1
|
Cheng Y, Hou W, Fang H, Yan Y, Lu Y, Meng T, Ma C, Liu Q, Zhou Z, Li H, Li H, Xiao N. SENP2-NDR2-p21 axis modulates lung cancer cell growth. Eur J Pharmacol 2024; 978:176761. [PMID: 38908669 DOI: 10.1016/j.ejphar.2024.176761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Sentrin/small ubiquitin-like modifier (SUMO)-specific proteases (SENPs) perform pivotal roles in SUMO maturation and recycling, which modulate the balance of SUMOylation/de-SUMOylation and spatiotemporal functions of SUMOylation targets. The malfunction of SENPs often results in cellular dysfunction and various diseases. However, studies rarely investigated the correlation between SENP2 and lung cancer. This study revealed that SENP2 is a required contributor to lung cancer-cell growth and targets nuclear Dbf2-related 2 (NDR2, also known as serine/threonine kinase 38L or STK38L) for de-SUMOylation, which improves NDR2 kinase activity. This condition leads to the instability of downstream target p21 in accelerating the G1/S cell cycle transition and suggests SENP2 as a promising therapeutic target for lung cancer in the future. Specifically, astragaloside IV, an active ingredient of Jinfukang Oral Liquid (JOL, a clinical combination antilung cancer drug approved by the National Food and Drug Administration (FDA) of China), can repress lung cancer-cell growth via the SENP2-NDR2-p21 axis, which provides new insights into the molecular mechanism of JOL for lung cancer treatment.
Collapse
Affiliation(s)
- Yixuan Cheng
- Institute of Traditional Chinese Medicine Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Longhua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wanxin Hou
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Houshun Fang
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yinjie Yan
- Department of Medical Affairs, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Lu
- Longhua Clinical Medical College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian Meng
- Department of Breast Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chunshuang Ma
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qinghai Liu
- Department of Performance Management, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhiyi Zhou
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Department of Oncology, Tianshan Hospital of Traditional Chinese Medicine in Changning District, Shanghai, China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Fujian Children's Hospital, Fujian Branch of Shanghai Children's Medical Center Affiliated to Shanghai Jiao Tong University School of Medicine, Fujian, China.
| | - Hegen Li
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Ning Xiao
- Institute of Traditional Chinese Medicine Surgery, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
2
|
Zhao Y, Sun B, Fu X, Zuo Z, Qin H, Yao K. YAP in development and disease: Navigating the regulatory landscape from retina to brain. Biomed Pharmacother 2024; 175:116703. [PMID: 38713948 DOI: 10.1016/j.biopha.2024.116703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/30/2024] [Accepted: 05/01/2024] [Indexed: 05/09/2024] Open
Abstract
The distinctive role of Yes-associated protein (YAP) in the nervous system has attracted widespread attention. This comprehensive review strategically uses the retina as a vantage point, embarking on an extensive exploration of YAP's multifaceted impact from the retina to the brain in development and pathology. Initially, we explore the crucial roles of YAP in embryonic and cerebral development. Our focus then shifts to retinal development, examining in detail YAP's regulatory influence on the development of retinal pigment epithelium (RPE) and retinal progenitor cells (RPCs), and its significant effects on the hierarchical structure and functionality of the retina. We also investigate the essential contributions of YAP in maintaining retinal homeostasis, highlighting its precise regulation of retinal cell proliferation and survival. In terms of retinal-related diseases, we explore the epigenetic connections and pathophysiological regulation of YAP in diabetic retinopathy (DR), glaucoma, and proliferative vitreoretinopathy (PVR). Lastly, we broaden our exploration from the retina to the brain, emphasizing the research paradigm of "retina: a window to the brain." Special focus is given to the emerging studies on YAP in brain disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD), underlining its potential therapeutic value in neurodegenerative disorders and neuroinflammation.
Collapse
Affiliation(s)
- Yaqin Zhao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Bin Sun
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Xuefei Fu
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhuan Zuo
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Huan Qin
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Kai Yao
- Institute of Visual Neuroscience and Stem Cell Engineering, Wuhan University of Science and Technology, Wuhan 430065, China; College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
3
|
Palamiuc L, Johnson JL, Haratipour Z, Loughran RM, Choi WJ, Arora GK, Tieu V, Ly K, Llorente A, Crabtree S, Wong JC, Ravi A, Wiederhold T, Murad R, Blind RD, Emerling BM. Hippo and PI5P4K signaling intersect to control the transcriptional activation of YAP. Sci Signal 2024; 17:eado6266. [PMID: 38805583 PMCID: PMC11283293 DOI: 10.1126/scisignal.ado6266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/09/2024] [Indexed: 05/30/2024]
Abstract
Phosphoinositides are essential signaling molecules. The PI5P4K family of phosphoinositide kinases and their substrates and products, PI5P and PI4,5P2, respectively, are emerging as intracellular metabolic and stress sensors. We performed an unbiased screen to investigate the signals that these kinases relay and the specific upstream regulators controlling this signaling node. We found that the core Hippo pathway kinases MST1/2 phosphorylated PI5P4Ks and inhibited their signaling in vitro and in cells. We further showed that PI5P4K activity regulated several Hippo- and YAP-related phenotypes, specifically decreasing the interaction between the key Hippo proteins MOB1 and LATS and stimulating the YAP-mediated genetic program governing epithelial-to-mesenchymal transition. Mechanistically, we showed that PI5P interacted with MOB1 and enhanced its interaction with LATS, thereby providing a signaling connection between the Hippo pathway and PI5P4Ks. These findings reveal how these two important evolutionarily conserved signaling pathways are integrated to regulate metazoan development and human disease.
Collapse
Affiliation(s)
| | - Jared L. Johnson
- Weill Cornell Medicine, Meyer Cancer Center, New York, NY 10021
- Weill Cornell Medicine, Department of Medicine, New York, NY 10021
| | - Zeinab Haratipour
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN 37232
- Austin Peay State University, Clarksville, TN, 37044
| | | | - Woong Jae Choi
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN 37232
| | | | | | - Kyanh Ly
- Sanford Burnham Prebys, La Jolla, CA 92037
| | | | | | - Jenny C.Y. Wong
- Weill Cornell Medicine, Meyer Cancer Center, New York, NY 10021
- New York University Grossman School of Medicine, Department of Cell Biology, New York, NY 10016, USA
| | | | | | - Rabi Murad
- Sanford Burnham Prebys, La Jolla, CA 92037
| | - Raymond D. Blind
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Nashville, TN 37232
| | | |
Collapse
|
4
|
Jonischkies K, del Angel M, Demiray YE, Loaiza Zambrano A, Stork O. The NDR family of kinases: essential regulators of aging. Front Mol Neurosci 2024; 17:1371086. [PMID: 38803357 PMCID: PMC11129689 DOI: 10.3389/fnmol.2024.1371086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Aging is defined as a progressive decline of cognitive and physiological functions over lifetime. Since the definition of the nine hallmarks of aging in 2013 by López-Otin, numerous studies have attempted to identify the main regulators and contributors in the aging process. One interesting group of proteins whose participation has been implicated in several aging hallmarks are the nuclear DBF2-related (NDR) family of serine-threonine AGC kinases. They are one of the core components of the Hippo signaling pathway and include NDR1, NDR2, LATS1 and LATS2 in mammals, along with its highly conserved metazoan orthologs; Trc in Drosophila melanogaster, SAX-1 in Caenorhabditis elegans, CBK1, DBF20 in Saccharomyces cerevisiae and orb6 in Saccharomyces pombe. These kinases have been independently linked to the regulation of widely diverse cellular processes disrupted during aging such as the cell cycle progression, transcription, intercellular communication, nutrient homeostasis, autophagy, apoptosis, and stem cell differentiation. However, a comprehensive overview of the state-of-the-art knowledge regarding the post-translational modifications of and by NDR kinases in aging has not been conducted. In this review, we summarize the current understanding of the NDR family of kinases, focusing on their relevance to various aging hallmarks, and emphasize the growing body of evidence that suggests NDR kinases are essential regulators of aging across species.
