51
|
Scherr AL, Mock A, Gdynia G, Schmitt N, Heilig CE, Korell F, Rhadakrishnan P, Hoffmeister P, Metzeler KH, Schulze-Osthoff K, Illert AL, Boerries M, Trojan J, Waidmann O, Falkenhorst J, Siveke J, Jost PJ, Bitzer M, Malek NP, Vecchione L, Jelas I, Brors B, Glimm H, Stenzinger A, Grekova SP, Gehrig T, Schulze-Bergkamen H, Jäger D, Schirmacher P, Heikenwalder M, Goeppert B, Schneider M, Fröhling S, Köhler BC. Identification of BCL-XL as highly active survival factor and promising therapeutic target in colorectal cancer. Cell Death Dis 2020; 11:875. [PMID: 33070156 PMCID: PMC7568722 DOI: 10.1038/s41419-020-03092-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022]
Abstract
Since metastatic colorectal cancer (CRC) is a leading cause of cancer-related death, therapeutic approaches overcoming primary and acquired therapy resistance are an urgent medical need. In this study, the efficacy and toxicity of high-affinity inhibitors targeting antiapoptotic BCL-2 proteins (BCL-2, BCL-XL, and MCL-1) were evaluated. By RNA sequencing analysis of a pan-cancer cohort comprising >1500 patients and subsequent prediction of protein activity, BCL-XL was identified as the only antiapoptotic BCL-2 protein that is overactivated in CRC. Consistently, pharmacologic and genetic inhibition of BCL-XL induced apoptosis in human CRC cell lines. In a combined treatment approach, targeting BCL-XL augmented the efficacy of chemotherapy in vitro, in a murine CRC model, and in human ex vivo derived CRC tissue cultures. Collectively, these data show that targeting of BCL-XL is efficient and safe in preclinical CRC models, observations that pave the way for clinical translation.
Collapse
Affiliation(s)
- Anna-Lena Scherr
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Andreas Mock
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120, Heidelberg, Germany.,Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Georg Gdynia
- Institute of Pathology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Nathalie Schmitt
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Christoph E Heilig
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Felix Korell
- Department of Medicine V, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Praveen Rhadakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, 69120, Heidelberg, Germany
| | - Paula Hoffmeister
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Klaus H Metzeler
- Department of Medicine III, University Hospital, LMU Munich, 81377, Munich, Germany
| | - Klaus Schulze-Osthoff
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.,Department of Molecular Medicine, Interfaculty Institute for Biochemistry, University of Tübingen, 72076, Tübingen, Germany
| | - Anna L Illert
- Department of Internal Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Melanie Boerries
- German Cancer Consortium (DKTK) Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Comprehensive Cancer Center Freiburg (CCCF), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany.,Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Jörg Trojan
- Department of Medicine 1, University Hospital Frankfurt, 60590, Frankfurt, Germany.,Universitäres Centrum für Tumorerkrankungen (UCT), University Hospital Frankfurt, 60590, Frankfurt, Germany
| | - Oliver Waidmann
- Department of Medicine 1, University Hospital Frankfurt, 60590, Frankfurt, Germany.,Universitäres Centrum für Tumorerkrankungen (UCT), University Hospital Frankfurt, 60590, Frankfurt, Germany
| | - Johanna Falkenhorst
- Depārtment of Medical Oncology, Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, 45147, Essen, Germany.,DKTK partner site Essen and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Jens Siveke
- Institute for Developmental Cancer Therapeutics, West German Cancer Center, University Hospital Essen, 45147, Essen, Germany.,Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK, partner site Essen) and German Cancer Research Center, DKFZ, 69120, Heidelberg, Germany
| | - Philipp J Jost
- Medical Department III for Hematology and Oncology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, 81675, Munich, Germany.,Central Institute for Translational Cancer Research (Translatum), Technical University of Munich, 81675, Munich, Germany.,German Consortium for Translational Cancer Research (DKTK) partner site TUM, German Cancer Research Center Heidelberg (DKFZ), 69120, Heidelberg, Germany
| | - Michael Bitzer
- Department of Internal Medicine I, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Nisar P Malek
- Department of Internal Medicine I, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Loredana Vecchione
- Charité Comprehensive Cancer Center, 10117, Berlin, Germany.,Department of Hematology, Oncology and Tumor Immunology (CCM) Charité - Universitaetsmedizin Berlin, 10117, Berlin, Germany
| | - Ivan Jelas
- Charité Comprehensive Cancer Center, 10117, Berlin, Germany
| | - Benedikt Brors
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.,Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Hanno Glimm
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden and German Cancer Research Center (DKFZ), 01307, Dresden, Germany.,Center for Personalized Oncology, University Hospital Carl Gustav Carus Dresden at TU Dresden, 01307, Dresden, Germany.,German Cancer Consortium (DKTK) Dresden, 01307, Dresden, Germany
| | - Albrecht Stenzinger
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Svetlana P Grekova
- Institute of Pathology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Tobias Gehrig
- Department of General and Visceral Surgery, Spital Linth, 8730, Uznach, Switzerland
| | | | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120, Heidelberg, Germany.,German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Peter Schirmacher
- German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Benjamin Goeppert
- Institute of Pathology, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, 69120, Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Bruno C Köhler
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, 69120, Heidelberg, Germany. .,German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.
| |
Collapse
|
52
|
Zhang X, Wang Z, Guo Z, Song T, He N, Zhu J, Cao K, Zhang Z. A "Three-in-One" Multifunctional Probe for Bcl-2/Mcl-1 Profiling and Visualizing in Situ. Chembiochem 2020; 22:326-329. [PMID: 32881291 DOI: 10.1002/cbic.202000441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 08/23/2020] [Indexed: 11/10/2022]
Abstract
Bcl-2 and Mcl-1, the two arms of the anti-apoptotic Bcl-2 family proteins, have been identified as key regulators of apoptosis and effective therapeutic targets of cancer. However, no small-molecular probe is capable of profiling and visualizing both Bcl-2 and Mcl-1 simultaneously in situ. Herein, we report a multifunctional molecular probe (BnN3 -OPD-Alk) by a "three-in-one" molecular designing strategy, which integrated the Bcl-2/Mcl-1 binding ligand, fluorescent reporter group and photoreactive group azido into the same scaffold. BnN3 -OPD-Alk exhibited sub-micromolar affinities to Bcl-2/Mcl-1 and bright green self-fluorescence. It was then successfully applied for Bcl-2/Mcl-1 labeling, capturing, enriching, and bioimaging both in vitro and in cells. This strategy could facilitate the precise early diagnosis and effective therapy of dual Bcl-2/Mcl-1-related diseases.
Collapse
Affiliation(s)
- Xiaodong Zhang
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, No. 2 LingGong Road, Dalian, 116023, P. R. China
| | - Ziqian Wang
- Zhang Dayu School of Chemistry, Dalian University of Technology, No. 2 LingGong Road, Dalian, P. R. China
| | - Zongwei Guo
- School of Life Science and Technology, Dalian University of Technology, No. 2 LingGong Road, Dalian, P. R. China
| | - Ting Song
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, No. 2 LingGong Road, Dalian, 116023, P. R. China
| | - Nianzhe He
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, No. 2 LingGong Road, Dalian, 116023, P. R. China.,Department of Chemistry, Technical University of Denmark, Kemitorvet, 2800 Kgs., Lyngby, Denmark
| | - Junjie Zhu
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, No. 2 LingGong Road, Dalian, 116023, P. R. China
| | - Keke Cao
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, No. 2 LingGong Road, Dalian, 116023, P. R. China
| | - Zhichao Zhang
- State Key Laboratory of Fine Chemicals, School of Chemistry, Dalian University of Technology, No. 2 LingGong Road, Dalian, 116023, P. R. China
| |
Collapse
|
53
|
Shahar N, Larisch S. Inhibiting the inhibitors: Targeting anti-apoptotic proteins in cancer and therapy resistance. Drug Resist Updat 2020; 52:100712. [DOI: 10.1016/j.drup.2020.100712] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
|
54
|
Barrera CC, Groot H, Vargas WL, Narváez DM. Efficacy and Molecular Effects of a Reduced Graphene Oxide/Fe 3O 4 Nanocomposite in Photothermal Therapy Against Cancer. Int J Nanomedicine 2020; 15:6421-6432. [PMID: 32922009 PMCID: PMC7457756 DOI: 10.2147/ijn.s256760] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/19/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Expanded research on the biomedical applications of graphene has shown promising results, although interactions between cells and graphene are still unclear. The current study aims to dissect the cellular and molecular effects of graphene nanocomposite in photothermal therapy against cancer, and to evaluate its efficacy. METHODS In this study, a reduced graphene oxide and iron oxide (rGO-Fe3O4) nanocomposite was obtained by chemical synthesis. The nanocomposite was fully characterized by Raman spectroscopy, TEM, VSM and thermal profiling. Cell-nanocomposite interaction was evaluated by confocal microscopy and viability assays on cancer cell line HeLa. The efficacy of the thermal therapy and changes in gene expression of Bcl-2 and Hsp70 was assessed. RESULTS The resulting rGO-Fe3O4 nanocomposite exhibited superparamagnetic properties and the capacity to increase the surrounding temperature by 18-20°C with respect to the initial temperature. The studies of cell-nanocomposite interaction showed that rGO-Fe3O4 attaches to cell membrane but there is a range of concentration at which the nanomaterial preserves cell viability. Photothermal therapy reduced cell viability to 32.6% and 23.7% with 50 and 100 µg/mL of nanomaterial, respectively. The effect of treatment on the molecular mechanism of cell death demonstrated an overexpression of anti-apoptotic proteins Hsp70 and Bcl-2 as an initial response to the therapy and depending on the aggressiveness of the treatment. CONCLUSION The results of this study contribute to understanding the interactions between cell and graphene and support its application in photothermal therapy against cancer due to its promising results.
Collapse
Affiliation(s)
- Claudia C Barrera
- Human Genetics Laboratory, Department of Biological Sciences, Universidad de Los Andes, Bogotá, Colombia
| | - Helena Groot
- Human Genetics Laboratory, Department of Biological Sciences, Universidad de Los Andes, Bogotá, Colombia
| | - Watson L Vargas
- Department of Chemical Engineering, Universidad de Los Andes, Bogotá, Colombia
| | - Diana M Narváez
- Human Genetics Laboratory, Department of Biological Sciences, Universidad de Los Andes, Bogotá, Colombia
| |
Collapse
|
55
|
Wu X, Luo Q, Liu Z. Ubiquitination and deubiquitination of MCL1 in cancer: deciphering chemoresistance mechanisms and providing potential therapeutic options. Cell Death Dis 2020; 11:556. [PMID: 32699213 PMCID: PMC7376237 DOI: 10.1038/s41419-020-02760-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
Abstract
MCL1 is an important antiapoptotic member of the BCL-2 family that is distinguishable from other family members based on its relatively short half-life. Emerging studies have revealed the crucial role of MCL1 in the chemoresistance of cancer cells. The antiapoptotic function of MCL1 makes it a popular therapeutic target, although specific inhibitors have begun to emerge only recently. Notably, emerging studies have reported that several E3 ligases and deubiquitinases modulate MCL1 stability, providing an alternate means of targeting MCL1 activity. In addition, the emergence and development of proteolysis-targeting chimeras, the function of which is based on ubiquitination-mediated degradation, has shown great potential. In this review, we provide an overview of the studies investigating the ubiquitination and deubiquitination of MCL1, summarize the latest evidence regarding the development of therapeutic strategies targeting MCL1 in cancer treatment, and discuss the promising future of targeting MCL1 via the ubiquitin–proteasome system in clinical practice.
Collapse
Affiliation(s)
- Xiaowei Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Qingyu Luo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| |
Collapse
|
56
|
Thapa B, Kc R, Uludağ H. TRAIL therapy and prospective developments for cancer treatment. J Control Release 2020; 326:335-349. [PMID: 32682900 DOI: 10.1016/j.jconrel.2020.07.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/01/2020] [Accepted: 07/11/2020] [Indexed: 12/22/2022]
Abstract
Tumor Necrosis Factor (TNF) Related Apoptosis-Inducing Ligand (TRAIL), an immune cytokine of TNF-family, has received much attention in late 1990s as a potential cancer therapeutics due to its selective ability to induce apoptosis in cancer cells. TRAIL binds to cell surface death receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5) and facilitates formation of death-inducing signaling complex (DISC), eventually activating the p53-independent apoptotic cascade. This unique mechanism makes the TRAIL a potential anticancer therapeutic especially for p53-mutated tumors. However, recombinant human TRAIL protein (rhTRAIL) and TRAIL-R agonist monoclonal antibodies (mAb) failed to exert robust anticancer activities due to inherent and/or acquired resistance, poor pharmacokinetics and weak potencies for apoptosis induction. To get TRAIL back on track as a cancer therapeutic, multiple strategies including protein modification, combinatorial approach and TRAIL gene therapy are being extensively explored. These strategies aim to enhance the half-life and bioavailability of TRAIL and synergize with TRAIL action ultimately sensitizing the resistant and non-responsive cells. We summarize emerging strategies for enhanced TRAIL therapy in this review and cover a wide range of recent technologies that will provide impetus to rejuvenate the TRAIL therapeutics in the clinical realm.
