51
|
Lee YM, Liou YC. Gears-In-Motion: The Interplay of WW and PPIase Domains in Pin1. Front Oncol 2018; 8:469. [PMID: 30460195 PMCID: PMC6232885 DOI: 10.3389/fonc.2018.00469] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/04/2018] [Indexed: 01/22/2023] Open
Abstract
Pin1 belongs to the family of the peptidyl-prolyl cis-trans isomerase (PPIase), which is a class of enzymes that catalyze the cis/trans isomerization of the Proline residue. Pin1 is unique and only catalyzes the phosphorylated Serine/Threonine-Proline (S/T-P) motifs of a subset of proteins. Since the discovery of Pin1 as a key protein in cell cycle regulation, it has been implicated in numerous diseases, ranging from cancer to neurodegenerative diseases. The main features of Pin1 lies in its two main domains: the WW (two conserved tryptophan) domain and the PPIase domain. Despite extensive studies trying to understand the mechanisms of Pin1 functions, how these two domains contribute to the biological roles of Pin1 in cellular signaling requires more investigations. The WW domain of Pin1 is known to have a higher affinity to its substrate than that of the PPIase domain. Yet, the WW domain seems to prefer the trans configuration of phosphorylated S/T-P motif, while the PPIase catalyzes the cis to trans isomerasion. Such contradicting information has generated much confusion as to the actual mechanism of Pin1 function. In addition, dynamic allostery has been suggested to be important for Pin1 function. Henceforth, in this review, we will be looking at the progress made in understanding the function of Pin1, and how these understandings can aid us in overcoming the diseases implicated by Pin1 such as cancer during drug development.
Collapse
Affiliation(s)
- Yew Mun Lee
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| |
Collapse
|
52
|
Chen YJ, Huang CH, Shi YJ, Lee YC, Wang LJ, Chang LS. The suppressive effect of arsenic trioxide on TET2-FOXP3-Lyn-Akt axis-modulated MCL1 expression induces apoptosis in human leukemia cells. Toxicol Appl Pharmacol 2018; 358:43-55. [PMID: 30213730 DOI: 10.1016/j.taap.2018.09.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/02/2018] [Accepted: 09/07/2018] [Indexed: 01/11/2023]
Abstract
Arsenic trioxide (ATO) has been reported to inhibit the activity of Ten-eleven translocation methylcytosine dioxygenase (TET). TET modulates FOXP3 expression, while dysregulation of FOXP3 expression promotes the malignant progression of leukemia cells. We examined the role of TET-FOXP3 axis in the cytotoxic effects of ATO on the human acute myeloid leukemia cell line, U937. ATO-induced apoptosis in U937 cells was characterized by activation of caspase-3/-9, mitochondrial depolarization, and MCL1 downregulation. In addition, ATO-treated U937 cells showed ROS-mediated inhibition of TET2 transcription, leading to downregulation of FOXP3 expression and in turn, suppression of FOXP3-mediated activation of Lyn and Akt. Overexpression of FOXP3 or Lyn minimized the suppressive effect of ATO on Akt activation and MCL1 expression. Promoter luciferase activity and chromatin immunoprecipitation assays revealed the crucial role of Akt-mediated CREB phosphorylation in MCL1 transcription. Further, ATO-induced Akt inactivation promoted GSK3β-mediated degradation of MCL1. Transfection of constitutively active Akt expression abrogated ATO-induced MCL1 downregulation. MCL1 overexpression lessened the ATO-induced depolarization of mitochondrial membrane and increased the viability of ATO-treated cells. Thus, our data suggest that ATO induces mitochondria-mediated apoptosis in U937 cells through its suppressive effect on TET2-FOXP3-Lyn-Akt axis-modulated MCL1 transcription and protein stabilization. Our findings also indicate that the same pathway underlies ATO-induced death in human leukemia HL-60 cells.
Collapse
Affiliation(s)
- Ying-Jung Chen
- Department of Fragrance and Cosmetic Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Hui Huang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yi-Jun Shi
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yuan-Chin Lee
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Liang-Jun Wang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Long-Sen Chang
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|
53
|
Prolyl isomerase Pin1: a promoter of cancer and a target for therapy. Cell Death Dis 2018; 9:883. [PMID: 30158600 PMCID: PMC6115400 DOI: 10.1038/s41419-018-0844-y] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/15/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022]
Abstract
Pin1 is the only known peptidyl-prolyl cis–trans isomerase (PPIase) that specifically recognizes and isomerizes the phosphorylated Serine/Threonine-Proline (pSer/Thr-Pro) motif. The Pin1-mediated structural transformation posttranslationally regulates the biofunctions of multiple proteins. Pin1 is involved in many cellular processes, the aberrance of which lead to both degenerative and neoplastic diseases. Pin1 is highly expressed in the majority of cancers and its deficiency significantly suppresses cancer progression. According to the ground-breaking summaries by Hanahan D and Weinberg RA, the hallmarks of cancer comprise ten biological capabilities. Multiple researches illuminated that Pin1 contributes to these aberrant behaviors of cancer via promoting various cancer-driving pathways. This review summarized the detailed mechanisms of Pin1 in different cancer capabilities and certain Pin1-targeted small-molecule compounds that exhibit anticancer activities, expecting to facilitate anticancer therapies by targeting Pin1.
Collapse
|
54
|
Hu Y, Yagüe E, Zhao J, Wang L, Bai J, Yang Q, Pan T, Zhao H, Liu J, Zhang J. Sabutoclax, pan-active BCL-2 protein family antagonist, overcomes drug resistance and eliminates cancer stem cells in breast cancer. Cancer Lett 2018; 423:47-59. [DOI: 10.1016/j.canlet.2018.02.036] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/18/2018] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
|
55
|
Russo Spena C, De Stefano L, Palazzolo S, Salis B, Granchi C, Minutolo F, Tuccinardi T, Fratamico R, Crotti S, D'Aronco S, Agostini M, Corona G, Caligiuri I, Canzonieri V, Rizzolio F. Liposomal delivery of a Pin1 inhibitor complexed with cyclodextrins as new therapy for high-grade serous ovarian cancer. J Control Release 2018; 281:1-10. [PMID: 29746956 DOI: 10.1016/j.jconrel.2018.04.055] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/28/2018] [Accepted: 04/30/2018] [Indexed: 12/28/2022]
Abstract
Pin1, a prolyl isomerase that sustains tumor progression, is overexpressed in different types of malignancies. Functional inactivation of Pin1 restrains tumor growth and leaves normal cells unaffected making it an ideal pharmaceutical target. Although many studies on Pin1 have focused on malignancies that are influenced by sex hormones, studies in ovarian cancer have lagged behind. Here, we show that Pin1 is an important therapeutic target in high-grade serous epithelial ovarian cancer. Knock down of Pin1 in ovarian cancer cell lines induces apoptosis and restrains tumor growth in a syngeneic mouse model. Since specific and non-covalent Pin1 inhibitors are still limited, the first liposomal formulation of a Pin1 inhibitor was designed. The drug was efficiently encapsulated in modified cyclodextrins and remotely loaded into pegylated liposomes. This liposomal formulation accumulates preferentially in the tumor and has a desirable pharmacokinetic profile. The liposomal inhibitor was able to alter Pin1 cancer driving-pathways trough the induction of proteasome-dependent degradation of Pin1 and was found to be effective in curbing ovarian tumor growth in vivo.
Collapse
Affiliation(s)
- Concetta Russo Spena
- Department of Translational Research, Experimental and Clinical Pharmacology, Center for Molecular Biomedicine - CRO, National Cancer Institute, Aviano, Italy; Doctoral School in Chemistry, University of Trieste, Italy
| | - Lucia De Stefano
- Department of Translational Research, Experimental and Clinical Pharmacology, Center for Molecular Biomedicine - CRO, National Cancer Institute, Aviano, Italy; Doctoral School in Chemistry, University of Trieste, Italy
| | - Stefano Palazzolo
- Department of Translational Research, Experimental and Clinical Pharmacology, Center for Molecular Biomedicine - CRO, National Cancer Institute, Aviano, Italy
| | - Barbara Salis
- Doctoral School in Molecular Biomedicine, University of Trieste, Italy; Department of Molecular Biology and Translational Research, Pathology Unit, Center for Molecular Biomedicine - CRO, National Cancer Institute, Aviano, Italy
| | | | | | | | - Roberto Fratamico
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sara Crotti
- Città della Speranza, Institute of Pediatric Research, Padova, Italy
| | - Sara D'Aronco
- Città della Speranza, Institute of Pediatric Research, Padova, Italy; Department of Surgical, Oncological and Gastroenterological Sciences, First Surgical Clinic Section, University of Padova, Italy
| | - Marco Agostini
- Città della Speranza, Institute of Pediatric Research, Padova, Italy; Department of Surgical, Oncological and Gastroenterological Sciences, First Surgical Clinic Section, University of Padova, Italy
| | - Giuseppe Corona
- Department of Molecular Biology and Translational Research, Immunopathology and Cancer Biomarkers Unit, Center for Molecular Biomedicine - CRO, National Cancer Institute, Aviano, Italy
| | - Isabella Caligiuri
- Department of Molecular Biology and Translational Research, Pathology Unit, Center for Molecular Biomedicine - CRO, National Cancer Institute, Aviano, Italy
| | - Vincenzo Canzonieri
- Department of Molecular Biology and Translational Research, Pathology Unit, Center for Molecular Biomedicine - CRO, National Cancer Institute, Aviano, Italy
| | - Flavio Rizzolio
- Department of Translational Research, Experimental and Clinical Pharmacology, Center for Molecular Biomedicine - CRO, National Cancer Institute, Aviano, Italy; Department of Molecular Sciences and Nanosystems, Ca' Foscari University, Venezia-Mestre, Italy.
| |
Collapse
|
56
|
Zheng M, Xu H, Liao XH, Chen CP, Zhang AL, Lu W, Wang L, Yang D, Wang J, Liu H, Zhou XZ, Lu KP. Inhibition of the prolyl isomerase Pin1 enhances the ability of sorafenib to induce cell death and inhibit tumor growth in hepatocellular carcinoma. Oncotarget 2018; 8:29771-29784. [PMID: 28404959 PMCID: PMC5444702 DOI: 10.18632/oncotarget.15967] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer, but is the second leading cause of cancer deaths, partially due to its heterogeneity and drug resistance. Sorafenib is the only medical treatment with a proven efficacy against advanced HCC, but its overall clinical efficacy is still modest. Therefore, a major challenge is how to improve its therapeutic efficacy. The unique prolyl isomerase Pin1 regulates numerous cancer-driving pathways. Notably, Pin1 is overexpressed in about 70% HBV-positive HCC patients and contributes to HCC tumorigenesis. However, the role of Pin1 in the efficacy of sorafenib against HCC is unknown. Here we found that sorafenib down-regulated Pin1 mRNA and protein expression, likely through inhibition of Pin1 transcription by the Rb/E2F pathway. Importantly, Pin1 knockdown potently enhanced the ability of sorafenib to induce cell death in HCC, which was further supported by the findings that Pin1 knockdown led to stabilization of Fbxw7 and destabilization of Mcl-1. Furthermore, all-trans retinoic acid (ATRA), a known anticancer drug that inhibits and ultimately induces degradation of active Pin1 in cancer cells, also potently sensitized HCC cells to sorafenib-induced cell death at least in part through a caspase-dependent manner. Moreover, ATRA also synergistically enhanced the ability of sorafenib to reduce Pin1 and inhibit tumor growth of HCC in mouse xenograft models. Collectively, these results not only demonstrate that Pin1 down-regulation is a key event underlying the anti-tumor effects of sorafenib, but also uncover that Pin1 inhibitors offer a novel approach to enhance the therapeutic efficacy of sorafenib against HCC.
