51
|
Liu J, Liu W, Lu Y, Tian H, Duan C, Lu L, Gao G, Wu X, Wang X, Yang H. Piperlongumine restores the balance of autophagy and apoptosis by increasing BCL2 phosphorylation in rotenone-induced Parkinson disease models. Autophagy 2018; 14:845-861. [PMID: 29433359 PMCID: PMC6070010 DOI: 10.1080/15548627.2017.1390636] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 08/08/2017] [Accepted: 10/06/2017] [Indexed: 01/25/2023] Open
Abstract
Parkinson disease (PD) is the second most common neurodegenerative disorder after Alzheimer disease and is caused by genetics, environmental factors and aging, with few treatments currently available. Apoptosis and macroautophagy/autophagy play critical roles in PD pathogenesis; as such, modulating their balance is a potential treatment strategy. BCL2 (B cell leukemia/lymphoma 2) is a key molecule regulating this balance. Piperlongumine (PLG) is an alkaloid extracted from Piper longum L. that has antiinflammatory and anticancer effects. The present study investigated the protective effects of PLG in rotenone-induced PD cell and mouse models. We found that PLG administration (2 and 4 mg/kg) for 4 wk attenuated motor deficits in mice and prevented the loss of dopaminergic neurons in the substantia nigra induced by oral administration of rotenone (10 mg/kg) for 6 wk. PLG improved cell viability and enhanced mitochondrial function in primary neurons and SK-N-SH cells. These protective effects were exerted via inhibition of apoptosis and induction of autophagy through enhancement of BCL2 phosphorylation at Ser70. These results demonstrate that PLG exerts therapeutic effects in a rotenone-induced PD models by restoring the balance between apoptosis and autophagy. ABBREVIATIONS 6-OHDA, 6-hydroxydopamine; ACTB, actin, beta; BafA1, bafilomycin A1; BAK1, BCL2-antagonist/killer 1; BAX, BCL2-associated X protein; BCL2, B cell leukemia/lymphoma2; BECN1, Beclin 1, autophagy related; CoQ10, coenzyme Q10; COX4I1/COX IV, cytochrome c oxidase subunit 4I1; CsA, cyclosporine A; ED50, 50% effective dose; FITC, fluorescein isothiocyanate; GFP, green fluorescent protein; HPLC, high-performance liquid chromatography; JC-1, tetraethylbenz-imidazolylcarbocyanine iodide; LC3, microtubule-associated protein 1 light chain3; LC-MS/MS, liquid chromatography-tandem mass spectrometry; LDH, lactate dehydrogenase; l-dopa, 3, 4-dihydroxyphenyl-l-alanine; MAPK8/JNK1, mitogen-activated protein kinase 8; MMP, mitochondrial membrane potential; mPTP, mitochondrial permeability transition pore; mRFP, monomeric red fluorescent protein; MPTP, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; NFE2L2/NRF2, nuclear factor, erythroid derived 2, like 2; PD, Parkinson disease; PLG, piperlongumine; pNA, p-nitroanilide; PI, propidium iodide; PtdIns3K, phosphatidylinositol 3-kinase; PtdIns3P, phosphatidylinositol-3-phosphate; PTX, paclitaxel; Rap, rapamycin; SQSTM1/p62, sequestosome 1; TH, tyrosine hydroxylase; TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling; WIPI2, WD repeat domain, phosphoinositide interacting 2; ZFYVE1/DFCP1, zinc finger, FYVE domain containing 1.
Collapse
Affiliation(s)
- Jia Liu
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| | - Weijin Liu
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| | - Yongquan Lu
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| | - Hao Tian
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| | - Chunli Duan
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| | - Lingling Lu
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| | - Ge Gao
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| | - Xia Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiaomin Wang
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| | - Hui Yang
- Center of Parkinson Disease Beijing Institute for Brain Disorders, Beijing Key Laboratory on Parkinson Disease, Key Laboratory for Neurodegenerative Disease of the Ministry of Education, Beijing Center of Neural Regeneration and Repair, Department of Neurobiology, Capital Medical University, Beijing, China
| |
Collapse
|
52
|
Bodo J, Zhao X, Durkin L, Souers AJ, Phillips DC, Smith MR, Hsi ED. Acquired resistance to venetoclax (ABT-199) in t(14;18) positive lymphoma cells. Oncotarget 2018; 7:70000-70010. [PMID: 27661108 PMCID: PMC5342530 DOI: 10.18632/oncotarget.12132] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/10/2016] [Indexed: 12/03/2022] Open
Abstract
The chromosomal translocation t(14;18) in follicular lymphoma (FL) is a primary oncogenic event resulting in BCL-2 over-expression. This study investigates activity of the BH3 mimetic venetoclax (ABT-199), which targets BCL-2, and mechanisms of acquired resistance in FL. The sensitivity of FL cells to venetoclax treatment correlated with BCL-2/BIM ratio. Cells with similar expression of anti-apoptotic proteins, but with higher levels of BIM were more sensitive to the treatment. Venetoclax induced dissociation of BCL-2/BIM complex and a decrease in mitochondrial potential. Interestingly the population of cells that survived venetoclax treatment showed increased p-ERK1/2 and p-BIM (S69), as well as a decrease in total BIM levels. Venetoclax resistant cells initially showed elevated levels of p-AKT and p-Foxo1/3a, a dissociation of BIM/BCL-2/BECLIN1 complex, and a decrease in SQSTM1/p62 level (indicating increased autophagy) together with a slight decline in BIM expression. After stable resistant cell lines were established, a significant reduction of BCL-2 levels and almost total absence of BIM was observed. The acquisition of these resistance phenotypes could be prevented via selective ERK/AKT inhibition or anti-CD20 antibody treatment, thus highlighting possible combination therapies for FL patients.