Collapse
Affiliation(s)
- Bindu Thapa
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
| | - Remant Kc
- Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada.
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada; Department of Chemical & Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
57
|
Wu W, Xu C, Zhang X, Yu A, Shu L. Shrimp miR-965 induced the human melanoma stem-like cell apoptosis and inhibited their stemness by disrupting the MCL-1-ER stress-XBP1 feedback loop in a cross-species manner. Stem Cell Res Ther 2020; 11:248. [PMID: 32586376 PMCID: PMC7318764 DOI: 10.1186/s13287-020-01734-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 04/29/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Melanoma is a type of aggressive skin cancer with a poor survival rate. The resistance to conventional therapy of this disease is, at least in part, attributed to its cancer stem cell population. However, the mechanism of survival and stemness maintenance of cancer stem cells remains to be investigated. METHODS Tumorsphere formation assay was used to study the stem-like property of melanoma stem-like cells (MSLC). Chromatin immunoprecipitation (ChIP), promoter luciferase reporter assay were included for exploring the role of MCL-1 in MSLC and electrophoretic mobility shift assay were used to evaluate the interaction between shrimp miR-965 and human Ago2 protein. Melanoma xenograft nude mice were used to study the inhibition of tumor development. RESULTS In the present study, our results showed that myeloid cell leukemia sequence 1 (MCL-1) knocking down induced ER stress and apoptosis, and the expression reduction of stemness associated genes in MSLC, which implied a significant role of MCL-1 in MSLC. Further study indicated that ER stress agonist (tunicamycin) treatment in MSLC results in the translocation of XBP1, an ER stress sensor, into the nucleus to induce MCL-1 expression through direct binding to the - 313- to - 308-bp region of MCL-1 promoter. In addition, we found that a shrimp-derived miRNA (shrimp miR-965) could interact with the human Ago2 protein and suppressed the human MCL-1 expression by binding to the 3' UTR of MCL-1 mRNA, thereby inhibiting the MSLC proliferation and stemness in vitro and in vivo in a cross-species manner. CONCLUSION In conclusion, we identified an important role of MCL-1-ER stress-XBP1 feedback loop in the stemness and survival maintenance of MSLC, and shrimp miR-965, a natural food derived miRNA, could regulate MSLC stemness and survival by targeting MCL-1 and disrupting the balance of MCL-1-ER stress-XBP1 feedback loop. In conclusion, this study indicated an important mechanism of the regulation of MSLC stemness and survival, otherwise it also demonstrated the significance of cross-species-derived miRNA as promising natural drugs in melanoma therapy.
Collapse
Affiliation(s)
- Wenlin Wu
- College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou, 36200, Fujian Province, People's Republic of China
| | - Chenxi Xu
- College of Life Science, Zhejiang University, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Xiaobo Zhang
- College of Life Science, Zhejiang University, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - An Yu
- Huffington Centre on Aging, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Le Shu
- College of Life Science, Zhejiang University, Hangzhou, 310058, Zhejiang Province, People's Republic of China.
| |
Collapse
|
58
|
Masilamani AP, Dettmer-Monaco V, Monaco G, Cathomen T, Kuckuck I, Schultze-Seemann S, Huber N, Wolf P. An Anti-PSMA Immunotoxin Reduces Mcl-1 and Bcl2A1 and Specifically Induces in Combination with the BAD-Like BH3 Mimetic ABT-737 Apoptosis in Prostate Cancer Cells. Cancers (Basel) 2020; 12:cancers12061648. [PMID: 32580291 PMCID: PMC7352695 DOI: 10.3390/cancers12061648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 12/25/2022] Open
Abstract
Background: Upregulation of anti-apoptotic Bcl-2 proteins in advanced prostate cancer leads to therapeutic resistance by prevention of cell death. New therapeutic approaches aim to target the Bcl-2 proteins for the restoration of apoptosis. Methods: The immunotoxin hD7-1(VL-VH)-PE40 specifically binds to the prostate specific membrane antigen (PSMA) on prostate cancer cells and inhibits protein biosynthesis. It was tested with respect to its effects on the expression of anti-apoptotic Bcl-2 proteins. Combination with the BAD-like mimetic ABT-737 was examined on prostate cancer cells and 3D spheroids and in view of tumor growth and survival in the prostate cancer SCID mouse xenograft model. Results: The immunotoxin led to a specific inhibition of Mcl-1 and Bcl2A1 expression in PSMA expressing target cells. Its combination with ABT-737, which inhibits Bcl-2, Bcl-xl, and Bcl-w, led to an induction of the intrinsic apoptotic pathway and to a synergistic cytotoxicity in prostate cancer cells and 3D spheroids. Furthermore, combination therapy led to a significantly prolonged survival of mice bearing prostate cancer xenografts based on an inhibition of tumor growth. Conclusion: The combination therapy of anti-PSMA immunotoxin plus ABT-737 represents the first tumor-specific therapeutic approach on the level of Bcl-2 proteins for the induction of apoptosis in prostate cancer.
Collapse
Affiliation(s)
- Anie P. Masilamani
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
| | - Viviane Dettmer-Monaco
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106 Freiburg, Germany
| | - Gianni Monaco
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106 Freiburg, Germany
| | - Toni Cathomen
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
- Institute for Transfusion Medicine and Gene Therapy, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Center for Chronic Immunodeficiency, University of Freiburg, 79106 Freiburg, Germany
| | - Irina Kuckuck
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
| | - Susanne Schultze-Seemann
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
| | - Nathalie Huber
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
| | - Philipp Wolf
- Department of Urology, Medical Center—University of Freiburg, 79106 Freiburg, Germany; (A.P.M.); (I.K.); (S.S.-S.); (N.H.)
- Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (V.D.-M.); (G.M.); (T.C.)
- Correspondence: ; Tel.: +49-761-270-28921
| |
Collapse
|
59
|
Chang CY, Pan PH, Li JR, Ou YC, Wang JD, Liao SL, Chen WY, Wang WY, Chen CJ. Aspirin Induced Glioma Apoptosis through Noxa Upregulation. Int J Mol Sci 2020; 21:4219. [PMID: 32545774 PMCID: PMC7352791 DOI: 10.3390/ijms21124219] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023] Open
Abstract
Clinically, high cyclooxygenase-2 expression in malignant glioma correlates well with poor prognosis and the use of aspirin is associated with a reduced risk of glioma. To extend the current understanding of the apoptotic potential of aspirin in most cell types, this study provides evidence showing that aspirin induced glioma cell apoptosis and inhibited tumor growth, in vitro and in vivo. We found that the human H4 glioma cell-killing effects of aspirin involved mitochondria-mediated apoptosis accompanied by endoplasmic reticulum (ER) stress, Noxa upregulation, Mcl-1 downregulation, Bax mitochondrial distribution and oligomerization, and caspase 3/caspase 8/caspase 9 activation. Genetic silencing of Noxa or Bax attenuated aspirin-induced viability loss and apoptosis, while silencing Mcl-1 augmented the effects of aspirin. Data from genetic and pharmacological studies revealed that the axis of ER stress comprised an apoptotic cascade leading to Noxa upregulation and apoptosis. The apoptotic programs and mediators triggered by aspirin in H4 cells were duplicated in human U87 glioma cell line as well as in tumor-bearing BALB/c nude mice. The involvement of ER stress in indomethacin-induced Mcl-1 downregulation was reported in our previous study on glioma cells. Therefore, the aforementioned phenomena indicate that ER stress may be a valuable target for intervention in glioma apoptosis.
Collapse
Affiliation(s)
- Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung City 420, Taiwan;
| | - Ping-Ho Pan
- Department of Pediatrics, Tungs’ Taichung Metro Harbor Hospital, Taichung City 435, Taiwan;
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 402, Taiwan;
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Yen-Chuan Ou
- Department of Urology, Tungs’ Taichung Metro Harbor Hospital, Taichung City 435, Taiwan;
| | - Jiaan-Der Wang
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung City 407, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 402, Taiwan;
| | - Wen-Yi Wang
- Department of Nursing, HungKuang University, Taichung City 433, Taiwan;
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan
- Ph.D. Program in Translational Medicine, College of Life Sciences, National Chung Hsing University, Taichung City 402, Taiwan
| |
Collapse
|
60
|
Lin HC, Lin JY. GSF3, a polysaccharide from guava (Psidium guajava L.) seeds, inhibits MCF-7 breast cancer cell growth via increasing Bax/Bcl-2 ratio or Fas mRNA expression levels. Int J Biol Macromol 2020; 161:1261-1271. [PMID: 32531360 DOI: 10.1016/j.ijbiomac.2020.06.057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 01/26/2023]
Abstract
Guava seed polysaccharide fraction 3 (GSF3) is an immunomodulatory polysaccharide from guava (Psidium guajava L.) seed polysaccharides. However, effects of GSF3 on the growth of breast cancer cells were not understood, yet. To clarify the GSF3 effects on breast cancer cell growth, GSF3 was subjected to treat MCF-7 cells using direct action or indirect immunotherapy using immune cells conditioned media, respectively. The viabilities of MCF-7 cells were measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Changes in pro-(Bax)/anti-apoptotic (Bcl-2) and Fas mRNA expression levels in the treated MCF-7 cells were measured using two-step reverse transcription quantitative polymerase chain reaction. Our results showed that GSF3 inhibited MCF-7 cell growth through either direct action or indirect immunotherapy. GSF3 direct action significantly (P < 0.05) decreased Bcl-2 mRNA expression amount but increased pro-(Bax)/anti-apoptotic (Bcl-2) mRNA expression ratios in the treated cells. The splenocytes conditioned media cultured with GSF3 increased Fas mRNA expression amounts in the treated MCF-7 cells. There was a significant negative correlation between Th2-polarized cytokines secreted by immune cells and Fas mRNA expression levels in the corresponding treated MCF-7 cells. Our findings suggested that GSF3 is a potent anti-cancerous polysaccharide by direct action or indirectly modulating immune cell cytokine secretion profiles.
Collapse
Affiliation(s)
- Hsiao-Chien Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung City 402, Taiwan, ROC
| | - Jin-Yuarn Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung City 402, Taiwan, ROC.
| |
Collapse
|
61
|
Jozkowiak M, Skupin-Mrugalska P, Nowicki A, Borys-Wojcik S, Wierzchowski M, Kaczmarek M, Ramlau P, Jodynis-Liebert J, Piotrowska-Kempisty H. The Effect of 4'-hydroxy-3,4,5-trimetoxystilbene, the Metabolite of Resveratrol Analogue DMU-212, on Growth, Cell Cycle and Apoptosis in DLD-1 and LOVO Colon Cancer Cell Lines. Nutrients 2020; 12:nu12051327. [PMID: 32392733 PMCID: PMC7285027 DOI: 10.3390/nu12051327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/10/2020] [Accepted: 05/01/2020] [Indexed: 12/25/2022] Open
Abstract
Resveratrol is a phytoalexin that naturally occurs in grapes, blueberries, cranberries, peanuts and many other plants. Although resveratrol inhibits carcinogenesis in all three stages, its clinical application is restricted due to poor pharmacokinetics. The methylated analogues of resveratrol have been found to have higher bioavailability and cytotoxic activity than that of the prototupe compound. Among the various methoxy derivatives of resveratrol, 3,4,5,4′-tetrametoxystilbene (DMU-212) is suggested to be one of the strongest activators of cytotoxicity and apoptosis. DMU-212 has been shown to exert anti-tumor activity in DLD-1 and LOVO colon cancer cells. Since colorectal cancer is the third most common cause of cancer-related deaths worldwide, the development of new anticancer agents is nowadays of high significance. The aim of the present study was to assess the anticancer activity of 4′-hydroxy-3,4,5-trimetoxystilbene (DMU-281), the metabolite of DMU-212, in DLD-1 and LOVO cell lines. We showed for the first time the cytotoxic activity of DMU-281 triggered via cell cycle arrest at G2/M phase and apoptosis induction accompanied by the activation of caspases-9, -8, -3/7. Furthermore, DMU-281 has been found to change the expression pattern of genes and proteins related to intrinsic as well as extrinsic apoptosis. Since the activation of these pathways of apoptosis is still the most desired strategy in anticancer research, DMU-281 seems to provide a promising approach to the treatment of colon cancer.