Collapse
Affiliation(s)
- Min Zheng
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Huijuan Xu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Xin-Hua Liao
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Champ Peng Chen
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Arina Li Zhang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Wenxian Lu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Long Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Dayun Yang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Jichuang Wang
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Hekun Liu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China
| | - Xiao Zhen Zhou
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China.,Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Kun Ping Lu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350108, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian 350108, China.,Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
57
|
Marmé F, Gomez-Roca C, Graudenz K, Huang F, Lettieri J, Peña C, Trnkova ZJ, Eucker J. Phase 1, open-label, dose-escalation study of sorafenib in combination with eribulin in patients with advanced, metastatic, or refractory solid tumors. Cancer Chemother Pharmacol 2018; 81:727-737. [PMID: 29468456 DOI: 10.1007/s00280-018-3540-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/08/2018] [Indexed: 11/24/2022]
Abstract
Combining sorafenib and eribulin mesylate may provide synergistic antitumor activities with limited overlapping toxicities. This phase 1b, open-label, dose-escalation study evaluated safety, pharmacokinetics, maximum tolerated dose/recommended phase 2 dose (MTD/RP2D), and preliminary efficacy of sorafenib plus standard-dose eribulin mesylate in patients with advanced, metastatic, or refractory tumors. Patients received sorafenib 200 mg twice daily (BID; n = 5), 600 mg/day (n = 8), and 400 mg BID (MTD; n = 27). Dose-limiting toxicities were increased alanine aminotransferase and acute coronary syndrome (both grade 3) in the 400-mg BID dose-escalation and expansion cohorts, respectively. No significant increase in mean QTcF duration was observed with eribulin plus sorafenib versus eribulin alone; there were no drug-drug interactions. Five patients achieved partial response; 16 achieved stable disease. The combination of sorafenib and eribulin mesylate presented no unexpected safety concerns and no significant impact on QT/QTc intervals or drug-drug interactions. Sorafenib 400 mg BID plus standard-dose eribulin is the RP2D.
Collapse
Affiliation(s)
- Frederik Marmé
- National Center for Tumor Diseases, Gynecologic Oncology, Heidelberg, Germany
| | - Carlos Gomez-Roca
- Institut Universitaire du Cancer-Oncopole and Institut Claudius Regaud, Toulouse, France
| | | | - Funan Huang
- Bayer HealthCare Pharmaceuticals, Whippany, NJ, USA
| | | | - Carol Peña
- Bayer HealthCare Pharmaceuticals, Whippany, NJ, USA
| | | | - Jan Eucker
- Department of Oncology and Hematology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12209, Berlin, Germany.
| |
Collapse
|
58
|
Park SH, Lee DH, Kim JL, Kim BR, Na YJ, Jo MJ, Jeong YA, Lee SY, Lee SI, Lee YY, Oh SC. Metformin enhances TRAIL-induced apoptosis by Mcl-1 degradation via Mule in colorectal cancer cells. Oncotarget 2018; 7:59503-59518. [PMID: 27517746 PMCID: PMC5312327 DOI: 10.18632/oncotarget.11147] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 07/06/2016] [Indexed: 01/09/2023] Open
Abstract
Metformin is an anti-diabetic drug with a promising anti-cancer potential. In this study, we show that subtoxic doses of metformin effectively sensitize human colorectal cancer (CRC) cells to tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), which induces apoptosis. Metformin alone did not induce apoptosis, but significantly potentiated TRAIL-induced apoptosis in CRC cells. CRC cells treated with metformin and TRAIL showed activation of the intrinsic and extrinsic pathways of caspase activation. We attempted to elucidate the underlying mechanism, and found that metformin significantly reduced the protein levels of myeloid cell leukemia 1 (Mcl-1) in CRC cells and, the overexpression of Mcl-1 inhibited cell death induced by metformin and/or TRAIL. Further experiments revealed that metformin did not affect mRNA levels, but increased proteasomal degradation and protein stability of Mcl-1. Knockdown of Mule triggered a significant decrease of Mcl-1 polyubiquitination. Metformin caused the dissociation of Noxa from Mcl-1, which allowed the binding of the BH3-containing ubiquitin ligase Mule followed by Mcl-1ubiquitination and degradation. The metformin-induced degradation of Mcl-1 required E3 ligase Mule, which is responsible for the polyubiquitination of Mcl-1. Our study is the first report indicating that metformin enhances TRAIL-induced apoptosis through Noxa and favors the interaction between Mcl-1 and Mule, which consequently affects Mcl-1 ubiquitination.
Collapse
Affiliation(s)
- Seong Hye Park
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | - Dae-Hee Lee
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea.,Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jung Lim Kim
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Bo Ram Kim
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yoo Jin Na
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | - Min Jee Jo
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea
| | - Yoon A Jeong
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Suk-Young Lee
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sun Il Lee
- Department of Surgery, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yong Yook Lee
- The Korean Ginseng Research Institute, Daejeon, Republic of Korea
| | - Sang Cheul Oh
- Brain Korea 21 Program for Biomedicine Science, Korea University College of Medicine, Korea University, Seoul, Republic of Korea.,Division of Oncology/Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
59
|
Young AI, Timpson P, Gallego-Ortega D, Ormandy CJ, Oakes SR. Myeloid cell leukemia 1 (MCL-1), an unexpected modulator of protein kinase signaling during invasion. Cell Adh Migr 2017; 12:513-523. [PMID: 29166822 PMCID: PMC6363037 DOI: 10.1080/19336918.2017.1393591] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Myeloid cell leukemia-1 (MCL-1), closely related to B-cell lymphoma 2 (BCL-2), has a well-established role in cell survival and has emerged as an important target for cancer therapeutics. We have demonstrated that inhibiting MCL-1 is efficacious in suppressing tumour progression in pre-clinical models of breast cancer and revealed that in addition to its role in cell survival, MCL-1 modulated cellular invasion. Utilizing a MCL-1-specific genetic antagonist, we found two possible mechanisms; firstly MCL-1 directly binds to and alters the phosphorylation of the cytoskeletal remodeling protein, Cofilin, a protein important for cytoskeletal remodeling during invasion, and secondly MCL-1 modulates the levels SRC family kinases (SFKs) and their targets. These data provide evidence that MCL-1 activities are not limited to endpoints of extracellular and intracellular signaling culminating in cell survival as previously thought, but can directly modulate the output of SRC family kinases signaling during cellular invasion. Here we review the pleotropic roles of MCL-1 and discuss the implications of this newly discovered effect on protein kinase signaling for the development of cancer therapeutics.
Collapse
Affiliation(s)
- Adelaide Ij Young
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia
| | - Paul Timpson
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia.,b St. Vincent's Clinical School, UNSW Medicine , Victoria Street, Darlinghurst , NSW , Australia
| | - David Gallego-Ortega
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia.,b St. Vincent's Clinical School, UNSW Medicine , Victoria Street, Darlinghurst , NSW , Australia
| | - Christopher J Ormandy
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia.,b St. Vincent's Clinical School, UNSW Medicine , Victoria Street, Darlinghurst , NSW , Australia
| | - Samantha R Oakes
- a Cancer Research Division , Garvan Institute of Medical Research and the Kinghorn Cancer Centre , 384 Victoria Street, Darlinghurst , NSW , Australia.,b St. Vincent's Clinical School, UNSW Medicine , Victoria Street, Darlinghurst , NSW , Australia
| |
Collapse
|
60
|
Senichkin VV, Kopeina GS, Prokhorova EA, Zamaraev AV, Lavrik IN, Zhivotovsky B. Modulation of Mcl-1 transcription by serum deprivation sensitizes cancer cells to cisplatin. Biochim Biophys Acta Gen Subj 2017; 1862:557-566. [PMID: 29203282 DOI: 10.1016/j.bbagen.2017.11.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/19/2017] [Accepted: 11/29/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND The development of approaches that increase therapeutic effects of anti-cancer drugs is one of the most important tasks of oncology. Caloric restriction in vivo or serum deprivation (SD) in vitro has been shown to be an effective tool for sensitizing cancer cells to chemotherapeutic drugs. However, the detailed mechanisms underlying the enhancement of apoptosis in cancer cells by SD remain to be elucidated. METHODS Flow cytometry, caspase activity assay and western blotting were used for cell death rate evaluation. Western blotting, gel-filtration, siRNA approach and qRT-PCR were used to elucidate the mechanism underlying cell death potentiation upon SD. RESULTS We demonstrated that SD sensitizes cancer cells to treatment with chemotherapeutic agent cisplatin. This effect is independent on activation of caspases-2 and -8, apical caspases triggering apoptosis in response to genotoxic stress. SD potentiates cell death via downregulation of the anti-apoptotic protein Mcl-1. In fact, SD reduces the Mcl-1 mRNA level, which consequently decreases the Mcl-1 protein level and renders cells more susceptible to apoptosis induction via the formation of apoptosome. CONCLUSIONS Mcl-1 protein is an important regulator of sensitivity of cancer cells to apoptotic stimuli upon SD. GENERAL SIGNIFICANCE This study identifies Mcl-1 as a new target for the sensitization of human cancer cells to cell death by SD, which is of great significance for the development of efficient anti-cancer therapies.
Collapse
Affiliation(s)
| | - Gelina S Kopeina
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia
| | | | - Alexey V Zamaraev
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia
| | - Inna N Lavrik
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia; Department of Translational Inflammation, Institute of Experimental Internal Medicine, Otto von Guericke University, Magdeburg, Germany
| | - Boris Zhivotovsky
- Faculty of Basic Medicine, MV Lomonosov Moscow State University, Moscow, Russia; Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
61
|
Singh PK, Roukounakis A, Frank DO, Kirschnek S, Das KK, Neumann S, Madl J, Römer W, Zorzin C, Borner C, Haimovici A, Garcia-Saez A, Weber A, Häcker G. Dynein light chain 1 induces assembly of large Bim complexes on mitochondria that stabilize Mcl-1 and regulate apoptosis. Genes Dev 2017; 31:1754-1769. [PMID: 28982759 PMCID: PMC5666674 DOI: 10.1101/gad.302497.117] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 09/05/2017] [Indexed: 12/17/2022]
Abstract
In this study, Singh et al. investigated Bim structure and activity and show that Bim is regulated by the formation of large protein complexes containing dynein light chain 1 (DLC1). Their findings demonstrate that control of apoptosis at mitochondria extends beyond the interaction of monomers of proapoptotic and anti-apoptotic Bcl-2 family members and involves more complex structures of proteins at the mitochondrial outer membrane. The Bcl-2 family protein Bim triggers mitochondrial apoptosis. Bim is expressed in nonapoptotic cells at the mitochondrial outer membrane, where it is activated by largely unknown mechanisms. We found that Bim is regulated by formation of large protein complexes containing dynein light chain 1 (DLC1). Bim rapidly inserted into cardiolipin-containing membranes in vitro and recruited DLC1 to the membrane. Bim binding to DLC1 induced the formation of large Bim complexes on lipid vesicles, on isolated mitochondria, and in intact cells. Native gel electrophoresis and gel filtration showed Bim-containing mitochondrial complexes of several hundred kilodaltons in all cells tested. Bim unable to form complexes was consistently more active than complexed Bim, which correlated with its substantially reduced binding to anti-apoptotic Bcl-2 proteins. At endogenous levels, Bim surprisingly bound only anti-apoptotic Mcl-1 but not Bcl-2 or Bcl-XL, recruiting only Mcl-1 into large complexes. Targeting of DLC1 by RNAi in human cell lines induced disassembly of Bim–Mcl-1 complexes and the proteasomal degradation of Mcl-1 and sensitized the cells to the Bcl-2/Bcl-XL inhibitor ABT-737. Regulation of apoptosis at mitochondria thus extends beyond the interaction of monomers of proapoptotic and anti-apoptotic Bcl-2 family members but involves more complex structures of proteins at the mitochondrial outer membrane, and targeting complexes may be a novel therapeutic strategy.