Collapse
Affiliation(s)
- Juraj Bodo
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Xiaoxian Zhao
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lisa Durkin
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | - Mitchell R Smith
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric D Hsi
- Department of Laboratory Medicine, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
53
|
Huang S, Tang R, Poon RYC. BCL-W is a regulator of microtubule inhibitor-induced mitotic cell death. Oncotarget 2018; 7:38718-38730. [PMID: 27231850 PMCID: PMC5122423 DOI: 10.18632/oncotarget.9586] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/28/2016] [Indexed: 02/06/2023] Open
Abstract
Microtubule inhibitors including taxanes and vinca alkaloids are among the most widely used anticancer agents. Disrupting the microtubules activates the spindle-assembly checkpoint and traps cells in mitosis. Whether cells subsequently undergo mitotic cell death is an important factor for the effectiveness of the anticancer agents. Given that apoptosis accounts for the majority of mitotic cell death induced by microtubule inhibitors, we performed a systematic study to determine which members of the anti-apoptotic BCL-2 family are involved in determining the duration of mitotic block before cell death or slippage. Depletion of several anti-apoptotic BCL-2-like proteins significantly shortened the time before apoptosis. Among these proteins, BCL-W has not been previously characterized to play a role in mitotic cell death. Although the expression of BCL-W remained constant during mitotic block, it varied significantly between different cell lines. Knockdown of BCL-W with siRNA or disruption of the BCL-W gene with CRISPR-Cas9 speeded up mitotic cell death. Conversely, overexpression of BCL-W delayed mitotic cell death, extending the mitotic block to allow mitotic slippage. Taken together, these results showed that BCL-W contributes to the threshold of anti-apoptotic activity during mitosis.
Collapse
Affiliation(s)
- Shan Huang
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Rui Tang
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| | - Randy Y C Poon
- Division of Life Science, Center for Cancer Research, and State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong
| |
Collapse
|
54
|
Deng J, Park D, Wang M, Nooka A, Deng Q, Matulis S, Kaufman J, Lonial S, Boise LH, Galipeau J, Deng X. BCL2-BH4 antagonist BDA-366 suppresses human myeloma growth. Oncotarget 2017; 7:27753-63. [PMID: 27049723 PMCID: PMC5053685 DOI: 10.18632/oncotarget.8513] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 03/28/2016] [Indexed: 11/25/2022] Open
Abstract
Multiple myeloma (MM) is a heterogeneous plasma cell malignancy and remains incurable. B-cell lymphoma-2 (BCL2) protein correlates with the survival and the drug resistance of myeloma cells. BH3 mimetics have been developed to disrupt the binding between BCL2 and its pro-apoptotic BCL2 family partners for the treatment of MM, but with limited therapeutic efficacy. We recently identified a small molecule BDA-366 as a BCL2 BH4 domain antagonist, converting it from an anti-apoptotic into a pro-apoptotic molecule. In this study, we demonstrated that BDA-366 induces robust apoptosis in MM cell lines and primary MM cells by inducing BCL2 conformational change. Delivery of BDA-366 substantially suppressed the growth of human MM xenografts in NOD-scid/IL2Rγnull mice, without significant cytotoxic effects on normal hematopoietic cells or body weight. Thus, BDA-366 functions as a novel BH4-based BCL2 inhibitor and offers an entirely new tool for MM therapy.
Collapse
Affiliation(s)
- Jiusheng Deng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Dongkyoo Park
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Mengchang Wang
- The First Affiliated Hospital, Xi'An Jiaotong University, Xi'An, China
| | - Ajay Nooka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Qiaoya Deng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Shannon Matulis
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Jonathan Kaufman
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Lawrence H Boise
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Jacques Galipeau
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| | - Xingming Deng
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, USA
| |
Collapse
|
55
|
Apoptosis signaling and BCL-2 pathways provide opportunities for novel targeted therapeutic strategies in hematologic malignances. Blood Rev 2017; 32:8-28. [PMID: 28802908 DOI: 10.1016/j.blre.2017.08.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 08/05/2017] [Accepted: 08/06/2017] [Indexed: 12/14/2022]
Abstract
Apoptosis is an essential biological process involved in tissue homeostasis and immunity. Aberrations of the two main apoptotic pathways, extrinsic and intrinsic, have been identified in hematological malignancies; many of these aberrations are associated with pathogenesis, prognosis and resistance to standard chemotherapeutic agents. Targeting components of the apoptotic pathways, especially the chief regulatory BCL-2 family in the intrinsic pathway, has proved to be a promising therapeutic approach for patients with hematological malignances, with the expectation of enhanced efficacy and reduced adverse events. Continuous investigations regarding the biological importance of each of the BCL-2 family components and the clinical rationale to achieve optimal therapeutic outcomes, using either monotherapy or in combination with other targeted agents, have generated inspiring progress in the field. Genomic, epigenomic and biological analyses including BH3 profiling facilitate effective evaluation of treatment response, cancer recurrence and drug resistance. In this review, we summarize the biological features of each of the components in the BCL-2 apoptotic pathways, analyze the regulatory mechanisms and the pivotal roles of BCL-2 family members in the pathogenesis of major types of hematologic malignances, and evaluate the potential of apoptosis- and BCL-2-targeted strategies as effective approaches in anti-cancer therapies.
