351
|
Two Opposing Faces of Retinoic Acid: Induction of Stemness or Induction of Differentiation Depending on Cell-Type. Biomolecules 2019; 9:biom9100567. [PMID: 31590252 PMCID: PMC6843238 DOI: 10.3390/biom9100567] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
Stem cells have the capacity of self-renewal and, through proliferation and differentiation, are responsible for the embryonic development, postnatal development, and the regeneration of tissues in the adult organism. Cancer stem cells, analogous to the physiological stem cells, have the capacity of self-renewal and may account for growth and recurrence of tumors. Development and regeneration of healthy tissues and tumors depend on the balance of different genomic and nongenomic signaling pathways that regulate stem cell quiescence, proliferation, and differentiation. During evolution, this balance became dependent on all-trans retinoic acid (RA), a molecule derived from the environmental factor vitamin A. Here we summarize some recent findings on the prominent role of RA on the proliferation of stem and progenitor cells, in addition to its well-known function as an inductor of cell differentiation. A better understanding of the regulatory mechanisms of stemness and cell differentiation by RA may improve the therapeutic options of this molecule in regenerative medicine and cancer.
Collapse
|
352
|
Pibiri M. Liver regeneration in aged mice: new insights. Aging (Albany NY) 2019; 10:1801-1824. [PMID: 30157472 PMCID: PMC6128415 DOI: 10.18632/aging.101524] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023]
Abstract
The regenerative capacity of the liver after resection is reduced with aging. Recent studies on rodents revealed that both intracellular and extracellular factors are involved in the impairment of liver mass recovery during aging. Among the intracellular factors, age-dependent decrease of BubR1 (budding uninhibited by benzimidazole-related 1), YAP (Yes-associated protein) and SIRT1 (Sirtuin-1) have been associated to dampening of tissue reconstitution and inhibition of cell cycle genes following partial hepatectomy. Extra-cellular factors, such as age-dependent changes in hepatic stellate cells affect liver regeneration through inhibition of progenitor cells and reduction of liver perfusion. Furthermore, chronic release of pro-inflammatory proteins by senescent cells (SASP) affects cell proliferation suggesting that senescent cell clearance might improve tissue regeneration. Accordingly, young plasma restores liver regeneration in aged animals through autophagy re-establishment. This review will discuss how intracellular and extracellular factors cooperate to guarantee a proper liver regeneration and the possible causes of its impairment during aging. The possibility that an improvement of the liver regenerative capacity in elderly might be achieved through elimination of senescent cells via autophagy or by administration of direct mitogenic agents devoid of cytotoxicity will also be entertained.
Collapse
Affiliation(s)
- Monica Pibiri
- Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Cagliari 09124, Italy
| |
Collapse
|
353
|
Hu C, Sun J, Du J, Wen D, Lu H, Zhang H, Xue Y, Zhang A, Yang C, Zeng L, Jiang J. The Hippo-YAP pathway regulates the proliferation of alveolar epithelial progenitors after acute lung injury. Cell Biol Int 2019; 43:1174-1183. [PMID: 30632652 DOI: 10.1002/cbin.11098] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Regeneration of pulmonary epithelial cells plays an important role in the recovery of acute lung injury (ALI), which is defined by pulmonary epithelial cell death. However, the mechanism of the regenerative capacity of alveolar epithelial cells is unknown. Using a lung injury mouse model induced by hemorrhagic shock and lipopolysaccharide, a protein mass spectrometry-based high-throughput screening and linage tracing technology to mark alveolar epithelial type 2 cells (AEC2s), we analyzed the mechanism of alveolar epithelial cells proliferation. We demonstrated that the expression of Hippo-yes-associated protein 1 (YAP1) key proteins were highly consistent with the regularity of the proliferation of alveolar epithelial type 2 cells after ALI. Furthermore, the results showed that YAP1+ cells in lung tissue after ALI were mainly Sftpc lineage-labeled AEC2s. An in vitro proliferation assay of AEC2s demonstrated that AEC2 proliferation was significantly inhibited by both YAP1 small interfering RNA and Hippo inhibitor. These findings revealed that YAP functioned as a key regulator to promote AEC2s proliferation, with the Hippo signaling pathway playing a pivotal role in this process.
Collapse
Affiliation(s)
- Chen Hu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Jianhui Sun
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Juan Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Dalin Wen
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Hongxiang Lu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Huacai Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Yuqi Xue
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Anqiang Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Ce Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Traumatic Surgery, Institute of Surgery Research, Daping Hospital, Military Medical University, Chongqing, 400042, China
| |
Collapse
|
354
|
Yorkie and JNK Control Tumorigenesis in Drosophila Cells with Cytokinesis Failure. Cell Rep 2019; 23:1491-1503. [PMID: 29719260 DOI: 10.1016/j.celrep.2018.04.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 03/14/2018] [Accepted: 03/30/2018] [Indexed: 01/23/2023] Open
Abstract
Cytokinesis failure may result in the formation of polyploid cells, and subsequent mitosis can lead to aneuploidy and tumor formation. Tumor suppressor mechanisms limiting the oncogenic potential of these cells have been described. However, the universal applicability of these tumor-suppressive barriers remains controversial. Here, we use Drosophila epithelial cells to investigate the consequences of cytokinesis failure in vivo. We report that cleavage defects trigger the activation of the JNK pathway, leading to downregulation of the inhibitor of apoptosis DIAP1 and programmed cell death. Yorkie overcomes the tumor-suppressive role of JNK and induces neoplasia. Yorkie regulates the cell cycle phosphatase Cdc25/string, which drives tumorigenesis in a context of cytokinesis failure. These results highlight the functional significance of the JNK pathway in epithelial cells with defective cytokinesis and elucidate a mechanism used by emerging tumor cells to bypass this tumor-suppressive barrier and develop into tumors.
Collapse
|
355
|
Han Z, Yu Y, Cai B, Xu Z, Bao Z, Zhang Y, Bamba D, Ma W, Gao X, Yuan Y, Zhang L, Yu M, Liu S, Yan G, Jin M, Huang Q, Wang X, Hua B, Yang F, Pan Z, Liang H, Liu Y. YAP/TEAD3 signal mediates cardiac lineage commitment of human-induced pluripotent stem cells. J Cell Physiol 2019; 235:2753-2760. [PMID: 31541452 DOI: 10.1002/jcp.29179] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022]
Abstract
Cardiomyocytes differentiated from human-induced pluripotent stem cells (hiPSCs) hold great potential for therapy of heart diseases. However, the underlying mechanisms of its cardiac differentiation have not been fully elucidated. Hippo-YAP signal pathway plays important roles in cell differentiation, tissue homeostasis, and organ size. Here, we identify the role of Hippo-YAP signal pathway in determining cardiac differentiation fate of hiPSCs. We found that cardiac differentiation of hiPSCs were significantly inhibited after treatment with verteporfin (a selective and potent YAP inhibitor). During hiPSCs differentiation from mesoderm cells (MESs) into cardiomyocytes, verteporfin treatment caused the cells retained in the earlier cardiovascular progenitor cells (CVPCs) stage. Interestingly, during hiPSCs differentiation from CVPC into cardiomyocytes, verteporfin treatment induced cells dedifferentiation into the earlier CVPC stage. Mechanistically, we found that YAP interacted with transcriptional enhanced associate domain transcription factor 3 (TEAD3) to regulate cardiac differentiation of hiPSCs during the CVPC stage. Consistently, RNAi-based silencing of TEAD3 mimicked the phenotype as the cells treated with verteporfin. Collectively, our study suggests that YAP-TEAD3 signaling is important for cardiomyocyte differentiation of hiPSCs. Our findings provide new insight into the function of Hippo-YAP signal in cardiovascular lineage commitment.
Collapse
Affiliation(s)
- Zhenbo Han
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Ying Yu
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Benzhi Cai
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Zihang Xu
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Zhengyi Bao
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Ying Zhang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Djibril Bamba
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Wenya Ma
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Xinlu Gao
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Ye Yuan
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Lai Zhang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Meixi Yu
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Shenzhen Liu
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Gege Yan
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Mengyu Jin
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Qi Huang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Xiuxiu Wang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Bingjie Hua
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Fan Yang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Zhenwei Pan
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Haihai Liang
- The Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology at College of Pharmacy, Department of Pharmacy at the Affiliated Second Hospital, Harbin Medical University, Harbin, China
| | - Yu Liu
- Department of Clinical Laboratory at the Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
356
|
Abstract
The Hippo signalling pathway and its transcriptional co-activator targets Yorkie/YAP/TAZ first came to attention because of their role in tissue growth control. Over the past 15 years, it has become clear that, like other developmental pathways (e.g. the Wnt, Hedgehog and TGFβ pathways), Hippo signalling is a 'jack of all trades' that is reiteratively used to mediate a range of cellular decision-making processes from proliferation, death and morphogenesis to cell fate determination. Here, and in the accompanying poster, we briefly outline the core pathway and its regulation, and describe the breadth of its roles in animal development.
Collapse
Affiliation(s)
- John Robert Davis
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Nicolas Tapon
- Apoptosis and Proliferation Control Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
357
|
Gu L, Liu J, Xu D, Lu Y. Reciprocal Feedback Loop of the MALAT1-MicroRNA-194-YAP1 Pathway Regulates Progression of Acute Pancreatitis. Med Sci Monit 2019; 25:6894-6904. [PMID: 31518341 PMCID: PMC6756034 DOI: 10.12659/msm.915598] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Acute pancreatitis (AP) has a high mortality rate and often has serious complications. The Hippo-YAP signaling pathway is mainly involved in cell proliferation and stem cell self-renewal. Recent studies have reported that YAP1 plays a crucial role in pancreatic cancer initiation and acute and chronic pancreatitis (CP). However, the role of YAP1 in AP still needs to be clarified. Material/Methods To assess the role of YAP1 in the progression of AP, we established a cell model of AP in AR42J cells. AR42J, a rat pancreatic acinar cell line, was stimulated with caerulein to mimic AP-like acinar cell injury. Levels of interleukin (IL)-6 and tumor necrosis factor-α (TNF-α) were measured by ELISA to investigate the role of YAP1 in the progression of AP. Results The results showed that YAP1 and MALAT1 were the targets of miR-194 and were upregulated in caerulein-treated AR42J cells. Overexpression of MALAT1 or YAP1 can increase the levels of IL-6 and TNF-α secreted by AR42J cells, while miR-194 dramatically counteracts this enhancement effect. Conclusions Our results demonstrated a regulation loop among MATAL1, miR-194, and YAP1, which dynamically regulates the progression of AP, providing a new therapeutic target for treatment of this disease.
Collapse
Affiliation(s)
- Lina Gu
- Department of ICU (Intensive Care Unit), The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Jingyao Liu
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Dan Xu
- Department of Traditional Chinese Medicine, The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| | - Ying Lu
- Department of ICU (Intensive Care Unit), The First Hospital of Jilin University, Changchun, Jilin, China (mainland)
| |
Collapse
|
358
|
CCT3 acts upstream of YAP and TFCP2 as a potential target and tumour biomarker in liver cancer. Cell Death Dis 2019; 10:644. [PMID: 31501420 PMCID: PMC6733791 DOI: 10.1038/s41419-019-1894-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/16/2019] [Accepted: 08/11/2019] [Indexed: 02/06/2023]
Abstract
Although Yes-associated protein (YAP) is very important to liver cancer, its nuclear localisation prevents consideration as a promising therapeutic target and a diagnostic biomarker. Recently, we reported that the protumourigenic roles of YAP in liver cancer are indispensable for transcription factor CP2 (TFCP2) in a Hippo-independent manner; however, proteins that act upstream to simultaneously control YAP and TFCP2 remain unclear. The aim of this study was to uncover such proteins and evaluate whether they are potential YAP-associated therapeutic targets and diagnostic biomarkers. Mass spectrometry revealed that chaperonin containing TCP1 subunit 3 (CCT3) co-interact with YAP and TFCP2, and notably, CCT3 is a non-nuclear protein. CCT3 was elevated in liver cancer, and its higher expression was associated with poorer overall survival. Inhibiting CCT3 resulted in a suppressed transformative phenotype in liver cancer cells, suggesting that CCT3 might be a potential therapeutic target. CCT3 prolonged half-life of YAP and TFCP2 by blocking their ubiquitination caused by poly(rC) binding protein 2 (PCBP2) in a beta-transducin repeat containing E3 ubiquitin protein ligase (βTrCP)-independent manner. Interestingly, PCBP2 directly interacted with YAP via a WB motif-WW domain interaction, whereas indirectly interacted with TFCP2 via the aid of YAP. Furthermore, CCT3 was capable of separating PCBP2-YAP interactions, thereby preventing YAP and TFCP2 from PCBP2-induced ubiquitination. Moreover, YAP and TFCP2 were downstream of CCT3 to positively control tumourigenesis, yet such effects were inhibited by PCBP2. Clinically, CCT3 was positively correlated with YAP and TFCP2, and elevated levels of the CCT3-YAP-TFCP2 axis might be critical for liver malignancy. In addition, seral-CCT3 was proven to be a potential biomarker, and its diagnostic capacity was better than that of alpha fetoprotein (AFP) to a certain extent. Together, CCT3 acts as a trigger of YAP and TFCP2 to affect tumourigenesis and serves as a potential therapeutic target and biomarker in liver cancer.