Collapse
Affiliation(s)
- Kevin Jonischkies
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Miguel del Angel
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Yunus Emre Demiray
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Allison Loaiza Zambrano
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Center for Behavioral Brain Science, Magdeburg, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
- German Center for Mental Health (DZPG), Jena-Magdeburg-Halle, Germany
| |
Collapse
|
5
|
Bai Y, Sui X, Xuan Z, Du Y, Fu M, Zheng Z, Yang K, Xu C, Liu Y, Liu B, Zhong M, Zhang Z, Zheng J, Hu X, Zhang L, Sun H, Shao C. Discovery of a small-molecule NDR1 agonist for prostate cancer therapy. Front Pharmacol 2024; 15:1367358. [PMID: 38410130 PMCID: PMC10896269 DOI: 10.3389/fphar.2024.1367358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 01/30/2024] [Indexed: 02/28/2024] Open
Abstract
Prostatic cancer (PCa) is a common malignant neoplasm in men worldwide. Most patients develop castration-resistant prostate cancer (CRPC) after treatment with androgen deprivation therapy (ADT), usually resulting in death. Therefore, investigating new therapeutic targets and drugs for PCa patients is urgently needed. Nuclear Dbf2-related kinase 1 (NDR1), also known as STK38, is a serine/threonine kinase in the NDR/LATS kinase family that plays a critical role in cellular processes, including immunity, inflammation, metastasis, and tumorigenesis. It was reported that NDR1 inhibited the metastasis of prostate cancer cells by suppressing epithelial-mesenchymal transition (EMT), and decreased NDR1 expression might lead to a poorer prognosis, suggesting the enormous potential of NDR1 in antitumorigenesis. In this study, we characterized a small-molecule agonist named aNDR1, which specifically bound to NDR1 and potently promoted NDR1 expression, enzymatic activity and phosphorylation. aNDR1 exhibited drug-like properties, such as favorable stability, plasma protein binding capacity, cell membrane permeability, and PCa cell-specific inhibition, while having no obvious effect on normal prostate cells. Meanwhile, aNDR1 exhibited good antitumor activity both in vitro and in vivo. aNDR1 inhibited proliferation and migration of PCa cells and promoted apoptosis of PCa cells in vitro. We further found that aNDR1 inhibited subcutaneous tumors and lung metastatic nodules in vivo, with no obvious toxicity to the body. In summary, our study presents a potential small-molecule lead compound that targets NDR1 for clinical therapy of PCa patients.
Collapse
Affiliation(s)
- Yang Bai
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiuyuan Sui
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zuodong Xuan
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yifan Du
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Meiling Fu
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zeyuan Zheng
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Kunao Yang
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chunlan Xu
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yankuo Liu
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Bin Liu
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Min Zhong
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhengying Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jianzhong Zheng
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaoyan Hu
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Lei Zhang
- School of Public Health, Xiamen University, Xiamen, Fujian, China
| | - Huimin Sun
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chen Shao
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Urology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
6
|
Mokhtari RB, Ashayeri N, Baghaie L, Sambi M, Satari K, Baluch N, Bosykh DA, Szewczuk MR, Chakraborty S. The Hippo Pathway Effectors YAP/TAZ-TEAD Oncoproteins as Emerging Therapeutic Targets in the Tumor Microenvironment. Cancers (Basel) 2023; 15:3468. [PMID: 37444578 DOI: 10.3390/cancers15133468] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Various cancer cell-associated intrinsic and extrinsic inputs act on YAP/TAZ proteins to mediate the hyperactivation of the TEAD transcription factor-based transcriptome. This YAP/TAZ-TEAD activity can override the growth-limiting Hippo tumor-suppressor pathway that maintains normal tissue homeostasis. Herein, we provide an integrated summary of the contrasting roles of YAP/TAZ during normal tissue homeostasis versus tumor initiation and progression. In addition to upstream factors that regulate YAP/TAZ in the TME, critical insights on the emerging functions of YAP/TAZ in immune suppression and abnormal vasculature development during tumorigenesis are illustrated. Lastly, we discuss the current methods that intervene with the YAP/TAZ-TEAD oncogenic signaling pathway and the emerging applications of combination therapies, gut microbiota, and epigenetic plasticity that could potentiate the efficacy of chemo/immunotherapy as improved cancer therapeutic strategies.
Collapse
Affiliation(s)
- Reza Bayat Mokhtari
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Neda Ashayeri
- Division of Hematology and Oncology, Department of Pediatrics, Ali-Asghar Children Hospital, Iran University of Medical Science, Tehran 1449614535, Iran
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Manpreet Sambi
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Kosar Satari
- Division of Hematology and Oncology, Department of Pediatrics, Ali-Asghar Children Hospital, Iran University of Medical Science, Tehran 1449614535, Iran
| | - Narges Baluch
- Department of Immunology and Allergy, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Dmitriy A Bosykh
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Myron R Szewczuk
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Sayan Chakraborty
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| |
Collapse
|
7
|
Cellular Targets of HIV-1 Protease: Just the Tip of the Iceberg? Viruses 2023; 15:v15030712. [PMID: 36992421 PMCID: PMC10053624 DOI: 10.3390/v15030712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Human immunodeficiency virus 1 (HIV-1) viral protease (PR) is one of the most studied viral enzymes and a crucial antiviral target. Despite its well-characterized role in virion maturation, an increasing body of research is starting to focus on its ability to cleave host cell proteins. Such findings are apparently in contrast with the dogma of HIV-1 PR activity being restricted to the interior of nascent virions and suggest catalytic activity within the host cell environment. Given the limited amount of PR present in the virion at the time of infection, such events mainly occur during late viral gene expression, mediated by newly synthesized Gag-Pol polyprotein precursors, rather than before proviral integration. HIV-1 PR mainly targets proteins involved in three different processes: those involved in translation, those controlling cell survival, and restriction factors responsible for innate/intrinsic antiviral responses. Indeed, by cleaving host cell translation initiation factors, HIV-1 PR can impair cap-dependent translation, thus promoting IRES-mediated translation of late viral transcripts and viral production. By targeting several apoptotic factors, it modulates cell survival, thus promoting immune evasion and viral dissemination. Additionally, HIV-1 PR counteracts restriction factors incorporated in the virion that would otherwise interfere with nascent virus vitality. Thus, HIV-1 PR appears to modulate host cell function at different times and locations during its life cycle, thereby ensuring efficient viral persistency and propagation. However, we are far from having a complete picture of PR-mediated host cell modulation, which is emerging as a field that needs further investigation.
Collapse
|
8
|
Chen J, Liu F, Wu J, Yang Y, He J, Wu F, Yang K, Li J, Jiang Z, Jiang Z. Effect of STK3 on proliferation and apoptosis of pancreatic cancer cells via PI3K/AKT/mTOR pathway. Cell Signal 2023; 106:110642. [PMID: 36871796 DOI: 10.1016/j.cellsig.2023.110642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/12/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Pancreatic cancer, as a malignant tumor with a very poor prognosis, has a high mortality. It is imperative to clarify the mechanism of pancreatic cancer development and find suitable targets for diagnosis and treatment. Serine/threonine kinase 3 (STK3) is one of the core kinases of the Hippo pathway and has the ability to inhibit tumor growth. But the biological function of STK3 in pancreatic cancer remains unknown. Here, we confirmed that STK3 has an impact on the growth, apoptosis, and metastasis of pancreatic cancer cells and investigated the related molecular mechanisms. In our research, we found that STK3 is reduced in pancreatic cancer by RT-qPCR, IHC and IF, its expression level is correlated with the clinicopathological features. CCK-8 assay, colony formation assay and flow cytometry were used to detect the effect of STK3 on the proliferation and apoptosis of pancreatic cancer cells. In addition, the Transwell assay was used to detect the ability of cell migration and invasion. The results showed that STK3 promoted apoptosis and inhibited cell migration, invasion and proliferation in pancreatic cancer. Gene set enrichment analysis (GSEA) and western blotting are used to predict and verify the pathways related to STK3. Subsequently, we found that the effect of STK3 on proliferation and apoptosis is closely related to the PI3K/AKT/mTOR pathway. Moreover, the assistance of RASSF1 plays a significant role in the regulation of PI3K/AKT/mTOR pathway by STK3. The nude mouse xenograft experiment demonstrated the tumor suppressive ability of STK3 in vivo. Collectively, this study found that STK3 regulates pancreatic cancer cell proliferation and apoptosis by suppressing the PI3K/AKT/mTOR pathway with the assistance of RASSF1.
Collapse
Affiliation(s)
- Jun Chen
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fuqiang Liu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiao Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yichun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jin He
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Fan Wu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Kun Yang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Junfeng Li
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhongxiang Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zheng Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
9
|
Krasikova YS, Lavrik OI, Rechkunova NI. The XPA Protein-Life under Precise Control. Cells 2022; 11:cells11233723. [PMID: 36496984 PMCID: PMC9739396 DOI: 10.3390/cells11233723] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
Nucleotide excision repair (NER) is a central DNA repair pathway responsible for removing a wide variety of DNA-distorting lesions from the genome. The highly choreographed cascade of core NER reactions requires more than 30 polypeptides. The xeroderma pigmentosum group A (XPA) protein plays an essential role in the NER process. XPA interacts with almost all NER participants and organizes the correct NER repair complex. In the absence of XPA's scaffolding function, no repair process occurs. In this review, we briefly summarize our current knowledge about the XPA protein structure and analyze the formation of contact with its protein partners during NER complex assembling. We focus on different ways of regulation of the XPA protein's activity and expression and pay special attention to the network of post-translational modifications. We also discuss the data that is not in line with the currently accepted hypothesis about the functioning of the XPA protein.