Collapse
|
56
|
WeiΔ LM, Hugle M, Fulda S. Eribulin alone or in combination with the PLK1 inhibitor BI 6727 triggers intrinsic apoptosis in Ewing sarcoma cell lines. Oncotarget 2017; 8:52445-52456. [PMID: 28881742 PMCID: PMC5581041 DOI: 10.18632/oncotarget.17190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/02/2017] [Indexed: 11/25/2022] Open
Abstract
In this study, we investigated the molecular mechanisms of eribulin-induced cell death and its therapeutic potential in combination with the PLK1 inhibitor BI 6727 in Ewing sarcoma (ES). Here, we show that eribulin triggers cell death in a dose-dependent manner in a panel of ES cell lines. In addition, eribulin at subtoxic, low nanomolar concentrations acts in concert with BI 6727 to induce cell death and to suppress long-term clonogenic survival. Mechanistic studies reveal that eribulin monotherapy at cytotoxic concentrations and co-treatment with eribulin at subtoxic concentrations together with BI 6727 arrest cells in the M phase of the cell cycle prior to the onset of cell death. This mitotic arrest is followed by increased phosphorylation of BCL-2 and BCL-xL as well as downregulation of MCL-1, suggesting inactivation of these antiapoptotic BCL-2 family proteins. Consistently, eribulin monotherapy and eribulin/BI 6727 co-treatment trigger activation of BAX, a key proapoptotic BCL-2 family protein, and increase proteolytic activation of caspase-9 and -3. Importantly, overexpression of BCL-2 or addition of the broad-range caspase inhibitor zVAD.fmk significantly rescue eribulin- as well as eribulin/BI 6727-induced cell death. Together, these findings demonstrate that eribulin induces cell death via the intrinsic pathway of apoptosis in ES cells, both alone at cytotoxic concentrations and in combination with BI 6727 at subtoxic concentrations. Thus, our study highlights the therapeutic potential of eribulin for the treatment of ES alone or in rational combination therapies.
Collapse
Affiliation(s)
- Lilly Magdalena WeiΔ
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manuela Hugle
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Frankfurt, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
57
|
Jana T, Ghosh A, Das Mandal S, Banerjee R, Saha S. PPIMpred: a web server for high-throughput screening of small molecules targeting protein-protein interaction. ROYAL SOCIETY OPEN SCIENCE 2017; 4:160501. [PMID: 28484602 PMCID: PMC5414239 DOI: 10.1098/rsos.160501] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 03/20/2017] [Indexed: 05/31/2023]
Abstract
PPIMpred is a web server that allows high-throughput screening of small molecules for targeting specific protein-protein interactions, namely Mdm2/P53, Bcl2/Bak and c-Myc/Max. Three different kernels of support vector machine (SVM), namely, linear, polynomial and radial basis function (RBF), and two other machine learning techniques including Naive Bayes and Random Forest were used to train the models. A fivefold cross-validation technique was used to measure the performance of these classifiers. The RBF kernel of SVM outperformed and/or was comparable with all other methods with accuracy values of 83%, 79% and 90% for Mdm2/P53, Bcl2/Bak and c-Myc/Max, respectively. About 80% of the predicted SVM scores of training/testing datasets from Mdm2/P53 and Bcl2/Bak have significant IC50 values and docking scores. The proposed models achieved an accuracy of 66-90% with blind sets. The three mentioned (Mdm2/P53, Bcl2/Bak and c-Myc/Max) proposed models were screened in a large dataset of 265 242 small chemicals from National Cancer Institute open database. To further realize the robustness of this approach, hits with high and random SVM scores were used for molecular docking in AutoDock Vina wherein the molecules with high and random predicted SVM scores yielded moderately significant docking scores (p-values < 0.1). In addition to the above-mentioned classification scheme, this web server also allows users to get the structural and chemical similarities with known chemical modulators or drug-like molecules based on Tanimoto coefficient similarity search algorithm. PPIMpred is freely available at http://bicresources.jcbose.ac.in/ssaha4/PPIMpred/.
Collapse
Affiliation(s)
- Tanmoy Jana
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
| | - Abhirupa Ghosh
- Department of Bioinformatics, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
| | - Sukhen Das Mandal
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
| | - Raja Banerjee
- Department of Bioinformatics, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, India
| | - Sudipto Saha
- Bioinformatics Centre, Bose Institute, P 1/12, C.I.T. Road, Scheme-VII (M), Kolkata, West Bengal, India
| |
Collapse
|
58
|
Zheng R, Studzinski GP. Optimal AraC-Cytotoxicity to AML Cells Requires ERK5 Activity. J Cell Biochem 2017; 118:1583-1589. [DOI: 10.1002/jcb.25820] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 02/05/2023]
Affiliation(s)
- Ruifang Zheng
- Department of Pathology and Laboratory Medicine, New Jersey Medical School; Rutgers University; 185 South Orange Ave. Newark New Jersey 07103
| | - George P. Studzinski
- Department of Pathology and Laboratory Medicine, New Jersey Medical School; Rutgers University; 185 South Orange Ave. Newark New Jersey 07103
| |
Collapse
|
59
|
Abstract
The BCL2-selective BH3 mimetic venetoclax was recently approved for the treatment of relapsed, chromosome 17p-deleted chronic lymphocytic leukemia (CLL) and is undergoing extensive testing, alone and in combination, in lymphomas, acute leukemias, and solid tumors. Here we summarize recent advances in understanding of the biology of BCL2 family members that shed light on the action of BH3 mimetics, review preclinical and clinical studies leading to the regulatory approval of venetoclax, and discuss future investigation of this new class of antineoplastic agent.