Collapse
|
359
|
Pajtler KW, Wei Y, Okonechnikov K, Silva PBG, Vouri M, Zhang L, Brabetz S, Sieber L, Gulley M, Mauermann M, Wedig T, Mack N, Imamura Kawasawa Y, Sharma T, Zuckermann M, Andreiuolo F, Holland E, Maass K, Körkel-Qu H, Liu HK, Sahm F, Capper D, Bunt J, Richards LJ, Jones DTW, Korshunov A, Chavez L, Lichter P, Hoshino M, Pfister SM, Kool M, Li W, Kawauchi D. YAP1 subgroup supratentorial ependymoma requires TEAD and nuclear factor I-mediated transcriptional programmes for tumorigenesis. Nat Commun 2019; 10:3914. [PMID: 31477715 PMCID: PMC6718408 DOI: 10.1038/s41467-019-11884-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/07/2019] [Indexed: 01/22/2023] Open
Abstract
YAP1 fusion-positive supratentorial ependymomas predominantly occur in infants, but the molecular mechanisms of oncogenesis are unknown. Here we show YAP1-MAMLD1 fusions are sufficient to drive malignant transformation in mice, and the resulting tumors share histo-molecular characteristics of human ependymomas. Nuclear localization of YAP1-MAMLD1 protein is mediated by MAMLD1 and independent of YAP1-Ser127 phosphorylation. Chromatin immunoprecipitation-sequencing analyses of human YAP1-MAMLD1-positive ependymoma reveal enrichment of NFI and TEAD transcription factor binding site motifs in YAP1-bound regulatory elements, suggesting a role for these transcription factors in YAP1-MAMLD1-driven tumorigenesis. Mutation of the TEAD binding site in the YAP1 fusion or repression of NFI targets prevents tumor induction in mice. Together, these results demonstrate that the YAP1-MAMLD1 fusion functions as an oncogenic driver of ependymoma through recruitment of TEADs and NFIs, indicating a rationale for preclinical studies to block the interaction between YAP1 fusions and NFI and TEAD transcription factors. The molecular mechanisms driving proliferation in the pediatric brain cancer epdendymoma are poorly understood. Here the authors show that a YAP1- MAMLD1 fusion drives tumor formation in mice and show that the fusion protein can collaborate with the TEAD and NFI transcription factors.
Collapse
Affiliation(s)
- Kristian W Pajtler
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Yiju Wei
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Penn State Health Hershey Medical Center, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Konstantin Okonechnikov
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Patricia B G Silva
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Mikaella Vouri
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Lei Zhang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Penn State Health Hershey Medical Center, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Sebastian Brabetz
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Laura Sieber
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Melissa Gulley
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Penn State Health Hershey Medical Center, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Monika Mauermann
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Tatjana Wedig
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Norman Mack
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Yuka Imamura Kawasawa
- Department of Biochemistry and Molecular Biology, Penn State Health Hershey Medical Center, Penn State College of Medicine, Hershey, PA, 17033, USA.,Department of Pharmacology, Penn State Health Hershey Medical Center, Penn State College of Medicine, Hershey, PA, 17033, USA
| | - Tanvi Sharma
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Marc Zuckermann
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Felipe Andreiuolo
- Department of Neuropathology, Ste. Anne Hospital, 75014, Paris, France
| | - Eric Holland
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - Kendra Maass
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Huiqin Körkel-Qu
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Hai-Kun Liu
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Felix Sahm
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Department of Neuropathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - David Capper
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neuropathology, Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jens Bunt
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia
| | - Linda J Richards
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, Australia
| | - David T W Jones
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Andrey Korshunov
- Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Department of Neuropathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Lukas Chavez
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Peter Lichter
- Division of Molecular Genetics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Stefan M Pfister
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Marcel Kool
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Wei Li
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Penn State Health Hershey Medical Center, Penn State College of Medicine, Hershey, PA, 17033, USA. .,Department of Biochemistry and Molecular Biology, Penn State Health Hershey Medical Center, Penn State College of Medicine, Hershey, PA, 17033, USA.
| | - Daisuke Kawauchi
- Hopp-Children's Cancer Center Heidelberg (KiTZ), 69120, Heidelberg, Germany. .,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| |
Collapse
|
360
|
Valizadeh A, Majidinia M, Samadi-Kafil H, Yousefi M, Yousefi B. The roles of signaling pathways in liver repair and regeneration. J Cell Physiol 2019; 234:14966-14974. [PMID: 30770551 DOI: 10.1002/jcp.28336] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 12/23/2018] [Accepted: 01/10/2019] [Indexed: 01/24/2023]
Abstract
The liver has remarkable regeneration potency that restores liver mass and sustains body hemostasis. Liver regeneration through signaling pathways following resection or moderate damages are well studied. Various cell signaling, growth factors, cytokines, receptors, and cell types implicated in liver regeneration undergo controlled hypertrophy and proliferation. Some aspects of liver regeneration have been discovered and many investigations have been carried out to identify its mechanisms. However, for optimizing liver regeneration more should be understood about mechanisms that control the growth of hepatocytes and other liver cell types in adults. The current paper deals with the possible applicability of liver regeneration signaling pathways as a target for therapeutic approaches and preventing various liver damages. Furthermore, the latest findings of spectrum-specific signaling pathway mechanisms that underlie liver regeneration are briefly described.
Collapse
Affiliation(s)
- Amir Valizadeh
- Stem Cells Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Hossein Samadi-Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
361
|
Das D, Fletcher RB, Ngai J. Cellular mechanisms of epithelial stem cell self-renewal and differentiation during homeostasis and repair. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e361. [PMID: 31468728 DOI: 10.1002/wdev.361] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/14/2022]
Abstract
Epithelia in adult mammals exhibit remarkable regenerative capacities owing to the presence of adult stem cells, which self-renew and differentiate to replace cells lost to normal turnover or injury. The mechanisms supporting tissue homeostasis and injury-induced repair often differ from each other as well as from those used in embryonic development. Recent studies have also highlighted the phenomenon of cellular plasticity in adult tissues, in which differentiated cells can change fate and even give rise to new stem cell populations to complement the canonical stem cells in promoting repair following injury. Signaling pathways such as WNT, bone morphogenetic protein, and Sonic Hedgehog play critical roles in stem cell maintenance and cell fate decisions across diverse epithelia and conditions, suggesting that conserved mechanisms underlie the regenerative capacity of adult epithelial structures. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Adult Stem Cells, Tissue Renewal, and Regeneration > Tissue Stem Cells and Niches Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration.
Collapse
Affiliation(s)
- Diya Das
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Berkeley Institute for Data Science, University of California, Berkeley, California
| | - Russell B Fletcher
- Department of Molecular and Cell Biology, University of California, Berkeley, California
| | - John Ngai
- Department of Molecular and Cell Biology, University of California, Berkeley, California.,Helen Wills Neuroscience Institute, University of California, Berkeley, California.,QB3 Functional Genomics Laboratory, University of California, Berkeley, California
| |
Collapse
|
362
|
Metformin targets a YAP1-TEAD4 complex via AMPKα to regulate CCNE1/2 in bladder cancer cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:376. [PMID: 31455378 PMCID: PMC6712726 DOI: 10.1186/s13046-019-1346-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/24/2019] [Indexed: 12/14/2022]
Abstract
Background Metformin has been reported to function as the anti-tumor inhibiting the growth of different types of cancers, including bladder cancer. But there are few reports on the roles of Yap1, the key molecule of Hippo pathway, in the metformin induced inhibition of bladder cancer (BLCA). We are wondering if the inhibitory effect of metformin on bladder cancer is fulfilled via Yap1 and exploring the related mechanism. Methods MTS and colony formation assays were used to explore the cellular viabilities and proliferation of BLCA cells challenged by metformin at different concentrations, in vitro. Flow Cytometry (FCM) was used to analyze the cell cycle and the cellular apoptosis of the BLCA cells. Western Blot was performed to detect the expressions of AMPKα, Yap1, CCND1, CCNE1/2 and CDK2/4/6 in the metformin-treated BLCA cell lines. RNAi method was used for the related genetic functional analysis. The relationships among Yap1, TEADs and CCNE1/2 were predicted and evaluated using bioinformatics, dual-luciferase reporter and co-immunoprecipitation (Co-IP) assays. For in vivo experiments, a xenograft model was used to investigate the effects of metformin on the proliferation of BLCA cells. And Immunohistochemistry (IHC) assay was performed to assess the expressions of CCNE1/2 and Yap1 proteins in the tumor tissues from the model. Results Metformin could inhibit the proliferation of the BLCA cells via inducing the G1 cell cycle arrest without apoptosis. And metformin upregulated the phosphorylated AMPKα and decreased the expressions of Yap1 and CCND1, CCNE1/2 and CDK4/6. AMPK inhibition by compound C (CC) restored the cell proliferation and the G1 cell cycle arrest induced by metformin, in vivo. Knockdown of YAP1 inhibited the proliferation of BLCA cells and caused the cell cycle arrest at G1 phase by decreasing the expressions of CCNE1/2 and other G1 phase related molecules, which has been restored by the Yap 5SA mutant. Bioinformatics analysis showed that trans-factor TEAD4 was highly expressed and positively associated with the expressions of CCNE1 and CCNE2 in BLCA and only TEAD4 was precipitated by Yap1 in the BLCA cells. Further studies demonstrated that Yap1 positively regulated both CCNE1 and CCNE2 expressions via forming complex with TEAD4. Furthermore, we observed that metformin inhibited the cell proliferation by decreasing the expressions of Yap1 and both CCNE1 and CCNE2 in xenograft model. Conclusions The results of our study reveal a new potential regulatory pathway in which metformin inhibits cell proliferation via AMPKα/Yap1/TEAD4/CCNE1/2 axis in BLCA cells, providing new insights into novel molecular therapeutic targets for BLCA. Electronic supplementary material The online version of this article (10.1186/s13046-019-1346-1) contains supplementary material, which is available to authorized users.
Collapse
|
363
|
Angius A, Uva P, Pira G, Muroni MR, Sotgiu G, Saderi L, Uleri E, Caocci M, Ibba G, Cesaraccio MR, Serra C, Carru C, Manca A, Sanges F, Porcu A, Dolei A, Scanu AM, Rocca PC, De Miglio MR. Integrated Analysis of miRNA and mRNA Endorses a Twenty miRNAs Signature for Colorectal Carcinoma. Int J Mol Sci 2019; 20:E4067. [PMID: 31434359 PMCID: PMC6720928 DOI: 10.3390/ijms20164067] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) ranks as the most frequent carcinoma worldwide. CRC patients show strong prognostic differences and responses to treatment, and 20% have incurable metastatic disease at diagnosis. We considered it essential to investigate mechanisms that control cellular regulatory networks, such as the miRNA-mRNA interaction, known to be involved in cancer pathogenesis. We conducted a human miRNome analysis by TaqMan low density array, comparing CRC to normal colon tissue (NCT, and experimentally identified gene targets of miRNAs deregulated, by anti-correlation analysis, with the CRC whole-transcriptome profile obtained from RNASeq experiments. We identified an integrated signature of 20 deregulated miRNAs in CRC. Enrichment analyses of the gene targets controlled by these miRNAs brought to light 25 genes, members of pathways known to lead to cell growth and death (CCND1, NKD1, FZD3, MAD2L1, etc.), such as cell metabolism (ACSL6, PRPS1-2). A screening of prognosis-mediated miRNAs underlined that the overexpression of miR-224 promotes CRC metastasis, and is associated with high stage and poor survival. These findings suggest that the biology and progression of CRC depend on deregulation of multiple miRNAs that cause a complex dysfunction of cellular molecular networks. Our results have further established miRNA-mRNA interactions and defined multiple pathways involved in CRC pathogenesis.