Collapse
Affiliation(s)
- Prafull Kumar Singh
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center-University of Freiburg, 79104 Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Aristomenis Roukounakis
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center-University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Daniel O Frank
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center-University of Freiburg, 79104 Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Susanne Kirschnek
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center-University of Freiburg, 79104 Freiburg, Germany
| | - Kushal Kumar Das
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, 72076 Tübingen, Germany
| | - Simon Neumann
- Institute of Molecular Medicine and Cell Research, University of Freiburg, 79104 Freiburg, Germany
| | - Josef Madl
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Winfried Römer
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Carina Zorzin
- Institute of Pharmaceutical Technology and Biopharmacy, University of Freiburg, 79104 Freiburg, Germany
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Aladin Haimovici
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center-University of Freiburg, 79104 Freiburg, Germany
| | - Ana Garcia-Saez
- Interfaculty Institute of Biochemistry (IFIB), University of Tübingen, 72076 Tübingen, Germany
| | - Arnim Weber
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center-University of Freiburg, 79104 Freiburg, Germany
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Faculty of Medicine, Medical Center-University of Freiburg, 79104 Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
62
|
Hannen R, Hauswald M, Bartsch JW. A Rationale for Targeting Extracellular Regulated Kinases ERK1 and ERK2 in Glioblastoma. J Neuropathol Exp Neurol 2017; 76:838-847. [DOI: 10.1093/jnen/nlx076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
63
|
Seo SU, Kim TH, Kim DE, Min KJ, Kwon TK. NOX4-mediated ROS production induces apoptotic cell death via down-regulation of c-FLIP and Mcl-1 expression in combined treatment with thioridazine and curcumin. Redox Biol 2017; 13:608-622. [PMID: 28806703 PMCID: PMC5554966 DOI: 10.1016/j.redox.2017.07.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 07/25/2017] [Accepted: 07/30/2017] [Indexed: 10/24/2022] Open
Abstract
Thioridazine is known to have anti-tumor effects by inhibiting PI3K/Akt signaling, which is an important signaling pathway in cell survival. However, thioridazine alone does not induce apoptosis in head and neck squamous cell carcinoma (AMC-HN4), human breast carcinoma (MDA-MB231), and human glioma (U87MG) cells. Therefore, we investigated whether combined treatment with thioridazine and curcumin induces apoptosis. Combined treatment with thioridazine and curcumin markedly induced apoptosis in cancer cells without inducing apoptosis in human normal mesangial cells and human normal umbilical vein cells (EA.hy926). We found that combined treatment with thioridazine and curcumin had synergistic effects in AMC-HN4 cells. Among apoptosis-related proteins, thioridazine plus curcumin induced down-regulation of c-FLIP and Mcl-1 expression at the post-translational levels in a proteasome-dependent manner. Augmentation of proteasome activity was related to the up-regulation of proteasome subunit alpha 5 (PSMA5) expression in curcumin plus thioridazine-treated cells. Combined treatment with curcumin and thioridazine produced intracellular ROS in a NOX4-dependent manner, and ROS-mediated activation of Nrf2/ARE signaling played a critical role in the up-regulation of PSMA5 expression. Furthermore, ectopic expression of c-FLIP and Mcl-1 inhibited apoptosis in thioridazine and curcumin-treated cells. Therefore, we demonstrated that thioridazine plus curcumin induces proteasome activity by up-regulating PSMA5 expression via NOX4-mediated ROS production and that down-regulation of c-FLIP and Mcl-1 expression post-translationally is involved in apoptosis.
Collapse
Affiliation(s)
- Seung Un Seo
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Tae Hwan Kim
- Department of Otolaryngology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Dong Eun Kim
- Department of Otolaryngology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | - Kyoung-Jin Min
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea.
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, 2800 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea.
| |
Collapse
|
64
|
Shi P, Oh YT, Deng L, Zhang G, Qian G, Zhang S, Ren H, Wu G, Legendre B, Anderson E, Ramalingam SS, Owonikoko TK, Chen M, Sun SY. Overcoming Acquired Resistance to AZD9291, A Third-Generation EGFR Inhibitor, through Modulation of MEK/ERK-Dependent Bim and Mcl-1 Degradation. Clin Cancer Res 2017; 23:6567-6579. [PMID: 28765329 DOI: 10.1158/1078-0432.ccr-17-1574] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/06/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022]
Abstract
Purpose: The mechanisms accounting for anticancer activity of AZD9291 (osimertinib or TAGRISSO), an approved third-generation EGFR inhibitor, in EGFR-mutant non-small cell lung cancer (NSCLC) cells and particularly for the subsequent development of acquired resistance are unclear and thus are the focus of this study.Experimental Design: AZD9219-resistant cell lines were established by exposing sensitive cell lines to AZD9291. Protein alterations were detected with Western blotting. Apoptosis was measured with annexin V/flow cytometry. Growth-inhibitory effects of tested drugs were evaluated in vitro with cell number estimation and colony formation assay and in vivo with mouse xenograft models. Protein degradation was determined by comparing protein half-lives and inhibiting proteasome. Gene knockdown were achieved with siRNA or shRNA.Results: AZD9291 potently induced apoptosis in EGFR-mutant NSCLC cell lines, in which ERK phosphorylation was suppressed accompanied with Bim elevation and Mcl-1 reduction likely due to enhanced Mcl-1 degradation and increased Bim stability. Blocking Bim elevation by gene knockdown or enforcing Mcl-1 expression attenuated or abolished AZD9291-induced apoptosis. Moreover, AZD9291 lost its ability to modulate Bim and Mcl-1 levels in AZD9291-resistant cell lines. The combination of a MEK inhibitor with AZD9291 restores the sensitivity of AZD9291-resistant cells including those with C797S mutation to undergo apoptosis and growth regression in vitro and in vivoConclusions: Modulation of MEK/ERK-dependent Bim and Mcl-1 degradation critically mediates sensitivity and resistance of EGFR-mutant NSCLC cells to AZD9291 and hence is an effective strategy to overcome acquired resistance to AZD9291. Clin Cancer Res; 23(21); 6567-79. ©2017 AACR.
Collapse
Affiliation(s)
- Puyu Shi
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China.,Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - You-Take Oh
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Liang Deng
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Guojing Zhang
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Guoqing Qian
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Shuo Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China.,Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Hui Ren
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China
| | - Grant Wu
- Transgenomic, Inc., Omaha, Nebraska
| | | | | | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Taofeek K Owonikoko
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia
| | - Mingwei Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, P.R. China.
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia.
| |
Collapse
|
65
|
Decker T, Overkamp F, Rösel S, Nusch A, Göhler T, Indorf M, Sahlmann J, Trarbach T. A randomized phase II study of paclitaxel alone versus paclitaxel plus sorafenib in second- and third-line treatment of patients with HER2-negative metastatic breast cancer (PASO). BMC Cancer 2017; 17:499. [PMID: 28743247 PMCID: PMC5526236 DOI: 10.1186/s12885-017-3492-1] [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: 04/25/2016] [Accepted: 07/20/2017] [Indexed: 01/07/2023] Open
Abstract
Background We conducted an open-label, randomized, two-arm multi-center study to assess the efficacy and safety of paclitaxel versus paclitaxel + sorafenib in patients with locally advanced or metastatic HER2-negative breast cancer. Methods Patients were randomly assigned to receive either paclitaxel monotherapy (80 mg/m2) weekly (3 weeks on, 1 week off) plus sorafenib 400 mg orally, twice a day taken continuously throughout 28 day cycles. Sorafenib dose was gradually escalated from a starting dose of 200 mg twice a day. The primary endpoint was progression free survival (PFS). Results A pre-planned efficacy interim analysis was performed on the data of 60 patients, 30 patients in each treatment arm. Median PFS was estimated at 6.6 months (95% CI: 5.1 to 9.0) in patients randomized to single-agent paclitaxel (Arm A) and 5.6 months (95% CI: 3.8 to 6.5) in patients randomized to paclitaxel-sorafenib combination (Arm B) therapy. Contrary to the hypothesis, the treatment effect was statistically significant in favor of paclitaxel monotherapy (hazard ratio 1.80, 95% CI: 1.02 to 3.20; log-rank test P = 0.0409). It was decided to stop the trial early for futility. Median OS was also in favor of Arm A (20.7 months (95% CI: 16.4 to 26.7) versus 12.1 months (95% CI: 5.8 to 20.4) in Arm B. Clinical control was achieved in 28 patients (93.3%) in Arm A and in 21 patients 70.0% in Arm B. Overall response rate was met in 43.3% of patients in Arm A and in 40.0% in Arm B. Toxicities were increased in Arm B with higher rates of diarrhea, nausea, neutropenia, hand-foot skin reaction (HFSR) and anorexia, Grad 3 and 4 toxicities were rare. Conclusions In this pre-planned interim analysis, paclitaxel-sorafenib combination therapy was not found to be superior to paclitaxel monotherapy with regard to the primary end point, progression-free survival. The trial was therefore discontinued early. There was no indication of more favorable outcomes for combination therapy in secondary efficacy end points. As expected, the safety and toxicity profile of the combination therapy was less favorable compared to monotherapy. Overall, this trial did not demonstrate that adding sorafenib to second- or third-line paclitaxel provides any clinical benefit to patients with HER2-negative advanced or metastatic breast cancer. Cautious dosing using a sorafenib ramp up schedule might have contributed to negative results. Trial registration The study was registered at EudraCT (No 2009–018025-73) and retrospectively registered at Clinical trials.gov on March 17, 2011 (NCT01320111).
Collapse
Affiliation(s)
| | | | | | - Arnd Nusch
- Practice for Haematology and internal Oncology, Velbert, Germany
| | | | | | | | - Tanja Trarbach
- iOMEDICO, Freiburg, Germany.,Center for Tumor Biology and Integrative Medicine Clinics Wilhelmshaven, Wilhelmshaven, Germany
| |
Collapse
|
66
|
Campaner E, Rustighi A, Zannini A, Cristiani A, Piazza S, Ciani Y, Kalid O, Golan G, Baloglu E, Shacham S, Valsasina B, Cucchi U, Pippione AC, Lolli ML, Giabbai B, Storici P, Carloni P, Rossetti G, Benvenuti F, Bello E, D'Incalci M, Cappuzzello E, Rosato A, Del Sal G. A covalent PIN1 inhibitor selectively targets cancer cells by a dual mechanism of action. Nat Commun 2017; 8:15772. [PMID: 28598431 PMCID: PMC5472749 DOI: 10.1038/ncomms15772] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 04/27/2017] [Indexed: 12/21/2022] Open
Abstract
The prolyl isomerase PIN1, a critical modifier of multiple signalling pathways, is overexpressed in the majority of cancers and its activity strongly contributes to tumour initiation and progression. Inactivation of PIN1 function conversely curbs tumour growth and cancer stem cell expansion, restores chemosensitivity and blocks metastatic spread, thus providing the rationale for a therapeutic strategy based on PIN1 inhibition. Notwithstanding, potent PIN1 inhibitors are still missing from the arsenal of anti-cancer drugs. By a mechanism-based screening, we have identified a novel covalent PIN1 inhibitor, KPT-6566, able to selectively inhibit PIN1 and target it for degradation. We demonstrate that KPT-6566 covalently binds to the catalytic site of PIN1. This interaction results in the release of a quinone-mimicking drug that generates reactive oxygen species and DNA damage, inducing cell death specifically in cancer cells. Accordingly, KPT-6566 treatment impairs PIN1-dependent cancer phenotypes in vitro and growth of lung metastasis in vivo. PIN1 is a promising therapeutic target for cancer treatment. In this study, the authors identify a covalent inhibitor of PIN1 with anti-tumour and anti-metastatic properties thanks to PIN1 inactivation and to the release, after binding to PIN1, of a quinone-mimicking compound that elicits reactive oxygen generation and causes DNA damage.
Collapse
Affiliation(s)
- Elena Campaner
- National Laboratory CIB (LNCIB), Area Science Park Padriciano, Trieste 34149, Italy.,Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Alessandra Rustighi
- National Laboratory CIB (LNCIB), Area Science Park Padriciano, Trieste 34149, Italy
| | - Alessandro Zannini
- National Laboratory CIB (LNCIB), Area Science Park Padriciano, Trieste 34149, Italy.,Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Alberto Cristiani
- National Laboratory CIB (LNCIB), Area Science Park Padriciano, Trieste 34149, Italy
| | - Silvano Piazza
- National Laboratory CIB (LNCIB), Area Science Park Padriciano, Trieste 34149, Italy
| | - Yari Ciani
- National Laboratory CIB (LNCIB), Area Science Park Padriciano, Trieste 34149, Italy
| | - Ori Kalid
- Karyopharm Therapeutics, Newton, Massachusetts 02459, USA
| | - Gali Golan
- Karyopharm Therapeutics, Newton, Massachusetts 02459, USA
| | - Erkan Baloglu
- Karyopharm Therapeutics, Newton, Massachusetts 02459, USA
| | - Sharon Shacham
- Karyopharm Therapeutics, Newton, Massachusetts 02459, USA
| | | | - Ulisse Cucchi
- Nerviano Medical Sciences Srl, Nerviano 20014, Italy
| | | | - Marco Lucio Lolli
- Department of Science and Drug Technology, University of Torino, Torino 10125, Italy
| | - Barbara Giabbai
- Elettra Sincrotrone Trieste S.C.p.A., Area Science Park Basovizza, Trieste 34149, Italy
| | - Paola Storici
- Elettra Sincrotrone Trieste S.C.p.A., Area Science Park Basovizza, Trieste 34149, Italy
| | - Paolo Carloni
- Computational Biomedicine, Institute for Advanced Simulation (IAS-5) and Institute of Neuroscience and Medicine (INM-9), Forschungszentrum Jülich, Jülich 52425, Germany
| | - Giulia Rossetti
- Computational Biomedicine, Institute for Advanced Simulation (IAS-5) and Institute of Neuroscience and Medicine (INM-9), Forschungszentrum Jülich, Jülich 52425, Germany.,Jülich Supercomputing Center (JSC), Forschungszentrum Jülich, Jülich 52425, Germany.,Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Aachen 52074, Germany
| | - Federica Benvenuti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Area Science Park Padriciano, Trieste 34149, Italy
| | - Ezia Bello
- IRCCS-Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Maurizio D'Incalci
- IRCCS-Mario Negri Institute for Pharmacological Research, Milano 20156, Italy
| | - Elisa Cappuzzello
- Department of Surgery, Oncology and Gastroenterology, Oncology and Immunology Section, University of Padova, Padova 35128, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, Oncology and Immunology Section, University of Padova, Padova 35128, Italy.,Veneto Institute of Oncology (IOV)-IRCCS, Padova 35128, Italy
| | - Giannino Del Sal
- National Laboratory CIB (LNCIB), Area Science Park Padriciano, Trieste 34149, Italy.,Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| |
Collapse
|
67
|
Bronte G, Andreis D, Bravaccini S, Maltoni R, Cecconetto L, Schirone A, Farolfi A, Fedeli A, Serra P, Donati C, Amadori D, Rocca A. Sorafenib for the treatment of breast cancer. Expert Opin Pharmacother 2017; 18:621-630. [PMID: 28335647 DOI: 10.1080/14656566.2017.1309024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Breast cancer treatment includes many options depending on the tumor clinicopathological profile, which groups breast cancer into various subtypes. Bevacizumab is currently the only drug capable of targeting angiogenesis in breast cancer. Sorafenib has also been studied in combination with other agents. Areas covered: Pharmacological aspects of sorafenib, including results from preclinical studies on breast cancer cells; findings about clinical efficacy and safety in both single-arm and randomized clinical trials; ongoing trials. Expert opinion: Since sorafenib as a single agent has shown limited efficacy in breast cancer, its combination with other drugs is under investigation. Dose reduction is the main challenge when sorafenib is combined with chemotherapy or endocrine therapy. Although randomized phase-II trials on sorafenib plus chemotherapy versus chemotherapy alone have shown potential benefits in progression-free survival, preliminary results from a phase-III study in combination with capecitabine are negative. The definitive results of this trial and results from other ongoing phase-II trials will determine further developments of sorafenib in breast cancer. Although these additional data could help determine the most appropriate dose, drug combination and patient settings, a confirmation of the preliminary negative results reported in the phase-III trial are likely to discourage further use of sorafenib in breast cancer, given its non-negligible toxicity, lack of predicting markers, and the number of more promising drugs for breast cancer.