Collapse
Affiliation(s)
- Malgorzata Jozkowiak
- Department of Toxicology, Poznan University of Medical Sciences; Dojazd 30 St., PL-60-631 Poznan, Poland; (M.J.); (A.N.); (P.R.); (J.J.-L.)
| | - Paulina Skupin-Mrugalska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, Poland;
| | - Andrzej Nowicki
- Department of Toxicology, Poznan University of Medical Sciences; Dojazd 30 St., PL-60-631 Poznan, Poland; (M.J.); (A.N.); (P.R.); (J.J.-L.)
| | - Sylwia Borys-Wojcik
- Department of Anatomy, Poznan University of Medical Sciences, Swiecickiego 6 St., PL-60-781 Poznan, Poland;
| | - Marcin Wierzchowski
- Department of Chemical Technology of Drugs, Poznan University of Medical Sciences, Grunwaldzka 6 St., PL-60-780 Poznan, Poland;
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Garbary 15 St., PL-61-866 Poznan, Poland;
- Gene Therapy Unit, Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, Garbary 15 St., PL-61-866 Poznan, Poland
| | - Piotr Ramlau
- Department of Toxicology, Poznan University of Medical Sciences; Dojazd 30 St., PL-60-631 Poznan, Poland; (M.J.); (A.N.); (P.R.); (J.J.-L.)
| | - Jadwiga Jodynis-Liebert
- Department of Toxicology, Poznan University of Medical Sciences; Dojazd 30 St., PL-60-631 Poznan, Poland; (M.J.); (A.N.); (P.R.); (J.J.-L.)
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences; Dojazd 30 St., PL-60-631 Poznan, Poland; (M.J.); (A.N.); (P.R.); (J.J.-L.)
- Correspondence: ; Tel.: +48-61847-07-21
| |
Collapse
|
62
|
Liu L, Liu R, Yang X, Hou X, Fang H. Design, synthesis and biological evaluation of tyrosine derivatives as Mcl-1 inhibitors. Eur J Med Chem 2020; 191:112142. [DOI: 10.1016/j.ejmech.2020.112142] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 10/25/2022]
|
63
|
Sohn YK, Kim HS. Targeted delivery of a human Bcl-2-specific protein binder effectively induces apoptosis of cancer cells. Biochem Biophys Res Commun 2020; 526:447-452. [PMID: 32228885 DOI: 10.1016/j.bbrc.2020.03.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 03/20/2020] [Indexed: 01/02/2023]
Abstract
Bcl-2 family proteins are critical switches to control cell death and survival, and Bcl-2 is a key regulator in pro-survival signaling, causing various diseases including cancers. Bcl-2 has drawn a considerable attention as a potential target for developing a pro-apoptotic agent for cancers. We here present the development of a specific protein binder against human Bcl-2 and its cytosolic delivery to effectively induce apoptosis of cancer cells. The protein binder composed of leucine-rich repeat modules was selected for human Bcl-2, and its binding affinity was increased up to 60 nM through a modular evolution-based approach. The protein binder was efficiently delivered into cancer cells by an intracellular delivery system using a translocation domain from a bacterial exotoxin, resulting in a strong suppression of anti-apoptotic signaling in cancer cells. Our results demonstrate that the human Bcl-2-specific protein binder can act as a potent therapeutic agent for cancers.
Collapse
Affiliation(s)
- Yoo-Kyoung Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
| |
Collapse
|
64
|
Ngoi NYL, Choong C, Lee J, Bellot G, Wong ALA, Goh BC, Pervaiz S. Targeting Mitochondrial Apoptosis to Overcome Treatment Resistance in Cancer. Cancers (Basel) 2020; 12:E574. [PMID: 32131385 PMCID: PMC7139457 DOI: 10.3390/cancers12030574] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/23/2020] [Accepted: 02/27/2020] [Indexed: 01/09/2023] Open
Abstract
Deregulated cellular apoptosis is a hallmark of cancer and chemotherapy resistance. The B-cell lymphoma 2 (BCL-2) protein family members are sentinel molecules that regulate the mitochondrial apoptosis machinery and arbitrate cell fate through a delicate balance between pro- and anti-apoptotic factors. The recognition of the anti-apoptotic BCL2 gene as an oncogenic driver in hematological malignancies has directed attention toward unraveling the biological significance of each of the BCL-2 superfamily members in cancer progression and garnered interest in the targeting of apoptosis in cancer therapy. Accordingly, the approval of venetoclax (ABT-199), a small molecule BCL-2 inhibitor, in patients with chronic lymphocytic leukemia and acute myeloid leukemia has become the proverbial torchbearer for novel candidate drug approaches selectively targeting the BCL-2 superfamily. Despite the inspiring advances in this field, much remains to be learned regarding the optimal therapeutic context for BCL-2 targeting. Functional assays, such as through BH3 profiling, may facilitate prediction of treatment response, development of drug resistance and shed light on rational combinations of BCL-2 inhibitors with other branches of cancer therapy. This review summarizes the pathological roles of the BCL-2 family members in cancer, discusses the current landscape of their targeting in clinical practice, and highlights the potential for future therapeutic inroads in this important area.
Collapse
Affiliation(s)
- Natalie Yan Li Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
| | - Clarice Choong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
| | - Joanne Lee
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
| | - Gregory Bellot
- Department of Hand & Reconstructive Microsurgery, University Orthopedic, Hand & Reconstructive Microsurgery Cluster, National University Health System, Singapore 119228, Singapore;
| | - Andrea LA Wong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
- Cancer Science Institute, National University of Singapore, Singapore 117599, Singapore
| | - Boon Cher Goh
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore 119228, Singapore; (N.Y.L.N.); (C.C.); (J.L.); (A.L.W.); (B.C.G.)
- Cancer Science Institute, National University of Singapore, Singapore 117599, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 119077, Singapore
- National University Cancer Institute, National University Health System, Singapore 119228, Singapore
| |
Collapse
|
65
|
Whitaker RH, Placzek WJ. MCL1 binding to the reverse BH3 motif of P18INK4C couples cell survival to cell proliferation. Cell Death Dis 2020; 11:156. [PMID: 32111816 PMCID: PMC7048787 DOI: 10.1038/s41419-020-2351-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/11/2020] [Indexed: 12/20/2022]
Abstract
Commitment to cell cycle entry and cellular duplication is a tightly coordinated and regulated process. Once initiated, a series of multiple checkpoints ensure both accurate genomic replication and chromosomal separation. In the event of unsuccessful cell division, parallel pathways exist that induce the cell to undergo programmed cell death, or apoptosis. At the center of such stress-induced, intrinsic apoptotic regulation lies the BCL2 family of pro- and anti-apoptotic regulatory proteins. In a proliferative state the balance of pro- and anti-apoptotic signaling proteins would be expected to favor an excess population of anti-apoptotic members. While the anti-apoptotic BCL2 family member, MCL1, has been identified to oversee mitotic progression, direct communication between the BCL2 family and cell proliferation has not been observed. In this study, we demonstrate a direct protein–protein interaction between MCL1 and the G1/S checkpoint protein, P18INK4C. This interaction is mediated by a reverse BH3 (rBH3) motif located in P18INK4C’s C-terminal ankyrin repeat. MCL1 is further shown to decrease P18INK4C expression and thereby regulate cell cycle entry in a retinoblastoma (RB1)-dependent manner. Our findings establish a mechanism for translation independent and direct communication between the BCL2 family regulation of apoptosis and CDK4/6-RB regulation of early G1/S transition during cellular division/growth.
Collapse
Affiliation(s)
- Robert H Whitaker
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William J Placzek
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
66
|
Arunmanee W, Ecoy GAU, Khine HEE, Duangkaew M, Prompetchara E, Chanvorachote P, Chaotham C. Colicin N Mediates Apoptosis and Suppresses Integrin-Modulated Survival in Human Lung Cancer Cells. Molecules 2020; 25:E816. [PMID: 32069989 PMCID: PMC7070259 DOI: 10.3390/molecules25040816] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 01/26/2023] Open
Abstract
The inherent limitations, including serious side-effects and drug resistance, of current chemotherapies necessitate the search for alternative treatments especially for lung cancer. Herein, the anticancer activity of colicin N, bacteria-produced antibiotic peptide, was investigated in various human lung cancer cells. After 24 h of treatment, colicin N at 5-15 µM selectively caused cytotoxicity detected by MTT assay in human lung cancer H460, H292 and H23 cells with no noticeable cell death in human dermal papilla DPCs cells. Flow cytometry analysis of annexin V-FITC/propidium iodide indicated that colicin N primarily induced apoptosis in human lung cancer cells. The activation of extrinsic apoptosis evidenced with the reduction of c-FLIP and caspase-8, as well as the modulation of intrinsic apoptosis signaling proteins including Bax and Mcl-1 were observed via Western blot analysis in lung cancer cells cultured with colicin N (10-15 µM) for 12 h. Moreover, 5-15 µM of colicin N down-regulated the expression of activated Akt (p-Akt) and its upstream survival molecules, integrin β1 and αV in human lung cancer cells. Taken together, colicin N exhibits selective anticancer activity associated with suppression of integrin-modulated survival which potentiate the development of a novel therapy with high safety profile for treatment of human lung cancer.
Collapse
Affiliation(s)
- Wanatchaporn Arunmanee
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (W.A.); (G.A.U.E.); (H.E.E.K.); (M.D.)
- Vaccines and Therapeutic Proteins Research Group, the Special Task Force for Activating Research (STAR), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Gea Abigail U. Ecoy
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (W.A.); (G.A.U.E.); (H.E.E.K.); (M.D.)
- Department of Pharmacy, School of Health Care Professions, University of San Carlos, Cebu 6000, Philippines
| | - Hnin Ei Ei Khine
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (W.A.); (G.A.U.E.); (H.E.E.K.); (M.D.)
| | - Methawee Duangkaew
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (W.A.); (G.A.U.E.); (H.E.E.K.); (M.D.)
- Vaccines and Therapeutic Proteins Research Group, the Special Task Force for Activating Research (STAR), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Eakachai Prompetchara
- Vaccines and Therapeutic Proteins Research Group, the Special Task Force for Activating Research (STAR), Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Laboratory Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Vaccine Research and Development (Chula Vaccine Research Center-Chula VRC), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chatchai Chaotham
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (W.A.); (G.A.U.E.); (H.E.E.K.); (M.D.)
- Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
67
|
Enabling Combinatorial siRNA Delivery against Apoptosis-Related Proteins with Linoleic Acid and α-Linoleic Acid Substituted Low Molecular Weight Polyethylenimines. Pharm Res 2020; 37:46. [PMID: 32016611 DOI: 10.1007/s11095-020-2770-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Short interfering RNA (siRNA) therapy promises a new era in treatment of breast cancers but effective delivery systems are needed for clinical use. Since silencing complementary targets may offer improved efficacy, this study was undertaken to identify non-viral carriers for combinatorial siRNA delivery for more effective therapy. METHODS A library of lipid-substituted polymers from low molecular weight polyethyleneimine (PEI), linoleic acid (LA) and α-linoleic acid (αLA) with amide or thioester linkages was prepared and investigated for delivering Mcl-1, survivin and STAT5A siRNAs in breast cancer cells. RESULTS The effective polymers formed 80-190 nm particles with similar zeta-potentials, but the serum stability was greater for complexes formed with amide-linked lipid conjugates. The LA and αLA substitutions, with the low molecular weight PEI (1.2 kDa and 2.0 kDa) were able to deliver siRNA effectively to cells and retarded the growth of breast cancer cells. The amide-linked lipid substituents showed higher cellular delivery of siRNA as compared to thioester linkages. Upon combinational delivery of siRNAs, growth of MCF-7 cells was inhibited to a greater extent with 2.0PEI-LA9 mediated delivery of Mcl-1 combined survivin siRNAs as compared to individual siRNAs. The qRT-PCR analysis confirmed the decrease in mRNA levels of target genes with specific siRNAs and 2.0PEI-LA9 was the most effective polymer for delivering siRNAs (either single or in combination). CONCLUSIONS This study yielded effective siRNA carriers for combinational delivery of siRNAs. Careful choice of siRNA combinations will be critical since targeting individual genes might alter the expression of other critical mediators.