Collapse
Affiliation(s)
- Haiming Dai
- Division of Oncology Research , Mayo Clinic, Rochester, MN, 55905, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.,Center for Medical Physics and Technology, Hefei Institute of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - X Wei Meng
- Division of Oncology Research , Mayo Clinic, Rochester, MN, 55905, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Scott H Kaufmann
- Division of Oncology Research , Mayo Clinic, Rochester, MN, 55905, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
60
|
Meister MT, Boedicker C, Graab U, Hugle M, Hahn H, Klingebiel T, Fulda S. Arsenic trioxide induces Noxa-dependent apoptosis in rhabdomyosarcoma cells and synergizes with antimicrotubule drugs. Cancer Lett 2016; 381:287-95. [PMID: 27521572 DOI: 10.1016/j.canlet.2016.07.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/06/2016] [Accepted: 07/11/2016] [Indexed: 12/23/2022]
Abstract
The prognosis of metastatic or relapsed rhabdomyosarcoma (RMS) is poor, highlighting the need of new treatment options. In the present study, we evaluated the in vitro efficacy of arsenic trioxide (ATO) in RMS, a FDA-approved drug used in pediatric leukemia. Here, we report that ATO exerts antitumor activity against RMS cells both as single agent and in combination with microtubule-targeting drugs. Monotherapy with ATO reduces cell viability, triggers apoptosis and suppresses clonogenic survival of RMS cells, at least in part, by transcriptional induction of the proapoptotic BH3-only protein Noxa. siRNA-mediated knockdown of Noxa significantly rescues ATO-mediated cell death, demonstrating that Noxa is required for cell death. Also, ATO suppresses endogenous Hedgehog (Hh) signaling, as it significantly reduces Gli1 transcriptional activity and expression levels of several Hh target genes. Furthermore, we identify synergistic induction of apoptosis by ATO together with several antimicrotubule agents including vincristine (VCR), vinblastine and eribulin. The addition of the broad-range caspase inhibitor zVAD.fmk or overexpression of the antiapoptotic protein Bcl-2 significantly reduce ATO/VCR-induced cell death, indicating that the ATO/VCR combination triggers caspase-dependent apoptosis via the mitochondrial pathway. In summary, ATO exerts antitumor activity against RMS, especially in combination with antimicrotubule drugs. These findings have important implications for the development of novel therapeutic strategies for RMS.
Collapse
Affiliation(s)
- Michael Torsten Meister
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Cathinka Boedicker
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrike Graab
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany
| | - Manuela Hugle
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany
| | - Heidi Hahn
- Department of Human Genetics, University Medical Center Goettingen, Goettingen, Germany
| | - Thomas Klingebiel
- German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Division of Pediatric Hematology and Oncology, Hospital for Children and Adolescents, Johann Wolfgang Goethe-University, Frankfurt, Germany
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
61
|
The broken "Off" switch in cancer signaling: PP2A as a regulator of tumorigenesis, drug resistance, and immune surveillance. BBA CLINICAL 2016; 6:87-99. [PMID: 27556014 PMCID: PMC4986044 DOI: 10.1016/j.bbacli.2016.08.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 08/01/2016] [Accepted: 08/02/2016] [Indexed: 12/31/2022]
Abstract
Aberrant activation of signal transduction pathways can transform a normal cell to a malignant one and can impart survival properties that render cancer cells resistant to therapy. A diverse set of cascades have been implicated in various cancers including those mediated by serine/threonine kinases such RAS, PI3K/AKT, and PKC. Signal transduction is a dynamic process involving both "On" and "Off" switches. Activating mutations of RAS or PI3K can be viewed as the switch being stuck in the "On" position resulting in continued signaling by a survival and/or proliferation pathway. On the other hand, inactivation of protein phosphatases such as the PP2A family can be seen as the defective "Off" switch that similarly can activate these pathways. A problem for therapeutic targeting of PP2A is that the enzyme is a hetero-trimer and thus drug targeting involves complex structures. More importantly, since PP2A isoforms generally act as tumor suppressors one would want to activate these enzymes rather than suppress them. The elucidation of the role of cellular inhibitors like SET and CIP2A in cancer suggests that targeting these proteins can have therapeutic efficacy by mechanisms involving PP2A activation. Furthermore, drugs such as FTY-720 can activate PP2A isoforms directly. This review will cover the current state of knowledge of PP2A role as a tumor suppressor in cancer cells and as a mediator of processes that can impact drug resistance and immune surveillance.
Collapse
|
62
|
Zitterbart R, Seitz O. Parallele chemische Proteinsynthese auf der Oberfläche zur schnellen Analyse der Phosphoregulierung von SH3-Domänen. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601843] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Robert Zitterbart
- Institut für Chemie; Humboldt-Universität zu Berlin; Brook-Taylor-Straße 2 12489 Berlin Deutschland
| | - Oliver Seitz
- Institut für Chemie; Humboldt-Universität zu Berlin; Brook-Taylor-Straße 2 12489 Berlin Deutschland
| |
Collapse
|
63
|
Zitterbart R, Seitz O. Parallel Chemical Protein Synthesis on a Surface Enables the Rapid Analysis of the Phosphoregulation of SH3 Domains. Angew Chem Int Ed Engl 2016; 55:7252-6. [DOI: 10.1002/anie.201601843] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Robert Zitterbart
- Institut für Chemie; Humboldt-Universität zu Berlin; Brook-Taylor-Strasse 2 12489 Berlin Germany
| | - Oliver Seitz
- Institut für Chemie; Humboldt-Universität zu Berlin; Brook-Taylor-Strasse 2 12489 Berlin Germany
| |
Collapse
|
64
|
Dai H, Ding H, Meng XW, Peterson KL, Schneider PA, Karp JE, Kaufmann SH. Constitutive BAK activation as a determinant of drug sensitivity in malignant lymphohematopoietic cells. Genes Dev 2016; 29:2140-52. [PMID: 26494789 PMCID: PMC4617978 DOI: 10.1101/gad.267997.115] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mitochondrial outer membrane permeabilization (MOMP), a key step in the intrinsic apoptotic pathway, is incompletely understood. Current models emphasize the role of BH3-only BCL2 family members in BAX and BAK activation. Here we demonstrate concentration-dependent BAK autoactivation under cell-free conditions and provide evidence that this autoactivation plays a key role in regulating the intrinsic apoptotic pathway in intact cells. In particular, we show that up to 80% of BAK (but not BAX) in lymphohematopoietic cell lines is oligomerized and bound to anti-apoptotic BCL2 family members in the absence of exogenous death stimuli. The extent of this constitutive BAK oligomerization is diminished by BAK knockdown and unaffected by BIM or PUMA down-regulation. Further analysis indicates that sensitivity of cells to BH3 mimetics reflects the identity of the anti-apoptotic proteins to which BAK is constitutively bound, with extensive BCLXL•BAK complexes predicting navitoclax sensitivity, and extensive MCL1•BAK complexes predicting A1210477 sensitivity. Moreover, high BAK expression correlates with sensitivity of clinical acute myelogenous leukemia to chemotherapy, whereas low BAK levels correlate with resistance and relapse. Collectively, these results inform current understanding of MOMP and provide new insight into the ability of BH3 mimetics to induce apoptosis without directly activating BAX or BAK.