Collapse
Affiliation(s)
- Giuseppe Bronte
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Daniele Andreis
- b Unit of Biostatistics and Clinical Trials , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Sara Bravaccini
- c Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Roberta Maltoni
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Lorenzo Cecconetto
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Alessio Schirone
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Alberto Farolfi
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Anna Fedeli
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Patrizia Serra
- b Unit of Biostatistics and Clinical Trials , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Caterina Donati
- d Oncology Pharmacy , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Dino Amadori
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| | - Andrea Rocca
- a Department of Medical Oncology , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS , Meldola , Italy
| |
Collapse
|
68
|
Cheng CW, Leong KW, Tse E. Understanding the role of PIN1 in hepatocellular carcinoma. World J Gastroenterol 2016; 22:9921-9932. [PMID: 28018099 PMCID: PMC5143759 DOI: 10.3748/wjg.v22.i45.9921] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/26/2016] [Accepted: 10/30/2016] [Indexed: 02/06/2023] Open
Abstract
PIN1 is a peptidyl-prolyl cis/trans isomerase that binds and catalyses isomerization of the specific motif comprising a phosphorylated serine or threonine residue preceding a proline (pSer/Thr-Pro) in proteins. PIN1 can therefore induce conformational and functional changes of its interacting proteins that are regulated by proline-directed serine/threonine phosphorylation. Through this phosphorylation-dependent prolyl isomerization, PIN1 fine-tunes the functions of key phosphoproteins (e.g., cyclin D1, survivin, β-catenin and x-protein of hepatitis B virus) that are involved in the regulation of cell cycle progression, apoptosis, proliferation and oncogenic transformation. PIN1 has been found to be over-expressed in many cancers, including human hepatocellular carcinoma (HCC). It has been shown previously that overexpression of PIN1 contributes to the development of HCC in-vitro and in xenograft mouse model. In this review, we first discussed the aberrant transcription factor expression, miRNAs dysregulation, PIN1 gene promoter polymorphisms and phosphorylation of PIN1 as potential mechanisms underlying PIN1 overexpression in cancers. Furthermore, we also examined the role of PIN1 in HCC tumourigenesis by reviewing the interactions between PIN1 and various cellular and viral proteins that are involved in β-catenin, NOTCH, and PI3K/Akt/mTOR pathways, apoptosis, angiogenesis and epithelial-mesenchymal transition. Finally, the potential of PIN1 inhibitors as an anti-cancer therapy was explored and discussed.
Collapse
|
69
|
Rustighi A, Zannini A, Campaner E, Ciani Y, Piazza S, Del Sal G. PIN1 in breast development and cancer: a clinical perspective. Cell Death Differ 2016; 24:200-211. [PMID: 27834957 DOI: 10.1038/cdd.2016.122] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/19/2016] [Accepted: 09/20/2016] [Indexed: 12/12/2022] Open
Abstract
Mammary gland development, various stages of mammary tumorigenesis and breast cancer progression have the peptidyl-prolyl cis/trans isomerase PIN1 at their centerpiece, in virtue of the ability of this unique enzyme to fine-tune the dynamic crosstalk between multiple molecular pathways. PIN1 exerts its action by inducing conformational and functional changes on key cellular proteins, following proline-directed phosphorylation. Through this post-phosphorylation signal transduction mechanism, PIN1 controls the extent and direction of the cellular response to a variety of inputs, in physiology and disease. This review discusses PIN1's roles in normal mammary development and cancerous progression, as well as the clinical impact of targeting this enzyme in breast cancer patients.
Collapse
Affiliation(s)
- Alessandra Rustighi
- National Laboratory CIB (LNCIB), Area Science Park, Padriciano 99, Trieste 34149, Italy
| | - Alessandro Zannini
- National Laboratory CIB (LNCIB), Area Science Park, Padriciano 99, Trieste 34149, Italy.,Department of Life Sciences, University of Trieste, via Weiss 2, Trieste 34128, Italy
| | - Elena Campaner
- National Laboratory CIB (LNCIB), Area Science Park, Padriciano 99, Trieste 34149, Italy.,Department of Life Sciences, University of Trieste, via Weiss 2, Trieste 34128, Italy
| | - Yari Ciani
- National Laboratory CIB (LNCIB), Area Science Park, Padriciano 99, Trieste 34149, Italy
| | - Silvano Piazza
- National Laboratory CIB (LNCIB), Area Science Park, Padriciano 99, Trieste 34149, Italy.,Bioinformatics Core Facility, Centre for Integrative Biology, CIBIO, University of Trento, Via Sommarive 18, 38123, Povo, Trento, Italy
| | - Giannino Del Sal
- National Laboratory CIB (LNCIB), Area Science Park, Padriciano 99, Trieste 34149, Italy.,Department of Life Sciences, University of Trieste, via Weiss 2, Trieste 34128, Italy
| |
Collapse
|
70
|
Amsacrine-induced apoptosis of human leukemia U937 cells is mediated by the inhibition of AKT- and ERK-induced stabilization of MCL1. Apoptosis 2016; 22:406-420. [DOI: 10.1007/s10495-016-1307-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
71
|
Kawakami H, Huang S, Pal K, Dutta SK, Mukhopadhyay D, Sinicrope FA. Mutant BRAF Upregulates MCL-1 to Confer Apoptosis Resistance that Is Reversed by MCL-1 Antagonism and Cobimetinib in Colorectal Cancer. Mol Cancer Ther 2016; 15:3015-3027. [PMID: 27765849 DOI: 10.1158/1535-7163.mct-16-0017] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 08/26/2016] [Accepted: 09/27/2016] [Indexed: 01/19/2023]
Abstract
Oncogenic BRAFV600E mutations activate MAPK signaling and are associated with treatment resistance and poor prognosis in patients with colorectal cancer. In BRAFV600E-mutant colorectal cancers, treatment failure may be related to BRAFV600E-mediated apoptosis resistance that occurs by an as yet undefined mechanism. We found that BRAFV600E can upregulate anti-apoptotic MCL-1 in a gene dose-dependent manner using colorectal cancer cell lines isogenic for BRAF BRAFV600E-induced MCL-1 upregulation was confirmed by ectopic BRAFV600E expression that activated MEK/ERK signaling to phosphorylate (MCL-1Thr163) and stabilize MCL-1. Upregulation of MCL-1 was mediated by MEK/ERK shown by the ability of ERK siRNA to suppress MCL-1. Stabilization of MCL-1 by phosphorylation was shown by a phosphorylation-mimicking mutant and an unphosphorylated MCL-1 mutant that decreased or increased MCL-1 protein turnover, respectively. MEK/ERK inhibition by cobimetinib suppressed MCL-1 expression/phosphorylation and induced proapoptotic BIM to a greater extent than did vemurafenib in BRAFV600E cell lines. MCL-1 knockdown versus control shRNA significantly enhanced cobimetinib-induced apoptosis in vitro and in HT29 colon cancer xenografts. The small-molecule MCL-1 inhibitor, A-1210477, also enhanced cobimetinib-induced apoptosis in vitro that was due to disruption of the interaction of MCL-1 with proapoptotic BAK and BIM. Knockdown of BIM attenuated BAX, but not BAK, activation by cobimetinib plus A-1210477. In summary, BRAFV600E-mediated MEK/ERK activation can upregulate MCL-1 by phosphorylation/stabilization to confer apoptosis resistance that can be reversed by MCL-1 antagonism combined with cobimetinib, suggesting a novel therapeutic strategy against BRAFV600E-mutant CRCs. Mol Cancer Ther; 15(12); 3015-27. ©2016 AACR.
Collapse
Affiliation(s)
- Hisato Kawakami
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Shengbing Huang
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, Florida
| | | | - Frank A Sinicrope
- Departments of Medicine and Oncology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
72
|
Li RH, Zhang AM, Li S, Li TY, Wang LJ, Zhang HR, Shi JW, Liu XR, Chen Y, Chen YC, Wei TY, Gao Y, Li W, Tang HY, Tang MY. PAQR3 gene expression and its methylation level in colorectal cancer tissues. Oncol Lett 2016; 12:1773-1778. [PMID: 27588124 PMCID: PMC4998131 DOI: 10.3892/ol.2016.4843] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/11/2016] [Indexed: 01/21/2023] Open
Abstract
The aim of the present study was to investigate the PAQR3 gene expression and its methylation level in colorectal cancer tissues, as well as the association with colorectal cancer clinical data. In total, 54 cases of colorectal cancer tissue samples and normal adjacent tissue samples were collected between June, 2013 and July, 2014. RT-PCR and western blot analysis were used to detect the mRNA and protein levels of PAQR3 in colorectal samples, respectively. MSP was used to detect the methylation level of PAQR3 gene in colorectal samples, which was compared with colorectal data. The results showed that a decreased expression level of PAQR3 mRNA in colorectal cancer tissues and the expression reduction rate was 57.4% (31/54). Similarly, the expression level of PAQR3 protein was reduced in cancer tissues, and the reduction rate was 46.3% (25/54), while the protein expression reduction rate in cancer adjacent tissue was 5.6% (3/54), and the difference was statistically significant (P<0.05). Furthermore, the methylation rates of PAQR3 in cancer tissues and cancer adjacent tissues were 33.3% (18/54) and 5.6% (3/54), respectively. In addition, PAQR3 mRNA and protein levels in colorectal cancer tissues were associated with the differentiation degree, lymphatic metastasis and tumor infiltration depth. The methylation level of PAQR3 was associated with age, differentiated degree, lymphatic metastasis and tumor infiltration depth. In conclusion, the expression of PAQR3 mRNA and protein in colorectal cancer was reduced and methylation of PAQR3 occurred. Although the PAQR3 mRNA and protein levels were not associated with gender, age or the location of tumor, there was an association with differentiation degree, lymphatic metastasis and tumor infiltration depth. In addition, the methylation level of PAQR3 was not correlated with gender or tumor location, but was correlated with age, differentiation degree, lymphatic metastasis and tumor infiltration depth.