Collapse
Affiliation(s)
- Andrea Angius
- Istituto di Ricerca Genetica e Biomedica (IRGB), CNR, Cittadella Universitaria di Cagliari, 09042 Monserrato (CA), Italy
| | - Paolo Uva
- CRS4, Science and Technology Park Polaris, Piscina Manna, 09050 Pula (CA), Italy
| | - Giovanna Pira
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Maria Rosaria Muroni
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Giovanni Sotgiu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Laura Saderi
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Elena Uleri
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Maurizio Caocci
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Gabriele Ibba
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Maria Rosaria Cesaraccio
- Department of Prevention, Registro Tumori Provincia di Sassari, ASSL Sassari-ATS Sardegna, Via Rizzeddu 21, Sassari, Italy
| | - Caterina Serra
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Ciriaco Carru
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Alessandra Manca
- Department of Pathology, AOU Sassari, Via Matteotti 60, 07100 Sassari, Italy
| | - Francesca Sanges
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Alberto Porcu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| | - Antonia Dolei
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43-b, 07100 Sassari, Italy
| | - Antonio Mario Scanu
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
| | - Paolo Cossu Rocca
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy.
- Department of Diagnostic Services, "Giovanni Paolo II" Hospital, ASSL Olbia-ATS Sardegna, Via Bazzoni-Sircana, 07026 Olbia, Italy.
| | - Maria Rosaria De Miglio
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy
| |
Collapse
|
364
|
Islam M, Nishinakamura R. How to rebuild the kidney: recent advances in kidney organoids. J Biochem 2019; 166:7-12. [PMID: 30847486 DOI: 10.1093/jb/mvz021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 03/06/2019] [Indexed: 01/31/2023] Open
Abstract
It is difficult to restore kidney function once it has become severely impaired. Although kidney transplantation is a curative therapy, donor numbers remain limited. Thus, the generation of kidney organoids (mainly comprising glomeruli and renal tubules) from multipotent stem cells represents an important advance in regenerative medicine of the kidney. Recently, a protocol that can generate the higher-order structure of the mouse embryonic kidney was reported. Kidney organoids are now being used for disease modelling, and may eventually be applicable for clinical transplantation. In this review, we summarize the recent advances in kidney organoid research, and discuss the issues to be resolved toward kidney reconstruction.
Collapse
Affiliation(s)
- Mazharul Islam
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
365
|
DLC1 deficiency and YAP signaling drive endothelial cell contact inhibition of growth and tumorigenesis. Oncogene 2019; 38:7046-7059. [PMID: 31409902 DOI: 10.1038/s41388-019-0944-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/12/2019] [Accepted: 05/27/2019] [Indexed: 12/14/2022]
Abstract
Deleted in Liver Cancer 1 (DLC1) is a tumor suppressor gene deleted in many cancers, including angiosarcoma, an aggressive malignancy of endothelial cell derivation. DLC1-deficiency in primary endothelial cells causes the loss of cell contact inhibition of growth through incompletely defined mechanisms. We report that DLC1 is a regulator of YAP, a transcriptional coactivator of proliferation-promoting and tumor-promoting genes; when confluent, active/nuclear YAP was significantly more abundant in DLC1-deficient endothelial cells compared with control cells. We also found that YAP is a required effector of the loss of cell contact inhibition of growth manifested by DLC1-deficient endothelial cells, as the silencing of YAP prevents this loss. Consistently, human angiosarcomas specimens contained a significantly greater proportion of DLC1- tumor cells with nuclear YAP compared with the DLC1+ normal cells in the adjacent tissue. Verteporfin, an inhibitor of YAP, significantly reduced angiosarcoma growth in mice. These results identify YAP as a previously unrecognized effector of DLC1 deficiency-associated loss of cell contact growth inhibition in endothelial cells and a potential therapeutic target in angiosarcoma.
Collapse
|
366
|
Yin L, Li W, Wang G, Shi H, Wang K, Yang H, Peng B. NR1B2 suppress kidney renal clear cell carcinoma (KIRC) progression by regulation of LATS 1/2-YAP signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:343. [PMID: 31391070 PMCID: PMC6686564 DOI: 10.1186/s13046-019-1344-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Kidney Renal Clear Cell Carcinoma (KIRC) accounts for 75% of all renal cancers. Previous study had conflict evidences regarding NR1B2 role in cancer, and its expression and biological role in KIRC remained unclear. Our aims were to characterize the role of NR1B2 in KIRC. METHODS NR1B2 expression in TCGA database were analyzed. Clinical KIRC samples were examined by RT-PCR, western blot and tissue microarray (TMA). The relationship between NR1B2 expression and the clinical characteristics were evaluated. KIRC cell line were stably overexpressed NR1B2 or with an NR1B2 knocked down using lentivirus system. The cells were analyzed by migration and invasion assay, then injected into nude mice to assess tumor growth and metastasis. EMT marker expression and LATS 1/2-YAP pathway demonstration were detected by the TCGA database and western blot. RESULTS The expression of NR1B2 in KIRC was significantly down-regulated in the TCGA database and our clinical samples. Moreover, NR1B2 expression negatively correlated with tumor stage and positively correlated with overall and disease-free survival rate. Univariate and multivariate analyses indicated the expression level of NR1B2 could be used as an independent factor for predicting the prognosis of KIRC. Overexpression NR1B2 significantly inhibited and knockdown NR1B2 markedly promoted KIRC cell invasion and metastasis both in vitro and in vivo. Mechanistic investigations revealed that NR1B2 might be a tumor suppressor to inhibit EMT through the LATS1/2-YAP pathway. CONCLUSIONS our results defined NR1B2 as a tumor suppressor in KIRC that restricted EMT by the LATS1/2-YAP pathway.
Collapse
Affiliation(s)
- Lei Yin
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Wenjia Li
- Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangchun Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Heng Shi
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China.,Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, China
| | - Keyi Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Huan Yang
- Department of Urology, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China.
| |
Collapse
|
367
|
CSCs in Breast Cancer-One Size Does Not Fit All: Therapeutic Advances in Targeting Heterogeneous Epithelial and Mesenchymal CSCs. Cancers (Basel) 2019; 11:cancers11081128. [PMID: 31394796 PMCID: PMC6721464 DOI: 10.3390/cancers11081128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/02/2019] [Accepted: 08/04/2019] [Indexed: 02/06/2023] Open
Abstract
Unlike other breast cancer subtypes, triple-negative breast cancer (TNBC) has no specific targets and is characterized as one of the most aggressive subtypes of breast cancer that disproportionately accounts for the majority of breast cancer-related deaths. Current conventional chemotherapeutics target the bulk tumor population, but not the cancer stem cells (CSCs) that are capable of initiating new tumors to cause disease relapse. Recent studies have identified distinct epithelial-like (E) ALDH+ CSCs, mesenchymal-like (M) CD44+/CD24- CSCs, and hybrid E/M ALDH+/CD44+/CD24- CSCs. These subtypes of CSCs exhibit differential signal pathway regulations, possess plasticity, and respond differently to treatment. As such, co-inhibition of different subtypes of CSCs is key to viable therapy. This review serves to highlight different pathway regulations in E and M CSCs in TNBC, and to further describe their role in disease progression. Potential inhibitors targeting E and/or M CSCs based on clinical trials are summarized for further investigation. Since future research needs to adopt suitable tumor models and take into account the divergence of E and M CSCs for the development of effective treatments, TNBC models for clinically translatable studies are further discussed.
Collapse
|
368
|
Zheng Y, Pan D. The Hippo Signaling Pathway in Development and Disease. Dev Cell 2019; 50:264-282. [PMID: 31386861 PMCID: PMC6748048 DOI: 10.1016/j.devcel.2019.06.003] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/23/2019] [Accepted: 06/09/2019] [Indexed: 12/13/2022]
Abstract
The Hippo signaling pathway regulates diverse physiological processes, and its dysfunction has been implicated in an increasing number of human diseases, including cancer. Here, we provide an updated review of the Hippo pathway; discuss its roles in development, homeostasis, regeneration, and diseases; and highlight outstanding questions for future investigation and opportunities for Hippo-targeted therapies.
Collapse
Affiliation(s)
- Yonggang Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.
| |
Collapse
|
369
|
Fu CY, Chen MC, Tseng YS, Chen MC, Zhou Z, Yang JJ, Lin YM, Viswanadha VP, Wang G, Huang CY. Fisetin activates Hippo pathway and JNK/ERK/AP-1 signaling to inhibit proliferation and induce apoptosis of human osteosarcoma cells via ZAK overexpression. ENVIRONMENTAL TOXICOLOGY 2019; 34:902-911. [PMID: 31044527 DOI: 10.1002/tox.22761] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 03/28/2019] [Accepted: 04/14/2019] [Indexed: 06/09/2023]
Abstract
Osteosarcoma (OS) is a tumor entity that can cause a large number of cancer-related deaths. Although chemotherapy can decrease proliferation and increase apoptosis of human OS cells, the clinical prognosis remains poor. Fisetin is a flavonol found in fruits and vegetables and is reported to inhibit cell growth in numerous cancers. But the molecular mechanism underlying fisetin in human OS cells is not clear. It is known that sterile-alpha motif and leucine zipper containing kinase (ZAK), a kinase in the MAP3K family, is involved in various cell processes, including proliferation and apoptosis. In our lab, we have demonstrated that overexpression of ZAK can induce apoptosis in human OS cells. In the previous studies, MAP4K, the upstream of MAP3K, can act in parallel to MST1/2 to activate LATS1/2 in the Hippo pathway. Turning on the Hippo pathway can decrease proliferation and otherwise cause cell apoptosis in cancer cells. In this study, we found that fisetin can upregulate ZAK expression to induce the Hippo pathway and mediate the activation of JNK/ERK, the downstream of ZAK, to trigger cell apoptosis via AP-1 dependent manner in human OS cells. These findings reveal a novel molecular mechanism underlying fisetin effect on human OS cells.
Collapse
Affiliation(s)
- Chien-Yao Fu
- Department of Orthopaedics, National Defense Cental Medical Center, Taipei, Taiwan
- Department of Orthopaedics, Taichung Armed Forces General Hospital, Taichung, Taiwan
| | - Mei-Chih Chen
- Medical Center for Exosomes and Mitochondria Related Diseases, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Yan-Shen Tseng
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Ming-Cheng Chen
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Zhengtao Zhou
- Department of Oncological Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Jaw-Ji Yang
- School of Dentistry, Chung-Shan Medical University, Taichung, Taiwan
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
| | | | - Guiqing Wang
- Department of Orthopedics, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong, China
| | - Chih-Yang Huang
- College of Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
370
|
Yang Z, Joyner AL. YAP1 is involved in replenishment of granule cell precursors following injury to the neonatal cerebellum. Dev Biol 2019; 455:458-472. [PMID: 31376393 DOI: 10.1016/j.ydbio.2019.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 01/08/2023]
Abstract
The cerebellum undergoes major rapid growth during the third trimester and early neonatal stage in humans, making it vulnerable to injuries in pre-term babies. Experiments in mice have revealed a remarkable ability of the neonatal cerebellum to recover from injuries around birth. In particular, recovery following irradiation-induced ablation of granule cell precursors (GCPs) involves adaptive reprogramming of Nestin-expressing glial progenitors (NEPs). Sonic hedgehog signaling is required for the initial step in NEP reprogramming; however, the full spectrum of developmental signaling pathways that promote NEP-driven regeneration is not known. Since the growth regulatory Hippo pathway has been implicated in the repair of several tissue types, we tested whether Hippo signaling is involved in regeneration of the cerebellum. Using mouse models, we found that the Hippo pathway transcriptional co-activator YAP1 (Yes-associated protein 1) but not TAZ (transcriptional coactivator with PDZ binding motif, or WWTR1) is required in NEPs for full recovery of cerebellar growth following irradiation one day after birth. Although Yap1 plays only a minor role during normal development in differentiation of NEPs or GCPs, the size of the cerebellum, and in particular the internal granule cell layer produced by GCPs, is significantly reduced in Yap1 mutants after irradiation, and the organization of Purkinje cells and Bergmann glial fibers is disrupted. The initial proliferative response of Yap1 mutant NEPs to irradiation is normal and the cells migrate to the GCP niche, but subsequently there is increased cell death of GCPs and altered migration of granule cells, possibly due to defects in Bergmann glia. Moreover, loss of Taz along with Yap1 in NEPs does not abrogate regeneration or alter development of the cerebellum. Our study provides new insights into the molecular signaling underlying postnatal cerebellar development and regeneration.