Collapse
Affiliation(s)
- Haiming Dai
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55905, USA; Center for Medical Physics and Technology, Hefei Institutes of Physical Science, Chinese Academy of Science, Hefei 230031, China
| | - Husheng Ding
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - X Wei Meng
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Kevin L Peterson
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Paula A Schneider
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Judith E Karp
- Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland 21287, USA
| | - Scott H Kaufmann
- Division of Oncology Research, Mayo Clinic, Rochester, Minnesota 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
65
|
Abstract
Apoptosis is a regulated form of cell death that proceeds by defined biochemical pathways. Most apoptosis is controlled by interactions between pro-survival and pro-apoptotic Bcl-2 family proteins in which death is often the consequence of permeabilization of the mitochondrial outer membrane. Many drugs affect this equilibrium to favor apoptosis but this process is not completely understood. We show that the chemotherapeutic drug cisplatin initiates an apoptotic pathway by phosphorylation of a pro-survival Bcl-2 family member, Bcl-xL, by cyclin-dependent kinase 2. The phosphorylation occurred at a previously unreported site and its biologic significance was demonstrated by a phosphomimetic modification of Bcl-xL that was able to induce apoptosis without addition of cisplatin. The mechanism of cell death induction was similar to that initiated by pro-apoptotic Bcl-2 family proteins, that is, phosphorylated Bcl-xL translocated to the mitochondrial membrane, and formed pores in the membrane. This initiated cytochrome c release and caspase activation that resulted in cell death.
Collapse
|
66
|
Zhang L, Wang K, Lei Y, Li Q, Nice EC, Huang C. Redox signaling: Potential arbitrator of autophagy and apoptosis in therapeutic response. Free Radic Biol Med 2015; 89:452-65. [PMID: 26454086 DOI: 10.1016/j.freeradbiomed.2015.08.030] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 08/27/2015] [Accepted: 08/31/2015] [Indexed: 02/05/2023]
Abstract
Redox signaling plays important roles in the regulation of cell death and survival in response to cancer therapy. Autophagy and apoptosis are discrete cellular processes mediated by distinct groups of regulatory and executioner molecules, and both are thought to be cellular responses to various stress conditions including oxidative stress, therefore controlling cell fate. Basic levels of reactive oxygen species (ROS) may function as signals to promote cell proliferation and survival, whereas increase of ROS can induce autophagy and apoptosis by damaging cellular components. Growing evidence in recent years argues for ROS that below detrimental levels acting as intracellular signal transducers that regulate autophagy and apoptosis. ROS-regulated autophagy and apoptosis can cross-talk with each other. However, how redox signaling determines different cell fates by regulating autophagy and apoptosis remains unclear. In this review, we will focus on understanding the delicate molecular mechanism by which autophagy and apoptosis are finely orchestrated by redox signaling and discuss how this understanding can be used to develop strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Lu Zhang
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, P.R. China; Department of Neurology, the Affiliated Hospital of Hainan Medical College, Haikou, 570102, P.R. China
| | - Kui Wang
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, P.R. China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, P.R. China
| | - Qifu Li
- Department of Neurology, the Affiliated Hospital of Hainan Medical College, Haikou, 570102, P.R. China
| | - Edouard Collins Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Canhua Huang
- State Key Laboratory for Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, P.R. China.
| |
Collapse
|
67
|
Adem J, Hämäläinen A, Ropponen A, Eeva J, Eray M, Nuutinen U, Pelkonen J. ERK1/2 has an essential role in B cell receptor- and CD40-induced signaling in an in vitro model of germinal center B cell selection. Mol Immunol 2015; 67:240-7. [DOI: 10.1016/j.molimm.2015.05.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 02/02/2023]
|
68
|
Abstract
Reversible protein phosphorylation is critically important in biology and medicine. Hundreds of thousands of sites of protein phosphorylation have been discovered but our understanding of the functions of the vast majority of these post-translational modifications is lacking. This review describes several chemical and biochemical methods that are under development and in current use to install phospho-amino acids and their mimics site-specifically into proteins. The relative merits of total chemical synthesis, semisynthesis, and nonsense suppression strategies for studying protein phosphorylation are discussed in terms of technical simplicity, scope, and versatility.
Collapse
Affiliation(s)
- Zan Chen
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States
| | - Philip A Cole
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
69
|
Haschka MD, Soratroi C, Kirschnek S, Häcker G, Hilbe R, Geley S, Villunger A, Fava LL. The NOXA-MCL1-BIM axis defines lifespan on extended mitotic arrest. Nat Commun 2015; 6:6891. [PMID: 25922916 PMCID: PMC4423218 DOI: 10.1038/ncomms7891] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 03/09/2015] [Indexed: 11/09/2022] Open
Abstract
Cell death on extended mitotic arrest is considered arguably most critical for the efficacy of microtubule-targeting agents (MTAs) in anticancer therapy. While the molecular machinery controlling mitotic arrest on MTA treatment, the spindle assembly checkpoint (SAC), appears well defined, the molecular components executing cell death, as well as factors connecting both networks remain poorly understood. Here we conduct a mini screen exploring systematically the contribution of individual BCL2 family proteins at single cell resolution to death on extended mitotic arrest, and demonstrate that the mitotic phosphorylation of BCL2 and BCLX represent a priming event for apoptosis that is ultimately triggered by NOXA-dependent MCL1 degradation, enabling BIM-dependent cell death. Our findings provide a comprehensive model for the initiation of apoptosis in cells stalled in mitosis and provide a molecular basis for the increased efficacy of combinatorial treatment of cancer cells using MTAs and BH3 mimetics. Cells experiencing extended mitotic arrest often undergo cell death as a result of steadily declining levels of the apoptotic inhibitor MCL1, but the mechanism controlling this process is poorly understood. Here, Haschka et al. show that the BH3-only protein NOXA promotes the degradation of MCL1, enabling BIM-dependent cell death.
