1
|
Lee JK, Chatterjee A, Scarpa M, Bailey CM, Niyongere S, Singh P, Mustafa Ali MK, Kapoor S, Wang Y, Silvestri G, Baer MR. Pim Kinase Inhibitors Increase Gilteritinib Cytotoxicity in FLT3-ITD Acute Myeloid Leukemia Through GSK-3β Activation and c-Myc and Mcl-1 Proteasomal Degradation. CANCER RESEARCH COMMUNICATIONS 2024; 4:431-445. [PMID: 38284896 PMCID: PMC10870818 DOI: 10.1158/2767-9764.crc-23-0379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/12/2023] [Accepted: 01/24/2024] [Indexed: 01/30/2024]
Abstract
Acute myeloid leukemia (AML) with fms-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) has poor outcomes. FLT3-ITD drives constitutive and aberrant FLT3 signaling, activating STAT5 and upregulating the downstream oncogenic serine/threonine kinase Pim-1. FLT3 inhibitors are in clinical use, but with limited and transient efficacy. We previously showed that concurrent treatment with Pim and FLT3 inhibitors increases apoptosis induction in FLT3-ITD-expressing cells through posttranslational downregulation of Mcl-1. Here we further elucidate the mechanism of action of this dual targeting strategy. Cytotoxicity, apoptosis and protein expression and turnover were measured in FLT3-ITD-expressing cell lines and AML patient blasts treated with the FLT3 inhibitor gilteritinib and/or the Pim inhibitors AZD1208 or TP-3654. Pim inhibitor and gilteritinib cotreatment increased apoptosis induction, produced synergistic cytotoxicity, downregulated c-Myc protein expression, earlier than Mcl-1, increased turnover of both proteins, which was rescued by proteasome inhibition, and increased efficacy and prolonged survival in an in vivo model. Gilteritinib and Pim inhibitor cotreatment of Ba/F3-ITD cells infected with T58A c-Myc or S159A Mcl-1 plasmids, preventing phosphorylation at these sites, did not downregulate these proteins, increase their turnover or increase apoptosis induction. Moreover, concurrent treatment with gilteritinib and Pim inhibitors dephosphorylated (activated) the serine/threonine kinase glycogen synthase kinase-3β (GSK-3β), and GSK-3β inhibition prevented c-Myc and Mcl-1 downregulation and decreased apoptosis induction. The data are consistent with c-Myc T58 and Mcl-1 S159 phosphorylation by activated GSK-3β as the mechanism of action of gilteritinib and Pim inhibitor combination treatment, further supporting GSK-3β activation as a therapeutic strategy in FLT3-ITD AML. SIGNIFICANCE FLT3-ITD is present in 25% of in AML, with continued poor outcomes. Combining Pim kinase inhibitors with the FDA-approved FLT3 inhibitor gilteritinib increases cytotoxicity in vitro and in vivo through activation of GSK-3β, which phosphorylates and posttranslationally downregulates c-Myc and Mcl-1. The data support efficacy of GSK-3β activation in FLT3-ITD AML, and also support development of a clinical trial combining the Pim inhibitor TP-3654 with gilteritinib.
Collapse
Affiliation(s)
- Jonelle K. Lee
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Aditi Chatterjee
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mario Scarpa
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Christopher M. Bailey
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sandrine Niyongere
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Prerna Singh
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Moaath K. Mustafa Ali
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Shivani Kapoor
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| | - Yin Wang
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Giovannino Silvestri
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Maria R. Baer
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
2
|
Matsuura K, Shimizu T. Cardiac cell sheet engineering for regenerative medicine and tissue modeling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 199:179-197. [PMID: 37678971 DOI: 10.1016/bs.pmbts.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
Stem cell biology and tissue engineering are essential techniques for cardiac tissue construction. We have succeeded in fabricating human cardiac tissue using the mass production technology of human iPS cell-derived cardiomyocytes and cell sheet engineering, and we are developing regenerative medicine and tissue models to apply this tissue to heart disease research. Cardiac tissue fabrication and tissue functional evaluation technologies for contractile and electrophysiological function are indispensable, which lead to the functional improvement of bioengineered human cardiac tissue.
Collapse
Affiliation(s)
- Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University.