Collapse
Affiliation(s)
- Yuliya S. Krasikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
- Department of Natural Sciences, Novosibirsk State University, Novosibirsk 630090, Russia
| | - Nadejda I. Rechkunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Novosibirsk 630090, Russia
- Correspondence:
| |
Collapse
|
10
|
Yousefi H, Delavar MR, Piroozian F, Baghi M, Nguyen K, Cheng T, Vittori C, Worthylake D, Alahari SK. Hippo signaling pathway: A comprehensive gene expression profile analysis in breast cancer. Biomed Pharmacother 2022; 151:113144. [PMID: 35623167 DOI: 10.1016/j.biopha.2022.113144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/10/2022] [Accepted: 05/15/2022] [Indexed: 11/30/2022] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy in women and a major public health concern. The Hippo pathway is an evolutionarily conserved signaling pathway that serves as a key regulator for a wide variety of biological processes. Hippo signaling has been shown to have both oncogenic and tumor-suppressive functions in various cancers. Core components of the Hippo pathway consist of various kinases and downstream effectors such as YAP/TAZ. In the current report, differential expression of Hippo pathway elements as well as the correlation of Hippo pathway mRNAs with various clinicopathologic characteristics, including molecular subtypes, receptor status, and methylation status, has been investigated in BC using METABRIC and TCGA datasets. In this review, we note deregulation of several Hippo signaling elements in BC patients. Moreover, we see examples of negative correlations between methylation of Hippo genes and mRNA expression. The expression of Hippo genes significantly varies between different receptor subgroups. Because of the clear associations between mRNA expression and methylation status, DNA methylation may be one of the mechanisms that regulate the Hippo pathway in BC cells. Differential expression of Hippo genes among various BC molecular subtypes suggests that Hippo signaling may function differently in different subtypes of BC. Our data also highlights an interesting link between Hippo components' transcription and ER negativity in BC. In conclusion, substantial deregulation of Hippo signaling components suggests an important role of these genes in breast cancer.
Collapse
Affiliation(s)
- Hassan Yousefi
- Louisiana State University Health Science Center (LSUHSC), Biochemistry & Molecular Biology, New Orleans, LA, USA
| | - Mahsa Rostamian Delavar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | | | - Masoud Baghi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Khoa Nguyen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Thomas Cheng
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Cecilia Vittori
- Louisiana State University Health Sciences Center and Stanley S. Scott Cancer Center, New Orleans, LA, USA
| | - David Worthylake
- Louisiana State University Health Science Center (LSUHSC), Biochemistry & Molecular Biology, New Orleans, LA, USA
| | - Suresh K Alahari
- Louisiana State University Health Science Center (LSUHSC), Biochemistry & Molecular Biology, New Orleans, LA, USA.
| |
Collapse
|
11
|
Dubois F, Bazille C, Levallet J, Maille E, Brosseau S, Madelaine J, Bergot E, Zalcman G, Levallet G. Molecular Alterations in Malignant Pleural Mesothelioma: A Hope for Effective Treatment by Targeting YAP. Target Oncol 2022; 17:407-431. [PMID: 35906513 PMCID: PMC9345804 DOI: 10.1007/s11523-022-00900-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 01/11/2023]
Abstract
Malignant pleural mesothelioma is a rare and aggressive neoplasm, which has primarily been attributed to the exposure to asbestos fibers (83% of cases); yet, despite a ban of using asbestos in many countries, the incidence of malignant pleural mesothelioma failed to decline worldwide. While little progress has been made in malignant pleural mesothelioma diagnosis, bevacizumab at first, then followed by double immunotherapy (nivolumab plus ipilumumab), were all shown to improve survival in large phase III randomized trials. The morphological analysis of the histological subtyping remains the primary indicator for therapeutic decision making at an advanced disease stage, while a platinum-based chemotherapy regimen combined with pemetrexed, either with or without bevacizumab, is still the main treatment option. Consequently, malignant pleural mesothelioma still represents a significant health concern owing to poor median survival (12-18 months). Given this context, both diagnosis and therapy improvements require better knowledge of the molecular mechanisms underlying malignant pleural mesothelioma's carcinogenesis and progression. Hence, the Hippo pathway in malignant pleural mesothelioma initiation and progression has recently received increasing attention, as the aberrant expression of its core components may be closely related to patient prognosis. The purpose of this review was to provide a critical analysis of our current knowledge on these topics, the main focus being on the available evidence concerning the role of each Hippo pathway's member as a promising biomarker, enabling detection of the disease at earlier stages and thus improving prognosis.
Collapse
Affiliation(s)
- Fatéméh Dubois
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
- Department of Pathology, CHU de Caen, Caen, France
- Federative Structure of Cyto-Molecular Oncogenetics (SF-MOCAE), CHU de Caen, Caen, France
| | - Céline Bazille
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
- Department of Pathology, CHU de Caen, Caen, France
| | - Jérôme Levallet
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
| | - Elodie Maille
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
| | - Solenn Brosseau
- Department of Thoracic Oncology and CIC1425, Hospital Bichat-Claude Bernard, Assistance Publique Hôpitaux de Paris, Université Paris-Diderot, Paris, France
- U830 INSERM "Genetics and Biology of Cancers, A.R.T Group", Curie Institute, Paris, France
| | - Jeannick Madelaine
- Department of Pulmonology and Thoracic Oncology, CHU de Caen, Caen, France
| | - Emmanuel Bergot
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France
- Department of Pulmonology and Thoracic Oncology, CHU de Caen, Caen, France
| | - Gérard Zalcman
- Department of Thoracic Oncology and CIC1425, Hospital Bichat-Claude Bernard, Assistance Publique Hôpitaux de Paris, Université Paris-Diderot, Paris, France
- U830 INSERM "Genetics and Biology of Cancers, A.R.T Group", Curie Institute, Paris, France
| | - Guénaëlle Levallet
- Normandie University, UNICAEN, CNRS, ISTCT Unit, Avenue H. Becquerel, 14074, Caen, France.
- Department of Pathology, CHU de Caen, Caen, France.
- Federative Structure of Cyto-Molecular Oncogenetics (SF-MOCAE), CHU de Caen, Caen, France.
| |
Collapse
|
12
|
Zuko A, Mallik M, Thompson R, Spaulding EL, Wienand AR, Been M, Tadenev ALD, van Bakel N, Sijlmans C, Santos LA, Bussmann J, Catinozzi M, Das S, Kulshrestha D, Burgess RW, Ignatova Z, Storkebaum E. tRNA overexpression rescues peripheral neuropathy caused by mutations in tRNA synthetase. Science 2021; 373:1161-1166. [PMID: 34516840 DOI: 10.1126/science.abb3356] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Amila Zuko
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Moushami Mallik
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands.,Molecular Neurogenetics Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Robin Thompson
- Biochemistry and Molecular Biology, Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Emily L Spaulding
- The Jackson Laboratory, Bar Harbor, ME, USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - Anne R Wienand
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Marije Been
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands
| | | | - Nick van Bakel
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Céline Sijlmans
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands
| | - Leonardo A Santos
- Biochemistry and Molecular Biology, Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Julia Bussmann
- Molecular Neurogenetics Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Marica Catinozzi
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands.,Molecular Neurogenetics Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Sarada Das
- Biochemistry and Molecular Biology, Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Divita Kulshrestha
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands.,Molecular Neurogenetics Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Robert W Burgess
- The Jackson Laboratory, Bar Harbor, ME, USA.,Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - Zoya Ignatova
- Biochemistry and Molecular Biology, Department of Chemistry, University of Hamburg, Hamburg, Germany
| | - Erik Storkebaum
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, Netherlands.,Molecular Neurogenetics Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
13
|
Nuclear Dbf2-related (NDR1) functions as tumor suppressor in glioblastoma by phosphorylation of Yes-associated protein (YAP). Chin Med J (Engl) 2021; 134:2054-2065. [PMID: 34343153 PMCID: PMC8440018 DOI: 10.1097/cm9.0000000000001653] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background: The Nuclear Dbf2-related (NDR1) kinase is a member of the NDR/LATS family, which was a supplementary of Hippo pathway. However, whether NDR1 could inhibit glioblastoma (GBM) growth by phosphorylating Yes-associated protein (YAP) remains unknown. Meanwhile, the role of NDR1 in GBM was not clear. This study aimed to investigate the role of NDR1-YAP pathway in GBM. Methods: Bioinformation analysis and immunohistochemistry (IHC) were performed to identify the expression of NDR1 in GBM. The effect of NDR1 on cell proliferation and cell cycle was analyzed utilizing CCK-8, clone formation, immunofluorescence and flow cytometry, respectively. In addition, the xenograft tumor model was established as well. Protein interaction was examined by Co-immunoprecipitation and immunofluorescence to observe co-localization. Results: Bioinformation analysis and IHC of our patients’ tumor tissues showed that expression of NDR1 in tumor tissue was relatively lower than that in normal tissues and was positively related to a lower survival rate. NDR1 could markedly reduce the proliferation and colony formation of U87 and U251. Furthermore, the results of flow cytometry showed that NDR1 led to cell cycle arrest at the G1 phase. Tumor growth was also inhibited in xenograft nude mouse models in NDR1-overexpression group. Western blotting and immunofluorescence showed that NDR1 could integrate with and phosphorylate YAP at S127 site. Meanwhile, NDR1 could mediate apoptosis process. Conclusion: In summary, our findings point out that NDR1 functions as a tumor suppressor in GBM. NDR1 is identified as a novel regulator of YAP, which gives us an in-depth comprehension of the Hippo signaling pathway.
