular and Molecular MedicineLaboratory of Cell Death and TherapyKU LeuvenBelgium
| | - Jackie de Belleroche
- Neurogenetics GroupDivision of Brain SciencesFaculty of MedicineImperial College LondonUK
| | - Leif A. Eriksson
- Department of Chemistry and Molecular BiologyUniversity of GothenburgGöteborgSweden
| | - Simone Fulda
- Institute for Experimental Cancer Research in PaediatricsGoethe‐UniversityFrankfurtGermany
| | | | - Sandra Healy
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| | - Andrey Kozlov
- Ludwig Boltzmann Institute for Experimental and Clinical TraumatologyAUVA Research CentreViennaAustria
| | - Cristina Muñoz‐Pinedo
- Cell Death Regulation GroupOncobell ProgramBellvitge Biomedical Research Institute (IDIBELL)BarcelonaSpain
| | - Markus Rehm
- Institute of Cell Biology and ImmunologyUniversity of StuttgartGermany
| | - Eric Chevet
- INSERM U1242University of RennesFrance
- Centre de Lutte Contre le Cancer Eugène MarquisRennesFrance
| | - Afshin Samali
- Apoptosis Research CentreNational University of IrelandGalwayIreland
| |
Collapse
|
18
|
BH3-mimetic toolkit guides the respective use of BCL2 and MCL1 BH3-mimetics in myeloma treatment. Blood 2018; 132:2656-2669. [PMID: 30309889 DOI: 10.1182/blood-2018-03-836718] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 09/28/2018] [Indexed: 02/07/2023] Open
Abstract
BH3 mimetics are promising drugs for hematologic malignancies that trigger cell death by promoting the release of proapoptotic BCL2 family members from antiapoptotic proteins. Multiple myeloma is considered to be a disease dependent mainly on MCL1 for survival, based mostly on studies using cell lines. We used a BH3-mimetic toolkit to study the dependency on BCL2, BCLXL, or MCL1 in malignant plasma cells from 60 patients. Dependencies were analyzed using an unbiased BH3 mimetics cell-death clustering by k-means. In the whole cohort of patients, BCL2 dependency was mostly found in the CCND1 subgroup (83%). Of note, MCL1 dependence significantly increased from 33% at diagnosis to 69% at relapse, suggesting a plasticity of the cellular dependency favoring MCL1 dependencies at relapse. In addition, 35% of overall patient samples showed codependencies on either BCL2/MCL1 or BCLXL/MCL1. Finally, we identified a group of patients not targeted by any of the BH3 mimetics, predominantly at diagnosis in patients not presenting the common recurrent translocations. Mechanistically, we demonstrated that BAK is crucial for cell death induced by MCL1 mimetic A1210477, according to the protection from cell death observed by BAK knock-down, as well as the complete and early disruption of MCL1/BAK complexes on A1210477 treatment. Interestingly, this complex was also dissociated in A1210477-resistant cells, but free BAK was simultaneously recaptured by BCLXL, supporting the role of BCLXL in A1210477 resistance. In conclusion, our study opens the way to rationally use venetoclax and/or MCL1 BH3 mimetics for clinical evaluation in myeloma at both diagnosis and relapse.
Collapse
|
19
|
Huang Z, Lei W, Tan J, Hu HB. Long noncoding RNA LINC00961 inhibits cell proliferation and induces cell apoptosis in human non-small cell lung cancer. J Cell Biochem 2018; 119:9072-9080. [PMID: 30010215 DOI: 10.1002/jcb.27166] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 05/18/2018] [Indexed: 12/19/2022]
Abstract
Long noncoding RNAs (LncRNAs) have been identified in multiple human cancer types, including lung cancer. An increasing number of studies have indicated that lncRNAs can function as important gene regulators. However, the biological mechanism of LINC00961 in lung cancerremains poorly understood. In our current study, we recognized lncRNA LINC00961, and we observed that it was significantly reduced in human non-small cell lung cancer (NSCLC) tissues. LINC00961 was elevated by infecting LV-LINC00961, while decreased by LV-shLINC00961 in H226 and A549 cells. Furthermore, it was shown that LINC00961 overexpression greatly inhibited lung cancer cell proliferation, whereas downregulated LINC00961 induced cell proliferation. In addition, further experiments showed that restoration of LINC00961 could dramatically increase apoptotic ratios of NSCLC H226 and A549 cells, and knockdown of LINC00961 exhibited an opposite effect. Moreover, Western blot analysis showed that upregulation of LINC00961 repressed proliferating cell nuclear antigen expression and increased Bax expression, indicating that it acts as an important pro-apoptosis gene. Conversely, inhibition of LINC00961 induced proliferating cell nuclear antigen expression and restrained Bax protein levels. Taking these together, LINC00961 might play a tumor suppressive role in NSCLC progression, and it could serve as a novel prognostic biomarker in NSCLC diagnosis and treatment.