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University
| |
Collapse
|
3
|
Lin EH, Hsu JW, Lee TF, Hsu CF, Lin TH, Jan YH, Chang HY, Cheng CM, Hsu HJ, Chen WW, Chen BH, Tsai HF, Li JJ, Huang CY, Chuang SH, Chang JM, Hsiao M, Wu CW. Targeting cancer stemness mediated by BMI1 and MCL1 for non-small cell lung cancer treatment. J Cell Mol Med 2022; 26:4305-4321. [PMID: 35794816 PMCID: PMC9401641 DOI: 10.1111/jcmm.17453] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Lung cancer is the leading cause of cancer‐associated death, with a global 5‐year survival rate <20%. Early metastasis and recurrence remain major challenges for lung cancer treatment. The stemness property of cancer cells has been suggested to play a key role in cancer plasticity, metastasis and drug‐resistance, and is a potential target for drug development. In this study, we found that in non‐small cell lung cancer (NSCLC), BMI1 and MCL1 play crucial roles of cancer stemness including invasion, chemo‐resistance and tumour initiation. JNK signalling serves as a link between oncogenic pathway or genotoxicity to cancer stemness. The activation of JNK, either by mutant EGFR or chemotherapy agent, stabilized BMI1 and MCL1 proteins through suppressing the expression of E3‐ubiquitin ligase HUWE1. In lung cancer patient samples, high level of BMI1 is correlated with poor survival, and the expression of BMI1 is positively correlated with MCL1. A novel small‐molecule, BI‐44, was developed, which effectively suppressed BMI1/MCL1 expressions and inhibited tumour formation and progression in preclinical models. Targeting cancer stemness mediated by BMI1/MCL1 with BI‐44 provides the basis for a new therapeutic approach in NSCLC treatment.
Collapse
Affiliation(s)
- Erh-Hsuan Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jhen-Wei Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Fang Lee
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chiung-Fang Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Tsung-Hsien Lin
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hua Jan
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsiang-Yi Chang
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Ming Cheng
- Department of Pharmacology, Development Center for Biotechnology, Institute for Drug Evaluation Platform, Taipei, Taiwan
| | - Hui-Jan Hsu
- Department of Medicinal Chemistry, Development Center for Biotechnology, Institute of Pharmaceutics, Taipei, Taiwan
| | - Wei-Wei Chen
- Department of Pharmacology, Development Center for Biotechnology, Institute for Drug Evaluation Platform, Taipei, Taiwan
| | - Bo-Hung Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | - Jung-Jung Li
- Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Ying Huang
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Hsien Chuang
- Department of Medicinal Chemistry, Development Center for Biotechnology, Institute of Pharmaceutics, Taipei, Taiwan
| | - Jia-Ming Chang
- Department of Pharmacology, Development Center for Biotechnology, Institute for Drug Evaluation Platform, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Cheng-Wen Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.,Institute of Microbiology and Immunology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
4
|
Riess C, Koczan D, Schneider B, Linke C, Del Moral K, Classen CF, Maletzki C. Cyclin-dependent kinase inhibitors exert distinct effects on patient-derived 2D and 3D glioblastoma cell culture models. Cell Death Discov 2021; 7:54. [PMID: 33723248 PMCID: PMC7961149 DOI: 10.1038/s41420-021-00423-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/23/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Current therapeutic approaches have met limited clinical success for glioblastoma multiforme (GBM). Since GBM harbors genomic alterations in cyclin-dependent kinases (CDKs), targeting these structures with specific inhibitors (CDKis) is promising. Here, we describe the antitumoral potential of selective CDKi on low-passage GBM 2D- and 3D models, cultured as neurospheres (NSCs) or glioma stem-like cells (GSCs). By applying selective CDK4/6i abemaciclib and palbociclib, and the more global CDK1/2/5/9-i dinaciclib, different effects were seen. Abemaciclib and dinaciclib significantly affected viability in 2D- and 3D models with clearly visible changes in morphology. Palbociclib had weaker and cell line-specific effects. Motility and invasion were highly affected. Abemaciclib and dinaciclib additionally induced senescence. Also, mitochondrial dysfunction and generation of mitochondrial reactive oxygen species (ROS) were seen. While autophagy was predominantly visible after abemaciclib treatment, dinaciclib evoked γ-H2AX-positive double-strand breaks that were boosted by radiation. Notably, dual administration of dinaciclib and abemaciclib yielded synergistic effects in most cases, but the simultaneous combination with standard chemotherapeutic agent temozolomide (TMZ) was antagonistic. RNA-based microarray analysis showed that gene expression was significantly altered by dinaciclib: genes involved in cell-cycle regulation (different CDKs and their cyclins, SMC3), mitosis (PLK1, TTK), transcription regulation (IRX3, MEN1), cell migration/division (BCAR1), and E3 ubiquitination ligases (RBBP6, FBXO32) were downregulated, whereas upregulation was seen in genes mediating chemotaxis (CXCL8, IL6, CCL2), and DNA-damage or stress (EGR1, ARC, GADD45A/B). In a long-term experiment, resistance development was seen in 1/5 cases treated with dinaciclib, but this could be prevented by abemaciclib. Vice versa, adding TMZ abrogated therapeutic effects of dinaciclib and growth was comparable to controls. With this comprehensive analysis, we confirm the therapeutic activity of selective CDKi in GBM. In addition to the careful selection of individual drugs, the timing of each combination partner needs to be considered to prevent resistance.