Collapse
|
68
|
Baggio C, Udompholkul P, Gambini L, Jossart J, Salem AF, Håkansson M, Perry JJP, Pellecchia M. N-locking stabilization of covalent helical peptides: Application to Bfl-1 antagonists. Chem Biol Drug Des 2020; 95:412-426. [PMID: 31898401 DOI: 10.1111/cbdd.13661] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/16/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Recently, it was reported that tetrapeptides cyclized via lactam bond between the amino terminus and a glutamic residue in position 4 (termed here N-lock) can nucleate helix formation in longer peptides. We applied such strategy to derive N-locked covalent BH3 peptides that were designed to selectively target the anti-apoptotic protein Bfl-1. The resulting agents were soluble in aqueous buffer and displayed a remarkable (low nanomolar) affinity for Bfl-1 and cellular activity. The crystal structure of the complex between such N-locked covalent peptide and Bfl-1 provided insights on the geometry of the N-locking strategy and of the covalent bond between the agent and Bfl-1.
Collapse
Affiliation(s)
- Carlo Baggio
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Parima Udompholkul
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Luca Gambini
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | - Jennifer Jossart
- Department of Biochemistry, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA, USA
| | - Ahmed F Salem
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| | | | - J Jefferson P Perry
- Department of Biochemistry, College of Natural and Agricultural Sciences, University of California Riverside, Riverside, CA, USA
| | - Maurizio Pellecchia
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, USA
| |
Collapse
|
69
|
Chang CY, Li JR, Wu CC, Wang JD, Liao SL, Chen WY, Wang WY, Chen CJ. Endoplasmic Reticulum Stress Contributes to Indomethacin-Induced Glioma Apoptosis. Int J Mol Sci 2020; 21:557. [PMID: 31952288 PMCID: PMC7013513 DOI: 10.3390/ijms21020557] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/07/2020] [Accepted: 01/14/2020] [Indexed: 02/07/2023] Open
Abstract
The dormancy of cellular apoptotic machinery has been highlighted as a crucial factor in therapeutic resistance, recurrence, and poor prognosis in patients with malignancy, such as malignant glioma. Increasing evidence indicates that nonsteroidal anti-inflammatory drugs (NSAIDs) confer chemopreventive effects, and indomethacin has been shown to have a novel chemotherapeutic application targeting glioma cells. To extend these findings, herein, we studied the underlying mechanisms of apoptosis activation caused by indomethacin in human H4 and U87 glioma cells. We found that the glioma cell-killing effects of indomethacin involved both death receptor- and mitochondria-mediated apoptotic cascades. Indomethacin-induced glioma cell apoptosis was accompanied by a series of biochemical changes, including reactive oxygen species generation, endoplasmic reticulum (ER) stress, apoptosis signal-regulating kinase-1 (Ask1) activation, p38 hyperphosphorylation, protein phosphatase 2A (PP2A) activation, Akt dephosphorylation, Mcl-1 and FLICE-inhibiting protein (FLIP) downregulation, Bax mitochondrial distribution, and caspases 3/caspase 8/caspase 9 activation. Data on pharmacological inhibition related to oxidative stress, ER stress, free Ca2+, and p38 revealed that the axis of oxidative stress/ER stress/Ask1/p38/PP2A/Akt comprised an apoptotic cascade leading to Mcl-1/FLIP downregulation and glioma apoptosis. Since indomethacin is an emerging choice in chemotherapy and its antineoplastic effects have been demonstrated in glioma tumor-bearing models, the findings further strengthen the argument for turning on the aforementioned axis in order to activate the apoptotic machinery of glioma cells.
Collapse
Affiliation(s)
- Cheng-Yi Chang
- Department of Surgery, Feng Yuan Hospital, Taichung City 420, Taiwan;
| | - Jian-Ri Li
- Division of Urology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Financial Engineering, Providence University, Taichung City 433, Taiwan
- Department of Data Science and Big Data Analytics, Providence University, Taichung City 433, Taiwan
| | - Jiaan-Der Wang
- Children’s Medical Center, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Industrial Engineering and Enterprise Information, Tunghai University, Taichung City 407, Taiwan
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung Hsing University, Taichung City 402, Taiwan;
| | - Wen-Yi Wang
- Department of Nursing, HungKuang University, Taichung City 433, Taiwan;
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan
| |
Collapse
|
70
|
Rocha-Viegas L, Silbermins M, Ogara MF, Pellegrini JM, Nuñez SY, García VE, Vicent GP, Pecci A. Glucocorticoids uncover a critical role for ASH2L on BCL-X expression regulation in leukemia cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1863:194475. [PMID: 31870784 DOI: 10.1016/j.bbagrm.2019.194475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 12/23/2022]
Abstract
Targeting the apoptosis machinery is a promising therapeutic approach in myeloid malignancies. BCL2L1 is a well-known glucocorticoid-responsive gene and a key apoptosis regulator that, when over-expressed, can contribute to tumor development, progression and therapeutic resistance. Moreover, synthetic glucocorticoids, like dexamethasone, are frequently used in the treatment of hematopoietic diseases due to its pro-apoptotic properties. We report here that the trithorax protein ASH2L, considered one of the core subunits of H3K4-specific MLL/SET methyltransferase complexes, contributes to anti-apoptotic BCL-XL over-expression and cell survival in patient-derived myeloid leukemia cells. We find that the unliganded glucocorticoid receptor (uGR) and ASH2L interact in a common protein complex through a chromatin looping determined by uGR and ASH2L binding to BCL2L1 specific +58 HRE and promoter region, respectively. Upon addition of dexamethasone, GR and ASH2L recruitment is reduced, BCL-XL expression diminishes and apoptosis is induced consequently. Overall, our findings indicate that uGR and ASH2L may act as key regulatory players of BCL- XL upregulation in AML cells.
Collapse
Affiliation(s)
- Luciana Rocha-Viegas
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), C1428EHA, Buenos Aires, Argentina.
| | - Micaela Silbermins
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), C1428EHA, Buenos Aires, Argentina
| | - María Florencia Ogara
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), C1428EHA, Buenos Aires, Argentina
| | - Joaquín Miguel Pellegrini
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), C1428EHA, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, C1428EHA, Buenos Aires, Argentina
| | - Sol Yanel Nuñez
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), C1428ADN, Buenos Aires, Argentina
| | - Verónica Edith García
- CONICET-Universidad de Buenos Aires, Instituto de Química Biológica Ciencias Exactas y Naturales (IQUIBICEN), C1428EHA, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, C1428EHA, Buenos Aires, Argentina
| | - Guillermo Pablo Vicent
- Department of Molecular Genomics, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri Reixac 4-8, 08028, Barcelona, Spain
| | - Adali Pecci
- CONICET-Universidad de Buenos Aires, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), C1428EHA, Buenos Aires, Argentina; Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, C1428EHA, Buenos Aires, Argentina
| |
Collapse
|
71
|
Kønig SM, Rissler V, Terkelsen T, Lambrughi M, Papaleo E. Alterations of the interactome of Bcl-2 proteins in breast cancer at the transcriptional, mutational and structural level. PLoS Comput Biol 2019; 15:e1007485. [PMID: 31825969 PMCID: PMC6927658 DOI: 10.1371/journal.pcbi.1007485] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/23/2019] [Accepted: 10/12/2019] [Indexed: 12/11/2022] Open
Abstract
Apoptosis is an essential defensive mechanism against tumorigenesis. Proteins of the B-cell lymphoma-2 (Bcl-2) family regulate programmed cell death by the mitochondrial apoptosis pathway. In response to intracellular stress, the apoptotic balance is governed by interactions of three distinct subgroups of proteins; the activator/sensitizer BH3 (Bcl-2 homology 3)-only proteins, the pro-survival, and the pro-apoptotic executioner proteins. Changes in expression levels, stability, and functional impairment of pro-survival proteins can lead to an imbalance in tissue homeostasis. Their overexpression or hyperactivation can result in oncogenic effects. Pro-survival Bcl-2 family members carry out their function by binding the BH3 short linear motif of pro-apoptotic proteins in a modular way, creating a complex network of protein-protein interactions. Their dysfunction enables cancer cells to evade cell death. The critical role of Bcl-2 proteins in homeostasis and tumorigenesis, coupled with mounting insight in their structural properties, make them therapeutic targets of interest. A better understanding of gene expression, mutational profile, and molecular mechanisms of pro-survival Bcl-2 proteins in different cancer types, could help to clarify their role in cancer development and may guide advancement in drug discovery. Here, we shed light on the pro-survival Bcl-2 proteins in breast cancer using different bioinformatic approaches, linking -omics with structural data. We analyzed the changes in the expression of the Bcl-2 proteins and their BH3-containing interactors in breast cancer samples. We then studied, at the structural level, a selection of interactions, accounting for effects induced by mutations found in the breast cancer samples. We find two complexes between the up-regulated Bcl2A1 and two down-regulated BH3-only candidates (i.e., Hrk and Nr4a1) as targets associated with reduced apoptosis in breast cancer samples for future experimental validation. Furthermore, we predict L99R, M75R as damaging mutations altering protein stability, and Y120C as a possible allosteric mutation from an exposed surface to the BH3-binding site.
Collapse
Affiliation(s)
- Simon Mathis Kønig
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Vendela Rissler
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Thilde Terkelsen
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Matteo Lambrughi
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Elena Papaleo
- Computational Biology Laboratory, Danish Cancer Society Research Center, Copenhagen, Denmark
- Translational Disease Systems Biology, Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
72
|
Targeting Apoptotic Pathways in Acute Myeloid Leukaemia. Cancers (Basel) 2019; 11:cancers11111660. [PMID: 31717784 PMCID: PMC6895902 DOI: 10.3390/cancers11111660] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022] Open
Abstract
Acute Myeloid Leukaemia is a devastating disease that continues to have a poor outcome for the majority of patients. In recent years, however, a number of drugs have received FDA approval, following on from successful clinical trial results. This parallels the characterization of the molecular landscape of Acute Myeloid Leukaemia (AML) over the last decade, which has led to the development of drugs targeting newly identified recurring mutations. In addition, basic biological research into the pathobiology of AML has identified aberrant programmed cell death pathways in AML. Following on from successful outcomes in lymphoid malignancies, drugs targeting the B Cell Lymphoma 2 (BCL-2) family of anti-apoptotic proteins have been explored in AML. In this review, we will outline the preclinical and clinical work to date supporting the role of drugs targeting BCL-2, with Venetoclax being the most advanced to date. We will also highlight rationale combinations using Venetoclax, ongoing clinical trials and biomarkers of response identified from the early phase clinical trials performed.
Collapse
|
73
|
Denis C, Sopková-de Oliveira Santos J, Bureau R, Voisin-Chiret AS. Hot-Spots of Mcl-1 Protein. J Med Chem 2019; 63:928-943. [DOI: 10.1021/acs.jmedchem.9b00983] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Camille Denis
- Normandie Univiversité, UNICAEN, CERMN, 14000 Caen, France
| | | | - Ronan Bureau
- Normandie Univiversité, UNICAEN, CERMN, 14000 Caen, France
| | | |
Collapse
|
74
|
Gu X, Peng Y, Zhao Y, Liang X, Tang Y, Liu J. A novel derivative of artemisinin inhibits cell proliferation and metastasis via down-regulation of cathepsin K in breast cancer. Eur J Pharmacol 2019; 858:172382. [DOI: 10.1016/j.ejphar.2019.05.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 02/04/2023]
|
75
|
HUWE1 controls MCL1 stability to unleash AMBRA1-induced mitophagy. Cell Death Differ 2019; 27:1155-1168. [PMID: 31434979 DOI: 10.1038/s41418-019-0404-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 11/08/2022] Open
Abstract
Receptor-mediated mitophagy is a crucial process involved in mitochondria quality control. AMBRA1 is a mitophagy receptor for the selective removal of damaged mitochondria in mammalian cells. A critical unresolved issue is how AMBRA1-mediated mitophagy is controlled in response to cellular stress. Here, we investigated the role of BCL2-family proteins on AMBRA1-dependent mitophagy and showed that MCL1 delays AMBRA1-dependent mitophagy. Indeed, MCL1 overexpression is sufficient to inhibit recruitment to mitochondria of the E3 Ubiquitin ligase HUWE1, a crucial dynamic partner of AMBRA1, upon AMBRA1-mediated mitophagy induction. In addition, we found that during mitophagy induced by AMBRA1, MCL1 levels decreased but were sustained by inhibition of the GSK-3β kinase, which delayed AMBRA1-mediated mitophagy. Also, we showed that MCL1 was phosphorylated by GSK-3β at a conserved GSK-3 phosphorylation site (S159) during AMBRA1-mediated mitophagy and that this event was accompanied by HUWE1-dependent MCL1 degradation. Altogether, our results demonstrate that MCL1 stability is regulated by the kinase GSK-3β and the E3 ubiquitin ligase HUWE1 in regulating AMBRA1-mediated mitophagy. Our work thus defines MCL1 as an upstream stress-sensitive protein, functional in AMBRA1-mediated mitophagy.