Collapse
Affiliation(s)
- Manuel D Haschka
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Claudia Soratroi
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Susanne Kirschnek
- Institute for Medical Microbiology and Hygiene, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Georg Häcker
- Institute for Medical Microbiology and Hygiene, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Richard Hilbe
- Division of Molecular Pathophysiology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Stephan Geley
- Division of Molecular Pathophysiology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - Luca L Fava
- Division of Developmental Immunology, Biocenter, Medical University Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| |
Collapse
|
70
|
Correia C, Lee SH, Meng XW, Vincelette ND, Knorr KLB, Ding H, Nowakowski GS, Dai H, Kaufmann SH. Emerging understanding of Bcl-2 biology: Implications for neoplastic progression and treatment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1658-71. [PMID: 25827952 DOI: 10.1016/j.bbamcr.2015.03.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/20/2015] [Accepted: 03/22/2015] [Indexed: 02/07/2023]
Abstract
Bcl-2, the founding member of a family of apoptotic regulators, was initially identified as the protein product of a gene that is translocated and overexpressed in greater than 85% of follicular lymphomas (FLs). Thirty years later we now understand that anti-apoptotic Bcl-2 family members modulate the intrinsic apoptotic pathway by binding and neutralizing the mitochondrial permeabilizers Bax and Bak as well as a variety of pro-apoptotic proteins, including the cellular stress sensors Bim, Bid, Puma, Bad, Bmf and Noxa. Despite extensive investigation of all of these proteins, important questions remain. For example, how Bax and Bak breach the outer mitochondrial membrane remains poorly understood. Likewise, how the functions of anti-apoptotic Bcl-2 family members such as eponymous Bcl-2 are affected by phosphorylation or cancer-associated mutations has been incompletely defined. Finally, whether Bcl-2 family members can be successfully targeted for therapeutic advantage is only now being investigated in the clinic. Here we review recent advances in understanding Bcl-2 family biology and biochemistry that begin to address these questions.
Collapse
Affiliation(s)
- Cristina Correia
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Sun-Hee Lee
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - X Wei Meng
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicole D Vincelette
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Katherine L B Knorr
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Husheng Ding
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Grzegorz S Nowakowski
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Haiming Dai
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| | - Scott H Kaufmann
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
71
|
Dick TE, Hengst JA, Fox TE, Colledge AL, Kale VP, Sung SS, Sharma A, Amin S, Loughran TP, Kester M, Wang HG, Yun JK. The apoptotic mechanism of action of the sphingosine kinase 1 selective inhibitor SKI-178 in human acute myeloid leukemia cell lines. J Pharmacol Exp Ther 2015; 352:494-508. [PMID: 25563902 PMCID: PMC4352591 DOI: 10.1124/jpet.114.219659] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/05/2015] [Indexed: 12/20/2022] Open
Abstract
We previously developed SKI-178 (N'-[(1E)-1-(3,4-dimethoxyphenyl)ethylidene]-3-(4-methoxxyphenyl)-1H-pyrazole-5-carbohydrazide) as a novel sphingosine kinase-1 (SphK1) selective inhibitor and, herein, sought to determine the mechanism-of-action of SKI-178-induced cell death. Using human acute myeloid leukemia (AML) cell lines as a model, we present evidence that SKI-178 induces prolonged mitosis followed by apoptotic cell death through the intrinsic apoptotic cascade. Further examination of the mechanism of action of SKI-178 implicated c-Jun NH2-terminal kinase (JNK) and cyclin-dependent protein kinase 1 (CDK1) as critical factors required for SKI-178-induced apoptosis. In cell cycle synchronized human AML cell lines, we demonstrate that entry into mitosis is required for apoptotic induction by SKI-178 and that CDK1, not JNK, is required for SKI-178-induced apoptosis. We further demonstrate that the sustained activation of CDK1 during prolonged mitosis, mediated by SKI-178, leads to the simultaneous phosphorylation of the prosurvival Bcl-2 family members, Bcl-2 and Bcl-xl, as well as the phosphorylation and subsequent degradation of Mcl-1. Moreover, multidrug resistance mediated by multidrug-resistant protein1 and/or prosurvival Bcl-2 family member overexpression did not affect the sensitivity of AML cells to SKI-178. Taken together, these findings highlight the therapeutic potential of SKI-178 targeting SphK1 as a novel therapeutic agent for the treatment of AML, including multidrug-resistant/recurrent AML subtypes.