Collapse
|
14
|
Furry is required for cell movements during gastrulation and functionally interacts with NDR1. Sci Rep 2021; 11:6607. [PMID: 33758327 PMCID: PMC7987989 DOI: 10.1038/s41598-021-86153-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 03/11/2021] [Indexed: 11/09/2022] Open
Abstract
Gastrulation is a key event in animal embryogenesis during which germ layer precursors are rearranged and the embryonic axes are established. Cell polarization is essential during gastrulation, driving asymmetric cell division, cell movements, and cell shape changes. The furry (fry) gene encodes an evolutionarily conserved protein with a wide variety of cellular functions, including cell polarization and morphogenesis in invertebrates. However, little is known about its function in vertebrate development. Here, we show that in Xenopus, Fry plays a role in morphogenetic processes during gastrulation, in addition to its previously described function in the regulation of dorsal mesoderm gene expression. Using morpholino knock-down, we demonstrate a distinct role for Fry in blastopore closure and dorsal axis elongation. Loss of Fry function drastically affects the movement and morphological polarization of cells during gastrulation and disrupts dorsal mesoderm convergent extension, responsible for head-to-tail elongation. Finally, we evaluate a functional interaction between Fry and NDR1 kinase, providing evidence of an evolutionarily conserved complex required for morphogenesis.
Collapse
|
15
|
Martin AP, Aushev VN, Zalcman G, Camonis JH. The STK38-XPO1 axis, a new actor in physiology and cancer. Cell Mol Life Sci 2021; 78:1943-1955. [PMID: 33145612 PMCID: PMC11072208 DOI: 10.1007/s00018-020-03690-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/30/2020] [Accepted: 10/21/2020] [Indexed: 12/24/2022]
Abstract
The Hippo signal transduction pathway is an essential regulator of organ size during developmental growth by controlling multiple cellular processes such as cell proliferation, cell death, differentiation, and stemness. Dysfunctional Hippo signaling pathway leads to dramatic tissue overgrowth. Here, we will briefly introduce the Hippo tumor suppressor pathway before focusing on one of its members and the unexpected twists that followed our quest of its functions in its multifarious actions beside the Hippo pathway: the STK38 kinase. In this review, we will precisely discuss the newly identified role of STK38 on regulating the nuclear export machinery by phosphorylating and activating, the major nuclear export receptor XPO1. Finally, we will phrase STK38's role on regulating the subcellular distribution of crucial cellular regulators such as Beclin1 and YAP1 with its implication in cancer.
Collapse
Affiliation(s)
- Alexandre Pj Martin
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, USA.
| | - Vasily N Aushev
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gérard Zalcman
- Thoracic Oncology Department, CIC1425/CLIP2 Paris-Nord, Hopital Bichat-Claude-Bernard, Paris, France
- Inserm U830, Institut Curie, Centre de Recherche, Paris Sciences Et Lettres Research University, Paris, France
| | - Jacques H Camonis
- Inserm U830, Institut Curie, Centre de Recherche, Paris Sciences Et Lettres Research University, Paris, France
| |
Collapse
|
16
|
Xie Z, Wang Y, Yang G, Han J, Zhu L, Li L, Zhang S. The role of the Hippo pathway in the pathogenesis of inflammatory bowel disease. Cell Death Dis 2021; 12:79. [PMID: 33436549 PMCID: PMC7804279 DOI: 10.1038/s41419-021-03395-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 01/29/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic and recurrent inflammatory disorder that primarily comprises Crohn's disease (CD) and ulcerative colitis (UC). Owing to its increasing prevalence in Eastern countries and the intractable challenges faced during IBD treatment, extensive research on IBD has been carried out over the last few years. Although the precise aetiology of IBD is undefined, the currently accepted hypothesis for IBD pathogenesis considers it to be a combination of environment, genetic predisposition, gut microbiota, and abnormal immunity. A recently emerged signalling pathway, the Hippo pathway, acts as a key regulator of cell growth, tissue homoeostasis, organ size, and has been implicated in several human cancers. In the past few years, studies have revealed the importance of the Hippo pathway in gastrointestinal tract physiology and gastrointestinal diseases, such as colorectal cancer and IBD. However, the role of the Hippo pathway and its exact impact in IBD remains to be elucidated. This review summarises the latest scientific literature on the involvement of this pathway in IBD from the following perspectives that account for the IBD pathogenesis: intestinal epithelial cell regeneration, immune regulation, gut microbiota, and angiogenesis. A comprehensive understanding of the specific role of the Hippo pathway in IBD will provide novel insights into future research directions and clinical implications of the Hippo pathway.
Collapse
Affiliation(s)
- Zhuo Xie
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ying Wang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Guang Yang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jing Han
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liguo Zhu
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Li
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shenghong Zhang
- Division of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
17
|
Sahu MR, Mondal AC. Neuronal Hippo signaling: From development to diseases. Dev Neurobiol 2020; 81:92-109. [PMID: 33275833 DOI: 10.1002/dneu.22796] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/18/2020] [Accepted: 11/27/2020] [Indexed: 01/12/2023]
Abstract
Hippo signaling pathway is a highly conserved and familiar tissue growth regulator, primarily dealing with cell survival, cell proliferation, and apoptosis. The Yes-associated protein (YAP) is the key transcriptional effector molecule, which is under negative regulation of the Hippo pathway. Wealth of studies have identified crucial roles of Hippo/YAP signaling pathway during the process of development, including the development of neuronal system. We provide here, an overview of the contributions of this signaling pathway at multiple stages of neuronal development including, proliferation of neural stem cells (NSCs), migration of NSCs toward their destined niche, maintaining NSCs in the quiescent state, differentiation of NSCs into neurons, neuritogenesis, synaptogenesis, brain development, and in neuronal apoptosis. Hyperactivation of the neuronal Hippo pathway can also lead to a variety of devastating neurodegenerative diseases. Instances of aberrant Hippo pathway leading to neurodegenerative diseases along with the approaches utilizing this pathway as molecular targets for therapeutics has been highlighted in this review. Recent evidences suggesting neuronal repair and regenerative potential of this pathway has also been pointed out, that will shed light on a novel aspect of Hippo pathway in regenerative medicine. Our review provides a better understanding of the significance of Hippo pathway in the journey of neuronal system from development to diseases as a whole.
Collapse
Affiliation(s)
- Manas Ranjan Sahu
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
18
|
Delgado ILS, Carmona B, Nolasco S, Santos D, Leitão A, Soares H. MOB: Pivotal Conserved Proteins in Cytokinesis, Cell Architecture and Tissue Homeostasis. BIOLOGY 2020; 9:biology9120413. [PMID: 33255245 PMCID: PMC7761452 DOI: 10.3390/biology9120413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 01/08/2023]
Abstract
The MOB family proteins are constituted by highly conserved eukaryote kinase signal adaptors that are often essential both for cell and organism survival. Historically, MOB family proteins have been described as kinase activators participating in Hippo and Mitotic Exit Network/ Septation Initiation Network (MEN/SIN) signaling pathways that have central roles in regulating cytokinesis, cell polarity, cell proliferation and cell fate to control organ growth and regeneration. In metazoans, MOB proteins act as central signal adaptors of the core kinase module MST1/2, LATS1/2, and NDR1/2 kinases that phosphorylate the YAP/TAZ transcriptional co-activators, effectors of the Hippo signaling pathway. More recently, MOBs have been shown to also have non-kinase partners and to be involved in cilia biology, indicating that its activity and regulation is more diverse than expected. In this review, we explore the possible ancestral role of MEN/SIN pathways on the built-in nature of a more complex and functionally expanded Hippo pathway, by focusing on the most conserved components of these pathways, the MOB proteins. We discuss the current knowledge of MOBs-regulated signaling, with emphasis on its evolutionary history and role in morphogenesis, cytokinesis, and cell polarity from unicellular to multicellular organisms.