Collapse
Affiliation(s)
- Zhiwen Huang
- Department of Respiratory Medicine, Affiliated Renhe Hospital of China Three Gorges University, Yichang, Hubei, China
| | - Wei Lei
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin Tan
- Department of Thoracic Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, China
| | - Hai-Bo Hu
- Department of Thoracic Surgery, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| |
Collapse
|
20
|
Fang Z, Han B, Jung KH, Lee JH, El-Damasy AK, Gadhe CG, Kim SJ, Yan HH, Park JH, Lee JE, Kang YW, Pae AN, Keum G, Hong SS. A novel tropomyosin-related kinase A inhibitor, KK5101 to treat pancreatic cancer. Cancer Lett 2018; 426:25-36. [DOI: 10.1016/j.canlet.2018.03.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/15/2018] [Accepted: 03/26/2018] [Indexed: 12/31/2022]
|
21
|
Budhraja A, Turnis ME, Churchman ML, Kothari A, Yang X, Xu H, Kaminska E, Panetta JC, Finkelstein D, Mullighan CG, Opferman JT. Modulation of Navitoclax Sensitivity by Dihydroartemisinin-Mediated MCL-1 Repression in BCR-ABL + B-Lineage Acute Lymphoblastic Leukemia. Clin Cancer Res 2017; 23:7558-7568. [PMID: 28974549 DOI: 10.1158/1078-0432.ccr-17-1231] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/31/2017] [Accepted: 09/28/2017] [Indexed: 01/06/2023]
Abstract
Purpose: BCR-ABL+ B-ALL leukemic cells are highly dependent on the expression of endogenous antiapoptotic MCL-1 to promote viability and are resistant to BH3-mimetic agents such as navitoclax (ABT-263) that target BCL-2, BCL-XL, and BCL-W. However, the survival of most normal blood cells and other cell types is also dependent on Mcl-1 Despite the requirement for MCL-1 in these cell types, initial reports of MCL-1-specific BH3-mimetics have not described any overt toxicities associated with single-agent use, but these agents are still early in clinical development. Therefore, we sought to identify approved drugs that could sensitize leukemic cells to ABT-263.Experimental Design: A screen identified dihydroartemisinin (DHA), a water-soluble metabolite of the antimalarial artemisinin. Using mouse and human leukemic cell lines, and primary patient-derived xenografts, the effect of DHA on survival was tested, and mechanistic studies were carried out to discover how DHA functions. We further tested in vitro and in vivo whether combining DHA with ABT-263 could enhance the response of leukemic cells to combination therapy.Results: DHA causes the downmodulation of MCL-1 expression by triggering a cellular stress response that represses translation. The repression of MCL-1 renders leukemic cells highly sensitive to synergistic cell death induced by ABT-263 in a mouse model of BCR-ABL+ B-ALL both in vitro and in vivo Furthermore, DHA synergizes with ABT-263 in human Ph+ ALL cell lines, and primary patient-derived xenografts of Ph+ ALL in culture.Conclusions: Our findings suggest that combining DHA with ABT-263 can improve therapeutic response in BCR-ABL+ B-ALL. Clin Cancer Res; 23(24); 7558-68. ©2017 AACR.
Collapse
Affiliation(s)
- Amit Budhraja
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Meghan E Turnis
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michelle L Churchman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Anisha Kothari
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Xue Yang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Haiyan Xu
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ewa Kaminska
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
22
|
Guo X, Zhang L, Fan Y, Zhang D, Qin L, Dong S, Li G. Oxysterol-Binding Protein-Related Protein 8 Inhibits Gastric Cancer Growth Through Induction of ER Stress, Inhibition of Wnt Signaling, and Activation of Apoptosis. Oncol Res 2016; 25:799-808. [PMID: 27983927 PMCID: PMC7841135 DOI: 10.3727/096504016x14783691306605] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related mortality worldwide. Oxysterol-binding protein-related protein 8 (ORP8) functions as a sterol sensor that regulates a number of cellular functions. We showed that ORP8 expression was significantly lower in GC tissues and cells. Overexpression of ORP8 significantly inhibited GC cell proliferation in several GC cells. The formation of colonies in AGS cells was inhibited by the overexpression of ORP8. Moreover, overexpression of ORP8 significantly decreased implanted tumor growth in nude mice. Overexpression of ORP8 resulted in a significant increase in CHOP and GRP78 expression and the phosphorylation of PERK, indicating the occurrence of ER stress. Inhibition of ER stress by 4-PBA notably suppressed overexpression of ORP8-induced decrease of GC cell proliferation, formation of colonies, and implanted tumor growth. Overexpression of ORP8 resulted in a significant decrease in Wnt3a and β-catenin expression, and activation of Wnt signaling by HLY78 markedly blocked overexpression of ORP8-induced decrease in GC cell proliferation, formation of colonies, and implanted tumor growth. 4-PBA inhibited overexpression of ORP8-induced decrease in Wnt signaling. Furthermore, overexpression of ORP8 resulted in significant activation of mitochondrial apoptotic events and increase in apoptosis, which was inhibited by 4-PBA and HLY78. Induction of ER stress, inhibition of Wnt signaling, and apoptotic cell death were involved in ORP8-induced inhibition of GC cell proliferation. These findings indicate that downregulation of ORP8 plays a pivotal role in the progression of GC, and it may be a novel therapeutic target in the treatment of GC.
Collapse
|