Collapse
Affiliation(s)
- Zhaohui Yang
- Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, 10065, United States; Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, United States
| | - Alexandra L Joyner
- Biochemistry, Cell and Molecular Biology Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, 10065, United States; Developmental Biology Program, Sloan Kettering Institute, New York, NY, 10065, United States.
| |
Collapse
|
371
|
Dong X, Meng L, Liu P, Ji R, Su X, Xin Y, Jiang X. YAP/TAZ: a promising target for squamous cell carcinoma treatment. Cancer Manag Res 2019; 11:6245-6252. [PMID: 31360073 PMCID: PMC6625644 DOI: 10.2147/cmar.s197921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/04/2019] [Indexed: 12/03/2022] Open
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are two homologous transcriptional coactivators and the final effectors of the Hippo signaling transduction pathway. The transcriptional activity of YAP/TAZ is dependent on their recruitment to the nucleus, which promotes binding to the transcription factor of TEA domain family members 1–4 (TEAD1-4). In Hippo-signaling pathway, YAP/TAZ is inactivated and its translocation to the nucleus is blocked via a core kinase cascade stimulated by a variety of upstream signals, such as G-protein-coupled receptor signaling, mechanical pressure, and adherens junction signaling. This pathway plays a very important role in regulating organ size, tissue homeostasis, and tumor development. In recent years, many studies have reported upregulation or nuclear localization of YAP/TAZ in a number of human malignancies, such as breast cancer, melanoma, lung cancer, especially squamous cell carcinoma in different organs. A large number of experiments demonstrate that YAP/TAZ activation promotes cancer formation, progression, and metastasis. Therefore, in this review, we summarize the evidence of regulation and function of YAP/TAZ and discuss its role in squamous cell carcinoma. Collectively, this summary strongly suggests that targeting aberrant YAP/TAZ activation is a promising strategy for the suppression of squamous cell carcinoma.
Collapse
Affiliation(s)
- Xiaoming Dong
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China.,Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, People's Republic of China
| | - Lingbin Meng
- Department of Internal Medicine, Florida Hospital, Orlando, FL 32804, USA
| | - Pinyi Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, People's Republic of China
| | - Rui Ji
- Department of Biology, Valencia College, Orlando, FL 32804, USA
| | - Xuling Su
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, People's Republic of China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, People's Republic of China
| | - Xin Jiang
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| |
Collapse
|
372
|
Kaur H, Moreau R. Role of mTORC1 in intestinal epithelial repair and tumorigenesis. Cell Mol Life Sci 2019; 76:2525-2546. [PMID: 30944973 PMCID: PMC11105546 DOI: 10.1007/s00018-019-03085-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 03/08/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022]
Abstract
mTORC1 signaling is the prototypical pathway regulating protein synthesis and cell proliferation. mTORC1 is active in stem cells located at the base of intestinal crypts but silenced as transit-amplifying cells differentiate into enterocytes or secretory cells along the epithelium. After an insult or injury, self-limiting and controlled activation of mTORC1 is critical for the renewal and repair of intestinal epithelium. mTORC1 promotes epithelial cell renewal by driving cryptic stem cell division, and epithelial cell repair by supporting the dedifferentiation and proliferation of enterocytes or secretory cells. Under repeated insult or injury, mTORC1 becomes constitutively active, triggering an irreversible return to stemness, cell division, proliferation, and inflammation among dedifferentiated epithelial cells. Epithelium-derived cytokines promulgate inflammation within the lamina propria, which in turn releases inflammatory factors that act back on the epithelium where undamaged intestinal epithelial cells participate in the pervading state of inflammation and become susceptible to tumorigenesis.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
373
|
Zhang X, Tang JZ, Vergara IA, Zhang Y, Szeto P, Yang L, Mintoff C, Colebatch A, McIntosh L, Mitchell KA, Shaw E, Rizos H, Long GV, Hayward N, McArthur GA, Papenfuss AT, Harvey KF, Shackleton M. Somatic Hypermutation of the YAP Oncogene in a Human Cutaneous Melanoma. Mol Cancer Res 2019; 17:1435-1449. [PMID: 30833299 DOI: 10.1158/1541-7786.mcr-18-0407] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 10/25/2018] [Accepted: 02/28/2019] [Indexed: 11/16/2022]
Abstract
Melanoma is usually driven by mutations in BRAF or NRAS, which trigger hyperactivation of MAPK signaling. However, MAPK-targeted therapies are not sustainably effective in most patients. Accordingly, characterizing mechanisms that co-operatively drive melanoma progression is key to improving patient outcomes. One possible mechanism is the Hippo signaling pathway, which regulates cancer progression via its central oncoproteins YAP and TAZ, although is thought to be only rarely affected by direct mutation. As YAP hyperactivation occurs in uveal melanoma, we investigated this oncogene in cutaneous melanoma. YAP protein expression was elevated in most benign nevi and primary cutaneous melanomas but present at only very low levels in normal melanocytes. In patient-derived xenografts and melanoma cell lines, we observed variable reliance of cell viability on Hippo pathway signaling that was independent of TAZ activity and also of classical melanoma driver mutations such as BRAF and NRAS. Finally, in genotyping studies of melanoma, we observed the first ever hyperactivating YAP mutations in a human cancer, manifest as seven distinct missense point mutations that caused serine to alanine transpositions. Strikingly, these mutate four serine residues known to be targeted by the Hippo pathway and we show that they lead to hyperactivation of YAP. IMPLICATIONS: Our studies highlight the YAP oncoprotein as a potential therapeutic target in select subgroups of melanoma patients, although successful treatment with anti-YAP therapies will depend on identification of biomarkers additional to YAP protein expression.
Collapse
Affiliation(s)
- Xiaomeng Zhang
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jian Zhong Tang
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Olivia Newton John Cancer Research Institute & School of Cancer Medicine, La Trobe University, Heidelberg, Victoria, Australia
| | | | - Youfang Zhang
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| | - Pacman Szeto
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| | - Lie Yang
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- School of Medicine, Tsinghua University, Beijing, China
| | | | | | - Lachlan McIntosh
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- The Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Department of Mathematics and Statistics, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Evangeline Shaw
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Helen Rizos
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
- Melanoma Institute of Australia, Sydney, NSW, Australia
| | | | - Nicholas Hayward
- Melanoma Institute of Australia, Sydney, NSW, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Grant A McArthur
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Anthony T Papenfuss
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- The Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
- Department of Mathematics and Statistics, University of Melbourne, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mark Shackleton
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
374
|
Pocaterra A, Santinon G, Romani P, Brian I, Dimitracopoulos A, Ghisleni A, Carnicer-Lombarte A, Forcato M, Braghetta P, Montagner M, Galuppini F, Aragona M, Pennelli G, Bicciato S, Gauthier N, Franze K, Dupont S. F-actin dynamics regulates mammalian organ growth and cell fate maintenance. J Hepatol 2019; 71:130-142. [PMID: 30878582 DOI: 10.1016/j.jhep.2019.02.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/31/2019] [Accepted: 02/22/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS In vitro, cell function can be potently regulated by the mechanical properties of cells and of their microenvironment. Cells measure these features by developing forces via their actomyosin cytoskeleton, and respond accordingly by regulating intracellular pathways, including the transcriptional coactivators YAP/TAZ. Whether mechanical cues are relevant for in vivo regulation of adult organ homeostasis, and whether this occurs through YAP/TAZ, remains largely unaddressed. METHODS We developed Capzb conditional knockout mice and obtained primary fibroblasts to characterize the role of CAPZ in vitro. In vivo functional analyses were carried out by inducing Capzb inactivation in adult hepatocytes, manipulating YAP/Hippo activity by hydrodynamic tail vein injections, and treating mice with the ROCK inhibitor, fasudil. RESULTS We found that the F-actin capping protein CAPZ restrains actomyosin contractility: Capzb inactivation alters stress fiber and focal adhesion dynamics leading to enhanced myosin activity, increased traction forces, and increased liver stiffness. In vitro, this rescues YAP from inhibition by a small cellular geometry; in vivo, it induces YAP activation in parallel to the Hippo pathway, causing extensive hepatocyte proliferation and leading to striking organ overgrowth. Moreover, Capzb is required for the maintenance of the differentiated hepatocyte state, for metabolic zonation, and for gluconeogenesis. In keeping with changes in tissue mechanics, inhibition of the contractility regulator ROCK, or deletion of the Yap1 mechanotransducer, reverse the phenotypes emerging in Capzb-null livers. CONCLUSIONS These results indicate a previously unsuspected role for CAPZ in tuning the mechanical properties of cells and tissues, which is required in hepatocytes for the maintenance of the differentiated state and to regulate organ size. More generally, it indicates for the first time that mechanotransduction has a physiological role in maintaining liver homeostasis in mammals. LAY SUMMARY The mechanical properties of cells and tissues (i.e. whether they are soft or stiff) are thought to be important regulators of cell behavior. Herein, we found that inactivation of the protein CAPZ alters the mechanical properties of cells and liver tissues, leading to YAP hyperactivation. In turn, this profoundly alters liver physiology, causing organ overgrowth, defects in liver cell differentiation and metabolism. These results reveal a previously uncharacterized role for mechanical signals in the maintenance of adult liver homeostasis.
Collapse
Affiliation(s)
| | - Giulia Santinon
- Department of Molecular Medicine DMM, University of Padova, Italy
| | - Patrizia Romani
- Department of Molecular Medicine DMM, University of Padova, Italy
| | - Irene Brian
- Department of Molecular Medicine DMM, University of Padova, Italy
| | | | - Andrea Ghisleni
- Institute FIRC (Italian Foundation for Cancer Research) of Molecular Oncology (IFOM Institute FIRC for Molecular Oncology), Milan, Italy
| | | | - Mattia Forcato
- Department of Life Sciences, University of Modena and Reggio Emilia, Italy
| | - Paola Braghetta
- Department of Molecular Medicine DMM, University of Padova, Italy
| | - Marco Montagner
- Department of Molecular Medicine DMM, University of Padova, Italy
| | | | | | | | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Italy
| | - Nils Gauthier
- Institute FIRC (Italian Foundation for Cancer Research) of Molecular Oncology (IFOM Institute FIRC for Molecular Oncology), Milan, Italy
| | - Kristian Franze
- Department of Physiology Development and Neuroscience, University of Cambridge, UK
| | - Sirio Dupont
- Department of Molecular Medicine DMM, University of Padova, Italy.
| |
Collapse
|
375
|
D'Agostino L, Nie Y, Goswami S, Tong K, Yu S, Bandyopadhyay S, Flores J, Zhang X, Balasubramanian I, Joseph I, Sakamori R, Farrell V, Li Q, Yang CS, Gao B, Ferraris RP, Yehia G, Bonder EM, Goldenring JR, Verzi MP, Zhang L, Ip YT, Gao N. Recycling Endosomes in Mature Epithelia Restrain Tumorigenic Signaling. Cancer Res 2019; 79:4099-4112. [PMID: 31239271 DOI: 10.1158/0008-5472.can-18-4075] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/11/2019] [Accepted: 06/11/2019] [Indexed: 11/16/2022]
Abstract
The effects of polarized membrane trafficking in mature epithelial tissue on cell growth and cancer progression have not been fully explored in vivo. A majority of colorectal cancers have reduced and mislocalized Rab11, a small GTPase dedicated to trafficking of recycling endosomes. Patients with low Rab11 protein expression have poor survival rates. Using genetic models across species, we show that intact recycling endosome function restrains aberrant epithelial growth elicited by APC or RAS mutations. Loss of Rab11 protein led to epithelial dysplasia in early animal development and synergized with oncogenic pathways to accelerate tumor progression initiated by carcinogen, genetic mutation, or aging. Transcriptomic analysis uncovered an immediate expansion of the intestinal stem cell pool along with cell-autonomous Yki/Yap activation following disruption of Rab11a-mediated recycling endosomes. Intestinal tumors lacking Rab11a traffic exhibited marked elevation of nuclear Yap, upd3/IL6-Stat3, and amphiregulin-MAPK signaling, whereas suppression of Yki/Yap or upd3/IL6 reduced gut epithelial dysplasia and hyperplasia. Examination of Rab11a function in enteroids or cultured cell lines suggested that this endosome unit is required for suppression of the Yap pathway by Hippo kinases. Thus, recycling endosomes in mature epithelia constitute key tumor suppressors, loss of which accelerates carcinogenesis. SIGNIFICANCE: Recycling endosome traffic in mature epithelia constitutes a novel tumor suppressing mechanism.