Collapse
Affiliation(s)
- Ri-Heng Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Ai-Min Zhang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Shuang Li
- Department of Blood Transfusion, Neimenggu Xinganleague People's Hospital, Wulanhaote, Inner Mongolia Autonomous Region 137400, P.R. China
| | - Tian-Yang Li
- Clinical Medical College of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Lian-Jing Wang
- Clinical Medical College of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Hao-Ran Zhang
- Clinical Medical College of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Jian-Wei Shi
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Xiao-Rui Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Yuan Chen
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Ya-Chao Chen
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Teng-Yao Wei
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Ying Gao
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Wei Li
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Hong-Ying Tang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| | - Mei-Yu Tang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| |
Collapse
|
73
|
The role of Pin1 in the development and treatment of cancer. Arch Pharm Res 2016; 39:1609-1620. [PMID: 27572155 DOI: 10.1007/s12272-016-0821-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 08/18/2016] [Indexed: 12/16/2022]
Abstract
Protein phosphorylation and post-phosphorylation events regulate many cellular signaling pathways. Peptidyl-prolyl isomerase (Pin1) is the only peptidyl-prolyl cis/trans isomerase that interacts with numerous oncogenic or tumor suppressive phosphorylated proteins, causes conformational changes in target proteins, and eventually regulates the activities of such proteins. These alterations in activity play a pivotal role in tumorigenesis. Since Pin1 is overexpressed and/or activated in various types of cancers, and the dysregulation of proline-directed phosphorylation contributes to tumorigenesis, Pin1 represents an attractive target for cancer therapy. This review will describe the role of Pin1 in cancer and the current status of Pin1 inhibitor development.
Collapse
|
74
|
Garg SM, Falamarzian A, Vakili MR, Aliabadi HM, Uludağ H, Lavasanifar A. Polymeric micelles for MCL-1 gene silencing in breast tumors following systemic administration. Nanomedicine (Lond) 2016; 11:2319-39. [PMID: 27527491 DOI: 10.2217/nnm-2016-0178] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
AIM To develop delivery systems for efficient siRNA delivery to breast cancer. METHODS Poly(ethylene oxide)-block-poly(ϵ-caprolactone-grafted-spermine) (PEO-b-P(CL-g-SP)) micelles were modified with cholesterol group in their core and with RGD4C peptide on their shell. Transfection efficiency of complexed MCL-1 siRNA in MDA-MB-435 was investigated, in vitro and in vivo following intratumoral and intravenous injection. RESULTS Cholesteryl modification of the core significantly increased the transfection efficiency of PEO-b-P(CL-g-SP)-complexed siRNA, in vitro, but not following intratumoral or intravenous administration, in vivo. Instead, RGD4C modification of the micellar shell enhanced transfection efficiency of complexed MCL-1 siRNA in tumor upon intravenous administration. CONCLUSION RGD4C-PEO-b-P(CL-g-SP) micelles, without or with cholesterol modification, can provide efficient delivery of siRNA to breast tumors following systemic administration.
Collapse
Affiliation(s)
- Shyam M Garg
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Arash Falamarzian
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Mohammad Reza Vakili
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Hamidreza M Aliabadi
- Department of Biomedical & Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA 92618, USA
| | - Hasan Uludağ
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.,Department of Chemical & Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, T6G 2V4, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy & Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2E1, Canada.,Department of Chemical & Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, T6G 2V4, Canada
| |
Collapse
|
75
|
Choudhary GS, Tat TT, Misra S, Hill BT, Smith MR, Almasan A, Mazumder S. Cyclin E/Cdk2-dependent phosphorylation of Mcl-1 determines its stability and cellular sensitivity to BH3 mimetics. Oncotarget 2016. [PMID: 26219338 PMCID: PMC4627281 DOI: 10.18632/oncotarget.4857] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cyclin E/Cdk2 kinase activity is frequently deregulated in human cancers, resulting in impaired apoptosis. Here, we show that cyclin E/Cdk2 phosphorylates and stabilizes the pro-survival Bcl-2 family protein Mcl-1, a key cell death resistance determinant to the small molecule Bcl-2 family inhibitors ABT-199 and ABT-737, mimetics of the Bcl-2 homology domain 3 (BH3). Cyclin E levels were elevated and there was increased association of cyclin E/Cdk2 with Mcl-1 in ABT-737-resistant compared to parental cells. Cyclin E depletion in various human tumor cell-lines and cyclin E-/- mouse embryo fibroblasts showed decreased levels of Mcl-1 protein, with no change in Mcl-1 mRNA levels. In the absence of cyclin E, Mcl-1 ubiquitination was enhanced, leading to decreased protein stability. Studies with Mcl-1 phosphorylation mutants show that cyclin E/Cdk2-dependent phosphorylation of Mcl-1 residues on its PEST domain resulted in increased Mcl-1 stability (Thr92, and Thr163) and Bim binding (Ser64). Cyclin E knock-down restored ABT-737 sensitivity to acquired and inherently resistant Mcl-1-dependent tumor cells. CDK inhibition by dinaciclib resulted in Bim release from Mcl-1 in ABT-737-resistant cells. Dinaciclib in combination with ABT-737 and ABT-199 resulted in robust synergistic cell death in leukemic cells and primary chronic lymphocytic leukemia patient samples. Collectively, our findings identify a novel mechanism of cyclin E-mediated Mcl-1 regulation that provides a rationale for clinical use of Bcl-2 family and Cdk inhibitors for Mcl-1-dependent tumors.
Collapse
Affiliation(s)
- Gaurav S Choudhary
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Trinh T Tat
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Saurav Misra
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brian T Hill
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mitchell R Smith
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alexandru Almasan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Suparna Mazumder
- Department of Immunology, Lerner Research Institute, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
76
|
Kim G, Kim JY, Choi HS. Peptidyl-Prolyl cis/trans Isomerase NIMA-Interacting 1 as a Therapeutic Target in Hepatocellular Carcinoma. Biol Pharm Bull 2016; 38:975-9. [PMID: 26133706 DOI: 10.1248/bpb.b15-00245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Phosphorylation of proteins on serine or threonine residues preceding proline is a pivotal signaling mechanism regulating cell proliferation. The recent identification and characterization of the enzyme peptidyl-prolyl cis/trans isomerase never in mitosis A (NIMA)-interacting 1 (PIN1) has led to the discovery of a new mechanism regulating phosphorylation in cell signaling. PIN1 specifically binds phosphorylated serine or threonine residues immediately preceding proline (pSer/Thr-Pro) and then regulates protein functions, including catalytic activity, phosphorylation status, protein interactions, subcellular location, and protein stability, by promoting cis/trans isomerization of the peptide bond. Recent results have indicated that such conformational changes following phosphorylation represent a novel signaling mechanism in the regulation of many cellular functions. Understanding this mechanism also provides new insight into the pathogenesis and treatment of human hepatocellular carcinoma. A better understanding of the role of PIN1 in the pathogenesis of hepatocellular carcinoma may lead to the identification of molecular targets for prevention and therapeutic intervention.
Collapse
Affiliation(s)
- Garam Kim
- College of Pharmacy, Chosun University
| | | | | |
Collapse
|
77
|
Sakuma Y, Nishikiori H, Hirai S, Yamaguchi M, Yamada G, Watanabe A, Hasegawa T, Kojima T, Niki T, Takahashi H. Prolyl isomerase Pin1 promotes survival in EGFR-mutant lung adenocarcinoma cells with an epithelial-mesenchymal transition phenotype. J Transl Med 2016; 96:391-8. [PMID: 26752745 DOI: 10.1038/labinvest.2015.155] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/09/2015] [Accepted: 11/13/2015] [Indexed: 12/31/2022] Open
Abstract
The secondary epidermal growth factor receptor (EGFR) T790M mutation is the most prominent mechanism that confers resistance to first- or second-generation EGFR tyrosine kinase inhibitors (TKIs) in lung cancer treatment. Although third-generation EGFR TKIs can suppress the kinase activity of T790M-positive EGFR, they still cannot eradicate EGFR-mutated cancer cells. We previously reported that a subpopulation of EGFR-mutant lung adenocarcinomas depends on enhanced autophagy, instead of EGFR, for survival, and in this study we explore another mechanism that contributes to TKI resistance. We demonstrate here that an EGFR-mutant lung adenocarcinoma cell line, H1975 (L858R+T790M), has a subset of cells that exhibits an epithelial-mesenchymal transition (EMT) phenotype and can thrive in the presence of third-generation EGFR TKIs. These cells depend on not only autophagy but also on the isomerase Pin1 for survival in vitro, unlike their parental cells. The Pin1 protein was expressed in an EGFR-mutant lung cancer tissue that has undergone partial EMT and acquired resistance to EGFR TKIs, but not its primary tumor. These findings suggest that inhibition of Pin1 activity can be a novel strategy in lung cancer treatment.
Collapse
Affiliation(s)
- Yuji Sakuma
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirotaka Nishikiori
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Sachie Hirai
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Miki Yamaguchi
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Gen Yamada
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Watanabe
- Department of Thoracic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tadashi Hasegawa
- Department of Surgical Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshiro Niki
- Department of Pathology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Hiroki Takahashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
78
|
Pramchu-em C, Meksawan K, Chanvorachote P. Zinc Sensitizes Lung Cancer Cells to Anoikis through Down-Regulation of Akt and Caveolin-1. Nutr Cancer 2016; 68:312-9. [DOI: 10.1080/01635581.2016.1142582] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
79
|
Pin1 modulates chemo-resistance by up-regulating FoxM1 and the involvements of Wnt/β-catenin signaling pathway in cervical cancer. Mol Cell Biochem 2016; 413:179-87. [DOI: 10.1007/s11010-015-2651-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/23/2015] [Indexed: 01/17/2023]
|
80
|
Kim SY, Park SE, Shim SM, Park S, Kim KK, Jeong SY, Choi EK, Hwang JJ, Jin DH, Chung CD, Kim I. Bay 61-3606 Sensitizes TRAIL-Induced Apoptosis by Downregulating Mcl-1 in Breast Cancer Cells. PLoS One 2015; 10:e0146073. [PMID: 26720004 PMCID: PMC4697837 DOI: 10.1371/journal.pone.0146073] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/11/2015] [Indexed: 11/21/2022] Open
Abstract
Breast cancer cells generally develop resistance to TNF-Related Apoptosis-Inducing Ligand (TRAIL) and, therefore, assistance from sensitizers is required. In our study, we have demonstrated that Spleen tyrosine kinase (Syk) inhibitor Bay 61–3606 was identified as a TRAIL sensitizer. Amplification of TRAIL-induced apoptosis by Bay 61–3606 was accompanied by the strong activation of Bak, caspases, and DNA fragmentation. In mechanism of action, Bay 61–3606 sensitized cells to TRAIL via two mechanisms regulating myeloid cell leukemia sequence-1 (Mcl-1). First, Bay 61–3606 triggered ubiquitin-dependent degradation of Mcl-1 by regulating Mcl-1 phosphorylation. Second, Bay 61–3606 downregulates Mcl-1 expression at the transcription level. In this context, Bay 61–3606 acted as an inhibitor of Cyclin-Dependent Kinase (CDK) 9 rather than Syk. In summary, Bay 61–3606 downregulates Mcl-1 expression in breast cancer cells and sensitizes cancer cells to TRAIL-mediated apoptosis.
Collapse
Affiliation(s)
- So-Young Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
| | - Sang Eun Park
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
| | - Sang-Mi Shim
- Department of Biomedical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sojung Park
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
| | - Kyung Kon Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seong-Yun Jeong
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Institute for Innovative Cancer Research, ASAN Medical Center, Seoul, Republic of Korea
| | - Eun Kyung Choi
- Department of Radiation Oncology, ASAN Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Institute for Innovative Cancer Research, ASAN Medical Center, Seoul, Republic of Korea
| | - Jung Jin Hwang
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Institute for Innovative Cancer Research, ASAN Medical Center, Seoul, Republic of Korea
| | - Dong-Hoon Jin
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Institute for Innovative Cancer Research, ASAN Medical Center, Seoul, Republic of Korea
| | | | - Inki Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
81
|
Choe MH, Min JW, Jeon HB, Cho DH, Oh JS, Lee HG, Hwang SG, An S, Han YH, Kim JS. ERp57 modulates STAT3 activity in radioresistant laryngeal cancer cells and serves as a prognostic marker for laryngeal cancer. Oncotarget 2015; 6:2654-66. [PMID: 25605256 PMCID: PMC4413608 DOI: 10.18632/oncotarget.3042] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/12/2014] [Indexed: 01/01/2023] Open
Abstract
Although targeting radioresistant tumor cells is essential for enhancing the efficacy of radiotherapy, the signals activated in resistant tumors are still unclear. This study shows that ERp57 contributes to radioresistance of laryngeal cancer by activating STAT3. Increased ERp57 was associated with the radioresistant phenotype of laryngeal cancer cells. Interestingly, increased interaction between ERp57 and STAT3 was observed in radioresistant cells, compared to the control cells. This physical complex is required for the activation of STAT3 in the radioresistant cells. Among STAT3-regulatory genes, Mcl-1 was predominantly regulated by ERp57. Inhibition of STAT3 activity with a chemical inhibitor or siRNA-mediated depletion of Mcl-1 sensitized radioresistant cells to irradiation, suggesting that the ERp57-STAT3-Mcl-1 axis regulates radioresistance of laryngeal cancer cells. Furthermore, we observed a positive correlation between ERp57 and phosphorylated STAT3 or Mcl-1 and in vivo interactions between ERp57 and STAT3 in human laryngeal cancer. Importantly, we also found that increased ERp57-STAT3 complex was associated with poor prognosis in human laryngeal cancer, indicating the prognostic role of ERp57-STAT3 regulation. Overall, our data suggest that ERp57-STAT3 regulation functions in radioresistance of laryngeal cancer, and targeting the ERp57-STAT3 pathway might be important for enhancing the efficacy of radiotherapy in human laryngeal cancer.