Collapse
|
76
|
Lu P, Bowman KER, Brown SM, Joklik-Mcleod M, Mause ERV, Nguyen HTN, Lim CS. p53-Bad: A Novel Tumor Suppressor/Proapoptotic Factor Hybrid Directed to the Mitochondria for Ovarian Cancer Gene Therapy. Mol Pharm 2019; 16:3386-3398. [PMID: 31241338 PMCID: PMC10760809 DOI: 10.1021/acs.molpharmaceut.9b00136] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Clinical trials involving p53 gene therapy for ovarian cancer failed due to the dominant negative inhibition of wild-type p53 and multiple genetic aberrations in ovarian cancer. To overcome this problem, we have designed a more potent chimeric gene fusion, called p53-Bad, that combines p53 with the mitochondrial pro-apoptotic factor Bad. Unlike wild-type p53, which acts as a nuclear transcription factor, this novel p53-Bad construct has multiple unique mechanisms of action including a direct and rapid apoptotic effect at the mitochondria. The mitochondrial localization, transcription activity, and apoptotic activity of the constructs were tested. The results suggest that p53 can be effectively targeted to the mitochondria by controlling the phosphorylation of pro-apoptotic Bad, which can only localize to the mitochondria when Ser-112 and Ser-136 of Bad are unphosphorylated. By introducing S112A and S136A mutations, p53-Bad fusion cannot be phosphorylated at these two sites and always localizes to the mitochondria. p53-Bad constructs also have superior activity over p53 and Bad alone. The apoptotic activity is consistent in many ovarian cancer cell lines regardless of the endogenous p53 status. Both p53 and the BH3 domain of Bad contribute to the superior activity of p53-Bad. Our data suggests that p53-Bad fusions are capable of inducing apoptosis and should be further pursued for gene therapy for ovarian cancer.
Collapse
Affiliation(s)
- Phong Lu
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Katherine E. Redd Bowman
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Sarah M. Brown
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Madeline Joklik-Mcleod
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Erica R. Vander Mause
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Han T. N. Nguyen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Carol S. Lim
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
77
|
Akbarzadeh Khiavi M, Safary A, Aghanejad A, Barar J, Rasta SH, Golchin A, Omidi Y, Somi MH. Enzyme-conjugated gold nanoparticles for combined enzyme and photothermal therapy of colon cancer cells. Colloids Surf A Physicochem Eng Asp 2019. [DOI: 10.1016/j.colsurfa.2019.04.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
78
|
Chen C, Nie Y, Xu G, Yang X, Fang H, Hou X. Design, synthesis and preliminary bioactivity studies of indomethacin derivatives as Bcl-2/Mcl-1 dual inhibitors. Bioorg Med Chem 2019; 27:2771-2783. [DOI: 10.1016/j.bmc.2019.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/22/2019] [Accepted: 05/04/2019] [Indexed: 11/28/2022]
|
79
|
Lu P, Vander Mause ER, Redd Bowman KE, Brown SM, Ahne L, Lim CS. Mitochondrially targeted p53 or DBD subdomain is superior to wild type p53 in ovarian cancer cells even with strong dominant negative mutant p53. J Ovarian Res 2019; 12:45. [PMID: 31092272 PMCID: PMC6521536 DOI: 10.1186/s13048-019-0516-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/18/2019] [Indexed: 12/26/2022] Open
Abstract
Background While tumor suppressor p53 functions primarily as a transcription factor in the nucleus, cellular stress can cause p53 to translocate to the mitochondria and directly trigger a rapid apoptotic response. We have previously shown that fusing p53 (or its DNA binding domain, DBD, alone) to the mitochondrial targeting signal (MTS) from Bak or Bax can target p53 to the mitochondria and induce apoptosis in gynecological cancer cell lines including cervical cancer cells (HeLa; wt p53), ovarian cancer cells (SKOV-3; p53 267del non-expressing), and breast cancer cells (T47D; L194F p53 mutation). However, p53 with Bak or Bax MTSs have not been previously tested in cancers with strong dominant negative (DN) mutant p53 which are capable of inactivating wt p53 by homo-oligomerization. Since p53-Bak or Bax MTS constructs act as monomers, they are not subject to DN inhibition. For this study, the utility of p53-Bak or p53-Bax MTS constructs was tested for ovarian cancers which are known to have varying p53 statuses, including a strong DN contact mutant p53 (Ovcar-3 cells), a p53 DN structural mutant (Kuramochi cells), and a p53 wild type, low expressing cells (ID8). Results Our mitochondrial p53 constructs were tested for their ability to localize to the mitochondria in both mutant non-expressing p53 (Skov-3) and p53 structural mutant (Kuramochi) cell lines using fluorescence microscopy and a nuclear transcriptional activity assay. The apoptotic activity of these mitochondrial constructs was determined using a mitochondrial outer membrane depolarization assay (TMRE), caspase assay, and a late stage cell death assay (7-AAD). We also tested the possibility of using our constructs with paclitaxel, the current standard of care in ovarian cancer treatment. Our data indicates that our mitochondrial p53 constructs are able to effectively localize to the mitochondria in cancer cells with structural mutant p53 and induce apoptosis in many ovarian cancer cell lines with different p53 statuses. These constructs can also be used in combination with paclitaxel for an increased apoptotic effect. Conclusions The results suggest that targeting p53 to mitochondria can be a new strategy for ovarian cancer treatment. Electronic supplementary material The online version of this article (10.1186/s13048-019-0516-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Phong Lu
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S 2000 E Rm 301, Salt Lake City, UT, 84112, USA
| | - Erica R Vander Mause
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S 2000 E Rm 301, Salt Lake City, UT, 84112, USA
| | - Katherine E Redd Bowman
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S 2000 E Rm 301, Salt Lake City, UT, 84112, USA
| | - Sarah M Brown
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S 2000 E Rm 301, Salt Lake City, UT, 84112, USA
| | - Lisa Ahne
- Philipps-Universitat Marburg, Biegenstraße 10, Marburg, 35037, Germany
| | - Carol S Lim
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 S 2000 E Rm 301, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
80
|
Lee EF, Harris TJ, Tran S, Evangelista M, Arulananda S, John T, Ramnac C, Hobbs C, Zhu H, Gunasingh G, Segal D, Behren A, Cebon J, Dobrovic A, Mariadason JM, Strasser A, Rohrbeck L, Haass NK, Herold MJ, Fairlie WD. BCL-XL and MCL-1 are the key BCL-2 family proteins in melanoma cell survival. Cell Death Dis 2019; 10:342. [PMID: 31019203 PMCID: PMC6482196 DOI: 10.1038/s41419-019-1568-3] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/28/2019] [Accepted: 04/02/2019] [Indexed: 12/17/2022]
Abstract
Malignant melanoma is one of the most difficult cancers to treat due to its resistance to chemotherapy. Despite recent successes with BRAF inhibitors and immune checkpoint inhibitors, many patients do not respond or become resistant to these drugs. Hence, alternative treatments are still required. Due to the importance of the BCL-2-regulated apoptosis pathway in cancer development and drug resistance, it is of interest to establish which proteins are most important for melanoma cell survival, though the outcomes of previous studies have been conflicting. To conclusively address this question, we tested a panel of established and early passage patient-derived cell lines against several BH3-mimetic drugs designed to target individual or subsets of pro-survival BCL-2 proteins, alone and in combination, in both 2D and 3D cell cultures. None of the drugs demonstrated significant activity as single agents, though combinations targeting MCL-1 plus BCL-XL, and to a lesser extent BCL-2, showed considerable synergistic killing activity that was elicited via both BAX and BAK. Genetic deletion of BFL-1 in cell lines that express it at relatively high levels only had minor impact on BH3-mimetic drug sensitivity, suggesting it is not a critical pro-survival protein in melanoma. Combinations of MCL-1 inhibitors with BRAF inhibitors also caused only minimal additional melanoma cell killing over each drug alone, whilst combinations with the proteasome inhibitor bortezomib was more effective in multiple cell lines. Our data show for the first time that therapies targeting specific combinations of BCL-2 pro-survival proteins, namely MCL-1 plus BCL-XL and MCL-1 plus BCL-2, could have significant benefit for the treatment of melanoma.
Collapse
Affiliation(s)
- Erinna F Lee
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia. .,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia. .,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia.
| | - Tiffany J Harris
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - Sharon Tran
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Marco Evangelista
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - Surein Arulananda
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Thomas John
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Celeste Ramnac
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Chloe Hobbs
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Haoran Zhu
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Gency Gunasingh
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, QLD, 4102, Australia
| | - David Segal
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Andreas Behren
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Jonathan Cebon
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Alexander Dobrovic
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - John M Mariadason
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Leona Rohrbeck
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Karolinska Institute, Stockholm, Sweden
| | - Nikolas K Haass
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Brisbane, QLD, 4102, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - W Douglas Fairlie
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia. .,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia. .,School of Cancer Medicine, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
81
|
Myeloid Cell Leukemia-1 (MCL-1) siRNA Therapy Showed Cytotoxic Effect on T Cells Acute Lymphoblastic Leukemia. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2019. [DOI: 10.5812/ijcm.87773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
82
|
Lever JR, Fergason-Cantrell EA. Allosteric modulation of sigma receptors by BH3 mimetics ABT-737, ABT-263 (Navitoclax) and ABT-199 (Venetoclax). Pharmacol Res 2019; 142:87-100. [PMID: 30721730 DOI: 10.1016/j.phrs.2019.01.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/12/2018] [Accepted: 01/21/2019] [Indexed: 11/21/2022]
Abstract
ABT-737, ABT-263 (Navitoclax) and ABT-199 (Venetoclax) are under intensive preclinical and clinical investigation as treatments for hematologic and other malignancies. These small molecules mimic pro-death B-cell lymphoma-2 (Bcl-2) Homology 3 (BH3) domain-only proteins. They also bear a structural resemblance to certain sigma (σ) receptor ligands. Moreover, the Bcl-2 and σ receptor protein families are both located primarily at the endoplasmic reticulum, mediate cell death and survival through protein-protein interactions, and physically associate. Accordingly, we examined the ability of the ABT series of BH3 mimetics to interact with σ receptors using radioligand-binding techniques. Negative allosteric modulation of [3H](+)-pentazocine, an agonist, binding to σ1 receptors in guinea pig brain membranes was observed for ABT-737, ABT-263 and ABT-199. Findings included reduction of specific binding to distinct plateaus in concentration-dependent fashion, significant slowing of radioligand dissociation kinetics, and decreases in radioligand affinity with no or modest changes in maximal receptor densities. Using a ternary complex model, dissociation constants (KX) for modulator binding to the σ1 receptor ranged from 1 to 2.5 μM, while negative cooperativity factors (α), representing the changes in affinity of ligand and modulator when bound as a ternary complex with the receptor, ranged from 0.15 to 0.42. These observations were extended and reinforced by studies using intact small cell (NCI-H69) and non-small cell (NCI-H23) lung cancer cells, and by using an antagonist σ1 receptor radioligand, E-N-1-(3'-[125I]iodoallyl)-N'-4-(3″,4″-dimethoxyphenethyl)piperazine, in mouse brain membranes. By contrast, exploratory studies indicate marked enhancement of the σ2 receptor binding of [3H]1,3-di-(o-tolyl)guanidine/(+)-pentazocine in NCI-H23 cells and guinea pig brain membranes. These findings raise intriguing questions regarding mechanism and potential functional outcomes.