Collapse
Affiliation(s)
- Inês L. S. Delgado
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); or (S.N.); (D.S.); (A.L.)
- Faculdade de Medicina Veterinária, Universidade Lusófona de Humanidades e Tecnologias, 1749-024 Lisboa, Portugal
| | - Bruno Carmona
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; or
- Centro de Química Estrutural–Faculdade de Ciências da Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Sofia Nolasco
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); or (S.N.); (D.S.); (A.L.)
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; or
| | - Dulce Santos
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); or (S.N.); (D.S.); (A.L.)
| | - Alexandre Leitão
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 Lisboa, Portugal or (I.L.S.D.); or (S.N.); (D.S.); (A.L.)
| | - Helena Soares
- Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; or
- Centro de Química Estrutural–Faculdade de Ciências da Universidade de Lisboa, 1749-016 Lisboa, Portugal
- Correspondence: or
| |
Collapse
|
19
|
Ando N, Tanaka K, Otsubo K, Toyokawa G, Ikematsu Y, Ide M, Yoneshima Y, Iwama E, Inoue H, Ijichi K, Tagawa T, Nakanishi Y, Okamoto I. Association of Mps one binder kinase activator 1 (MOB1) expression with poor disease-free survival in individuals with non-small cell lung cancer. Thorac Cancer 2020; 11:2830-2839. [PMID: 32841529 PMCID: PMC7529568 DOI: 10.1111/1759-7714.13608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/12/2020] [Accepted: 07/21/2020] [Indexed: 11/30/2022] Open
Abstract
Background Mps one binder kinase activator 1 (MOB1) is a core component of the Hippo signaling pathway and has been implicated as a tumor suppressor. Here, we evaluated the possible relationship of MOB1 expression in non‐small cell lung cancer (NSCLC) to prognosis. Methods We retrospectively analyzed 205 lung adenocarcinoma patients treated at Kyushu University Hospital between November 2007 and October 2012. MOB1 expression in tumor cells of surgical specimens was evaluated by immunohistochemistry. Invasive activity of NSCLC cell lines in vitro was measured with a transwell assay. Results Expression of MOB1 was classified as high in 105 of the 205 (51.2%) tumor specimens, and such high expression was significantly associated with poor disease‐free survival (P = 0.0161). Among the various clinicopathologic parameters examined, high MOB1 expression was significantly associated only with intratumoral vascular invasion (P = 0.0005). Multivariate analysis also identified high MOB1 expression as a significant independent risk factor for disease‐free survival (P = 0.0319). The invasiveness of H1299 cells in vitro was increased or attenuated by overexpression or knockdown of MOB1, respectively. Conclusions Our results suggest that MOB1 might promote early recurrence of NSCLC by increasing vascular invasion by tumor cells. Key points Significant findings of the study We found that high MOB1 expression in surgical specimens of lung adenocarcinoma was associated with poor disease‐free survival and with intratumoral vascular invasion. MOB1 expression also promoted the invasiveness of NSCLC cells in vitro. What this study adds Our results thus suggest that high MOB1 expression is a risk factor for early postoperative recurrence in lung adenocarcinoma.
Collapse
Affiliation(s)
- Nobuhisa Ando
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kentaro Tanaka
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohei Otsubo
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Gouji Toyokawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuki Ikematsu
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Maako Ide
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasuto Yoneshima
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Iwama
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Inoue
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kayo Ijichi
- Pathophysiological and Experimental Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuzo Tagawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoichi Nakanishi
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Kitakyushu City Hospital Organization, Fukuoka, Japan
| | - Isamu Okamoto
- Research Institute for Diseases of the Chest, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
20
|
Mohammadi S, Arefnezhad R, Danaii S, Yousefi M. New insights into the core Hippo signaling and biological macromolecules interactions in the biology of solid tumors. Biofactors 2020; 46:514-530. [PMID: 32445262 DOI: 10.1002/biof.1634] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/26/2022]
Abstract
As an evolutionarily conserved pathway, Hippo signaling pathway impacts different pathology and physiology processes such as wound healing, tissue repair/size and regeneration. When some components of Hippo signaling dysregulated, it affects cancer cells proliferation. Moreover, the relation Hippo pathway with other signaling including Wnt, TGFβ, Notch, and EGFR signaling leaves effect on the proliferation of cancer cells. Utilizing a number of therapeutic approaches, such as siRNAs and long noncoding RNA (lncRNA) to prevent cancer cells through the targeting of Hippo pathways, can provide new insights into cancer target therapy. The purpose of present review, first of all, is to demonstrate the importance of Hippo signaling and its relation with other signaling pathways in cancer. It also tries to demonstrate targeting Hippo signaling progress in cancer therapy.
Collapse
Affiliation(s)
- Solmaz Mohammadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Shahla Danaii
- Gynecology Department, Eastern Azerbaijan ACECR ART Center, Eastern Azerbaijan Branch of ACECR, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Depatment of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Qin B, Yu J, Nowsheen S, Zhao F, Wang L, Lou Z. STK38 promotes ATM activation by acting as a reader of histone H4 ufmylation. SCIENCE ADVANCES 2020; 6:eaax8214. [PMID: 32537488 PMCID: PMC7269669 DOI: 10.1126/sciadv.aax8214] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 04/10/2020] [Indexed: 05/11/2023]
Abstract
The ATM (ataxia-telangiectasia mutated) kinase is rapidly activated following DNA damage and phosphorylates its downstream targets to launch DDR signaling. Recently, we and others showed that UFM1 signaling promotes ATM activation. We further discovered that monoufmylation of histone H4 at Lys31 by UFM1-specific ligase 1 (UFL1) is an important step in the amplification of ATM activation. However, how monoufmylated H4 enhances ATM activation is still unknown. Here, we report STK38, a kinase in the Hippo pathway, serves as a reader for histone H4 ufmylation to promote ATM activation in a kinase-independent manner. STK38 contains a potential UFM1 binding motif which recognizes ufmylated H4 and recruits the SUV39H1 to the double-strand breaks, resulting in H3K9 trimethylation and Tip60 activation to promote ATM activation. Together, STK38 is a previously unknown player in DNA damage signaling and functions as a reader of monoufmylated H4 at Lys31 to promote ATM activation.
Collapse
Affiliation(s)
- Bo Qin
- Division of Oncology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jia Yu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Somaira Nowsheen
- Division of Oncology, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Medical Scientist Training Program, Mayo Medical School and Mayo Graduate School, Mayo Clinic, Rochester, MN 55905, USA
| | - Fei Zhao
- Division of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhenkun Lou
- Division of Oncology, Mayo Clinic, Rochester, MN 55905, USA
- Corresponding author.
| |
Collapse
|
22
|
Ueda Y, Kondo N, Kinashi T. MST1/2 Balance Immune Activation and Tolerance by Orchestrating Adhesion, Transcription, and Organelle Dynamics in Lymphocytes. Front Immunol 2020; 11:733. [PMID: 32435241 PMCID: PMC7218056 DOI: 10.3389/fimmu.2020.00733] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 03/31/2020] [Indexed: 01/15/2023] Open
Abstract
The STE20-like serine/threonine kinases MST1 and MST2 (MST1/2) are mammalian homologs of Hippo in flies. MST1/2 regulate organ size by suppressing the transcription factor YAP, which promotes proliferation. MST1 is predominantly expressed in immune cells, where it plays distinct roles. Here, we review the functions of MST1/2 in immune cells, uncovered by a series of recent studies, and discuss the connection between MST1/2 function and immune responses. MST1/2 regulate lymphocyte development, trafficking, survival, and antigen recognition by naive T cells. MST1/2 also regulate the function of regulatory T cells and effector T cell differentiation, thus acting to balance immune activation and tolerance. Interestingly, MST1/2 elicit these functions not by the “canonical” Hippo pathway, but by the non-canonical Hippo pathway or alternative pathways. In these pathways, MST1/2 regulates cellular processes relating to immune response, such as chemotaxis, cell adhesion, immunological synapse, gene transcriptions. Recent advances in our understanding of the molecular mechanisms of these processes have revealed important roles of MST1/2 in regulating cytoskeleton remodeling, integrin activation, and vesicular transport in lymphocytes. We discuss the significance of the MST1/2 signaling in lymphocytes in the regulation of organelle dynamics.