Collapse
Affiliation(s)
- Luca D'Agostino
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Yingchao Nie
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Sayantani Goswami
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Kevin Tong
- Department of Genetics, Rutgers University, Piscataway, New Jersey
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | | | - Juan Flores
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Xiao Zhang
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | | | - Ivor Joseph
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Ryotaro Sakamori
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Victoria Farrell
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - Qi Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey
| | - Bin Gao
- Department of Internal Medicine, Taixing Chinese Medicine Hospital, Taixing, Jiangsu, China
| | - Ronaldo P Ferraris
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey
| | - Ghassan Yehia
- Rutgers Genome Editing Core Facility, Rutgers University, New Brunswick, New Jersey
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| | - James R Goldenring
- Department of Surgery, Cell and Developmental Biology, and Epithelial Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Michael P Verzi
- Department of Genetics, Rutgers University, Piscataway, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Lanjing Zhang
- Department of Biological Sciences, Rutgers University, Newark, New Jersey.,Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey.,Department of Pathology, Princeton Medical Center, Plainsboro, New Jersey
| | - Y Tony Ip
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey. .,Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
376
|
Kandilya D, Maskomani S, Shyamasundar S, Tambyah PA, Shiao Yng C, Lee RCH, Hande MP, Mallilankaraman K, Chu JJH, Dheen ST. Zika virus alters DNA methylation status of genes involved in Hippo signaling pathway in human neural progenitor cells. Epigenomics 2019; 11:1143-1161. [PMID: 31234652 DOI: 10.2217/epi-2018-0180] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aim: This study was aimed to understand if Zika virus (ZIKV) alters the DNA methylome of human neural progenitor cells (hNPCs). Materials & methods: Whole genome DNA methylation profiling was performed using human methylationEPIC array in control and ZIKV infected hNPCs. Results & conclusion: ZIKV infection altered the DNA methylation of several genes such as WWTR1 (TAZ) and RASSF1 of Hippo signaling pathway which regulates organ size during brain development, and decreased the expression of several centrosomal-related microcephaly genes, and genes involved in stemness and differentiation in human neural progenitor cells. Overall, ZIKV downregulated the Hippo signaling pathway genes which perturb the stemness and differentiation process in hNPCs, which could form the basis for ZIKV-induced microcephaly.
Collapse
Affiliation(s)
- Deepika Kandilya
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Silambarasan Maskomani
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Sukanya Shyamasundar
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Paul Anantharajah Tambyah
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Chan Shiao Yng
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Regina Ching Hua Lee
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Manoor Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Karthik Mallilankaraman
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Justin Jang Hann Chu
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - S Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| |
Collapse
|
377
|
Mussell A, Frangou C, Zhang J. Regulation of the Hippo signaling pathway by deubiquitinating enzymes in cancer. Genes Dis 2019; 6:335-341. [PMID: 31832513 PMCID: PMC6888741 DOI: 10.1016/j.gendis.2019.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/04/2019] [Accepted: 06/18/2019] [Indexed: 12/30/2022] Open
Abstract
Regulation of the Hippo signaling pathway is essential for normal organ growth and tissue homeostasis. The proteins that act to regulate this pathway are important for ensuring proper function and cellular location. Deubiquitinases (DUBs) are a family of proteases that act upon many proteins. While ubiquitinases add ubiquitin and target proteins for degradation, DUBs act by removing ubiquitin (Ub) moieties. Changes in ubiquitin chain topology results in the stabilization of proteins, membrane trafficking, and the alteration of cellular localization. While the roles of these proteins have been well established in a cancer setting, their convergence in cancer is still under investigation. In this review, we discuss the roles that DUBs play in the regulation of the Hippo signaling pathway for homeostasis and disease.
Collapse
Affiliation(s)
- Ashley Mussell
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14261, USA
| | - Costa Frangou
- Harvard TH Chan School of Public Health, Molecular and Integrative Physiological Sciences, Boston, MA 02115, USA
| | - Jianmin Zhang
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14261, USA
| |
Collapse
|
378
|
Zhao W, Li LW, Tian RF, Dong QF, Li PQ, Yan ZF, Yang X, Huo JL, Fei Z, Zhen HN. Truncated TEAD-binding protein of TAZ inhibits glioma survival through the induction of apoptosis and repression of epithelial-mesenchymal transition. J Cell Biochem 2019; 120:17337-17344. [PMID: 31209945 DOI: 10.1002/jcb.28997] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022]
Abstract
Transcriptional coactivator with PDZ-binding motif (TAZ), a Hippo pathway downstream effector, promotes tumor progression by serving as a transcriptional coactivator with TEAD. Here, we introduced a new construct which can express the TEAD-binding domain of TAZ protein (TAZBD), and determined its antitumor effect in malignant glioma both in vitro and in vivo. We first observed that TAZ was upregulated in glioma tissues and related to malignant clinicopathologic characteristic, indicating the crucial role of TAZ during glioma progression. In U87 and U251 cells, TAZBD expression increased the proportion of apoptotic cells, and suppressed the colony formation and tumorigenicity. Further, TAZBD also decreased cell metastasis through the repression of epithelial-mesenchymal transition. The mechanistic study showed that TAZBD suppression of glioma cells was predominantly through blocking the TAZ-TEAD complex formation by competing with endogenous TAZ. Thus, the gene therapy of malignant glioma through blocking TAZ-TEAD complex by TAZBD may provide a new way for the targeted therapy of glioma.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Medical Affairs, Ludaopei Hospital, Beijing, China
| | - Li-Wen Li
- Department of Bioscience, College of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Rui-Feng Tian
- Department of Infection, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qiu-Feng Dong
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Peng-Qi Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhi-Feng Yan
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xin Yang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jun-Li Huo
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hai-Ning Zhen
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
379
|
Hong L, Li Y, Liu Q, Chen Q, Chen L, Zhou D. The Hippo Signaling Pathway in Regenerative Medicine. Methods Mol Biol 2019; 1893:353-370. [PMID: 30565146 DOI: 10.1007/978-1-4939-8910-2_26] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The major role of Hippo signaling is to inhibit their downstream effectors YAP/TAZ for organ size control during development and regeneration (Nat Rev Drug Discov 13(1):63-79, 2014; Dev Cell 19(4):491-505, 2010; Cell 163(4):811-828, 2015). We and others have demonstrated that the genetic disruption of kinases Mst1 and Mst2 (Mst1/2), the core components of Hippo signaling, results in YAP activation and sustained liver growth, thereby leading to an eight- to tenfold increase in liver size within 3 months and occurrence of liver cancer within 5 months (Curr Biol 17(23):2054-2060, 2007; Cancer Cell 16(5):425-438, 2009; Cell 130(6):1120-1133, 2007; Cancer Cell 31(5):669-684 e667, 2017; Nat Commun 6:6239, 2015; Cell Rep 3(5):1663-1677, 2013). XMU-MP-1, an Mst1/2 inhibitor, is able to augment mouse liver and intestinal repair and regeneration in both acute and chronic injury mouse models (Sci Transl Med 8:352ra108, 2016).In addition, YAP-deficient mice show an impaired intestinal regenerative response after DSS treatment or gamma irradiation (Proc Natl Acad Sci U S A 108(49):E1312-1320, 2011; Nature 493(7430):106-110, 2013; Genes Dev 24(21):2383-2388, 2010; J Vis Exp (111), 2010). IBS008738, a TAZ activator, facilitates muscle repair after cardiotoxin-induced muscle injury (Mol Cell Biol. 2014;34(9):1607-21). Deletion of Salvador (Sav) in mouse hearts enhances cardiomyocyte regeneration with reduced fibrosis and recovery of pumping function after myocardial infarction (MI) or resection of mouse cardiac apex (Development 140(23):4683-4690, 2013; Sci Signal 8(375):ra41, 2015; Nature 550(7675):260-264, 2017). This chapter provides a detailed description of procedures and important considerations when performing the protocols for the respective assays used to determine the effects of Hippo signaling on tissue repair and regeneration.
Collapse
Affiliation(s)
- Lixin Hong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yuxi Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qingxu Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qinghua Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lanfen Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Dawang Zhou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
380
|
Hyun J, Oh SH, Premont RT, Guy CD, Berg CL, Diehl AM. Dysregulated activation of fetal liver programme in acute liver failure. Gut 2019; 68:1076-1087. [PMID: 30670575 PMCID: PMC6580749 DOI: 10.1136/gutjnl-2018-317603] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/03/2018] [Accepted: 12/20/2018] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Uncertainty about acute liver failure (ALF) pathogenesis limits therapy. We postulate that ALF results from excessive reactivation of a fetal liver programme that is induced in hepatocytes when acutely injured livers regenerate. To evaluate this hypothesis, we focused on two molecules with known oncofetal properties in the liver, Yes-associated protein-1 (YAP1) and Insulin-like growth factor-2 RNA-binding protein-3 (IGF2BP3). DESIGN We compared normal liver with explanted livers of patients with ALF to determine if YAP1 and IGF2BP3 were induced; assessed whether these factors are upregulated when murine livers regenerate; determined if YAP1 and IGF2BP3 cooperate to activate the fetal programme in adult hepatocytes; and identified upstream signals that control these factors and thereby hepatocyte maturity during recovery from liver injury. RESULTS Livers of patients with ALF were massively enriched with hepatocytes expressing IGF2BP3, YAP1 and other fetal markers. Less extensive, transient accumulation of similar fetal-like cells that were proliferative and capable of anchorage-independent growth occurred in mouse livers that were regenerating after acute injury. Fetal reprogramming of hepatocytes was YAP1-dependent and involved YAP1-driven reciprocal modulation of let7 microRNAs and IGF2BP3, factors that negatively regulate each other to control fate decisions in fetal cells. Directly manipulating IGF2BP3 expression controlled the fetal-like phenotype regardless of YAP1 activity, proving that IGF2BP3 is the proximal mediator of this YAP1-directed fate. CONCLUSION After acute liver injury, hepatocytes are reprogrammed to fetal-like cells by a YAP1-dependent mechanism that differentially regulates let7 and IGF2BP3, identifying novel therapeutic targets for ALF.
Collapse
Affiliation(s)
- Jeongeun Hyun
- Department of Medicine, Duke University, Durham, North Carolina, USA
- Regeneration Next, Duke University School of Medicine, Durham, North Carolina, USA
| | - Seh-Hoon Oh
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Richard T Premont
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Cynthia D Guy
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Carl L Berg
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Anna Mae Diehl
- Department of Medicine, Duke University, Durham, North Carolina, USA
| |
Collapse
|
381
|
Sugihara T, Isomoto H, Gores G, Smoot R. YAP and the Hippo pathway in cholangiocarcinoma. J Gastroenterol 2019; 54:485-491. [PMID: 30815737 PMCID: PMC6536462 DOI: 10.1007/s00535-019-01563-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 02/20/2019] [Indexed: 02/04/2023]
Abstract
Cholangiocarcinoma (CCA) has an increasing incidence and remains a difficult to treat malignancy. In a search for more effective treatment options, progress has been made in identifying molecular drivers of oncogenic signaling including IDH mutations and FGFR2 fusions. In addition, multiple investigators have identified increased activity of YAP, the effector protein of the Hippo pathway, in CCA. The Hippo pathway regulates organ size, cellular proliferation, and apoptosis via YAP, a transcriptional co-activator. Targeting of the pathway has been difficult due the lack of a dedicated cell-surface receptor. However, more recently, additional cross-regulatory pathways have been identified that are potentially targetable. In this review, we address the current treatment landscape for CCA, the Hippo pathway broadly, animal models of CCA with attention to Hippo-related models, and the current strategies for targeting YAP.