Collapse
Affiliation(s)
- Christin Riess
- University Children's Hospital, Rostock University Medical Centre, Ernst-Heydemann-Straße 8, 18057, Rostock, Germany.,Department of Medicine Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock University Medical Centre, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany
| | - Dirk Koczan
- Core Facility for Microarray Analysis, Institute for Immunology, Rostock University Medical Centre, 18057, Rostock, Germany
| | - Björn Schneider
- Institute of Pathology, Strempelstraße 14, 18055 Rostock, Rostock University Medical Centre, 18057, Rostock, Germany
| | - Charlotte Linke
- University Children's Hospital, Rostock University Medical Centre, Ernst-Heydemann-Straße 8, 18057, Rostock, Germany
| | - Katharina Del Moral
- University Children's Hospital, Rostock University Medical Centre, Ernst-Heydemann-Straße 8, 18057, Rostock, Germany
| | - Carl Friedrich Classen
- University Children's Hospital, Rostock University Medical Centre, Ernst-Heydemann-Straße 8, 18057, Rostock, Germany
| | - Claudia Maletzki
- Department of Medicine Clinic III - Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock University Medical Centre, Ernst-Heydemann-Str. 6, 18057, Rostock, Germany.
| |
Collapse
|
5
|
Wu X, Luo Q, Liu Z. Ubiquitination and deubiquitination of MCL1 in cancer: deciphering chemoresistance mechanisms and providing potential therapeutic options. Cell Death Dis 2020; 11:556. [PMID: 32699213 PMCID: PMC7376237 DOI: 10.1038/s41419-020-02760-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
Abstract
MCL1 is an important antiapoptotic member of the BCL-2 family that is distinguishable from other family members based on its relatively short half-life. Emerging studies have revealed the crucial role of MCL1 in the chemoresistance of cancer cells. The antiapoptotic function of MCL1 makes it a popular therapeutic target, although specific inhibitors have begun to emerge only recently. Notably, emerging studies have reported that several E3 ligases and deubiquitinases modulate MCL1 stability, providing an alternate means of targeting MCL1 activity. In addition, the emergence and development of proteolysis-targeting chimeras, the function of which is based on ubiquitination-mediated degradation, has shown great potential. In this review, we provide an overview of the studies investigating the ubiquitination and deubiquitination of MCL1, summarize the latest evidence regarding the development of therapeutic strategies targeting MCL1 in cancer treatment, and discuss the promising future of targeting MCL1 via the ubiquitin–proteasome system in clinical practice.