Collapse
Affiliation(s)
- Min Ho Choe
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Joong Won Min
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hong Bae Jeon
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul, Korea
| | - Dong-Hyung Cho
- Graduate School of East-West Medical Science, Kyung Hee University, Suwon, Korea
| | - Jeong Su Oh
- Department of Genetic Engineering, Sungkyunkwan University, Suwon, Korea
| | - Hyun Gyu Lee
- Department of Microbiology and Immunology, College of Medicine, Yonsei University, Seoul, Korea
| | - Sang-Gu Hwang
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Sungkwan An
- Molecular-Targeted Drug Research Center and Korea Institute for Skin and Clinical Sciences, Konkuk University, Seoul, Korea
| | - Young-Hoon Han
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Jae-Sung Kim
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| |
Collapse
|
82
|
Dikovskaya D, Cole JJ, Mason SM, Nixon C, Karim SA, McGarry L, Clark W, Hewitt RN, Sammons MA, Zhu J, Athineos D, Leach JDG, Marchesi F, van Tuyn J, Tait SW, Brock C, Morton JP, Wu H, Berger SL, Blyth K, Adams PD. Mitotic Stress Is an Integral Part of the Oncogene-Induced Senescence Program that Promotes Multinucleation and Cell Cycle Arrest. Cell Rep 2015; 12:1483-96. [PMID: 26299965 PMCID: PMC4562906 DOI: 10.1016/j.celrep.2015.07.055] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 06/22/2015] [Accepted: 07/27/2015] [Indexed: 12/18/2022] Open
Abstract
Oncogene-induced senescence (OIS) is a tumor suppression mechanism that blocks cell proliferation in response to oncogenic signaling. OIS is frequently accompanied by multinucleation; however, the origin of this is unknown. Here, we show that multinucleate OIS cells originate mostly from failed mitosis. Prior to senescence, mutant H-RasV12 activation in primary human fibroblasts compromised mitosis, concordant with abnormal expression of mitotic genes functionally linked to the observed mitotic spindle and chromatin defects. Simultaneously, H-RasV12 activation enhanced survival of cells with damaged mitoses, culminating in extended mitotic arrest and aberrant exit from mitosis via mitotic slippage. ERK-dependent transcriptional upregulation of Mcl1 was, at least in part, responsible for enhanced survival and slippage of cells with mitotic defects. Importantly, mitotic slippage and oncogene signaling cooperatively induced senescence and key senescence effectors p21 and p16. In summary, activated Ras coordinately triggers mitotic disruption and enhanced cell survival to promote formation of multinucleate senescent cells.
Collapse
Affiliation(s)
- Dina Dikovskaya
- Institute of Cancer Sciences, CR-UK Beatson Laboratories, University of Glasgow, Glasgow G61 1BD, UK.
| | - John J Cole
- Institute of Cancer Sciences, CR-UK Beatson Laboratories, University of Glasgow, Glasgow G61 1BD, UK
| | - Susan M Mason
- Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Colin Nixon
- Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Saadia A Karim
- Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Lynn McGarry
- Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - William Clark
- Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Rachael N Hewitt
- Institute of Cancer Sciences, CR-UK Beatson Laboratories, University of Glasgow, Glasgow G61 1BD, UK
| | - Morgan A Sammons
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | - Jiajun Zhu
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | | | - Joshua D G Leach
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Francesco Marchesi
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - John van Tuyn
- Institute of Cancer Sciences, CR-UK Beatson Laboratories, University of Glasgow, Glasgow G61 1BD, UK
| | - Stephen W Tait
- Institute of Cancer Sciences, CR-UK Beatson Laboratories, University of Glasgow, Glasgow G61 1BD, UK
| | - Claire Brock
- Institute of Cancer Sciences, CR-UK Beatson Laboratories, University of Glasgow, Glasgow G61 1BD, UK
| | | | - Hong Wu
- Fox Chase Cancer Center, Philadelphia, PA 19111-2497, USA
| | - Shelley L Berger
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6058, USA
| | - Karen Blyth
- Beatson Institute for Cancer Research, Glasgow G61 1BD, UK
| | - Peter D Adams
- Institute of Cancer Sciences, CR-UK Beatson Laboratories, University of Glasgow, Glasgow G61 1BD, UK.
| |
Collapse
|
83
|
Hornstein T, Lehmann S, Philipp D, Detmer S, Hoffmann M, Peter C, Wesselborg S, Unfried K, Windolf J, Flohé S, Paunel-Görgülü A. Staurosporine resistance in inflammatory neutrophils is associated with the inhibition of caspase- and proteasome-mediated Mcl-1 degradation. J Leukoc Biol 2015; 99:163-74. [PMID: 26310832 DOI: 10.1189/jlb.3a1114-537rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 07/28/2015] [Indexed: 11/24/2022] Open
Abstract
Apoptosis resistance in activated neutrophils is known to be associated with collateral damage of surrounding tissue, as well as immune and organ dysfunction. Thus, the safe removal of neutrophils by apoptosis induction represents a prerequisite for the resolution of inflammation. Here, we report that intrinsic apoptosis resistance in human neutrophils, isolated from severely injured patients, is based on enhanced stabilization of antiapoptotic myeloid cell leukemia 1 and subsequent impairment of downstream apoptotic pathways. Whereas extrinsic apoptosis induction by the activation of Fas death receptor on inflammatory neutrophils was accompanied by caspase- and proteasome-mediated myeloid cell leukemia 1 degradation, intrinsic apoptosis induction by staurosporine led to a significant stabilization of myeloid cell leukemia 1 protein, which impeded on truncated forms of B cell lymphoma 2-associated X protein and B cell lymphoma 2 homology domain 3-interacting domain death translocation and subsequent cytochrome c release from the mitochondria. We show further that profound inhibition of myeloid cell leukemia 1 degradation is based on the inhibition of caspases and sustained activation of kinases involved in cell survival, such as Akt. Accordingly, impeded myeloid cell leukemia 1 phosphorylation on Ser159 by glycogen synthase kinase 3 and protein ubiquitination has been demonstrated. Inhibition of myeloid cell leukemia 1 activity markedly increased sensitivity to staurosporine-induced cell death. Altogether, these results provide new insights into the mechanisms underlying myeloid cell leukemia 1-mediated apoptosis resistance to staurosporine under inflammatory situations and should be considered for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Tamara Hornstein
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sarah Lehmann
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Denise Philipp
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Susanne Detmer
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Michèle Hoffmann
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Christoph Peter
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sebastian Wesselborg
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Klaus Unfried
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joachim Windolf
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sascha Flohé
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Adnana Paunel-Görgülü
- *IUF Leibniz Research Institute of Environmental Medicine, Düsseldorf, Germany; Department of Trauma and Hand Surgery, University Hospital Düsseldorf, Düsseldorf, Germany; and Department of Urology and Institute of Molecular Medicine, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
84
|
Abstract
Dichloroacetate (DCA) is a metabolic reprogramming agent that reverses the Warburg effect, causing cancer cells to couple glycolysis to oxidative phosphorylation. This has been shown to induce apoptosis and reduce the growth of various types of cancer but not normal cells. Colorectal cancer cells HCT116, HCT116 p53(-/-), and HCT116 Bax(-/-), were treated with DCA in vitro. Response to treatment was determined by measuring PDH phosphorylation, apoptosis, proliferation, and cell cycle. Molecular changes associated with these responses were determined using western immunoblotting and quantitative PCR. Treatment with 20 mM DCA did not increase apoptosis, despite decreasing levels of anti-apoptotic protein Mcl-1 after 6 h, in any of the cell lines observed. Mcl-1 expression was stabilized with MG-132, an inhibitor of proteasomal degradation. A decrease in Mcl-1 correlated with a decrease in proliferation, both of which showed dose-dependence in DCA treated cells. Cells showed nuclear localization of Mcl-1, however cell cycle was unaffected by DCA treatment. These data suggest that a reduction in the prosurvival Bcl-2 family member Mcl-1 due to increased proteasomal degradation is correlated with the ability of DCA to reduce proliferation of HCT116 human colorectal cancer cells without causing apoptosis.
Collapse
|
85
|
Lee HE, Nam JS, Shin JA, Hong IS, Yang IH, You MJ, Cho SD. Convallaria keiskei as a novel therapeutic alternative for salivary gland cancer treatment by targeting myeloid cell leukemia-1. Head Neck 2015; 38 Suppl 1:E761-70. [PMID: 25914292 DOI: 10.1002/hed.24096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Various chemotherapeutic agents have been used largely for the treatment of salivary gland cancer. However, results are disappointing, and these agents can cause some serious side effects. Therefore, recent studies have focused on the possible roles of natural products to overcome these limitations. METHODS Salivary gland cancer cells treated with or without Convallaria keiskei (MECK) for 24 hours. Apoptotic changes were evaluated by live/dead assay, immunoblotting, and expression levels of caspase-3 and B-cell lymphoma-2 family member. RESULTS MECK significantly inhibited salivary gland cancer growth. At the molecular level, MECK dramatically reduced myeloid cell leukemia-1 (Mcl-1) in a translation-dependent manner and thereby induced apoptosis through Bax/Bid. Furthermore, we found that Mcl-1 could be a potential therapeutic target of MECK-induced apoptosis and its stability is regulated by extracellular signal-regulated kinases 1/2 (ERK1/2) signaling CONCLUSION MECK can be used as a safe and efficient therapeutic alternative for the treatment of salivary gland cancer. © 2015 Wiley Periodicals, Inc. Head Neck 38: E761-E770, 2016.
Collapse
Affiliation(s)
- Haeng-Eun Lee
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| | - Jeong-Seok Nam
- Laboratory of Tumor Suppressor, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| | - In-Sun Hong
- Laboratory of Tumor Suppressor, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, South Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, South Korea
| | - In-Hyoung Yang
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| | - Myung-Jo You
- Laboratory of Veterinary Parasitology, College of Veterinary Medicine and Bio-Safety Research Centre, Chonbuk National University, Jeonju, Republic of Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Institute of Biodegradable Material, Institute of Oral Bioscience, Brain Korea 21 Project, Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
86
|
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: 12.4] [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
|
87
|
Pin1: Intimate involvement with the regulatory protein kinase networks in the global phosphorylation landscape. Biochim Biophys Acta Gen Subj 2015; 1850:2077-86. [PMID: 25766872 DOI: 10.1016/j.bbagen.2015.02.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/25/2015] [Accepted: 02/27/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Protein phosphorylation is a universal regulatory mechanism that involves an extensive network of protein kinases. The discovery of the phosphorylation-dependent peptidyl-prolyl isomerase Pin1 added an additional layer of complexity to these regulatory networks. SCOPE OF REVIEW We have evaluated interactions between Pin1 and the regulatory kinome and proline-dependent phosphoproteome taking into consideration findings from targeted studies as well as data that has emerged from systematic phosphoproteomic workflows and from curated protein interaction databases. MAJOR CONCLUSIONS The relationship between Pin1 and the regulatory protein kinase networks is not restricted simply to the recognition of proteins that are substrates for proline-directed kinases. In this respect, Pin1 itself is phosphorylated in cells by protein kinases that modulate its functional properties. Furthermore, the phosphorylation-dependent targets of Pin1 include a number of protein kinases as well as other enzymes such as phosphatases and regulatory subunits of kinases that modulate the actions of protein kinases. GENERAL SIGNIFICANCE As a result of its interactions with numerous protein kinases and their substrates, as well as itself being a target for phosphorylation, Pin1 has an intricate relationship with the regulatory protein kinase and phosphoproteomic networks that orchestrate complex cellular processes and respond to environmental cues. This article is part of a Special Issue entitled Proline-directed Foldases: Cell Signaling Catalysts and Drug Targets.