Collapse
Affiliation(s)
- Takaaki Sugihara
- Division of Medicine and Clinical Science, Department of Multidisciplinary Internal Medicine, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Hajime Isomoto
- Division of Medicine and Clinical Science, Department of Multidisciplinary Internal Medicine, Tottori University Faculty of Medicine, Yonago, Tottori, Japan
| | - Gregory Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Rory Smoot
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic College of Medicine and Science, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
382
|
Liu C, Cheng X, Chen J, Wang Y, Wu X, Tian R, Liu B, Ding X, Sun Q, Gong W. Suppression of YAP/TAZ-Notch1-NICD axis by bromodomain and extraterminal protein inhibition impairs liver regeneration. Am J Cancer Res 2019; 9:3840-3852. [PMID: 31281517 PMCID: PMC6587347 DOI: 10.7150/thno.33370] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/06/2019] [Indexed: 12/16/2022] Open
Abstract
Background and aims: Biological mechanisms that control liver regeneration remain poorly defined. However, these mechanisms are remarkable issues in the clinic that affect management of hepatic loss caused by liver surgery, traumatic injury, chronic infection, or liver poisoning. Increasing evidence has shown that various growth factors, cytokines, and metabolic signaling pathways affect the liver regenerative process. Our aim is to study the effect of bromodomain and extraterminal (BET) protein inhibition on liver regeneration and its mechanism. Methods: We studied the role of BET protein inhibitor, JQ1, in liver regeneration in a mouse model after 70% partial hepatectomy (PH). We evaluated yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ) and Notch signaling pathways, which were affected by BET protein inhibitor in mouse hepatic tissues and primary hepatocytes in vivo and AML12 cell lines in vitro. We evaluated the relationship of YAP/TAZ and Notch signaling pathway using YAP/TAZ pathway inhibitor in liver regeneration in vivo. Moreover, we analyzed the relationship of YAP/TAZ and Notch signaling pathways via overexpression or RNA silencing of Yap in AML12 cells. Furthermore, we used Yap overexpression mouse model to examine whether it can rescue liver regeneration damage caused by inhibition of BET proteins. Results: In this study, we report that BET protein inhibitor JQ1 molecule impairs the early phase of liver regeneration in a mouse model after 70% PH. Mechanistically, YAP/TAZ and Notch1-NICD pathways were suppressed by BET protein inhibitor in mouse hepatic tissues and primary hepatocytes in vivo and mouse AML12 cell lines in vitro. By using YAP/TAZ pathway inhibitor, we confirmed that the liver regeneration and the activation of Notch pathway were impaired by the inhibition of YAP/TAZ pathway in vivo. Furthermore, the study showed that Yap knockdown by shRNA in normal mouse hepatic cell line downregulated Notch1 signal transduction, whereas Yap overexpression promoted Notch1-NICD signals. Specific overexpression of Yap in mouse liver could rescue the effect of BET protein inhibition on liver regeneration injury. Conclusion: These results revealed the crucial role of the YAP/TAZ-Notch1-NICD axis in liver regeneration. Therefore, BET protein inhibitors must be used in caution in the treatment of hepatic diseases by reason of its suppressive roles in liver regeneration.
Collapse
|
383
|
Yan B, Li T, Shen L, Zhou Z, Liu X, Wang X, Sun X. Simultaneous knockdown of YAP and TAZ increases apoptosis of hepatocellular carcinoma cells under hypoxic condition. Biochem Biophys Res Commun 2019; 515:275-281. [PMID: 31146919 DOI: 10.1016/j.bbrc.2019.05.143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the common malignant tumors of the digestive system and its five-year survival rate is low. Hypoxia is an important feature of HCC, which can promote cell death resistance. However, the key regulator of HCC cell survival remains elusive in the hypoxic condition. Emerging researches have showed that the Hippo signaling pathway is involved in the initiation and progression of HCC. Here, we provide evidence that key downstream effectors YAP and its paralog TAZ play vital role in apoptosis resistance of HCC cells under hypoxia. Knockdown of YAP or TAZ does not affect the survival of HCC cells in normoxic and hypoxic microenvironment. In addition, the rate of apoptosis by knockdown or inhibition of both YAP and TAZ under hypoxic condition is largely higher than which under normoxia. In conclusion, simultaneous knockdown or inhibition of YAP and TAZ promote apoptosis of HCC cells dramatically. To our knowledge, this is the first research to explore the role of both YAP and TAZ in HCC hypoxic microenvironment in vitro and in vivo. Therefore, it may be useful for establishing novel targeted therapies of HCC, especially subtypes with plenty of hypoxic areas.
Collapse
Affiliation(s)
- Bin Yan
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyi Li
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Shen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhijie Zhou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueni Liu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoliang Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xing Sun
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
384
|
Kim CL, Choi SH, Mo JS. Role of the Hippo Pathway in Fibrosis and Cancer. Cells 2019; 8:cells8050468. [PMID: 31100975 PMCID: PMC6562634 DOI: 10.3390/cells8050468] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/10/2019] [Accepted: 05/14/2019] [Indexed: 12/12/2022] Open
Abstract
The Hippo pathway is the key player in various signaling processes, including organ development and maintenance of tissue homeostasis. This pathway comprises a core kinases module and transcriptional activation module, representing a highly conserved mechanism from Drosophila to vertebrates. The central MST1/2-LATS1/2 kinase cascade in this pathway negatively regulates YAP/TAZ transcription co-activators in a phosphorylation-dependent manner. Nuclear YAP/TAZ bind to transcription factors to stimulate gene expression, contributing to the regenerative potential and regulation of cell growth and death. Recent studies have also highlighted the potential role of Hippo pathway dysfunctions in the pathology of several diseases. Here, we review the functional characteristics of the Hippo pathway in organ fibrosis and tumorigenesis, and discuss its potential as new therapeutic targets.
Collapse
Affiliation(s)
- Cho-Long Kim
- Department of Biomedical Sciences, Cancer Biology Graduate Program, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Sue-Hee Choi
- Department of Biomedical Sciences, Cancer Biology Graduate Program, Ajou University Graduate School of Medicine, Suwon 16499, Korea.
| | - Jung-Soon Mo
- Genomic Instability Research Center (GIRC), Ajou University School of Medicine, Suwon 16499, Korea.
| |
Collapse
|
385
|
Mohagheghi S, Geramizadeh B, Nikeghbalian S, Khodadadi I, Karimi J, Khajehahmadi Z, Gharekhanloo F, Tavilani H. Intricate role of yes‐associated protein1 in human liver cirrhosis: TGF‐β1 still is a giant player. IUBMB Life 2019; 71:1453-1464. [DOI: 10.1002/iub.2052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Sina Mohagheghi
- Department of Clinical Biochemistry, Faculty of MedicineHamadan University of Medical Sciences Hamadan Iran
| | - Bita Geramizadeh
- Transplant Research Center, Pathology DepartmentShiraz University of Medical Sciences Shiraz Iran
| | - Saman Nikeghbalian
- Shiraz Transplant Center, Namazi HospitalShiraz University of Medical Sciences Shiraz Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, Faculty of MedicineHamadan University of Medical Sciences Hamadan Iran
| | - Jamshid Karimi
- Department of Clinical Biochemistry, Faculty of MedicineHamadan University of Medical Sciences Hamadan Iran
| | - Zohreh Khajehahmadi
- Department of Clinical Biochemistry, Faculty of MedicineHamadan University of Medical Sciences Hamadan Iran
| | - Farideh Gharekhanloo
- Clinical Research Development Unit of Besat HospitalHamadan University of Medical Sciences Hamadan Iran
| | - Heidar Tavilani
- Department of Clinical Biochemistry, Faculty of MedicineHamadan University of Medical Sciences Hamadan Iran
| |
Collapse
|
386
|
Kitson SJ, Rosser M, Fischer DP, Marshall KM, Clarke RB, Crosbie EJ. Targeting Endometrial Cancer Stem Cell Activity with Metformin Is Inhibited by Patient-Derived Adipocyte-Secreted Factors. Cancers (Basel) 2019; 11:cancers11050653. [PMID: 31083574 PMCID: PMC6562824 DOI: 10.3390/cancers11050653] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/02/2019] [Accepted: 05/07/2019] [Indexed: 12/18/2022] Open
Abstract
Advanced endometrial cancer continues to have a poor prognosis, due to limited treatment options, which may be further adversely impacted by obesity. Endometrial cancer stem cells have been reported to drive metastasis, chemotherapy resistance and disease relapse, but have yet to be fully characterised and no specific targeted therapies have been identified. Here, we describe the phenotype and genotype of aldehyde dehydrogenase high (ALDHhigh) and CD133+ve endometrial cancer stem cells and how adipocyte secreted mediators block the inhibitory effect of metformin on endometrial cancer stem cell activity. Ishikawa and Hec-1a cell lines were used to characterise ALDHhigh and CD133+ve endometrial cancer cells using flow cytometry, functional sphere assays and quantitative-Polymerase Chain Reaction. The comparative effect of metformin on endometrial cancer stem cell activity and bulk tumour cell proliferation was determined using an Aldefluor and cytotoxicity assay. The impact of adipocyte secreted mediators on metformin response was established using patient-derived conditioned media. ALDHhigh cells demonstrated greater endometrial cancer stem cell activity than CD133+ve cells and had increased expression of stem cell and epithelial-mesenchymal transition genes. Treatment with 0.5-1 mM metformin reduced the proportion and activity of both endometrial cancer stem cell populations (p ≤ 0.05), without affecting cell viability. This effect was, however, inhibited by exposure to patient-derived adipocyte conditioned media. These results indicate a selective and specific effect of metformin on endometrial cancer stem cell activity, which is blocked by adipocyte secreted mediators. Future studies of metformin as an adjuvant therapy in endometrial cancer should be adequately powered to investigate the influence of body mass on treatment response.
Collapse
Affiliation(s)
- Sarah J Kitson
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester M13 9WL, UK.
| | - Matthew Rosser
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
| | - Deborah P Fischer
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
| | - Kay M Marshall
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PL, UK.
| | - Robert B Clarke
- Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M20 4GJ, UK.
| | - Emma J Crosbie
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester M13 9WL, UK.
- Department of Obstetrics and Gynaecology, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK.
| |
Collapse
|
387
|
Planas-Paz L, Sun T, Pikiolek M, Cochran NR, Bergling S, Orsini V, Yang Z, Sigoillot F, Jetzer J, Syed M, Neri M, Schuierer S, Morelli L, Hoppe PS, Schwarzer W, Cobos CM, Alford JL, Zhang L, Cuttat R, Waldt A, Carballido-Perrig N, Nigsch F, Kinzel B, Nicholson TB, Yang Y, Mao X, Terracciano LM, Russ C, Reece-Hoyes JS, Gubser Keller C, Sailer AW, Bouwmeester T, Greenbaum LE, Lugus JJ, Cong F, McAllister G, Hoffman GR, Roma G, Tchorz JS. YAP, but Not RSPO-LGR4/5, Signaling in Biliary Epithelial Cells Promotes a Ductular Reaction in Response to Liver Injury. Cell Stem Cell 2019; 25:39-53.e10. [PMID: 31080135 DOI: 10.1016/j.stem.2019.04.005] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 01/29/2019] [Accepted: 04/04/2019] [Indexed: 12/13/2022]
Abstract
Biliary epithelial cells (BECs) form bile ducts in the liver and are facultative liver stem cells that establish a ductular reaction (DR) to support liver regeneration following injury. Liver damage induces periportal LGR5+ putative liver stem cells that can form BEC-like organoids, suggesting that RSPO-LGR4/5-mediated WNT/β-catenin activity is important for a DR. We addressed the roles of this and other signaling pathways in a DR by performing a focused CRISPR-based loss-of-function screen in BEC-like organoids, followed by in vivo validation and single-cell RNA sequencing. We found that BECs lack and do not require LGR4/5-mediated WNT/β-catenin signaling during a DR, whereas YAP and mTORC1 signaling are required for this process. Upregulation of AXIN2 and LGR5 is required in hepatocytes to enable their regenerative capacity in response to injury. Together, these data highlight heterogeneity within the BEC pool, delineate signaling pathways involved in a DR, and clarify the identity and roles of injury-induced periportal LGR5+ cells.