Collapse
Affiliation(s)
- John R Lever
- Department of Radiology, University of Missouri, Columbia, MO 65211, USA; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA.
| | - Emily A Fergason-Cantrell
- Department of Radiology, University of Missouri, Columbia, MO 65211, USA; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO 65201, USA
| |
Collapse
|
83
|
Lee T, Christov PP, Shaw S, Tarr JC, Zhao B, Veerasamy N, Jeon KO, Mills JJ, Bian Z, Sensintaffar JL, Arnold AL, Fogarty SA, Perry E, Ramsey HE, Cook RS, Hollingshead M, Davis Millin M, Lee KM, Koss B, Budhraja A, Opferman JT, Kim K, Arteaga CL, Moore WJ, Olejniczak ET, Savona MR, Fesik SW. Discovery of Potent Myeloid Cell Leukemia-1 (Mcl-1) Inhibitors That Demonstrate in Vivo Activity in Mouse Xenograft Models of Human Cancer. J Med Chem 2019; 62:3971-3988. [DOI: 10.1021/acs.jmedchem.8b01991] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Taekyu Lee
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Plamen P. Christov
- Chemical Synthesis Core, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Subrata Shaw
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - James C. Tarr
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Bin Zhao
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Nagarathanam Veerasamy
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Kyu Ok Jeon
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Jonathan J. Mills
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Zhiguo Bian
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - John L. Sensintaffar
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Allison L. Arnold
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Stuart A. Fogarty
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Evan Perry
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Haley E. Ramsey
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee 37232, United States
| | - Rebecca S. Cook
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | | | | | - Kyung-min Lee
- Department of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Brian Koss
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Amit Budhraja
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Joseph T. Opferman
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105, United States
| | - Kwangho Kim
- Chemical Synthesis Core, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Carlos L. Arteaga
- Department of Hematology and Oncology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - William J. Moore
- Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, United States
| | - Edward T. Olejniczak
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| | - Michael R. Savona
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee 37232, United States
| | - Stephen W. Fesik
- Department of Biochemistry, Vanderbilt University School of Medicine, 2215 Garland Avenue, 607 Light Hall, Nashville, Tennessee 37232-0146, United States
| |
Collapse
|
84
|
Farmer T, O'Neill KL, Naslavsky N, Luo X, Caplan S. Retromer facilitates the localization of Bcl-xL to the mitochondrial outer membrane. Mol Biol Cell 2019; 30:1138-1146. [PMID: 30840537 PMCID: PMC6724524 DOI: 10.1091/mbc.e19-01-0044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The anti-apoptotic Bcl-2 family protein Bcl-xL plays a critical role in cell survival by protecting the integrity of the mitochondrial outer membrane (MOM). The mechanism through which Bcl-xL is recruited to the MOM has not been fully discerned. The retromer is a conserved endosomal scaffold complex involved in membrane trafficking. Here we identify VPS35 and VPS26, two core components of the retromer, as novel regulators of Bcl-xL. We observed interactions and colocalization between Bcl-xL, VPS35, VPS26, and MICAL-L1, a protein involved in recycling endosome biogenesis that also interacts with the retromer. We also found that upon VPS35 depletion, levels of nonmitochondrial Bcl-xL were increased. In addition, retromer-depleted cells displayed more rapid Bax activation and apoptosis. These results suggest that the retromer regulates apoptosis by facilitating Bcl-xL's transport to the MOM. Importantly, our studies suggest a previously uncharacterized relationship between the machineries of cell death/survival and endosomal trafficking.
Collapse
Affiliation(s)
- Trey Farmer
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Katelyn L O'Neill
- Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Naava Naslavsky
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Xu Luo
- Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| | - Steve Caplan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870.,Eppley Institute for Research in Cancer and Allied Diseases, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870
| |
Collapse
|
85
|
Potential Analysis and Preparation of Chitosan Oligosaccharides as Oral Nutritional Supplements of Cancer Adjuvant Therapy. Int J Mol Sci 2019; 20:ijms20040920. [PMID: 30791594 PMCID: PMC6412339 DOI: 10.3390/ijms20040920] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 02/07/2023] Open
Abstract
Cancer is considered to have an adverse influence on health around the world. Chitosan, a linear polysaccharide that contains copolymers of β-1-4 linked d-glucosamine and N-acetyl-d-glucosamine units, has been widely used in the field of biomedicine, owing to its nontoxicity, biocompatibility, biodegradability, and hemocompatibility. This study was aimed at preparing the chitosan oligosaccharides (COS) and examining its ability on suppressing lung cancer in vitro and in vivo. Human non-small-cell lung cancer A549 cells model and C57BL/6 mice bearing lung cancer model were adopted. COS showed inhibition on the viability and proliferation of lung carcinoma cells (A549) in time-dependent manners, but no cytotoxicity to human liver cell (HL-7702). Moreover, COS could significantly increase Bax expression of A549 cells while decreasing Bcl-2 expression. COS supplementation significantly inhibited the growth of Lewis tissues and promoted necrosis of tumor cells in vivo. After treatment with COS, significantly elevated concentrations of Bax and reduced expression of Bcl-2 in tumor tissues, as well as elevated levels of TNF-α, IL-2, Fas and Fas-L in mice serum were observed (p < 0.05). In conclusion, COS had certain anti-tumor effects and potential application as a synergic functional food ingredient to prevent cancer.
Collapse
|
86
|
Bittremieux M, La Rovere RM, Schuermans M, Luyten T, Mikoshiba K, Vangheluwe P, Parys JB, Bultynck G. Extracellular and ER-stored Ca 2+ contribute to BIRD-2-induced cell death in diffuse large B-cell lymphoma cells. Cell Death Discov 2018; 4:101. [PMID: 30416758 PMCID: PMC6214954 DOI: 10.1038/s41420-018-0118-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/26/2018] [Indexed: 11/28/2022] Open
Abstract
The anti-apoptotic protein Bcl-2 is upregulated in several cancers, including diffuse large B-cell lymphoma (DLBCL) and chronic lymphocytic leukemia (CLL). In a subset of these cancer cells, Bcl-2 blocks Ca2+-mediated apoptosis by suppressing the function of inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs) located at the endoplasmic reticulum (ER). A peptide tool, called Bcl-2/IP3 receptor disruptor-2 (BIRD-2), was developed to disrupt Bcl-2/IP3R complexes, triggering pro-apoptotic Ca2+ signals and killing Bcl-2-dependent cancer cells. In DLBCL cells, BIRD-2 sensitivity depended on the expression level of IP3R2 channels and constitutive IP3 signaling downstream of the B-cell receptor. However, other cellular pathways probably also contribute to BIRD-2-provoked cell death. Here, we examined whether BIRD-2-induced apoptosis depended on extracellular Ca2+ and more particularly on store-operated Ca2+ entry (SOCE), a Ca2+-influx pathway activated upon ER-store depletion. Excitingly, DPB162-AE, a SOCE inhibitor, suppressed BIRD-2-induced cell death in DLBCL cells. However, DPB162-AE not only inhibits SOCE but also depletes the ER Ca2+ store. Treatment of the cells with YM-58483 and GSK-7975A, two selective SOCE inhibitors, did not protect against BIRD-2-induced apoptosis. Similar data were obtained by knocking down STIM1 using small interfering RNA. Yet, extracellular Ca2+ contributed to BIRD-2 sensitivity in DLBCL, since the extracellular Ca2+ buffer ethylene glycol tetraacetic acid (EGTA) blunted BIRD-2-triggered apoptosis. The protective effects observed with DPB162-AE are likely due to ER Ca2+-store depletion, since a similar protective effect could be obtained using the sarco/endoplasmic reticulum Ca2+-ATPase inhibitor thapsigargin. Thus, both the ER Ca2+-store content and extracellular Ca2+, but not SOCE, are critical factors underlying BIRD-2-provoked cell death.
Collapse
Affiliation(s)
- Mart Bittremieux
- 1Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven and Leuven Kanker Instituut, Leuven, 3000 Belgium
| | - Rita M La Rovere
- 1Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven and Leuven Kanker Instituut, Leuven, 3000 Belgium
| | - Marleen Schuermans
- 2Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, 3000 Belgium
| | - Tomas Luyten
- 1Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven and Leuven Kanker Instituut, Leuven, 3000 Belgium
| | - Katsuhiko Mikoshiba
- 3The Laboratory for Developmental Neurobiology, Brain Science Institute, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198 Japan
| | - Peter Vangheluwe
- 2Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, 3000 Belgium
| | - Jan B Parys
- 1Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven and Leuven Kanker Instituut, Leuven, 3000 Belgium
| | - Geert Bultynck
- 1Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven and Leuven Kanker Instituut, Leuven, 3000 Belgium
| |
Collapse
|
87
|
Vallet S, Fan F, Malvestiti S, Pecherstorfer M, Sattler M, Schneeweiss A, Schulze-Bergkamen H, Opferman JT, Cardone MH, Jäger D, Podar K. Rationally derived drug combinations with the novel Mcl-1 inhibitor EU-5346 in breast cancer. Breast Cancer Res Treat 2018; 173:585-596. [PMID: 30374681 DOI: 10.1007/s10549-018-5022-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/20/2018] [Indexed: 01/19/2023]
Abstract
PURPOSE Recent studies have emphasized a key role for the anti-apoptotic Bcl-2 family member Mcl-1 in conferring tumor cell survival and drug resistance in breast cancer (BC). Mcl-1 inhibitors, such as the BH3-mimetic EU-5346, therefore represent an exciting new class of targeting agents and are a current focus of widespread cancer-drug development efforts. METHODS ONCOMINE analysis was utilized to compare expression profiles of Bcl-2 family members across all major BC subgroups. Potential toxicities of EU-5346 were evaluated using iPS-generated cardiomyocytes, blood cells and astrocytes. The anti-BC cell activity of EU-5346-based therapies was evaluated using [3H]-thymidine uptake and spheroid-forming assays as well as immunoblotting and the Chou-Talalay method. Protein level-based activity of EU-5346, the specific anti-Bcl-2 inhibitor ABT-199 and the specific anti-Bcl-xL inhibitor WEHI-539 was verified in Mcl-1Δ/null versus Mcl-1wt/wt MEFs. RESULTS We previously demonstrated significant anti-tumor activity of EU-5346 in all BC subtypes. Our present results go further and suggest that EU-5346 may induce limited adverse events such as cardiotoxicity, hematotoxicity, and neurotoxicity, frequently observed with other BH3 mimetics. As demonstrated by our mathematical scoring model, the prediction of EU-5643-induced IC50 not only relies on the protein level of Mcl-1 but also on Bak, Bim, and Noxa. Synergistic anti-BC activity of low-dose EU-5346 with the BH3 mimetics ABT-199 or WEHI-539 was observed only in those BC cells expressing Bcl-2 or Bcl-xL, respectively. Similarly, when combined with tamoxifen or trastuzumab, low-dose EU-5346 induced significant anti-BC activity in hormone receptor positive or Her2-positive BC cells, respectively. Finally, EU-5346 in combination with paclitaxel induced synergistic anti-BC activity in both paclitaxel-sensitive and paclitaxel-resistant TNBC cells. CONCLUSION These data strongly support the further clinical development of EU-5346 to improve BC patient survival.
Collapse
Affiliation(s)
- Sonia Vallet
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Fengjuan Fan
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Stefano Malvestiti
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
| | - Martin Pecherstorfer
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Andreas Schneeweiss
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
| | - Henning Schulze-Bergkamen
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
| | | | | | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Applied Tumor Immunity, Heidelberg, Germany
| | - Klaus Podar
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), University of Heidelberg, Heidelberg, Germany.
- Department of Internal Medicine II, University Hospital Krems, Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria.
| |
Collapse
|
88
|
Rossi A, Pizzo P, Filadi R. Calcium, mitochondria and cell metabolism: A functional triangle in bioenergetics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:1068-1078. [PMID: 30982525 DOI: 10.1016/j.bbamcr.2018.10.016] [Citation(s) in RCA: 297] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/19/2018] [Accepted: 10/21/2018] [Indexed: 12/18/2022]
Abstract
The versatility of mitochondrial metabolism and its fine adjustments to specific physiological or pathological conditions regulate fundamental cell pathways, ranging from proliferation to apoptosis. In particular, Ca2+ signalling has emerged as a key player exploited by mitochondria to tune their activity according with cell demand. The functional interaction between mitochondria and endoplasmic reticulum (ER) deeply impacts on the correct mitochondrial Ca2+ signal, thus modulating cell bioenergetics and functionality. Indeed, Ca2+ released by the ER is taken up by mitochondria where, both in the intermembrane space and in the matrix, it regulates the activity of transporters, enzymes and proteins involved in organelles' metabolism. In this review, we will briefly summarize Ca2+-dependent mechanisms involved in the regulation of mitochondrial activity. Moreover, we will discuss some recent reports, in which alterations in mitochondrial Ca2+ signalling have been associated with specific pathological conditions, such as neurodegeneration and cancer.