Collapse
Affiliation(s)
- Xiaowei Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Qingyu Luo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 100021, Beijing, China.
| |
Collapse
|
6
|
Kim A, Lee SY, Seo CS, Chung SK. Ethanol extract of Magnoliae cortex (EEMC) limits teratoma formation of pluripotent stem cells by selective elimination of undifferentiated cells through the p53-dependent mitochondrial apoptotic pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 69:153198. [PMID: 32151917 DOI: 10.1016/j.phymed.2020.153198] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 01/16/2020] [Accepted: 02/25/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) are regarded as the best potential cell source for cell-based regenerative medicine. To develop a safe and efficient iPSC-based cell therapy, it is very important to avoid possible teratoma formation, which can arise from undifferentiated iPSCs (USCs) remaining among differentiated cell products. Dried bark of Magnolia officinalis (Magnolia cortex, MC) has long been used in traditional medicine to treat gastrointestinal ailments and allergic diseases, and has shown have various pharmacological activities, including anti-bacterial, anti-inflammatory, and anti-cancer effects. However, its effects on iPSCs have not yet been examined. PURPOSE In this study, we investigated the selective cytotoxic effects of ethanol extract of MC (EEMC) on undifferentiated iPSCs and elucidated the underlying apoptotic mechanisms in detail. We also investigated the inhibitory effects of EEMC on teratoma formation via in ovo experiments. RESULTS We found that EEMC greatly reduced cell growth and induced apoptotic cell death in USCs, but not in differentiated or normal cells. EEMC caused G2/M cell cycle arrest, mitochondrial damage, and caspase activation of USCs, accompanied by p53 accumulation. In p53KO human iPSCs, EEMC had no cytotoxicity, reinforcing that EEMC-mediated apoptosis of USCs is p53-dependent. EEMC did not cause DNA damage in iPSC-derived differentiated cells. In ovo teratoma formation assay revealed that EEMC treatment before injection efficiently eliminated USCs and prevented teratoma formation. CONCLUSIONS These results collectively indicate that EEMC has potent anti-teratoma activity, and therefore can be used for the development of safe iPSC-based therapy.
Collapse
Affiliation(s)
- Aeyung Kim
- Division of Clinical Medicine, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Seo-Young Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Chang-Seob Seo
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Sun-Ku Chung
- Division of Clinical Medicine, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| |
Collapse
|
7
|
Prunellae Spica Extract Suppresses Teratoma Formation of Pluripotent Stem Cells through p53-Mediated Apoptosis. Nutrients 2020; 12:nu12030721. [PMID: 32182802 PMCID: PMC7146640 DOI: 10.3390/nu12030721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/04/2020] [Accepted: 03/05/2020] [Indexed: 12/31/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have similar properties to embryonic stem cells in terms of indefinite self-renewal and differentiation capacity. After in vitro differentiation of iPSCs, undifferentiated iPSCs (USCs) may exist in cell therapy material and can form teratomas after in vivo transplantation. Selective elimination of residual USCs is, therefore, very important. Prunellae Spica (PS) is a traditional medicinal plant that has been shown to exert anti-cancer, antioxidant, and anti-inflammatory activities; however, its effects on iPSCs have not been previously characterized. In this study, we find that ethanol extract of PS (EPS) effectively induces apoptotic cell death of USCs through G2/M cell cycle arrest, generation of intracellular reactive oxygen species, alteration of mitochondrial membrane potentials, and caspase activation of USCs. In addition, EPS increases p53 accumulation and expression of its downstream targets. In p53 knockout (KO) iPSCs, the EPS did not induce apoptosis, indicating that EPS-mediated apoptosis of USCs was p53-dependent. In addition, EPS was not genotoxic towards iPSCs-derived differentiated cells. EPS treatment before injection efficiently prevented in ovo teratoma formation of p53 wild-type (WT) iPSCs but not p53KO iPSCs. Collectively, these results indicate that EPS has potent anti-teratoma activity and no genotoxicity to differentiated cells. It can, therefore, be used in the development of safe and efficient iPSC-based cell therapies.
Collapse
|
8
|
Gao B, Matsuura K, Shimizu T. Recent progress in induced pluripotent stem cell-derived cardiac cell sheets for tissue engineering. Biosci Trends 2020; 13:292-298. [PMID: 31527326 DOI: 10.5582/bst.2019.01227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The past decade has witnessed remarkable development in tissue engineering technologies and stem cells. Our lab has developed a novel technology - "cell sheet technology" for tissue engineering. After the confluent cells are cultured on an innovative temperature-responsive culture dish, the cells can be harvested as an intact sheet by lowering temperature. We have successfully created multiple cell sheet-based tissues for therapies of a vast variety of diseases, in particular, myocardial diseases. On the other side, the discovery of human induced pluripotent stem cells (hiPSC) enables stable production of defined tissue-specific cell types and thus makes it possible to regenerate tissues or even organs for clinical application and in vitro drug screening/disease modeling. Recently, we have combined cell sheet technology and hiPSC-derived cardiac cells for fabrication of functional human cardiac tissues. This review summarizes ongoing challenges in this field and our progresses in solving issues, such as large scale culture of hiPSC-derived cardiac cells, elimination of undifferentiated iPSCs to decrease the risk of tumor formation as well as myocardial tissue fabrication technologies.