Collapse
Affiliation(s)
- Yoshihiro Ueda
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Naoyuki Kondo
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| | - Tatsuo Kinashi
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan
| |
Collapse
|
23
|
Fan S, Price T, Huang W, Plue M, Warren J, Sundaramoorthy P, Paul B, Feinberg D, MacIver N, Chao N, Sipkins D, Kang Y. PINK1-Dependent Mitophagy Regulates the Migration and Homing of Multiple Myeloma Cells via the MOB1B-Mediated Hippo-YAP/TAZ Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1900860. [PMID: 32154065 PMCID: PMC7055555 DOI: 10.1002/advs.201900860] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 11/23/2019] [Indexed: 05/07/2023]
Abstract
The roles of mitochondrial dysfunction in carcinogenesis remain largely unknown. The effects of PTEN-induced putative kinase 1 (PINK1)-dependent mitophagy on the pathogenesis of multiple myeloma (MM) are determined. The levels of the PINK1-dependent mitophagy markers PINK1 and parkin RBR E3 ubiquitin protein ligase (PARK2) in CD138+ plasma cells are reduced in patients with MM and correlate with clinical outcomes in myeloma patients. Moreover, the induction of PINK1-dependent mitophagy with carbonylcyanide-m-chlorophenylhydrazone (CCCP) or salinomycin, or overexpression of PINK1 leads to inhibition of transwell migration, suppression of myeloma cell homing to calvarium, and decreased osteolytic bone lesions. Furthermore, genetic deletion of pink1 accelerates myeloma development in a spontaneous X-box binding protein-1 spliced isoform (XBP-1s) transgenic myeloma mouse model and in VK*MYC transplantable myeloma recipient mice. Additionally, treatment with salinomycin shows significant antimyeloma activities in vivo in murine myeloma xenograft models. Finally, the effects of PINK1-dependent mitophagy on myeloma pathogenesis are driven by the activation of the Mps one binder kinase activator (MOB1B)-mediated Hippo pathway and the subsequent downregulation of Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ) expression. These data provide direct evidence that PINK1-dependent mitophagy plays a critical role in the pathogenesis of MM and is a potential therapeutic target.
Collapse
Affiliation(s)
- Shengjun Fan
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Trevor Price
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Wei Huang
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Michelle Plue
- Shared Materials Instrumentation FacilityPratt School of EngineeringDuke UniversityDurhamNC27708USA
| | | | - Pasupathi Sundaramoorthy
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Barry Paul
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Daniel Feinberg
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | | | - Nelson Chao
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Dorothy Sipkins
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular TherapyDepartment of MedicineDuke University Medical CenterDurhamNC27710USA
| |
Collapse
|
24
|
Wang S, Zhou L, Ling L, Meng X, Chu F, Zhang S, Zhou F. The Crosstalk Between Hippo-YAP Pathway and Innate Immunity. Front Immunol 2020; 11:323. [PMID: 32174922 PMCID: PMC7056731 DOI: 10.3389/fimmu.2020.00323] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 02/10/2020] [Indexed: 12/12/2022] Open
Abstract
Recognition of pathogen-associated molecular patterns (PAMPs) triggers expression of antiviral interferons and proinflammatory cytokines, which functions as the frontier of host defense against microbial pathogen invasion. Hippo-YAP pathway regulates cell proliferation, survival, differentiation and is involved in diverse life processes, including tissue homeostasis and tumor suppression. Emerging discoveries elucidated that the components of Hippo-YAP pathway, such as MST1/2, NDR1/2, and YAP/TAZ played crucial regulatory roles in innate immunity. Meanwhile the innate immune signaling also exhibited regulatory effect on Hippo-YAP pathway. As for the importance of these two pathways, it would be interesting to figure out the deeper biological implications of their interplays. This review focuses on the regulation between Hippo-YAP pathway and innate immune signaling. We also propose the possible contribution of these interplays to tumor development.
Collapse
Affiliation(s)
- Shuai Wang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Lili Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Li Ling
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xuli Meng
- Department of Breast Surgery, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Feng Chu
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Suping Zhang
- Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
25
|
García-Gutiérrez L, McKenna S, Kolch W, Matallanas D. RASSF1A Tumour Suppressor: Target the Network for Effective Cancer Therapy. Cancers (Basel) 2020; 12:cancers12010229. [PMID: 31963420 PMCID: PMC7017281 DOI: 10.3390/cancers12010229] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
The RASSF1A tumour suppressor is a scaffold protein that is involved in cell signalling. Increasing evidence shows that this protein sits at the crossroad of a complex signalling network, which includes key regulators of cellular homeostasis, such as Ras, MST2/Hippo, p53, and death receptor pathways. The loss of expression of RASSF1A is one of the most common events in solid tumours and is usually caused by gene silencing through DNA methylation. Thus, re-expression of RASSF1A or therapeutic targeting of effector modules of its complex signalling network, is a promising avenue for treating several tumour types. Here, we review the main modules of the RASSF1A signalling network and the evidence for the effects of network deregulation in different cancer types. In particular, we summarise the epigenetic mechanism that mediates RASSF1A promoter methylation and the Hippo and RAF1 signalling modules. Finally, we discuss different strategies that are described for re-establishing RASSF1A function and how a multitargeting pathway approach selecting druggable nodes in this network could lead to new cancer treatments.
Collapse
Affiliation(s)
- Lucía García-Gutiérrez
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; (L.G.-G.); (S.M.); (W.K.)
| | - Stephanie McKenna
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; (L.G.-G.); (S.M.); (W.K.)
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; (L.G.-G.); (S.M.); (W.K.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; (L.G.-G.); (S.M.); (W.K.)
- School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Correspondence:
| |
Collapse
|
26
|
Rausch V, Hansen CG. The Hippo Pathway, YAP/TAZ, and the Plasma Membrane. Trends Cell Biol 2019; 30:32-48. [PMID: 31806419 DOI: 10.1016/j.tcb.2019.10.005] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/13/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022]
Abstract
The plasma membrane allows the cell to sense and adapt to changes in the extracellular environment by relaying external inputs via intracellular signaling networks. One central cellular signaling pathway is the Hippo pathway, which regulates homeostasis and plays chief roles in carcinogenesis and regenerative processes. Recent studies have found that mechanical stimuli and diffusible chemical components can regulate the Hippo pathway primarily through receptors embedded in the plasma membrane. Morphologically defined structures within the plasma membrane, such as cellular junctions, focal adhesions, primary cilia, caveolae, clathrin-coated pits, and plaques play additional key roles. Here, we discuss recent evidence highlighting the importance of these specialized plasma membrane domains in cellular feedback via the Hippo pathway.
Collapse
Affiliation(s)
- Valentina Rausch
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Carsten G Hansen
- Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh bioQuarter, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh bioQuarter, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| |
Collapse
|
27
|
Martin APJ, Jacquemyn M, Lipecka J, Chhuon C, Aushev VN, Meunier B, Singh MK, Carpi N, Piel M, Codogno P, Hergovich A, Parrini MC, Zalcman G, Guerrera IC, Daelemans D, Camonis JH. STK38 kinase acts as XPO1 gatekeeper regulating the nuclear export of autophagy proteins and other cargoes. EMBO Rep 2019; 20:e48150. [PMID: 31544310 PMCID: PMC6832005 DOI: 10.15252/embr.201948150] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/15/2019] [Accepted: 09/03/2019] [Indexed: 01/19/2023] Open
Abstract
STK38 (also known as NDR1) is a Hippo pathway serine/threonine protein kinase with multifarious functions in normal and cancer cells. Using a context-dependent proximity-labeling assay, we identify more than 250 partners of STK38 and find that STK38 modulates its partnership depending on the cellular context by increasing its association with cytoplasmic proteins upon nutrient starvation-induced autophagy and with nuclear ones during ECM detachment. We show that STK38 shuttles between the nucleus and the cytoplasm and that its nuclear exit depends on both XPO1 (aka exportin-1, CRM1) and STK38 kinase activity. We further uncover that STK38 modulates XPO1 export activity by phosphorylating XPO1 on serine 1055, thus regulating its own nuclear exit. We expand our model to other cellular contexts by discovering that XPO1 phosphorylation by STK38 regulates also the nuclear exit of Beclin1 and YAP1, key regulator of autophagy and transcriptional effector, respectively. Collectively, our results reveal STK38 as an activator of XPO1, behaving as a gatekeeper of nuclear export. These observations establish a novel mechanism of XPO1-dependent cargo export regulation by phosphorylation of XPO1's C-terminal auto-inhibitory domain.