Collapse
Affiliation(s)
- Taryn E Dick
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Jeremy A Hengst
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Todd E Fox
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Ashley L Colledge
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Vijay P Kale
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Shen-Shu Sung
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Arun Sharma
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Shantu Amin
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Thomas P Loughran
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Mark Kester
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Hong-Gang Wang
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| | - Jong K Yun
- Department of Pharmacology (T.E.D., J.A.H., A.L.C., V.P.K., S.-S.S., A.S., S.A., H.-G.W., J.K.Y.) and The Jake Gittlen Laboratories for Cancer Research (T.E.D., J.A.H., A.L.C., V.P.K., J.K.Y.), The Pennsylvania State University College of Medicine, Hershey, Pennsylvania; and Department of Pharmacology (T.E.F., M.K.), and University of Virginia Cancer Center (T.P.L.), University of Virginia, Charlottesville, Virginia
| |
Collapse
|
72
|
Hugle M, Fulda S. Dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235 synergizes with chloroquine to induce apoptosis in embryonal rhabdomyosarcoma. Cancer Lett 2015; 360:1-9. [PMID: 25637161 DOI: 10.1016/j.canlet.2014.12.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/05/2014] [Accepted: 12/05/2014] [Indexed: 11/28/2022]
Abstract
Aberrant activation of the phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathway has been reported for rhabdomyosarcoma (RMS) and is implicated in survival of tumor cells as well as therapeutic resistance. In the present study, we searched for combination therapies with the dual PI3K/mTOR inhibitor NVP-BEZ235 (BEZ235) in RMS. Here, we identify a synthetic lethal interaction of BEZ235 together with the lysosomotropic agent chloroquine (CQ), which is effective against embryonal rhabdomyosarcoma (ERMS). BEZ235 and CQ at subtoxic concentrations synergize to induce apoptosis in ERMS cells, as confirmed by calculation of combination index (CI). BEZ235 and CQ cooperate to activate caspase-9, -3 and -8, which is crucial for apoptosis induction given that the broad-range caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (zVAD.fmk) blocks BEZ235/CQ-induced apoptosis. Additionally, pharmacological inhibition of lysosomal enzymes significantly reduces BEZ235/CQ-induced apoptosis, indicating concomitant activation of the lysosomal compartment. Importantly, BEZ235/CQ-induced apoptosis is significantly inhibited by antioxidants, implying that increased oxidative stress contributes to BEZ235/CQ-induced cell death. Importantly, our molecular studies reveal that BEZ235/CQ-induced apoptosis is mediated by cooperative downregulation of the antiapoptotic BCL-2 family protein MCL-1, since stabilization of MCL-1 by expression of a non-degradable MCL-1 phospho-defective mutant significantly decreases BEZ235/CQ-induced apoptosis. Also, overexpression of antiapoptotic BCL-2 leads to a significant reduction of BEZ235/CQ-induced apoptosis, emphasizing that an intact mitochondrial pathway of apoptosis is required for BEZ235/CQ-induced cell death. This identification of a synthetic lethality of BEZ235 and CQ has important implications for the development of molecular targeted therapies for RMS.
Collapse
Affiliation(s)
- Manuela Hugle
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany
| | - Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
73
|
Zhang K, Song H, Yang P, Dai X, Li Y, Wang L, Du J, Pan K, Zhang T. Silencing dishevelled-1 sensitizes paclitaxel-resistant human ovarian cancer cells via AKT/GSK-3β/β-catenin signalling. Cell Prolif 2015; 48:249-58. [PMID: 25643607 DOI: 10.1111/cpr.12161] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 09/30/2014] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES Expression of dishevelled-1 (DVL1) has recently been linked to cancer progression, however, its role in resistance to cancer therapy is unclear. In this study, we aimed to explore the function of DVL1 in paclitaxel-resistant human ovarian cancer cells. MATERIALS AND METHODS The MTT assay was used to assess effects of DVL1 silencing on sensitivity of cells that were otherwise resistant to paclitaxel (Taxol). Western blotting and immunofluorescence staining were used to examine effects of DVL1 on AKT/GSK-3β/β-catenin signalling. RESULTS Dishevelled-1 was found to be over-expressed in a paclitaxel-resistant cell line derived from human ovarian cancer cell line A2780 (A2780/Taxol line) as well as parental A2780 cells. Down-regulation of DVL1 (using the inhibitor 3289-8625 or siRNA (siDVL1) against DVL1) sensitized A2780/Taxol cells to paclitaxel. Over-expression of DVL1 in A2780 cells increased protein levels of P-gp, BCRP and Bcl-2, which are known targets of β-catenin. Silencing DVL1 in A2780/Taxol cells also reduced levels of these proteins, and led to accumulation of β-catenin. In addition, DVL1 aberrantly activated AKT/GSK-3β/β-catenin signalling. Inactivation of AKT signalling attenuated DVL1-mediated inhibition of GSK-3β and accumulation of β-catenin, in both A2780 and A2780/Taxol cells. CONCLUSIONS Taken together, these results suggest that silencing DVL1 sensitized A2780/Taxol cells to paclitaxel, by down-regulating AKT/GSK-3β/β-catenin signalling, providing a novel strategy for chemosensitization of ovarian cancer to paclitaxel-induced cytotoxicity.
Collapse
Affiliation(s)
- Kun Zhang
- School of Biomedicine Sciences, Chengdu Medical College, Chengdu, 610500, China
| | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Song T, Chai G, Liu Y, Xie M, Chen Q, Yu X, Sheng H, Zhang Z. Mechanism of synergy of BH3 mimetics and paclitaxel in chronic myeloid leukemia cells: Mcl-1 inhibition. Eur J Pharm Sci 2015; 70:64-71. [PMID: 25596561 DOI: 10.1016/j.ejps.2015.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 01/06/2015] [Accepted: 01/06/2015] [Indexed: 12/16/2022]
Abstract
Paclitaxel is an alternative chemotherapeutic agent for chronic myelogenous leukemia (CML) when primary or secondary resistance of tyrosine kinase inhibitors (TKI) is emerging, because paclitaxel could bypass the apoptotic deficiencies linked to p53 and fas ligand pathways in CML. However, high levels of Bcl-2 family proteins in CML could resist paclitaxel-induced apoptosis. Herein, we utilized two BH3 mimetics ABT-737 and S1 to study the potential of BH3 mimetics in combination with paclitaxel in treatment of CML cells and illustrated the mechanism by which BH3 mimetics synergize with paclitaxel. As a single agent, S1 could induce apoptosis in CML-derived cell line K562, whereas ABT-737 was largely ineffective. However, both of the two agents could efficiently synergize with paclitaxel through intrinsic apoptosis pathway. By using Bcl-2 siRNA, Bcl-XL siRNA or Mcl-1 siRNA, we found although each of the three members exhibited activities to block paclitaxel-induced apoptosis, Mcl-1 was the determinant for the synergistic effect between paclitaxel and ABT-737 or S1. Furthermore, paclitaxel/ABT737 synergized to drastically upregulate Bim to displace Bak from Mcl-1, whereas S1 directly binds Mcl-1 to release both Bim and Bak. As such, ABT-737 and S1 sensitized CML to paclitaxel by Mcl-1 inhibition, indirect inhibition through Bim antagonizing Mcl-1, or direct inhibition through binding to Mcl-1 itself. Finally, activation of JNK/Bim pathway was identified as the apical mechanism for ABT-737/paclitaxel synergism. Together, our results demonstrated potent synergy between BH3 mimetics and paclitaxel in the killing of CML cells and revealed an important role for Mcl-1 in mediating synergism by these agents.