Collapse
Affiliation(s)
- Botao Gao
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University
| |
Collapse
|
9
|
Matsuura K, Ito K, Shiraki N, Kume S, Hagiwara N, Shimizu T. Induced Pluripotent Stem Cell Elimination in a Cell Sheet by Methionine-Free and 42°C Condition for Tumor Prevention. Tissue Eng Part C Methods 2019; 24:605-615. [PMID: 30234460 DOI: 10.1089/ten.tec.2018.0228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Pluripotent stem cells, including induced pluripotent stem (iPS) cells, are promising cell sources for regenerative medicine to replace injured tissues, and tissue engineering technologies enable engraftment of functional iPS cell-derived cells in vivo for prolonged periods. However, the risk of tumor formation is a concern for the use of iPS cells. Bioengineered tissues provide a suitable environment for cell survival, which requires vigorous efforts to eliminate remaining iPS cells and prevent tumor formation. We recently reported three iPS cell elimination strategies, including methionine-free medium, TRPV1 activation through 42°C cultivation, and dinaciclib, a cyclin-dependent kinase 1/9 inhibitor. However, it remains unclear how many iPS cells in bioengineered tissues can be eliminated using these strategies alone or in combination, as well as the mode of subsequent tumor prevention. In the present study, we found that 2 days of cultivation at 42°C sufficiently eliminated 1 × 102 iPS cells in fibroblast sheets and prevented tumor formation. After screening for suitable combinations of these strategies based on Lin28 expression in co-cultures of fibroblasts and 1 × 104 iPS cells, we found that 1 day of cultivation at 42°C in methionine-free culture medium with or without dinaciclib remarkably decreased Lin28 expression and prevented tumor formation. Furthermore, these culture strategies did not affect spontaneous beating or the cell number of human iPS cell-derived cardiomyocytes. These quantitative findings may contribute to decreasing tumor formation risk and development of regenerative medicine using iPS cells.
Collapse
Affiliation(s)
- Katsuhisa Matsuura
- 1 Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University , Tokyo, Japan .,2 Department of Cardiology, Tokyo Women's Medical University , Tokyo, Japan
| | - Kyoji Ito
- 1 Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University , Tokyo, Japan .,3 Division of Hepato-Biliary Pancreatic Surgery, Department of Surgery, Graduate School of Medicine, The University of Tokyo , Tokyo, Japan
| | - Nobuaki Shiraki
- 4 School and Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology , Yokohama, Japan
| | - Shoen Kume
- 4 School and Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology , Yokohama, Japan
| | - Nobuhisa Hagiwara
- 2 Department of Cardiology, Tokyo Women's Medical University , Tokyo, Japan
| | - Tatsuya Shimizu
- 1 Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University , Tokyo, Japan
| |
Collapse
|
10
|
Xu J, Huang F, Yao Z, Jia C, Xiong Z, Liang H, Lin N, Deng M. Inhibition of cyclin E1 sensitizes hepatocellular carcinoma cells to regorafenib by mcl-1 suppression. Cell Commun Signal 2019; 17:85. [PMID: 31349793 PMCID: PMC6660968 DOI: 10.1186/s12964-019-0398-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/15/2019] [Indexed: 12/22/2022] Open
Abstract
Background To clarify the effects of cylcin E1 expression on HCC tumor progression, we studied the expression of cyclin E1 and inhibitory efficacy of regorafenib and sorafenib in HCC cells, and investigated a potential therapy that combines regorafenib treatment with cyclin E1 inhibition. Methods Western blotting for caspase-3 and Hoechst 33225 staining was used to measure the expression level of apoptosis-related proteins under drug treatment. Results Our results showed that enhanced expression of cyclin E1 after transfection compromised apoptosis in HCC cells induced by regorafenib or sorafenib. Conversely, down-regulation of cyclin E1 gene expression or inhibition of cyclin E1 by the cyclin-dependent kinase (CDK) inhibitors dinaciclib (DIN) or flavopiridol sensitized HCC cells to regorafenib and sorafenib by inducing apoptosis. The expression of Mcl-1, which is modulated by STAT3, plays a key role in regulating the therapeutic effects of CDK inhibitors. Xenograft experiments conducted to test the efficacy of regorafenib combined with DIN showed dramatic tumor inhibitory effects due to induction of apoptosis. Our results suggested that the level of cyclin E1 expression in HCCs may be used as a pharmacodynamic biomarker to assess the antitumor effects of regorafenib or sorafenib. Conclusions Combining regorafenib and CDK inhibitors may enhance the clinical efficiency of the treatment of HCCs. Electronic supplementary material The online version of this article (10.1186/s12964-019-0398-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianliang Xu
- Hepatobilliary Surgery Department, The Third affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, Guangdong, China
| | - Fei Huang
- Anesthesiology Department, The third affiliated hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhicheng Yao
- General surgery, The Third affiliated hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, 510630, Guangdong, China.