Collapse
Affiliation(s)
- Alice Rossi
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy
| | - Paola Pizzo
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy; Neuroscience Institute - Italian National Research Council (CNR), 35131 Padova, Italy.
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/B, 35131 Padova, Italy
| |
Collapse
|
89
|
Arachchige D, Holub JM. Synthesis and Biological Activity of Scyllatoxin-Based BH3 Domain Mimetics Containing Two Disulfide Linkages. Protein J 2018; 37:428-443. [PMID: 30128635 DOI: 10.1007/s10930-018-9791-9] [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] [Indexed: 11/25/2022]
Abstract
The B cell lymphoma 2 (BCL2) proteins are a family of evolutionarily related proteins that act as positive or negative regulators of the intrinsic apoptosis pathway. Overexpression of anti-apoptotic BCL2 proteins in cells is associated with apoptotic resistance, which can result in cancerous phenotypes and pathogenic cell survival. Consequently, anti-apoptotic BCL2 proteins have attracted considerable interest as therapeutic targets. We recently reported the development of a novel class of synthetic protein based on scyllatoxin (ScTx) designed to mimic the helical BH3 interaction domain of the pro-apoptotic BCL2 protein Bax. These studies showed that the number and position of native disulfide linkages contained within the ScTx-Bax structure significantly influences the ability for these constructs to target anti-apoptotic BCL2 proteins in vitro. The goal of the present study is to investigate the contribution of two disulfide linkages in the folding and biological activity of ScTx-Bax proteins. Here, we report the full chemical synthesis of three ScTx-Bax sequence variants, each presenting two native disulfide linkages at different positions within the folded structure. It was observed that two disulfide linkages were sufficient to fold ScTx-Bax proteins into native-like architectures reminiscent of wild-type ScTx. Furthermore, we show that select (bis)disulfide ScTx-Bax variants can target Bcl-2 (proper) in vitro and that the position of the disulfide bonds significantly influences binding affinity. Despite exhibiting only modest binding to Bcl-2, the successful synthesis of ScTx-Bax proteins containing two disulfide linkages represents a viable route to ScTx-based BH3 domain mimetics that preserve native-like conformations. Finally, structural models of ScTx-Bax proteins in complex with Bcl-2 indicate that these helical mimetics bind in similar configurations as wild-type Bax BH3 domains. Taken together, these results suggest that ScTx-Bax proteins may serve as potent lead compounds that expand the repertoire of "druggable" protein-protein interactions.
Collapse
Affiliation(s)
- Danushka Arachchige
- Department of Chemistry and Biochemistry, Ohio University, Biochemistry Research Facility 108, 350 W. State St., Athens, OH, 45701, USA
| | - Justin M Holub
- Department of Chemistry and Biochemistry, Ohio University, Biochemistry Research Facility 108, 350 W. State St., Athens, OH, 45701, USA.
- Molecular and Cellular Biology Program, Ohio University, Athens, OH, 45701, USA.
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
90
|
Distelhorst CW. Targeting Bcl-2-IP 3 receptor interaction to treat cancer: A novel approach inspired by nearly a century treating cancer with adrenal corticosteroid hormones. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1795-1804. [PMID: 30053503 DOI: 10.1016/j.bbamcr.2018.07.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022]
Abstract
Bcl-2 inhibits cell death by at least two different mechanisms. On the one hand, its BH3 domain binds to pro-apoptotic proteins such as Bim and prevents apoptosis induction. On the other hand, the BH4 domain of Bcl-2 binds to the inositol 1,4,5-trisphosphate receptor (IP3R), preventing Ca2+ signals that mediate cell death. In normal T-cells, Bcl-2 levels increase during the immune response, protecting against cell death, and then decline as apoptosis ensues and the immune response dissipates. But in many cancers Bcl-2 is aberrantly expressed and exploited to prevent cell death by inhibiting IP3R-mediated Ca2+ elevation. This review summarizes what is known about the mechanism of Bcl-2's control over IP3R-mediated Ca2+ release and cell death induction. Early insights into the role of Ca2+ elevation in corticosteroid-mediated cell death serves as a model for how targeting IP3R-mediated Ca2+ elevation can be a highly effective therapeutic approach for different types of cancer. Moreover, the successful development of ABT-199 (Venetoclax), a small molecule targeting the BH3 domain of Bcl-2 but without effects on Ca2+, serves as proof of principle that targeting Bcl-2 can be an effective therapeutic approach. BIRD-2, a synthetic peptide that inhibits Bcl-2-IP3R interaction, induces cell death induction in ABT-199 (Venetoclax)-resistant cancer models, attesting to the value of developing therapeutic agents that selectively target Bcl-2-IP3R interaction, inducing Ca2+-mediated cell death.
Collapse
Affiliation(s)
- Clark W Distelhorst
- Case Western University School of Medicine, Case Comprehensive Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, United States of America.
| |
Collapse
|
91
|
TRIM17 and TRIM28 antagonistically regulate the ubiquitination and anti-apoptotic activity of BCL2A1. Cell Death Differ 2018; 26:902-917. [PMID: 30042493 DOI: 10.1038/s41418-018-0169-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/28/2018] [Accepted: 07/06/2018] [Indexed: 01/20/2023] Open
Abstract
BCL2A1 is an anti-apoptotic member of the BCL-2 family that contributes to chemoresistance in a subset of tumors. BCL2A1 has a short half-life due to its constitutive processing by the ubiquitin-proteasome system. This constitutes a major tumor-suppressor mechanism regulating BCL2A1 function. However, the enzymes involved in the regulation of BCL2A1 protein stability are currently unknown. Here, we provide the first insight into the regulation of BCL2A1 ubiquitination. We present evidence that TRIM28 is an E3 ubiquitin-ligase for BCL2A1. Indeed, endogenous TRIM28 and BCL2A1 bind to each other at the mitochondria and TRIM28 knock-down decreases BCL2A1 ubiquitination. We also show that TRIM17 stabilizes BCL2A1 by blocking TRIM28 from binding and ubiquitinating BCL2A1, and that GSK3 is involved in the phosphorylation-mediated inhibition of BCL2A1 degradation. BCL2A1 and its close relative MCL1 are thus regulated by common factors but with opposite outcome. Finally, overexpression of TRIM28 or knock-out of TRIM17 reduced BCLA1 protein levels and restored sensitivity of melanoma cells to BRAF-targeted therapy. Therefore, our data describe a molecular rheostat in which two proteins of the TRIM family antagonistically regulate BCL2A1 stability and modulate cell death.
Collapse
|
92
|
Yang IH, Jung W, Kim LH, Shin JA, Cho NP, Hong SD, Hong KO, Cho SD. Nitidine chloride represses Mcl-1 protein via lysosomal degradation in oral squamous cell carcinoma. J Oral Pathol Med 2018; 47:823-829. [PMID: 29924888 DOI: 10.1111/jop.12755] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/15/2018] [Accepted: 06/18/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND We have shown previously that nitidine chloride (NC) induces apoptosis via inhibition of signal transducer and activator of transcription 3 (STAT3). However, its downstream molecules are not fully understood yet. Here, we report that NC as STAT3 inhibitor downregulates myeloid cell leukemia-1 (Mcl-1) protein in HSC-3 and HSC-4 human oral squamous cell carcinoma (OSCC) cells and a nude mouse tumor xenograft model. METHODS This study investigated the effects of NC on Mcl-1 expression in HSC-3 and HSC-4 cells using Western blotting, RT-PCR, and dual-luciferase assay. Immunohistochemistry was employed to evaluate Mcl-1 expression levels in mouse tumor tissues. Construction of Mcl-1 overexpression vector and transient transfection was done to test the apoptosis of HSC-3 cells. RESULTS Nitidine chloride did not affect either mRNA level or promoter activity of Mcl-1, and the decrease in Mcl-1 protein by NC was caused by lysosome-dependent degradation, but not proteasome-dependent degradation. The overexpression of Mcl-1 protein in OSCC cell lines was sufficient to block the induction of apoptosis. In addition, NC strongly reduced the expression level of Mcl-1 protein compared with other STAT3 inhibitors such as cryptotanshione and S3I-201 in OSCCs. CONCLUSIONS Our findings suggest that NC triggers apoptosis via lysosome-dependent Mcl-1 protein degradation and could be chosen as a promising chemotherapeutic candidate against human OSCCs.
Collapse
Affiliation(s)
- In-Hyoung Yang
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable Material, Institute of Oral Bioscience, Chonbuk National University, Jeonju, Korea
| | - Won Jung
- Department of Oral medicine, School of Dentistry, Chonbuk National University, Jeonju, Korea
| | - Lee-Han Kim
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable Material, Institute of Oral Bioscience, Chonbuk National University, Jeonju, Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Nam-Pyo Cho
- Department of Oral Pathology, School of Dentistry, Institute of Biodegradable Material, Institute of Oral Bioscience, Chonbuk National University, Jeonju, Korea
| | - Seong Doo Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Kyoung-Ok Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
93
|
Overcoming Resistance of Human Non-Hodgkin's Lymphoma to CD19-CAR CTL Therapy by Celecoxib and Histone Deacetylase Inhibitors. Cancers (Basel) 2018; 10:cancers10060200. [PMID: 29904021 PMCID: PMC6025421 DOI: 10.3390/cancers10060200] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/14/2018] [Accepted: 06/12/2018] [Indexed: 12/16/2022] Open
Abstract
Patients with B-cell non-Hodgkin’s lymphoma (B-NHL) who fail to respond to first-line treatment regimens or develop resistance, exhibit poor prognosis. This signifies the need to develop alternative treatment strategies. CD19-chimeric antigen receptor (CAR) T cell-redirected immunotherapy is an attractive and novel option, which has shown encouraging outcomes in phase I clinical trials of relapsed/refractory NHL. However, the underlying mechanisms of, and approaches to overcome, acquired anti-CD19CAR CD8+ T cells (CTL)-resistance in NHL remain elusive. CD19CAR transduced primary human CTLs kill CD19+ human NHLs in a CD19- and caspase-dependent manner, mainly via the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) apoptotic pathway. To understand the dynamics of the development of resistance, we analyzed several anti-CD19CAR CTL-resistant NHL sublines (R-NHL) derived by serial exposure of sensitive parental lines to excessive numbers of anti-CD19CAR CTLs followed by a limiting dilution analysis. The R-NHLs retained surface CD19 expression and were efficiently recognized by CD19CAR CTLs. However, R-NHLs developed cross-resistance to CD19CAR transduced human primary CTLs and the Jurkat human T cell line, activated Jurkat, and lymphokine activated killer (LAK) cells, suggesting the acquisition of resistance is independent of CD19-loss and might be due to aberrant apoptotic machinery. We hypothesize that the R-NHL refractoriness to CD19CAR CTL killing could be partially rescued by small molecule sensitizers with apoptotic-gene regulatory effects. Chromatin modifiers and Celecoxib partially reversed the resistance of R-NHL cells to the cytotoxic effects of anti-CD19CAR CTLs and rhTRAIL. These in vitro results, though they require further examination, may provide a rational biological basis for combination treatment in the management of CD19CAR CTL-based therapy of NHL.