Collapse
Affiliation(s)
- Ting Song
- School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China
| | - Gaobo Chai
- School of Life Science and Technology, Dalian University of Technology, Dalian, China
| | - Yubo Liu
- School of Life Science and Technology, Dalian University of Technology, Dalian, China
| | - Mingzhou Xie
- School of Life Science and Technology, Dalian University of Technology, Dalian, China
| | - Qingbin Chen
- School of Life Science and Technology, Dalian University of Technology, Dalian, China
| | - Xiaoyan Yu
- School of Life Science and Technology, Dalian University of Technology, Dalian, China
| | - Hongkun Sheng
- School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China
| | - Zhichao Zhang
- School of Chemistry, State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China.
| |
Collapse
|
75
|
Adem J, Ropponen A, Eeva J, Eray M, Pelkonen J, Nuutinen U. Rituximab-induced early and late signaling have opposite effects on dexamethasone-induced apoptosis in human follicular lymphoma cells. Leuk Lymphoma 2015; 56:2448-57. [PMID: 25563557 DOI: 10.3109/10428194.2014.1001983] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The addition of rituximab (RTX) to standard chemotherapy has improved the treatment of B-cell malignancies. We show here that RTX and dexamethasone (Dex) induced synergistic apoptosis in follicular lymphoma cell lines. However, apoptosis was delayed by RTX-induced early protective signaling. RTX-induced early signaling also decreased Dex-induced apoptosis and led to phosphorylation of ERK1/2, Bcl-2 (at serine 70) and phosphorylation/degradation of BimL/EL. All these events were prevented by the MEK inhibitor, UO126. Therefore, we suggest that RTX-induced ERK-mediated signaling events lead to protection from apoptosis during early signaling and that blocking of Bim and Bcl-2 phosphorylation might be used as a novel strategy for lymphoma treatment.
Collapse
Affiliation(s)
- Jemal Adem
- a Department of Clinical Microbiology , Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland.,e Cancer Center of University of Eastern Finland , Kuopio , Finland
| | - Antti Ropponen
- a Department of Clinical Microbiology , Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland
| | - Jonna Eeva
- a Department of Clinical Microbiology , Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland
| | - Mine Eray
- b Fimlab Laboratories Oy, Tampere University Hospital , Tampere , Finland.,c Department of Medicine,University of Tampere , Tampere , Finland
| | - Jukka Pelkonen
- a Department of Clinical Microbiology , Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland.,d Eastern Finland Laboratory Centre (ISLAB) , Kuopio , Finland.,e Cancer Center of University of Eastern Finland , Kuopio , Finland
| | - Ulla Nuutinen
- a Department of Clinical Microbiology , Institute of Clinical Medicine, University of Eastern Finland , Kuopio , Finland
| |
Collapse
|
76
|
BCL2 mutations are associated with increased risk of transformation and shortened survival in follicular lymphoma. Blood 2014; 125:658-67. [PMID: 25452615 DOI: 10.1182/blood-2014-04-571786] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Follicular lymphoma (FL), an indolent neoplasm caused by a t(14;18) chromosomal translocation that juxtaposes the BCL2 gene and immunoglobulin locus, has a variable clinical course and frequently undergoes transformation to an aggressive lymphoma. Although BCL2 mutations have been previously described, their relationship to FL progression remains unclear. In this study, we evaluated the frequency and nature of BCL2 mutations in 2 independent cohorts of grade 1 and 2 FLs, along with the correlation between BCL2 mutations, transformation risk, and survival. The prevalence of BCL2 coding sequence mutations was 12% in FL at diagnosis and 53% at transformation (P < .0001). The presence of these BCL2 mutations at diagnosis correlated with an increased risk of transformation (hazard ratio 3.6; 95% CI, 2.0-6.2; P < .0001) and increased risk of death due to lymphoma (median survival of 9.5 years with BCL2 mutations vs 20.4 years without; P = .012). In a multivariate analysis, BCL2 mutations and high FL international prognostic index were independent risk factors for transformation and death due to lymphoma. Some mutant Bcl-2 proteins exhibited enhanced antiapoptotic capacity in vitro. Accordingly, BCL2 mutations can affect antiapoptotic Bcl-2 function, are associated with increased activation-induced cytidine deaminase expression, and correlate with increased risk of transformation and death due to lymphoma.
Collapse
|
77
|
Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy. Nat Rev Mol Cell Biol 2014; 15:49-63. [PMID: 24355989 DOI: 10.1038/nrm3722] [Citation(s) in RCA: 2299] [Impact Index Per Article: 209.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The BCL-2 protein family determines the commitment of cells to apoptosis, an ancient cell suicide programme that is essential for development, tissue homeostasis and immunity. Too little apoptosis can promote cancer and autoimmune diseases; too much apoptosis can augment ischaemic conditions and drive neurodegeneration. We discuss the biochemical, structural and genetic studies that have clarified how the interplay between members of the BCL-2 family on mitochondria sets the apoptotic threshold. These mechanistic insights into the functions of the BCL-2 family are illuminating the physiological control of apoptosis, the pathological consequences of its dysregulation and the promising search for novel cancer therapies that target the BCL-2 family.
Collapse
|