| | - Changchang Jia
- Cell & Gene therapy center, The Third affiliated Hospital of Sun Yat-sen Uuniversity, Guangzhou, Guangdong, China
| | - Zhiyong Xiong
- General surgery, The Third affiliated hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Hao Liang
- General surgery, The Third affiliated hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, 510630, Guangdong, China
| | - Nan Lin
- Hepatobilliary Surgery Department, The Third affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, Guangdong, China
| | - Meihai Deng
- Hepatobilliary Surgery Department, The Third affiliated Hospital of Sun Yat-sen University, No. 600, Tianhe District, Guangzhou, Guangdong, China.
| |
Collapse
|
11
|
Wang J, Merino DM, Light N, Murphy BL, Wang YD, Guo X, Hodges AP, Chau LQ, Liu KW, Dhall G, Asgharzadeh S, Kiehna EN, Shirey RJ, Janda KD, Taylor MD, Malkin D, Ellison DW, VandenBerg SR, Eberhart CG, Sears RC, Roussel MF, Gilbertson RJ, Wechsler-Reya RJ. Myc and Loss of p53 Cooperate to Drive Formation of Choroid Plexus Carcinoma. Cancer Res 2019; 79:2208-2219. [PMID: 30885981 PMCID: PMC6497574 DOI: 10.1158/0008-5472.can-18-2565] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 02/05/2019] [Accepted: 03/13/2019] [Indexed: 02/03/2023]
Abstract
Choroid plexus carcinoma (CPC) is a rare brain tumor that occurs most commonly in very young children and has a dismal prognosis despite intensive therapy. Improved outcomes for patients with CPC depend on a deeper understanding of the mechanisms underlying the disease. Here we developed transgenic models of CPCs by activating the Myc oncogene and deleting the Trp53 tumor suppressor gene in murine neural stem cells or progenitors. Murine CPC resembled their human counterparts at a histologic level, and like the hypodiploid subset of human CPC, exhibited multiple whole-chromosome losses, particularly of chromosomes 8, 12, and 19. Analysis of murine and human CPC gene expression profiles and copy number changes revealed altered expression of genes involved in cell cycle, DNA damage response, and cilium function. High-throughput drug screening identified small molecule inhibitors that decreased the viability of CPC. These models will be valuable tools for understanding the biology of choroid plexus tumors and for testing novel approaches to therapy. SIGNIFICANCE: This study describes new mouse models of choroid plexus carcinoma and uses them to investigate the biology and therapeutic responsiveness of this highly malignant pediatric brain tumor.