Collapse
|
94
|
Constitutive IP 3 signaling underlies the sensitivity of B-cell cancers to the Bcl-2/IP 3 receptor disruptor BIRD-2. Cell Death Differ 2018; 26:531-547. [PMID: 29899382 PMCID: PMC6370760 DOI: 10.1038/s41418-018-0142-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 12/03/2022] Open
Abstract
Anti-apoptotic Bcl-2 proteins are upregulated in different cancers, including diffuse large B-cell lymphoma (DLBCL) and chronic lymphocytic leukemia (CLL), enabling survival by inhibiting pro-apoptotic Bcl-2-family members and inositol 1,4,5-trisphosphate (IP3) receptor (IP3R)-mediated Ca2+-signaling. A peptide tool (Bcl-2/IP3R Disruptor-2; BIRD-2) was developed to abrogate the interaction of Bcl-2 with IP3Rs by targeting Bcl-2′s BH4 domain. BIRD-2 triggers cell death in primary CLL cells and in DLBCL cell lines. Particularly, DLBCL cells with high levels of IP3R2 were sensitive to BIRD-2. Here, we report that BIRD-2-induced cell death in DLBCL cells does not only depend on high IP3R2-expression levels, but also on constitutive IP3 signaling, downstream of the tonically active B-cell receptor. The basal Ca2+ level in SU-DHL-4 DLBCL cells was significantly elevated due to the constitutive IP3 production. This constitutive IP3 signaling fulfilled a pro-survival role, since inhibition of phospholipase C (PLC) using U73122 (2.5 µM) caused cell death in SU-DHL-4 cells. Milder inhibition of IP3 signaling using a lower U73122 concentration (1 µM) or expression of an IP3 sponge suppressed both BIRD-2-induced Ca2+ elevation and apoptosis in SU-DHL-4 cells. Basal PLC/IP3 signaling also fulfilled a pro-survival role in other DLBCL cell lines, including Karpas 422, RI-1 and SU-DHL-6 cells, whereas PLC inhibition protected these cells against BIRD-2-evoked apoptosis. Finally, U73122 treatment also suppressed BIRD-2-induced cell death in primary CLL, both in unsupported systems and in co-cultures with CD40L-expressing fibroblasts. Thus, constitutive IP3 signaling in lymphoma and leukemia cells is not only important for cancer cell survival, but also represents a vulnerability, rendering cancer cells dependent on Bcl-2 to limit IP3R activity. BIRD-2 seems to switch constitutive IP3 signaling from pro-survival into pro-death, presenting a plausible therapeutic strategy.
Collapse
|
95
|
Procacci P. Myeloid Cell Leukemia 1 Inhibition: An in Silico Study Using Non-equilibrium Fast Double Annihilation Technology. J Chem Theory Comput 2018; 14:3890-3902. [DOI: 10.1021/acs.jctc.8b00305] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Piero Procacci
- Department of Chemistry, University of Florence, Via Lastruccia No. 3, Sesto Fiorentino I-50019, Italy
| |
Collapse
|
96
|
PTBP1 enhances miR-101-guided AGO2 targeting to MCL1 and promotes miR-101-induced apoptosis. Cell Death Dis 2018; 9:552. [PMID: 29748555 PMCID: PMC5945587 DOI: 10.1038/s41419-018-0551-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/16/2018] [Accepted: 03/22/2018] [Indexed: 12/22/2022]
Abstract
Myeloid cell leukemia 1 (MCL1) is a key anti-apoptotic protein belonging to the BCL-2 protein family. To preserve normal cellular homeostasis, cells must maintain strict control over MCL1 expression. Overexpression of MCL1 has been identified as a key contributor to tumorigenesis, and further enables resistance to a number of anti-cancer chemotherapies. Thus, there is an ongoing interest to develop selective MCL1 inhibitors. In order to better target MCL1, it is essential to understand the molecular mechanisms that regulate MCL1 expression in cells. While MCL1 expression is tightly controlled by multiple mechanisms, the post-transcriptional regulation of MCL1 mRNA is poorly studied. Our previous work identified that polypyrimidine tract binding protein 1 (PTBP1) binds to MCL1 mRNA and represses MCL1 expression by destabilizing MCL1 mRNA. In this report, we show that PTBP1 modulates MCL1 expression by regulating the microRNA (miRNA) direction of the miRNA-induced silencing complex (miRISC) to MCL1. We demonstrate that PTBP1 enhances miR-101-guided AGO2 interaction with MCL1, thereby regulating miR-101-induced apoptosis and clonogenic cell survival inhibition in cells. Taken together, not only do these studies expand our understanding on the regulation of MCL1, they also demonstrate that PTBP1 and miRNAs can function cooperatively on a shared target mRNA.
Collapse
|
97
|
Merino D, Whittle JR, Vaillant F, Serrano A, Gong JN, Giner G, Maragno AL, Chanrion M, Schneider E, Pal B, Li X, Dewson G, Gräsel J, Liu K, Lalaoui N, Segal D, Herold MJ, Huang DCS, Smyth GK, Geneste O, Lessene G, Visvader JE, Lindeman GJ. Synergistic action of the MCL-1 inhibitor S63845 with current therapies in preclinical models of triple-negative and HER2-amplified breast cancer. Sci Transl Med 2018; 9:9/401/eaam7049. [PMID: 28768804 DOI: 10.1126/scitranslmed.aam7049] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/13/2017] [Accepted: 06/29/2017] [Indexed: 12/15/2022]
Abstract
The development of BH3 mimetics, which antagonize prosurvival proteins of the BCL-2 family, represents a potential breakthrough in cancer therapy. Targeting the prosurvival member MCL-1 has been an area of intense interest because it is frequently deregulated in cancer. In breast cancer, MCL-1 is often amplified, and high expression predicts poor patient outcome. We tested the MCL-1 inhibitor S63845 in breast cancer cell lines and patient-derived xenografts with high expression of MCL-1. S63845 displayed synergistic activity with docetaxel in triple-negative breast cancer and with trastuzumab or lapatinib in HER2-amplified breast cancer. Using S63845-resistant cells combined with CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 (CRISPR-associated 9) technology, we identified deletion of BAK and up-regulation of prosurvival proteins as potential mechanisms that confer resistance to S63845 in breast cancer. Collectively, our findings provide a strong rationale for the clinical evaluation of MCL-1 inhibitors in breast cancer.
Collapse
Affiliation(s)
- Delphine Merino
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - James R Whittle
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - François Vaillant
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Antonin Serrano
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jia-Nan Gong
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Cancer and Haematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Goknur Giner
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Ana Leticia Maragno
- Institut de Recherches Servier Oncology R&D Unit, Croissy Sur Seine 78290, France
| | - Maïa Chanrion
- Institut de Recherches Servier Oncology R&D Unit, Croissy Sur Seine 78290, France
| | - Emilie Schneider
- Institut de Recherches Servier Oncology R&D Unit, Croissy Sur Seine 78290, France
| | - Bhupinder Pal
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Xiang Li
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Grant Dewson
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Julius Gräsel
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Kevin Liu
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Najoua Lalaoui
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Cell Signalling and Cell Death Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - David Segal
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Cancer and Haematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Marco J Herold
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Molecular Genetics of Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - David C S Huang
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Cancer and Haematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Gordon K Smyth
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,School of Mathematics and Statistics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Olivier Geneste
- Institut de Recherches Servier Oncology R&D Unit, Croissy Sur Seine 78290, France
| | - Guillaume Lessene
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia.,Chemical Biology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia.,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jane E Visvader
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Geoffrey J Lindeman
- ACRF Stem Cells and Cancer Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia. .,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria 3010, Australia.,Parkville Familial Cancer Centre, Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Parkville, Victoria 3050, Australia
| |
Collapse
|
98
|
The observed alteration in BCL2 expression following lithium treatment is influenced by the choice of normalization method. Sci Rep 2018; 8:6399. [PMID: 29686228 PMCID: PMC5913222 DOI: 10.1038/s41598-018-24546-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 03/26/2018] [Indexed: 01/07/2023] Open
Abstract
Upregulation of B-cell CLL/lymphoma (BCL)2 expression following lithium treatment is seemingly well established and has been related to the neuroprotective property of the drug. However, while demonstrated by some (but not all) studies based on low-throughput techniques (e.g. qPCR) this effect is not reflected in high-throughput studies, such as microarrays and RNAseq. This manuscript presents a systematic review of currently available reports of lithium's effect on BCL2 expression. To our surprise, we found that the majority of the literature does not support the effect of lithium on BCL2 transcript or protein levels. Moreover, among the positive reports, several used therapeutically irrelevant lithium doses while others lack statistical power. We also noticed that numerous low-throughput studies normalized the signal using genes/proteins affected by lithium, imposing possible bias. Using wet bench experiments and reanalysis of publicly available microarray data, here we show that the reference gene chosen for normalization critically impacts the outcome of qPCR analyses of lithium's effect on BCL2 expression. Our findings suggest that experimental results might be severely affected by the choice of normalizing genes, and emphasize the need to re-evaluate stability of these genes in the context of the specific experimental conditions.
Collapse
|
99
|
Shaw S, Bian Z, Zhao B, Tarr JC, Veerasamy N, Jeon KO, Belmar J, Arnold AL, Fogarty SA, Perry E, Sensintaffar JL, Camper DV, Rossanese OW, Lee T, Olejniczak ET, Fesik SW. Optimization of Potent and Selective Tricyclic Indole Diazepinone Myeloid Cell Leukemia-1 Inhibitors Using Structure-Based Design. J Med Chem 2018; 61:2410-2421. [PMID: 29323899 DOI: 10.1021/acs.jmedchem.7b01155] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Myeloid cell leukemia 1 (Mcl-1), an antiapoptotic member of the Bcl-2 family of proteins, has emerged as an attractive target for cancer therapy. Mcl-1 upregulation is often found in many human cancers and is associated with high tumor grade, poor survival, and resistance to chemotherapy. Here, we describe a series of potent and selective tricyclic indole diazepinone Mcl-1 inhibitors that were discovered and further optimized using structure-based design. These compounds exhibit picomolar binding affinity and mechanism-based cellular efficacy, including growth inhibition and caspase induction in Mcl-1-sensitive cells. Thus, they represent useful compounds to study the implication of Mcl-1 inhibition in cancer and serve as potentially useful starting points toward the discovery of anti-Mcl-1 therapeutics.
Collapse
Affiliation(s)
- Subrata Shaw
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Zhiguo Bian
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Bin Zhao
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - James C Tarr
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Nagarathanam Veerasamy
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Kyu Ok Jeon
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Johannes Belmar
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Allison L Arnold
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Stuart A Fogarty
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Evan Perry
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - John L Sensintaffar
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - DeMarco V Camper
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Olivia W Rossanese
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Taekyu Lee
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Edward T Olejniczak
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| | - Stephen W Fesik
- Department of Biochemistry , Vanderbilt University School of Medicine , 2215 Garland Avenue, 607 Light Hall , Nashville , Tennessee 37232-0146 , United States
| |
Collapse
|
100
|
Kulsoom B, Shamsi TS, Afsar NA, Memon Z, Ahmed N, Hasnain SN. Bax, Bcl-2, and Bax/Bcl-2 as prognostic markers in acute myeloid leukemia: are we ready for Bcl-2-directed therapy? Cancer Manag Res 2018. [PMID: 29535553 PMCID: PMC5841349 DOI: 10.2147/cmar.s154608] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Purpose Many anticancer drugs induce apoptosis in malignant cells, and resistance to apoptosis could lead to suboptimal or no therapeutic benefit. Two cytoplasmic proteins, B-cell lymphoma protein 2 (Bcl-2)-associated X (Bax) and Bcl-2, act as a promoter and an inhibitor of apoptosis, respectively. Both Bax and Bcl-2 as well as their ratio have been regarded as prognostic markers in various cancers. However, conflicting results have been reported. A clear understanding of apoptosis has also become crucial due to reports about anti-Bcl-2 chemotherapy. We explored the relationship of Bax and Bcl-2 gene expression and their ratio with the therapeutic response in acute myeloid leukemia (AML) patients. Patients and methods Bone marrow and/or blood samples from 90 AML patients treated with cytarabine and daunorubicin were included. Expression of Bax and Bcl-2 was determined through real-time polymerase chain reaction by using ΔΔCt method of relative expression. Results Bax and Bcl-2 expression among marrow and blood samples correlated with each other (rs=0.5, p<0.01). Although bone marrow expression of Bax and Bcl-2 tended to remain higher among responders (median 1.01 and 0.29, respectively) as compared to non-responders (median 0.66 and 0.24, respectively), the difference failed to reach statistical significance (U=784.5 and 733; p=0.68 and 0.28, respectively). Conversely, Bax/Bcl-2 ratio was higher among poor responders (median 3.07 vs 1.78), though again failed to reach statistical significance (U=698.5, p=0.07). Conclusion Expression of Bax and Bcl-2 does not differ significantly among AML patients treated with cytarabine and daunorubicin in terms of remission, relapse, resistance, overall survival, and disease-free survival, thus questioning the utility of emerging anti-Bcl-2 therapy.
Collapse
Affiliation(s)
- Bibi Kulsoom
- National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Tahir Sultan Shamsi
- National Institute of Blood Diseases and Bone Marrow Transplantation, Karachi, Pakistan
| | - Nasir Ali Afsar
- Jinnah Medical and Dental College, Karachi, Pakistan.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | | | | | |
Collapse
|