Collapse
Affiliation(s)
- Alexandre PJ Martin
- ART GroupInserm U830ParisFrance
- Institut CurieCentre de RechercheParis Sciences et Lettres Research UniversityParisFrance
| | - Maarten Jacquemyn
- Laboratory of Virology and ChemotherapyKU Leuven Department of Microbiology, Immunology and TransplantationRega Institute for Medical ResearchKU LeuvenLeuvenBelgium
| | - Joanna Lipecka
- Inserm U894Center of Psychiatry and NeuroscienceParisFrance
- Université Paris DescartesSorbonne Paris CitéParisFrance
| | - Cerina Chhuon
- Université Paris DescartesSorbonne Paris CitéParisFrance
- Proteomics Platform 3P5‐NeckerUniversité Paris Descartes ‐ Structure Fédérative de Recherche NeckerINSERM US24/CNRS UMS3633ParisFrance
| | | | - Brigitte Meunier
- ART GroupInserm U830ParisFrance
- Institut CurieCentre de RechercheParis Sciences et Lettres Research UniversityParisFrance
| | - Manish K Singh
- ART GroupInserm U830ParisFrance
- Institut CurieCentre de RechercheParis Sciences et Lettres Research UniversityParisFrance
| | - Nicolas Carpi
- Institut CurieCentre de RechercheParis Sciences et Lettres Research UniversityParisFrance
- CNRSUMR 144ParisFrance
| | - Matthieu Piel
- Institut CurieCentre de RechercheParis Sciences et Lettres Research UniversityParisFrance
- CNRSUMR 144ParisFrance
| | - Patrice Codogno
- Université Paris DescartesSorbonne Paris CitéParisFrance
- Inserm U1151/CNRS UMR 8253Institut Necker Enfants‐MaladesParisFrance
| | | | - Maria Carla Parrini
- ART GroupInserm U830ParisFrance
- Institut CurieCentre de RechercheParis Sciences et Lettres Research UniversityParisFrance
| | - Gerard Zalcman
- ART GroupInserm U830ParisFrance
- Institut CurieCentre de RechercheParis Sciences et Lettres Research UniversityParisFrance
- Sorbonne Paris CitéUniversité Paris DiderotParisFrance
| | - Ida Chiara Guerrera
- Université Paris DescartesSorbonne Paris CitéParisFrance
- Proteomics Platform 3P5‐NeckerUniversité Paris Descartes ‐ Structure Fédérative de Recherche NeckerINSERM US24/CNRS UMS3633ParisFrance
| | - Dirk Daelemans
- Laboratory of Virology and ChemotherapyKU Leuven Department of Microbiology, Immunology and TransplantationRega Institute for Medical ResearchKU LeuvenLeuvenBelgium
| | - Jacques H Camonis
- ART GroupInserm U830ParisFrance
- Institut CurieCentre de RechercheParis Sciences et Lettres Research UniversityParisFrance
| |
Collapse
|
28
|
SIRT1 and p300/CBP regulate the reversible acetylation of serine-threonine kinase NDR2. Biochem Biophys Res Commun 2019; 518:396-401. [PMID: 31427083 DOI: 10.1016/j.bbrc.2019.08.069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 08/12/2019] [Indexed: 11/21/2022]
Abstract
Nuclear Dbf2-related kinase 2 (NDR2) is a highly conserved kinase that belongs to the NDR/LATS serine-threonine kinase family. NDR2 is involved in many cellular processes as a kinase or a scaffolding protein. As a known kinase, NDR2 requires self-phosphorylation and trans-phosphorylation to become fully active. However, beside phosphorylation, little is known about other posttranslational modifications of NDR2. In this study, we found that NDR2 can be specially acetylated at K463 in cells. In addition, SIRT1 acts as the major deacetylase for NDR2, while p300 and CBP function as specific acetyltransferases for NDR2. Interestingly, in SIRT1 deficient cells HDAC6 and HDAC1/2 can deacetylate NDR2, which provides a novel insight in deacetylation regulation. Our results demonstrate that NDR2 is a reversible acetylated kinase regulated by SIRT1 and p300/CBP.
Collapse
|
29
|
Klimek C, Jahnke R, Wördehoff J, Kathage B, Stadel D, Behrends C, Hergovich A, Höhfeld J. The Hippo network kinase STK38 contributes to protein homeostasis by inhibiting BAG3-mediated autophagy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2019; 1866:1556-1566. [PMID: 31326538 PMCID: PMC6692498 DOI: 10.1016/j.bbamcr.2019.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/18/2019] [Accepted: 07/14/2019] [Indexed: 12/31/2022]
Abstract
Chaperone-assisted selective autophagy (CASA) initiated by the cochaperone Bcl2-associated athanogene 3 (BAG3) represents an important mechanism for the disposal of misfolded and damaged proteins in mammalian cells. Under mechanical stress, the cochaperone cooperates with the small heat shock protein HSPB8 and the cytoskeleton-associated protein SYNPO2 to degrade force-unfolded forms of the actin-crosslinking protein filamin. This is essential for muscle maintenance in flies, fish, mice and men. Here, we identify the serine/threonine protein kinase 38 (STK38), which is part of the Hippo signaling network, as a novel interactor of BAG3. STK38 was previously shown to facilitate cytoskeleton assembly and to promote mitophagy as well as starvation and detachment induced autophagy. Significantly, our study reveals that STK38 exerts an inhibitory activity on BAG3-mediated autophagy. Inhibition relies on a disruption of the functional interplay of BAG3 with HSPB8 and SYNPO2 upon binding of STK38 to the cochaperone. Of note, STK38 attenuates CASA independently of its kinase activity, whereas previously established regulatory functions of STK38 involve target phosphorylation. The ability to exert different modes of regulation on central protein homeostasis (proteostasis) machineries apparently allows STK38 to coordinate the execution of diverse macroautophagy pathways and to balance cytoskeleton assembly and degradation.
Collapse
Affiliation(s)
- Christina Klimek
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Ricarda Jahnke
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Judith Wördehoff
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Barbara Kathage
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Daniela Stadel
- Institute of Biochemistry II, Goethe University Medical School, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Christian Behrends
- Munich Cluster for Systems Neurology, Ludwig-Maximilians-University Munich, Feodor-Lynen Strasse 17, 81377 München, Germany
| | | | - Jörg Höhfeld
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany.
| |
Collapse
|
30
|
Gundogdu R, Hergovich A. MOB (Mps one Binder) Proteins in the Hippo Pathway and Cancer. Cells 2019; 8:cells8060569. [PMID: 31185650 PMCID: PMC6627106 DOI: 10.3390/cells8060569] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/22/2022] Open
Abstract
The family of MOBs (monopolar spindle-one-binder proteins) is highly conserved in the eukaryotic kingdom. MOBs represent globular scaffold proteins without any known enzymatic activities. They can act as signal transducers in essential intracellular pathways. MOBs have diverse cancer-associated cellular functions through regulatory interactions with members of the NDR/LATS kinase family. By forming additional complexes with serine/threonine protein kinases of the germinal centre kinase families, other enzymes and scaffolding factors, MOBs appear to be linked to an even broader disease spectrum. Here, we review our current understanding of this emerging protein family, with emphases on post-translational modifications, protein-protein interactions, and cellular processes that are possibly linked to cancer and other diseases. In particular, we summarise the roles of MOBs as core components of the Hippo tissue growth and regeneration pathway.
Collapse
Affiliation(s)
- Ramazan Gundogdu
- Vocational School of Health Services, Bingol University, 12000 Bingol, Turkey.
| | - Alexander Hergovich
- UCL Cancer Institute, University College London, WC1E 6BT, London, United Kingdom.
| |
Collapse
|
31
|
Zhang P, Wang T, Zhang D, Zhang Z, Yuan S, Zhang J, Cao J, Li H, Li X, Shen H, Chen G. Exploration of MST1-Mediated Secondary Brain Injury Induced by Intracerebral Hemorrhage in Rats via Hippo Signaling Pathway. Transl Stroke Res 2019; 10:729-743. [PMID: 30941717 DOI: 10.1007/s12975-019-00702-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 01/28/2023]
Abstract
Intracerebral hemorrhage (ICH) is a serious public health problem which causes high rates of disability and mortality in adults. Cell apoptosis is a sign of secondary brain injury (SBI) following ICH. Mammalian sterile 20-like kinase-1 (MST1), an apoptosis-promoting kinase, is a part of the Hippo signaling pathway and involved in cell death, oxidative stress, and inflammation. However, the role and underlying mechanism of MST1 in SBI induced by ICH have not yet been fully explained. The main purpose of present research was to explore the role of MST1 and its potential mechanism in SBI after ICH. An ICH model was established by injecting autologous blood into the right basal ganglia in male SD rats. We found that MST1 phosphorylation was significantly increased in brain tissues of rats after ICH. Additionally, inhibition of MST1 phosphorylation by a chemical inhibitor (Xmu-mp-1) and genetic knockdown could effectively reduce the activation of P-LATS1 and P-YAP which are downstream proteins of MST1 and decrease neuronal cell death and inflammation reaction in ICH rats. Furthermore, the decreased of MST1 phosphorylation reduced brain edema, blood-brain barrier (BBB) damage, and neurobehavioral impairment during ICH. Over-expression of MST1 resulted in opposite effects. Finally, deletion of MST1 significantly reduced neuronal apoptosis in vitro. In summary, our study revealed that MST1 played an important role in the SBI following ICH, and inhibition of MST1 could alleviate ICH-induced SBI. Therefore, MST1 may be considered as a potential therapeutic target for SBI following ICH.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Tianyi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Dongping Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Zhuwei Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Shuai Yuan
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Juyi Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Jie Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
32
|
Liu Z, Wu C, Pan Y, Liu H, Wang X, Yang Y, Gu M, Zhang Y, Wang X. NDR2 promotes the antiviral immune response via facilitating TRIM25-mediated RIG-I activation in macrophages. SCIENCE ADVANCES 2019; 5:eaav0163. [PMID: 30775439 PMCID: PMC6365120 DOI: 10.1126/sciadv.aav0163] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 12/20/2018] [Indexed: 06/09/2023]
Abstract
Retinoic acid-inducible gene I (RIG-I), a pivotal cytosolic sensor, recognizes viral RNAs to initiate antiviral innate immunity. However, posttranslational regulation of RIG-I signaling is not well understood. We report here that nuclear Dbf2-related kinase 2 (NDR2) functions as a crucial positive regulator of the RIG-I-mediated antiviral immune response. Overexpression of NDR2 or its kinase-inactive mutants potentiates RNA virus-induced production of type I interferons and proinflammatory cytokines and dampens viral replication. NDR2 conditional knockout mice (Lysm+NDR2f/f) show an impaired antiviral immune response. Mechanistically, NDR2 directly associates with RIG-I and TRIM25, thus facilitating the RIG-I/TRIM25 complex and enhancing the TRIM25-mediated K63-linked polyubiquitination of RIG-I, which is required for the RIG-I-mediated antiviral immune response. Furthermore, NDR2 expression is notably down-regulated in peripheral blood from respiratory syncytial virus-infected patients and in virus-infected macrophages. Collectively, these findings provide insights into the function of NDR2 in antiviral immunity and its related clinical significance.