Collapse
Affiliation(s)
- Jun Wang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Diana M Merino
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicholas Light
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Brian L Murphy
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Yong-Dong Wang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Xiaohui Guo
- Bioinformatics Core Facility, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Andrew P Hodges
- Bioinformatics Core Facility, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Lianne Q Chau
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Kun-Wei Liu
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Girish Dhall
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles
| | - Shahab Asgharzadeh
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles
| | - Erin N Kiehna
- Division of Hematology, Oncology and Blood & Marrow Transplantation, Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles
| | - Ryan J Shirey
- Department of Chemistry, The Scripps Research Institute, La Jolla, California
- Department of Immunology, The Scripps Research Institute, La Jolla, California
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California
| | - Kim D Janda
- Department of Chemistry, The Scripps Research Institute, La Jolla, California
- Department of Immunology, The Scripps Research Institute, La Jolla, California
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California
| | - Michael D Taylor
- Division of Neurosurgery and Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - David Malkin
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - David W Ellison
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Scott R VandenBerg
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rosalie C Sears
- Molecular and Medical Genetics Department, Oregon Health and Sciences University, Portland, Oregon
| | - Martine F Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Richard J Gilbertson
- Cancer Research UK Cambridge Centre, CRUK Cambridge Institute, Li Ka Shing Centre, Cambridge, United Kingdom
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California.
| |
Collapse
|
12
|
Song KA, Hosono Y, Turner C, Jacob S, Lochmann TL, Murakami Y, Patel NU, Ham J, Hu B, Powell KM, Coon CM, Windle BE, Oya Y, Koblinski JE, Harada H, Leverson JD, Souers AJ, Hata AN, Boikos S, Yatabe Y, Ebi H, Faber AC. Increased Synthesis of MCL-1 Protein Underlies Initial Survival of EGFR-Mutant Lung Cancer to EGFR Inhibitors and Provides a Novel Drug Target. Clin Cancer Res 2018; 24:5658-5672. [PMID: 30087143 DOI: 10.1158/1078-0432.ccr-18-0304] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/29/2018] [Accepted: 08/01/2018] [Indexed: 11/16/2022]
Abstract
Purpose: EGFR inhibitors (EGFRi) are effective against EGFR-mutant lung cancers. The efficacy of these drugs, however, is mitigated by the outgrowth of resistant cells, most often driven by a secondary acquired mutation in EGFR, T790M We recently demonstrated that T790M can arise de novo during treatment; it follows that one potential therapeutic strategy to thwart resistance would be identifying and eliminating these cells [referred to as drug-tolerant cells (DTC)] prior to acquiring secondary mutations like T790M Experimental Design: We have developed DTCs to EGFRi in EGFR-mutant lung cancer cell lines. Subsequent analyses of DTCs included RNA-seq, high-content microscopy, and protein translational assays. Based on these results, we tested the ability of MCL-1 BH3 mimetics to combine with EGFR inhibitors to eliminate DTCs and shrink EGFR-mutant lung cancer tumors in vivo Results: We demonstrate surviving EGFR-mutant lung cancer cells upregulate the antiapoptotic protein MCL-1 in response to short-term EGFRi treatment. Mechanistically, DTCs undergo a protein biosynthesis enrichment resulting in increased mTORC1-mediated mRNA translation of MCL-1, revealing a novel mechanism in which lung cancer cells adapt to short-term pressures of apoptosis-inducing kinase inhibitors. Moreover, MCL-1 is a key molecule governing the emergence of early EGFR-mutant DTCs to EGFRi, and we demonstrate it can be effectively cotargeted with clinically emerging MCL-1 inhibitors both in vitro and in vivo Conclusions: Altogether, these data reveal that this novel therapeutic combination may delay the acquisition of secondary mutations, therefore prolonging therapy efficacy. Clin Cancer Res; 24(22); 5658-72. ©2018 AACR.
Collapse
Affiliation(s)
- Kyung-A Song
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Yasuyuki Hosono
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Crystal Turner
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Sheeba Jacob
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Timothy L Lochmann
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Yoshiko Murakami
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Neha U Patel
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Jungoh Ham
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Bin Hu
- Department of Pathology, VCU School of Medicine, Richmond, Virginia
| | - Krista M Powell
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Colin M Coon
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Brad E Windle
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | - Yuko Oya
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | | | - Hisashi Harada
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia
| | | | | | - Aaron N Hata
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Sosipatros Boikos
- Division of Hematology, Oncology and Palliative Care, Virginia Commonwealth University, Massey Cancer Center, Richmond, Virginia
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan.,Precision Medicine Center, Aichi Cancer Center, Nagoya, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan. .,Precision Medicine Center, Aichi Cancer Center, Nagoya, Japan
| | - Anthony C Faber
- Philips Institute for Oral Health Research, VCU School of Dentistry and Massey Cancer Center, Richmond, Virginia.
| |
Collapse
|