Collapse
Affiliation(s)
- Lara Planas-Paz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Tianliang Sun
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Monika Pikiolek
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Nadire R Cochran
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Sebastian Bergling
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Vanessa Orsini
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Zinger Yang
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Frederic Sigoillot
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Jasna Jetzer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Maryam Syed
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Marilisa Neri
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Sven Schuierer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Lapo Morelli
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Philipp S Hoppe
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Wibke Schwarzer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Carlos M Cobos
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland; Hospital Aleman, Buenos Aires, Argentina
| | - John L Alford
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Le Zhang
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Rachel Cuttat
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Annick Waldt
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | - Florian Nigsch
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Bernd Kinzel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Thomas B Nicholson
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Yi Yang
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Xiaohong Mao
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | | | - Carsten Russ
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - John S Reece-Hoyes
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | | | - Andreas W Sailer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Tewis Bouwmeester
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Linda E Greenbaum
- Novartis Institutes for Biomedical Research, Novartis Pharma AG, East Hanover, NJ, USA
| | - Jesse J Lugus
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Feng Cong
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Gregory McAllister
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Gregory R Hoffman
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Cambridge, MA, USA
| | - Guglielmo Roma
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Jan S Tchorz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
388
|
Gupta R, Bhatt LK, Johnston TP, Prabhavalkar KS. Colon cancer stem cells: Potential target for the treatment of colorectal cancer. Cancer Biol Ther 2019; 20:1068-1082. [PMID: 31050577 DOI: 10.1080/15384047.2019.1599660] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Despite incessant research, colon cancer still is one of the most common causes of fatalities in both men and women worldwide. Also, nearly 50% of patients with colorectal cancer show tumor recurrence. Recent investigations have highlighted the involvement of colon cancer stem cells (CCSCs) in cancer relapse and chemoresistance. CCSCs deliver a significant protumorigenic niche through persistent overexpression of self-renewal capabilities. Moreover, CSCs cross network with stromal cells, immune infiltrates, and cyotokine-chemokine, which potentiate their aggressive proliferative potential. Targeting CCSCs through small molecule inhibitors, miRNAs, and monoclonal antibodies (mAbs) in in vivo studies has generated compelling evidence for the effectiveness of these various treatments. This review effectively compiles the role of CCSC surface markers and dysregulated and/or upregulated pathways in the pathogenesis of colorectal cancer that can be used to target CCSCs for effective colorectal cancer treatment.
Collapse
Affiliation(s)
- Riya Gupta
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Lokesh Kumar Bhatt
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| | - Thomas P Johnston
- b Division of Pharmacology and Pharmaceutical Sciences , University of Missouri-Kansas City , Kansas City , MO , USA
| | - Kedar S Prabhavalkar
- a Department of Pharmacology , SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai , India
| |
Collapse
|
389
|
Serra D, Mayr U, Boni A, Lukonin I, Rempfler M, Challet Meylan L, Stadler MB, Strnad P, Papasaikas P, Vischi D, Waldt A, Roma G, Liberali P. Self-organization and symmetry breaking in intestinal organoid development. Nature 2019; 569:66-72. [PMID: 31019299 PMCID: PMC6544541 DOI: 10.1038/s41586-019-1146-y] [Citation(s) in RCA: 360] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 03/27/2019] [Indexed: 01/08/2023]
Abstract
Intestinal organoids are complex three-dimensional structures that mimic the cell-type composition and tissue organization of the intestine by recapitulating the self-organizing ability of cell populations derived from a single intestinal stem cell. Crucial in this process is a first symmetry-breaking event, in which only a fraction of identical cells in a symmetrical sphere differentiate into Paneth cells, which generate the stem-cell niche and lead to asymmetric structures such as the crypts and villi. Here we combine single-cell quantitative genomic and imaging approaches to characterize the development of intestinal organoids from single cells. We show that their development follows a regeneration process that is driven by transient activation of the transcriptional regulator YAP1. Cell-to-cell variability in YAP1, emerging in symmetrical spheres, initiates Notch and DLL1 activation, and drives the symmetry-breaking event and formation of the first Paneth cell. Our findings reveal how single cells exposed to a uniform growth-promoting environment have the intrinsic ability to generate emergent, self-organized behaviour that results in the formation of complex multicellular asymmetric structures.
Collapse
Affiliation(s)
- Denise Serra
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Urs Mayr
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Andrea Boni
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- Viventis Microscopy Sàrl, EPFL Innovation Park, Lausanne, Switzerland
| | - Ilya Lukonin
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Markus Rempfler
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | | | - Michael B Stadler
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Petr Strnad
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- Viventis Microscopy Sàrl, EPFL Innovation Park, Lausanne, Switzerland
| | - Panagiotis Papasaikas
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Dario Vischi
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | - Annick Waldt
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Guglielmo Roma
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
390
|
van Soldt BJ, Qian J, Li J, Tang N, Lu J, Cardoso WV. Yap and its subcellular localization have distinct compartment-specific roles in the developing lung. Development 2019; 146:dev.175810. [PMID: 30944105 DOI: 10.1242/dev.175810] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/28/2019] [Indexed: 12/11/2022]
Abstract
Although the Hippo-yes-associated protein (Yap) pathway has been implicated in lung development, the specific roles for Yap and its nucleocytoplasmic shuttling in the developing airway and alveolar compartments remain elusive. Moreover, conflicting results from expression studies and differences in the lung phenotypes of Yap and Hippo kinase null mutants caused controversy over the dynamics and significance of Yap subcellular localization in the developing lung. Here, we show that the aberrant morphogenesis of Yap-deficient lungs results from the disruption of developmental events specifically in distal epithelial progenitors. We also show that activation of nuclear Yap is enough to fulfill the Yap requirements to rescue abnormalities in these lungs. Remarkably, we found that Yap nucleocytoplasmic shuttling is largely dispensable in epithelial progenitors for both branching morphogenesis and sacculation. However, if maintained transcriptionally active in airways, nuclear Yap profoundly alters proximal-distal identity and halts epithelial differentiation. Taken together, these observations provide novel insights into the crucial importance of Hippo-Yap signaling in the lung prenatally.
Collapse
Affiliation(s)
- Benjamin J van Soldt
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, and Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| | - Jun Qian
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, and Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| | - Jiao Li
- National Institute of Biological Sciences, Beijing 102206, China
| | - Nan Tang
- National Institute of Biological Sciences, Beijing 102206, China
| | - Jining Lu
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, and Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| | - Wellington V Cardoso
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, and Department of Genetics and Development, Columbia University Medical Center, New York, NY, 10032, USA
| |
Collapse
|
391
|
Boopathy GTK, Hong W. Role of Hippo Pathway-YAP/TAZ Signaling in Angiogenesis. Front Cell Dev Biol 2019; 7:49. [PMID: 31024911 PMCID: PMC6468149 DOI: 10.3389/fcell.2019.00049] [Citation(s) in RCA: 247] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a highly coordinated process of formation of new blood vessels from pre-existing blood vessels. The process of development of the proper vascular network is a complex process that is crucial for the vertebrate development. Several studies have defined essential roles of Hippo pathway-YAP/TAZ in organ size control, tissue regeneration, and self-renewal. Thus Hippo pathway is one of the central components in tissue homeostasis. There are mounting evidences on the eminence of Hippo pathway-YAP/TAZ in angiogenesis in multiple model organisms. Hippo pathway-YAP/TAZ is now demonstrated to regulate endothelial cell proliferation, migration and survival; subsequently regulating vascular sprouting, vascular barrier formation, and vascular remodeling. Major intracellular signaling programs that regulate angiogenesis concomitantly activate YAP/TAZ to regulate key events in angiogenesis. In this review, we provide a brief overview of the recent findings in the Hippo pathway and YAP/TAZ signaling in angiogenesis.
Collapse
Affiliation(s)
- Gandhi T K Boopathy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
392
|
Francis D, Chanana B, Fernandez B, Gordon B, Mak T, Palacios IM. YAP/Yorkie in the germline modulates the age-related decline of germline stem cells and niche cells. PLoS One 2019; 14:e0213327. [PMID: 30943201 PMCID: PMC6447158 DOI: 10.1371/journal.pone.0213327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 11/19/2022] Open
Abstract
The properties and behaviour of stem cells rely heavily on signaling from the local microenvironment. At the apical end of Drosophila testis, self-renewal and differentiation of germline stem cells (GSCs) are tightly controlled by distinct somatic cells that comprise a specialised stem cell niche known as the hub. The hub maintains GSC homeostasis through adhesion and cell signaling. The Salvador/Warts/Hippo (SWH) pathway, which suppresses the transcriptional co-activator YAP/Yki via a kinase cascade, is a known regulator of stem cell proliferation and differentiation. Here, we show that increasing YAP/Yki expression in the germline, as well as reducing Warts levels, blocks the decrease of GSC numbers observed in aging flies, with only a small increase on their proliferation. An increased expression of YAP/Yki in the germline or a reduction in Warts levels also stymies an age-related reduction in hub cell number, suggesting a bilateral relationship between GSCs and the hub. Conversely, RNAi-based knockdown of YAP/Yki in the germline leads to a significant drop in hub cell number, further suggesting the existence of such a SC-to-niche relationship. All together, our data implicate the SWH pathway in Drosophila GSC maintenance and raise questions about its role in stem cell homeostasis in aging organisms.
Collapse
Affiliation(s)
| | | | - Beatriz Fernandez
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, United Kingdom
| | | | - Tiffany Mak
- University of Cambridge, Cambridge, United Kingdom
| | - Isabel M. Palacios
- University of Cambridge, Cambridge, United Kingdom
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London, United Kingdom
| |
Collapse
|
393
|
Lodge EJ, Santambrogio A, Russell JP, Xekouki P, Jacques TS, Johnson RL, Thavaraj S, Bornstein SR, Andoniadou CL. Homeostatic and tumourigenic activity of SOX2+ pituitary stem cells is controlled by the LATS/YAP/TAZ cascade. eLife 2019; 8:43996. [PMID: 30912742 PMCID: PMC6461440 DOI: 10.7554/elife.43996] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/25/2019] [Indexed: 12/14/2022] Open
Abstract
SOX2 positive pituitary stem cells (PSCs) are specified embryonically and persist throughout life, giving rise to all pituitary endocrine lineages. We have previously shown the activation of the STK/LATS/YAP/TAZ signalling cascade in the developing and postnatal mammalian pituitary. Here, we investigate the function of this pathway during pituitary development and in the regulation of the SOX2 cell compartment. Through loss- and gain-of-function genetic approaches, we reveal that restricting YAP/TAZ activation during development is essential for normal organ size and specification from SOX2+ PSCs. Postnatal deletion of LATS kinases and subsequent upregulation of YAP/TAZ leads to uncontrolled clonal expansion of the SOX2+ PSCs and disruption of their differentiation, causing the formation of non-secreting, aggressive pituitary tumours. In contrast, sustained expression of YAP alone results in expansion of SOX2+ PSCs capable of differentiation and devoid of tumourigenic potential. Our findings identify the LATS/YAP/TAZ signalling cascade as an essential component of PSC regulation in normal pituitary physiology and tumourigenesis. The pituitary is a gland inside the head that releases hormones that control major processes in the body including growth, fertility and stress. Diseases of the pituitary gland can prevent the body from producing the appropriate amounts of hormones, and also include tumours. A population of stem cells in the pituitary known as SOX2 cells divide to make the specialist cells that produce the hormones. This population forms as the pituitary develops in the embryo and continues to contribute new hormone-producing cells throughout life. Signals from inside and outside the gland control how the pituitary develops and maintain the correct balance of different types of cells in the gland in adults. In 2016, Lodge et al. reported that a cascade of signals known as the Hippo pathway is active in mouse and human pituitary glands, but its role remained unclear. Here, Lodge et al. use genetic approaches to study this signalling pathway in the pituitary of mice. The results of the experiments show that the Hippo pathway is essential for the pituitary gland to develop normally in mouse embryos. Furthermore, in adult mice the Hippo pathway is required to maintain the population of SOX2 cells in the pituitary and to regulate their cell numbers. Increasing the level of Hippo signalling in mouse embryos and adult mice led to an expansion of SOX2 stem cells that could generate new specialist cell types, but a further increase generated aggressive tumours that originated from the uncontrolled growth of SOX2 cells. These findings are the first step to understanding how the Hippo pathway works in the pituitary, which may eventually lead to new treatments for tumours and other diseases that affect this gland. The next step towards such treatments will be to carry out further experiments that use drugs to control this pathway and alter the fate of pituitary cells in mice and other animals.