Collapse
|
88
|
Williams MM, Cook RS. Bcl-2 family proteins in breast development and cancer: could Mcl-1 targeting overcome therapeutic resistance? Oncotarget 2015; 6:3519-30. [PMID: 25784482 PMCID: PMC4414133 DOI: 10.18632/oncotarget.2792] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/20/2014] [Indexed: 02/07/2023] Open
Abstract
Apoptosis, cell death executed by caspases, is essential to normal breast development and homeostasis. Pro-apoptotic and anti-apoptotic signals are tightly regulated in normal breast epithelial cells. Dysregulation of this balance is required for breast tumorigenesis and increases acquired resistance to treatments, including molecularly targeted therapies, radiation and chemotherapies. The pro-apoptotic or anti-apoptotic Bcl-2 family members interact with each other to maintain mitochondrial integrity and regulate cellular commitment to apoptosis. Among the anti-apoptotic Bcl-2 family members, Mcl-1 is uniquely regulated by numerous oncogenic signaling pathways. This review will focus on the role of Bcl-2 family proteins in normal breast development, breast tumorigenesis and acquired resistance to breast cancer treatment strategies, while highlighting Mcl-1 as a promising target to improve breast cancer tumor cell killing.
Collapse
Affiliation(s)
- Michelle M. Williams
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville TN 27232, USA
| | - Rebecca S. Cook
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville TN 27232, USA
- Department of Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
| |
Collapse
|
89
|
Abstract
FBW7 (F-box and WD repeat domain-containing 7) or Fbxw7 is a tumor suppressor, which promotes the ubiquitination and subsequent degradation of numerous oncoproteins including Mcl-1, Cyclin E, Notch, c- Jun, and c-Myc. In turn, FBW7 is regulated by multiple upstream factors including p53, C/EBP-δ, EBP2, Pin1, Hes-5 and Numb4 as well as by microRNAs such as miR-223, miR-27a, miR-25, and miR-129-5p. Given that the Fbw7 tumor suppressor is frequently inactivated or deleted in various human cancers, targeting FBW7 regulators is a promising anti-cancer therapeutic strategy.
Collapse
|
90
|
Wang JZ, Liu BG, Zhang Y. Pin1-based diagnostic and therapeutic strategies for breast cancer. Pharmacol Res 2014; 93:28-35. [PMID: 25553719 DOI: 10.1016/j.phrs.2014.12.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 01/12/2023]
Abstract
Pin1 is the only known cis-to-trans isomerase that recognizes the phosphorylated pThr/pSer-Pro motifs in many signaling molecules, playing unique roles in the pathogenesis of breast cancer. First, Pin1 is prevalently over-expressed in kinds of breast cancer cell lines and tissues, such as MDA-MB-231 cell, MCF-7 cell, Her2+, ERα+, and basal-like breast cancer subtypes. Second, Pin1 amplifies many oncogenic signaling pathways, inhibits multiple tumor suppressors, promotes the angiogenesis and metastasis of breast cancer cells, and enhances the resistance of breast cancer cells to anti-tumor medicines. Third, inhibiting Pin1 blocks most of these detrimental effects in a great number of breast cancer cell lines. These findings suggest Pin1 as a promising diagnostic biomarker as well as an efficient therapeutic target for breast cancer. It is strongly expected that a Pin1-positive subtype of breast cancers should be extremely concerned and that the therapeutic efficacy of Pin1 inhibitors on breast cancer patients should be evaluated as soon as possible. Nonetheless, Pin1-based therapeutic strategies for breast cancer still deserve some debates. Hence, we give the predictions of several important issues, such as application precondition, side effects, and personalized medication, when Pin1 inhibitors are used in the breast cancer therapy. These proposals are meaningful for the further development of Pin1-based diagnostic and therapeutic strategies in order to conquer breast cancer.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- Department of Medical Technology, Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan 056002, PR China.
| | - Bao-Guo Liu
- Department of Medical Technology, Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan 056002, PR China
| | - Yong Zhang
- Department of Medical Technology, Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan 056002, PR China
| |
Collapse
|
91
|
Sorrentino G, Comel A, Mantovani F, Del Sal G. Regulation of mitochondrial apoptosis by Pin1 in cancer and neurodegeneration. Mitochondrion 2014; 19 Pt A:88-96. [PMID: 25132079 DOI: 10.1016/j.mito.2014.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 11/15/2022]
Abstract
Mitochondria are sensitive and efficient organelles that regulate essential biological processes including: energy metabolism, decoding and transduction of intracellular signals, and balance between cell death and survival. Of note, dysfunctions in mitochondrial physiology are a general hallmark of cancer cells, leading to transformation-related features such as altered cellular metabolism, survival under stress conditions and reduced apoptotic response to chemotherapy. Mitochondrial apoptosis is a finely regulated process that derives from activation of multiple signaling networks. A crucial biochemical requirement for transducing pro-apoptotic stimuli is represented by kinase-dependent phosphorylation cascades. In this context a pivotal role is played by the prolyl-isomerase Pin1, which translates Ser/Thr-Pro phosphorylation into conformational changes able to modify the activities of its substrates. In this review we will discuss the impact of Pin1 in regulating various aspects of apoptosis in different biological contexts with particular emphasis on cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Giovanni Sorrentino
- Laboratorio Nazionale CIB Area Science Park, Trieste Italy; Dipartimento di Scienze della Vita Università degli Studi di Trieste- Trieste Italy
| | - Anna Comel
- Laboratorio Nazionale CIB Area Science Park, Trieste Italy; Dipartimento di Scienze della Vita Università degli Studi di Trieste- Trieste Italy
| | - Fiamma Mantovani
- Laboratorio Nazionale CIB Area Science Park, Trieste Italy; Dipartimento di Scienze della Vita Università degli Studi di Trieste- Trieste Italy
| | - Giannino Del Sal
- Laboratorio Nazionale CIB Area Science Park, Trieste Italy; Dipartimento di Scienze della Vita Università degli Studi di Trieste- Trieste Italy.
| |
Collapse
|
92
|
Abstract
Proline-directed phosphorylation is a posttranslational modification that is instrumental in regulating signaling from the plasma membrane to the nucleus, and its dysregulation contributes to cancer development. Protein interacting with never in mitosis A1 (Pin1), which is overexpressed in many types of cancer, isomerizes specific phosphorylated Ser/Thr-Pro bonds in many substrate proteins, including glycolytic enzyme, protein kinases, protein phosphatases, methyltransferase, lipid kinase, ubiquitin E3 ligase, DNA endonuclease, RNA polymerase, and transcription activators and regulators. This Pin1-mediated isomerization alters the structures and activities of these proteins, thereby regulating cell metabolism, cell mobility, cell cycle progression, cell proliferation, cell survival, apoptosis and tumor development.
Collapse
Affiliation(s)
- Zhimin Lu
- 1] Brain Tumor Center and Department of Neuro-Oncology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA [2] Department of Molecular and Cellular Oncology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA [3] Cancer Biology Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
93
|
Yu HJ, Shin JA, Jung JY, Nam JS, Hong IS, Cho NP, Cho SD. Inhibition of myeloid cell leukemia-1: Association with sorafenib-induced apoptosis in human mucoepidermoid carcinoma cells and tumor xenograft. Head Neck 2014; 37:1326-35. [PMID: 25043125 DOI: 10.1002/hed.23749] [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: 11/11/2013] [Revised: 03/24/2014] [Accepted: 05/07/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The purpose of our study was to investigate the anticancer effect of sorafenib on mucoepidermoid carcinoma (MEC) and find its new molecular mechanism. METHODS The apoptotic effects of sorafenib were performed using MTS assay, diamidino-phenylindole (DAPI) staining, Western blotting, reverse transcription-polymerase chain reaction (RT-PCR), siRNA, and xenograft. RESULTS Sorafenib had apoptotic effects on MC-3 and YD15 cells and decreased myeloid cell leukemia-1 (Mcl-1) through proteasome-dependent protein degradation and the inhibition of protein synthesis. Sorafenib significantly affected truncated bid (t-Bid) and siMcl-1 resulting in the upregulation of t-Bid to induce apoptosis. Signal transducer and activator of transcription 3 (STAT3) phosphorylation was also blocked by sorafenib and a potent STAT3 inhibitor, cryptotanshinone clearly induced poly ADP-ribose polymerase (PARP) cleavage by inhibiting Mcl-1 and increasing t-Bid. Finally, administration of sorafenib significantly suppressed tumor growth and induced apoptosis in tumor xenograft model in association with downregulation of Mcl-1 without any side effects. CONCLUSION Taken together, these findings suggest that sorafenib can be a good anticancer drug candidate for the treatment of MEC.
Collapse
Affiliation(s)
- Hyun-Ju Yu
- Department of Oral Pathology, School of Dentistry, and Institute of Oral Bioscience, Chonbuk National University, Jeonju, Republic of Korea
| | - Ji-Ae Shin
- Department of Oral Pathology, School of Dentistry, and Institute of Oral Bioscience, Chonbuk National University, Jeonju, Republic of Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Republic of Korea
| | - Jeong-Seok Nam
- Lee Gil Ya Cancer and Diabetes Institute, Inchon, Republic of Korea
| | - In-Sun Hong
- Department of Molecular Medicine, Gachon University, Incheon, Republic of Korea
| | - Nam-Pyo Cho
- Department of Oral Pathology, School of Dentistry, and Institute of Oral Bioscience, Chonbuk National University, Jeonju, Republic of Korea
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry, and Institute of Oral Bioscience, Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
94
|
Abstract
Members of the Bcl-2 protein family are frequently deregulated in tumors as they critically control cell death induction in mammalian cells. Alterations of these proteins may cause resistance to chemotherapy-induced cell death and immune responses. By serendipity we cloned a variant of the anti-apoptotic Bcl2-family member Myeloid cell leukemia-1 (Mcl1) from human neuroblastoma and leukemia cells. This Mcl1L variant lacks a 45 bp sequence that codes for 15 highly conserved amino acids ranging from Gly158 to Asp172. This region is part of the so called PEST-sequence of Mcl1L and contains two phosphorylation sites (Ser159 and Thr163) that regulate Mcl1L stability. A caspase 3/caspase 8 cleavage site at Asp157 which has been reported to be critical for death-receptor-induced apoptosis and for the conversion of Mcl1L into a pro-apoptotic protein is also missing in this novel variant. Importantly, Mcl1LdelGly158-Asp172 bound significantly more pro-apoptotic Bim compared to Mcl1L and showed increased anti-proliferative and anti-apoptotic activity compared to Mcl1L during death receptor-induced cell death. This suggests that this novel Mcl1L variant efficiently protects tumor cells against extrinsic death signalling and therefore may provide a survival advantage for highly aggressive tumors.
Collapse
|
95
|
Mcl-1 ubiquitination: unique regulation of an essential survival protein. Cells 2014; 3:418-37. [PMID: 24814761 PMCID: PMC4092850 DOI: 10.3390/cells3020418] [Citation(s) in RCA: 149] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 04/16/2014] [Accepted: 04/29/2014] [Indexed: 01/26/2023] Open
Abstract
Mcl-1 is an anti-apoptotic protein of the Bcl-2 family that is essential for the survival of multiple cell lineages and that is highly amplified in human cancer. Under physiological conditions, Mcl-1 expression is tightly regulated at multiple levels, involving transcriptional, post-transcriptional and post-translational processes. Ubiquitination of Mcl-1, that targets it for proteasomal degradation, allows for rapid elimination of the protein and triggering of cell death, in response to various cellular events. In the last decade, a number of studies have elucidated different pathways controlling Mcl-1 ubiquitination and degradation. Four different E3 ubiquitin-ligases (e.g., Mule, SCFβ-TrCP, SCFFbw7 and Trim17) and one deubiquitinase (e.g., USP9X), that respectively mediate and oppose Mcl-1 ubiquitination, have been formerly identified. The interaction between Mule and Mcl-1 can be modulated by other Bcl-2 family proteins, while recognition of Mcl-1 by the other E3 ubiquitin-ligases and deubiquitinase is influenced by phosphorylation of specific residues in Mcl-1. The protein kinases and E3 ubiquitin-ligases that are involved in the regulation of Mcl-1 stability vary depending on the cellular context, highlighting the complexity and pivotal role of Mcl-1 regulation. In this review, we attempt to recapitulate progress in understanding Mcl-1 regulation by the ubiquitin-proteasome system.
Collapse
|
96
|
Bu Q, Cui L, Li J, Du X, Zou W, Ding K, Pan J. SAHA and S116836, a novel tyrosine kinase inhibitor, synergistically induce apoptosis in imatinib-resistant chronic myelogenous leukemia cells. Cancer Biol Ther 2014; 15:951-62. [PMID: 24759597 DOI: 10.4161/cbt.28931] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Limited treatment options are available for chronic myelogenous leukemia (CML) patients who develop imatinib mesylate (IM) resistance. Here we proposed a novel combination regimen, a co-administration of S116836, a novel small molecule multi-targeted tyrosine kinase inhibitor that was synthesized by rational design, and histone deacetylases inhibitor (HDACi) suberoylanilide hydroxamic acid (SAHA), to overcome IM resistance in CML. S116836 at low concentrations used in the present study mildly downregulates auto-tyrosine phosphorylation of Bcr-Abl. SAHA, an FDA-approved HDACi drug, at 1 μM has modest anti-tumor activity in treating CML. However, we found a synergistic interaction between SAHA and S116836 in Bcr-Abl-positive CML cells that were sensitive or resistant to IM. Exposure of KBM5 and KBM5-T315I cells to minimal or non-toxic concentrations of SAHA and S116836 synergistically reduced cell viability and induced cell death. Co-treatment with SAHA and S116838 repressed the expressions of anti-apoptosis proteins, such as Mcl-1 and XIAP, but promoted Bim expression and mitochondrial damage. Of importance, treatment with both drugs significantly reduced cell viability of primary human CML cells, as compared with either agent alone. Taken together, our findings suggest that SAHA exerts synergistically with S116836 at a non-toxic concentration to promote apoptosis in the CML, including those resistant to imatinib or dasatinib.
Collapse
Affiliation(s)
- Qiangui Bu
- Department of Pathophysiology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou, PR China
| | - Lijing Cui
- Department of Pathophysiology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou, PR China
| | - Juan Li
- Department of Hematology; The First Affiliated Hospital; Sun Yat-sen University; Guangzhou, PR China
| | - Xin Du
- Department of Hematology; Guangdong Provincial People's Hospital; Guangzhou, PR China
| | - Waiyi Zou
- Department of Hematology; The First Affiliated Hospital; Sun Yat-sen University; Guangzhou, PR China
| | - Ke Ding
- Key Laboratory of Regenerative Biology and Institute of Chemical Biology; Guangzhou Institute of Biomedicine and Health; Chinese Academy of Sciences; Guangzhou, PR China
| | - Jingxuan Pan
- Department of Pathophysiology; Zhongshan School of Medicine; Sun Yat-sen University; Guangzhou, PR China; State Key Laboratory of Ophthalmology; Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou, PR China; Collaborative Innovation Center for Cancer Medicine; State Key Laboratory of Oncology in South China; Sun Yat-Sen University Cancer Center; Guangzhou, PR China
| |
Collapse
|
97
|
Yoshida A, Ookura M, Zokumasu K, Ueda T. Gö6976, a FLT3 kinase inhibitor, exerts potent cytotoxic activity against acute leukemia via inhibition of survivin and MCL-1. Biochem Pharmacol 2014; 90:16-24. [PMID: 24735609 DOI: 10.1016/j.bcp.2014.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/03/2014] [Accepted: 04/04/2014] [Indexed: 02/07/2023]
Abstract
Mutations of the FMS-like tyrosine kinase 3 (FLT3) have been reported in about a third of patients with acute myeloid leukemia (AML). The presence of FLT3 mutations confers a poor prognosis. Thus, pharmacological inhibitors of FLT3 are of therapeutic interest for AML. Gö6976 is an indolocarbazole with a similar structural backbone to staurosporine. In the present study, we demonstrated that Gö6976 displays a potent inhibitory activity against recombinant FLT3 using an in vitro kinase assay, with an IC50 value of 0.7nM. Gö6976 markedly inhibited the proliferation of human leukemia cells having FLT3-ITD such as MV4-11 and MOLM13. We also observed that Gö6976 showed minimal toxicity for human normal CD34(+) cells. Gö6976 suppressed the phosphorylation of FLT3 and downstream signaling molecules such as STAT3/5, Erk1/2, and Akt in MV4-11 and MOLM13 cells. Interestingly, induction of apoptosis by Gö6976 was associated with rapid and pronounced down-regulation of the anti-apoptotic protein survivin and MCL-1. Suppression of survivin protein expression by Gö6976 was due to the inhibition of transcription via the suppression of STAT3/5. On the other hand, Gö6976 induced proteasome-mediated degradation of MCL-1. Previously described FLT3 inhibitors such as PKC412 are bound by the human plasma protein, α1-acid glycoprotein, resulting in diminished inhibitory activity against FLT3. In contrast, we found that Gö6976 potently inhibited phosphorylation of FLT3 and exerted cytotoxicity in the presence of human serum. In conclusion, Gö6976 is a potent FLT3 inhibitor that displays a significant antiproliferative activity against leukemia cells with FLT3-ITD through the profound down-regulation of survivin and MCL-1.
Collapse
Affiliation(s)
- Akira Yoshida
- Department of Hematology and Oncology, Faculty of Medicine, University of Fukui, Shimoaizuki 23-3, Mastuoka, Eiheiji-Chou, Fukui 910-1193, Japan; Translational Research Center, University of Fukui, Japan.
| | - Miyuki Ookura
- Department of Hematology and Oncology, Faculty of Medicine, University of Fukui, Shimoaizuki 23-3, Mastuoka, Eiheiji-Chou, Fukui 910-1193, Japan
| | - Kouichi Zokumasu
- Department of Hematology and Oncology, Faculty of Medicine, University of Fukui, Shimoaizuki 23-3, Mastuoka, Eiheiji-Chou, Fukui 910-1193, Japan
| | - Takanori Ueda
- Department of Hematology and Oncology, Faculty of Medicine, University of Fukui, Shimoaizuki 23-3, Mastuoka, Eiheiji-Chou, Fukui 910-1193, Japan
| |
Collapse
|
98
|
Chen J, Tian CX, Yu M, Lv Q, Cheng NS, Wang Z, Wu X. Efficacy and Safety Profile of Combining Sorafenib with Chemotherapy in Patients with HER2-Negative Advanced Breast Cancer: A Meta-analysis. J Breast Cancer 2014; 17:61-8. [PMID: 24744799 PMCID: PMC3988344 DOI: 10.4048/jbc.2014.17.1.61] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Accepted: 01/04/2014] [Indexed: 02/05/2023] Open
Abstract
Purpose The aim of the study was to evaluate the efficacy and safety of combining sorafenib with chemotherapy in patients with human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer. Methods MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, American Society for Clinical Oncology abstracts, and European Society for Medical Oncology abstracts were searched. Randomized clinical trials that compared the efficacy and safety of sorafenib plus chemotherapy in patients with HER2-negative advanced breast cancer with placebo plus chemotherapy were eligible. The endpoints were progression-free survival (PFS), overall survival (OS), time to progression (TTP), duration of response (DOR), overall response rate (ORR), clinical benefits, and adverse effects. The meta-analysis was performed using Review Manager 5.2.6 (The Nordic Cochrane Centre), and the fixed-effect model weighted by the Mantel-Haenszel method was used. When considerable heterogeneity was found (p<0.1), further analysis (subgroup analysis, sensitivity analysis, or random-effect model) was performed to identify the potential cause. The results are expressed as hazard ratios or risk ratios, with their corresponding 95% confidence intervals. Results The final analysis included four trials comprising 844 patients. The results revealed longer PFS and TTP, and higher ORR and clinical benefit rates in patients receiving sorafenib combined with chemotherapy compared to those receiving chemotherapy and placebo. OS and DOR were similar in the two groups. Meanwhile, the incidence of some adverse effects, including hand-foot skin reaction/hand-foot syndrome, diarrhea, rash, and hypertension, were significantly higher in the sorafenib arm. Conclusion Sorafenib combined with chemotherapy may prolong PFS and TTP. This treatment was associated with manageable toxicities, but frequent dose interruptions and reductions were required.
Collapse
Affiliation(s)
- Jie Chen
- Department of Breast and Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chun-Xiang Tian
- Department of Breast and Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Miao Yu
- Department of Breast Disease, The Second People's Hospital of Sichuan, Chengdu, China
| | - Qing Lv
- Department of Breast and Thyroid Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Nan-Sheng Cheng
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zu Wang
- Tumor Molecular Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Wu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
99
|
Chen CH, Chen MC, Wang JC, Tsai AC, Chen CS, Liou JP, Pan SL, Teng CM. Synergistic interaction between the HDAC inhibitor, MPT0E028, and sorafenib in liver cancer cells in vitro and in vivo. Clin Cancer Res 2014; 20:1274-1287. [PMID: 24520095 DOI: 10.1158/1078-0432.ccr-12-3909] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE To investigate the antitumor activities of a histone deacetylase (HDAC) inhibitor, MPT0E028, plus sorafenib in liver cancer cells in vitro and in vivo. EXPERIMENTAL DESIGN Different liver cancer cell lines were exposed to sorafenib in the presence or absence of MPT0E028, and cell viability was determined by MTT assay. Effects of combined treatment on cell cycle and intracellular signaling pathways were assessed by flow cytometry and Western blot analysis. The Hep3B xenograft model was used to examine the antitumor activity in vivo. RESULTS Our data indicate that sorafenib and MPT0E028 synergistically reduced cell viability in liver cancer cells, and also markedly induced apoptotic cell death in these cells, as evidenced by the cleavage of caspase-3, PARP, and DNA fragmentation. MPT0E028 altered the global modifications of histone and nonhistone proteins regardless of the presence of sorafenib. However, sorafenib blocked MPT0E028-induced Erk activation and its downstream signaling cascades, such as Stat3 phosphorylation (Ser(727)) and Mcl-1 upregulation. Ectopic expression of constitutively active Mek successively reversed the apoptosis triggered by the combined treatment. Pharmacologic inhibition of Mek by PD98059 potentiated MPT0E028-induced apoptosis, suggesting that the synergistic interaction between MPT0E028 and sorafenib occurs at least partly through inhibition of Erk signaling. The data demonstrated that transcriptional activation of fibroblast growth factor receptor 3 (FGFR3) contributes to MPT0E028-mediated Erk phosphorylation. Finally, MPT0E028 plus sorafenib significantly improved the tumor growth delay (TGD) in a Hep3B xenograft model. CONCLUSIONS These findings suggest that MPT0E028 in combination with sorafenib has significant anti-hepatocellular carcinoma activity in preclinical models, potentially suggesting a novel therapeutic strategy for patients with advanced hepatocellular carcinoma.
Collapse
Affiliation(s)
- Chun-Han Chen
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Mei-Chuan Chen
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jing-Chi Wang
- The Ph.D. program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - An-Chi Tsai
- School of Pharmacy, College of Pharmacy, Taipei, Taiwan
| | - Ching-Shih Chen
- Division of Medicinal Chemistry, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | | | - Shiow-Lin Pan
- The Ph.D. program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Che-Ming Teng
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
100
|
Simonin K, N'Diaye M, Lheureux S, Loussouarn C, Dutoit S, Briand M, Giffard F, Brotin E, Blanc-Fournier C, Poulain L. Platinum compounds sensitize ovarian carcinoma cells to ABT-737 by modulation of the Mcl-1/Noxa axis. Apoptosis 2014; 18:492-508. [PMID: 23344663 DOI: 10.1007/s10495-012-0799-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the leading cause of death from gynecological cancer. The anti-apoptotic protein Bcl-x(L) is frequently overexpressed in ovarian carcinoma which correlates with chemotherapy resistance. It has been demonstrated that Bcl-x(L) cooperates with another anti-apoptotic protein, Mcl-1, to protect ovarian cancer cells against apoptosis, and that their concomitant inhibition induces massive cell death. Here, we examined the interest of ABT-737, a potent BH3-mimetic molecule targeting Bcl-x(L), both alone and in combination with Mcl-1 modulators, in ovarian cancer cell lines. As a single agent, ABT-737 was ineffective at promoting cell death in the four cell lines we tested in vitro. However, the specific inhibition of Mcl-1 by siRNA dramatically increased the sensitivity of chemoresistant cells to ABT-737. Platinum compounds also sensitize to ABT-737 by dose-dependently decreasing Mcl-1 expression or by increasing the expression of pro-apoptotic BH3-only proteins Noxa and, to a lower extent, Bim. Furthermore, we demonstrated that Noxa accumulation was involved in apoptosis occurring in response to the combination of ABT-737 and platinum compounds, since cells were protected from apoptosis by its silencing. Moreover, the combination was also highly cytotoxic ex vivo in sliced SKOV3 tumor nodes. However we observed in these slices a strong basal expression of Noxa and apoptotic cell death in response to ABT-737 alone. Therefore, we have revealed that the modulation of the Mcl-1/Noxa axis by platinum compounds results in a strong sensitization of chemoresistant ovarian carcinoma cells to ABT-737, which could constitute a promising therapeutic in these cancers.
Collapse
Affiliation(s)
- Karin Simonin
- Unité Biologie et Thérapies Innovantes des Cancers Localement Agressifs (EA 4656, Université de Caen Basse-Normandie et SF 4206 ICORE), Centre de Lutte Contre le Cancer François Baclesse, 3 Avenue du Général Harris, BP 5026, 14076 Caen Cedex 05, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|