Collapse
Affiliation(s)
- Emily J Lodge
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.,Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Alice Santambrogio
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.,Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - John P Russell
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom
| | - Paraskevi Xekouki
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.,Department of Endocrinology, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Thomas S Jacques
- UCL GOS Institute of Child Health and Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Randy L Johnson
- Department of Cancer Biology, The University of Texas, MD Anderson Cancer Center, Houston, United States
| | - Selvam Thavaraj
- Centre for Oral, Clinical and Translational Sciences, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom
| | - Stefan R Bornstein
- Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom.,Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Cynthia Lilian Andoniadou
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, United Kingdom.,Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
394
|
Yeung YT, Guerrero-Castilla A, Cano M, Muñoz MF, Ayala A, Argüelles S. Dysregulation of the Hippo pathway signaling in aging and cancer. Pharmacol Res 2019; 143:151-165. [PMID: 30910741 DOI: 10.1016/j.phrs.2019.03.018] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/04/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
Human beings are facing emerging degenerative and cancer diseases, in large part, as a consequence of increased life expectancy. In the near future, researchers will have to put even more effort into fighting these new challenges, one of which will be prevention of cancer while continuing to improve the aging process through this increased life expectancy. In the last few decades, relevance of the Hippo pathway on cancer has become an important study since it is a major regulator of organ size control and proliferation. However, its deregulation can induce tumors throughout the body by regulating cell proliferation, disrupting cell polarity, releasing YAP and TAZ from the Scribble complexes and facilitating survival gene expression via activation of TEAD transcription factors. This pathway is also involved in some of the most important mechanisms that control the aging processes, such as the AMP-activated protein kinase and sirtuin pathways, along with autophagy and oxidative stress response/antioxidant defense. This could be the link between two tightly connected processes that could open a broader range of targeted molecular therapies to fight aging and cancer. Therefore, available knowledge of the processes involved in the Hippo pathway during aging and cancer must necessarily be well understood.
Collapse
Affiliation(s)
- Yiu To Yeung
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | | | - Mercedes Cano
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Mario F Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Antonio Ayala
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Sandro Argüelles
- Department of Physiology, Faculty of Pharmacy, University of Seville, Seville, Spain.
| |
Collapse
|
395
|
Manmadhan S, Ehmer U. Hippo Signaling in the Liver - A Long and Ever-Expanding Story. Front Cell Dev Biol 2019; 7:33. [PMID: 30931304 PMCID: PMC6423448 DOI: 10.3389/fcell.2019.00033] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/25/2019] [Indexed: 12/27/2022] Open
Abstract
The first description of Hippo signaling in mammals a little more than 10 years ago showed a striking phenotype in the liver, linking the role of this signaling pathway to organ size control and carcinogenesis. Even though Hippo signaling has been extensively studied in the liver and other organs over the recent years, many open questions remain in our understanding of its role in hepatic physiology and disease. The functions of Hippo signaling extend well beyond cancer and organ size determination: components of upstream Hippo signaling and the downstream effectors YAP and TAZ are involved in a multitude of cell and non-cell autonomous functions including cell proliferation, survival, development, differentiation, metabolism, and cross-talk with the immune system. Moreover, regulation and biological functions of Hippo signaling are often organ or even cell type specific – making its role even more complex. Here, we give a concise overview of the role of Hippo signaling in the liver with a focus on cell-type specific functions. We outline open questions and future research directions that will help to improve our understanding of this important pathway in liver disease.
Collapse
Affiliation(s)
- Saumya Manmadhan
- Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Ursula Ehmer
- Department of Medicine II, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
396
|
Khatri A, Gu JJ, McKernan CM, Xu X, Pendergast AM. ABL kinase inhibition sensitizes primary lung adenocarcinomas to chemotherapy by promoting tumor cell differentiation. Oncotarget 2019; 10:1874-1886. [PMID: 30956771 PMCID: PMC6443011 DOI: 10.18632/oncotarget.26740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 02/15/2019] [Indexed: 01/28/2023] Open
Abstract
Lung cancer is the leading cause of cancer mortality in the United States, with an overall five-year survival rate of ~16%. Non-small cell lung cancer (NSCLC) accounts for ~80% of all lung cancer cases, and the majority (40%) of these are adenocarcinomas. Loss of function point mutations in TP53 (46%) and activating mutations in KRAS (33%) are the most common mutations in human lung adenocarcinomas. Because neither of these genetic alterations are clinically actionable, chemotherapy remains the mainstay of treatment in patients with oncogenic KRAS driver mutations. However, chemoresistance to genotoxic agents such as docetaxel remains a major clinical challenge facing lung cancer patients. Here we show that ABL kinase allosteric inhibitors can be effectively used for the treatment of KrasG12D/+; p53-/- lung adenocarcinomas in an autochthonous mouse model. Unexpectedly, we found that treatment of tumor-bearing mice with an ABL allosteric inhibitor promoted differentiation of lung adenocarcinomas from poorly differentiated tumors expressing basal cell markers to tumors expressing terminal differentiation markers in vivo, which rendered lung adenocarcinomas susceptible to chemotherapy. These findings uncover a novel therapeutic approach for the treatment of lung adenocarcinomas with poor response to chemotherapy.
Collapse
Affiliation(s)
- Aaditya Khatri
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Jing Jin Gu
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Courtney M. McKernan
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Xia Xu
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
397
|
Tsuchiya A, Lu WY. Liver stem cells: Plasticity of the liver epithelium. World J Gastroenterol 2019; 25:1037-1049. [PMID: 30862993 PMCID: PMC6406190 DOI: 10.3748/wjg.v25.i9.1037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/21/2019] [Accepted: 01/26/2019] [Indexed: 02/06/2023] Open
Abstract
The liver has a high regenerative capacity after acute liver injury, but this is often impaired during chronic liver injury. The existence of a dedicated liver stem cell population that acts as a source of regeneration during chronic liver injury has been controversial. Recent advances in transgenic models and cellular reprogramming have provided new insights into the plasticity of the liver epithelium and directions for the development of future therapies. This article will highlight recent findings about the cellular source of regeneration during liver injury and the advances in promoting liver regeneration.
Collapse
Affiliation(s)
- Atsunori Tsuchiya
- Division of Gastroenterology and Hepatology, Graduate school of medical and dental sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Wei-Yu Lu
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, the University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
398
|
Ou H, Chen Z, Xiang L, Fang Y, Xu Y, Liu Q, Hu Z, Li X, Huang Y, Yang D. Frizzled 2-induced epithelial-mesenchymal transition correlates with vasculogenic mimicry, stemness, and Hippo signaling in hepatocellular carcinoma. Cancer Sci 2019; 110:1169-1182. [PMID: 30677195 PMCID: PMC6447835 DOI: 10.1111/cas.13949] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 01/16/2019] [Accepted: 01/20/2019] [Indexed: 12/21/2022] Open
Abstract
Prior observation has indicated that Frizzled 2 (FZD2)‐induced epithelial‐mesenchymal transition (EMT) could be a key step in metastasis and early recurrence of hepatocellular carcinoma (HCC). However, the mechanism underlying tumor development and progression due to aberrant FZD2 expression is poorly defined. Here, we provide evidence that FZD2 is a driver for EMT, cancer stem cell properties, and vasculogenic mimicry (VM) in HCC. We found that FZD2 was highly expressed in two cohorts of Chinese hepatitis B virus‐related HCC patients, and that high FZD2 expression was associated with poor prognosis. Concerning the mechanism, gain‐ and loss‐of‐function experiments showed the oncogenic action of FZD2 in HCC cell proliferation, apoptosis, migration, and invasion. Further investigations in vitro and in vivo suggested that FZD2 promotes the EMT process, enhances stem‐like properties, and confers VM capacity to HCC cells. Notably, integrative RNA sequencing analysis of FZD2‐knockdown cells indicated the enrichment of Hippo signaling pathway. Taken together, our data suggest for the first time that FZD2 could promote clinically relevant EMT, CD44+ stem‐like properties, and the VM phenotype in HCC involving a potential Hippo signaling pathway‐dependent mechanism, and should be considered as a promising therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Huohui Ou
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zhanjun Chen
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Leyang Xiang
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yinghao Fang
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yuyan Xu
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Qin Liu
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zhigang Hu
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Xianghong Li
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yu Huang
- Department of Laboratory Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Dinghua Yang
- Department of Hepatobiliary Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
399
|
Cofre J, Saalfeld K, Abdelhay E. Cancer as an Embryological Phenomenon and Its Developmental Pathways: A Hypothesis regarding the Contribution of the Noncanonical Wnt Pathway. ScientificWorldJournal 2019; 2019:4714781. [PMID: 30940992 PMCID: PMC6421044 DOI: 10.1155/2019/4714781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 12/18/2018] [Accepted: 01/29/2019] [Indexed: 02/07/2023] Open
Abstract
For gastrulation to occur in human embryos, a mechanism that simultaneously regulates many different processes, such as cell differentiation, proliferation, migration, and invasion, is required to consistently and effectively create a human being during embryonic morphogenesis. The striking similarities in the processes of cancer and gastrulation have prompted speculation regarding the developmental pathways involved in their regulation. One of the fundamental requirements for the developmental pathways in gastrulation and cancer is the ability to respond to environmental stimuli, and it has been proposed that the Kaiso and noncanonical Wnt pathways participate in the mechanisms regulating these developmental pathways. In particular, these pathways might also explain the notable differences in invasive capacity between cancers of endodermal and mesodermal origins and cancers of ectodermal origin. Nevertheless, the available information indicates that cancer is an abnormal state of adult human cells in which developmental pathways are reactivated in inappropriate temporal and spatial contexts.
Collapse
Affiliation(s)
- Jaime Cofre
- Laboratório de Embriologia Molecular e Câncer, Universidade Federal de Santa Catarina, Sala 313b, 88040-900 Florianópolis, SC, Brazil
| | - Kay Saalfeld
- Laboratório de Filogenia Animal, Universidade Federal de Santa Catarina, Brazil
| | - Eliana Abdelhay
- Divisão de Laboratórios do CEMO, Instituto Nacional do Câncer, Rio de Janeiro, Brazil
| |
Collapse
|
400
|
He C, Lv X, Huang C, Hua G, Ma B, Chen X, Angeletti PC, Dong J, Zhou J, Wang Z, Rueda BR, Davis JS, Wang C. YAP1-LATS2 feedback loop dictates senescent or malignant cell fate to maintain tissue homeostasis. EMBO Rep 2019; 20:e44948. [PMID: 30755404 PMCID: PMC6399607 DOI: 10.15252/embr.201744948] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/16/2022] Open
Abstract
Dysfunction of the homeostasis-maintaining systems in specific cell types or tissues renders the organism susceptible to a range of diseases, including cancers. One of the emerging mechanisms for maintaining tissue homeostasis is cellular senescence. Here, we report that the Hippo pathway plays a critical role in controlling the fate of ovarian cells. Hyperactivation of Yes-associated protein 1 (YAP1), the major effector of the Hippo pathway, induces senescence in cultured primary human ovarian surface epithelial cells (hOSEs). Large tumor suppressor 2 (LATS2), the primary upstream negative regulator of YAP1, is elevated in both YAP1-induced and natural replicative-triggered senescence. Deletion of LATS2 in hOSEs prevents these cells from natural replicative and YAP1-induced senescence. Most importantly, loss of LATS2 switches ovarian cells from YAP-induced senescence to malignant transformation. Our results demonstrate that LATS2 and YAP1, two major components of the Hippo/YAP signaling pathway, form a negative feedback loop to control YAP1 activity and prevent ovarian cells from malignant transformation. Human cancer genomic data extracted from TCGA datasets further confirm the clinical relevance of our finding.
Collapse
Affiliation(s)
- Chunbo He
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xiangmin Lv
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Cong Huang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Guohua Hua
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bowen Ma
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peter C Angeletti
- Nebraska Center for Virology, School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Jixin Dong
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jin Zhou
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Department of Obstetrics and gynecology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Zhengfeng Wang
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Omaha Veterans Affairs Medical Center, Omaha, NE, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|