Collapse
Affiliation(s)
- Zhiyong Liu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Cheng Wu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yueyun Pan
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Huan Liu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Xiumei Wang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yuting Yang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Meidi Gu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Yuanyuan Zhang
- The Children’s Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Xiaojian Wang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
| |
Collapse
|
33
|
Hergovich A. Measuring the Kinase Activities of the LATS/NDR Protein Kinases. Methods Mol Biol 2019; 1893:305-317. [PMID: 30565143 DOI: 10.1007/978-1-4939-8910-2_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The Hippo tissue growth control and regeneration pathway is a main regulator of the YAP/TAZ effectors. In this regard, the LATS/NDR serine/threonine protein kinases can function as central components of the Hippo core module. More specifically, LATS/NDR-mediated phosphorylation of YAP/TAZ on different residues can regulate the subcellular localization and/or stability of YAP/TAZ. Therefore, the assessment of LATS/NDR activities can serve as readout for the activity status of the Hippo pathway. Here, we describe our preferred methodology regarding the measurement of the activities of LATS/NDR kinases.
Collapse
|
34
|
Zhuang Q, Chen Z, Shen J, Fan M, Xue D, Lu H, Xu R, He X. RASSF1A promoter methylation correlates development, progression, and poor cancer-specific survival of renal cell carcinoma: trial sequential analysis. Onco Targets Ther 2018; 12:119-134. [PMID: 30588036 PMCID: PMC6304251 DOI: 10.2147/ott.s183142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Background This meta-analysis evaluated the clinicopathologic and prognostic significance of RASSF1A promoter methylation in renal cell carcinoma (RCC). Materials and methods The ORs or HRs and their 95% CIs were calculated. Trial sequential analysis was conducted. Results Twenty-two articles that included 1,421 patients with RCC and 724 controls were identified. RASSF1A promoter methylation correlated with RCC in tissue, blood, and urine samples. On multivariate analysis, RASSF1A promoter methylation was associated with tumor grade (grade 3–4 vs 1–2: OR=3.59), clinical stage (stage 3–4 vs 1–2: OR=2.15), T classification (pT2–4 vs pT1: OR=2.66), histologic subtypes (papillary vs clear cell: OR=2.91), and cancer-specific survival (HR=1.78), but it was not linked to age, gender, lymph node status, distant metastasis, or overall survival. The Cancer Genome Atlas data also showed that RASSF1A methylation was significantly more likely to be seen in papillary vs clear-cell RCC (OR=23.19). Conclusion RASSF1A promoter methylation may be associated with the development and progression of RCC, as well as poor cancer-specific survival. Methylation was more frequent in papillary vs clear-cell RCC. More studies are needed to confirm these findings in blood or urine samples.
Collapse
Affiliation(s)
- Qianfeng Zhuang
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China,
| | - Zhen Chen
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China,
| | - Jie Shen
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China,
| | - Min Fan
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China,
| | - Dong Xue
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China,
| | - Hao Lu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China,
| | - Renfang Xu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China,
| | - Xiaozhou He
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China,
| |
Collapse
|
35
|
Targeting the Hippo Pathway for Breast Cancer Therapy. Cancers (Basel) 2018; 10:cancers10110422. [PMID: 30400599 PMCID: PMC6266939 DOI: 10.3390/cancers10110422] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 12/31/2022] Open
Abstract
Breast cancer (BC) is one of the most prominent diseases in the world, and the treatments for BC have many limitations, such as resistance and a lack of reliable biomarkers. Currently the Hippo pathway is emerging as a tumor suppressor pathway with its four core components that regulate downstream transcriptional targets. In this review, we introduce the present targeted therapies of BC, and then discuss the roles of the Hippo pathway in BC. Finally, we summarize the evidence of the small molecule inhibitors that target the Hippo pathway, and then discuss the possibilities and future direction of the Hippo-targeted drugs for BC therapy.
Collapse
|
36
|
Uehara Y, Temma K, Kobayashi Y, Irie N, Yamaguchi T. Reduction of Thermotolerance by Heat Shock Protein 90 Inhibitors in Murine Erythroleukemia Cells. Biol Pharm Bull 2018; 41:1393-1400. [PMID: 30175776 DOI: 10.1248/bpb.b18-00190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cells induce heat shock proteins (HSPs) against various stress. However, murine erythroleukemia (MEL) cells do not express HSP72, a heat-inducible member of HSP70 family. So, it is of interest to examine how MEL cells respond to heat stress (44°C, 30 min). Heat stress-induced apoptosis was suppressed by pretreatment of heat shock (44°C, 10 min). Such suppressive effects were maximal at 6 h after heat shock and remained up to 12 h. Interestingly, such effects of heat shock were abrogated by specific inhibitors of HSP90 such as 17-allylamino-17-demethoxygeldanamycin (17-AAG) and novobiocin. From flow cytometric analysis, it was found that MEL cells arrest in G2 phase at 6 h after heat shock, but restore original cell cycle at 12 h. High expression level of HSP90 was maintained before and after heat shock. Phosphorylation of HSP90α was observed in apoptotic cells induced by heat stress, but inhibited by pretreatment of heat shock. Such inhibition was abrogated by 17-AAG. Moreover, c-Jun NH2-terminal kinase (JNK) was activated in heat stress-induced apoptotic cells. Taken together, these results suggest that HSP90α in MEL cells plays an important role in the thermotolerance, i.e., suppression of heat stress-induced apoptosis by heat shock.
Collapse
Affiliation(s)
- Yousuke Uehara
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Kazunari Temma
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Yuuya Kobayashi
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Nobuyuki Irie
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Takeo Yamaguchi
- Department of Chemistry, Faculty of Science, Fukuoka University
| |
Collapse
|
37
|
Xiao N, Li H, Yu W, Gu C, Fang H, Peng Y, Mao H, Fang Y, Ni W, Yao M. SUMO‐specific protease 2 (SENP2) suppresses keratinocyte migration by targeting NDR1 for de‐SUMOylation. FASEB J 2018; 33:163-174. [DOI: 10.1096/fj.201800353r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ning Xiao
- Cancer Institute of Traditional Chinese MedicineLonghua HospitalShanghai University of Traditional Chinese Medicine Shanghai China
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Hui Li
- Key Laboratory of Pediatric Hematology and OncologyMinistry of Health and Pediatric Translational Medicine InstituteShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Weirong Yu
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Chuan Gu
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Houshun Fang
- Key Laboratory of Pediatric Hematology and OncologyMinistry of Health and Pediatric Translational Medicine InstituteShanghai Children's Medical CenterShanghai Jiao Tong University School of Medicine Shanghai China
| | - Yinbo Peng
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Heshui Mao
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Yong Fang
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Wei Ni
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| | - Min Yao
- Department of Burns and Plastic SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
- Institute of Traumatic MedicineShanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
38
|
Iwasa H, Hossain S, Hata Y. Tumor suppressor C-RASSF proteins. Cell Mol Life Sci 2018; 75:1773-1787. [PMID: 29353317 PMCID: PMC11105443 DOI: 10.1007/s00018-018-2756-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/05/2018] [Accepted: 01/17/2018] [Indexed: 12/13/2022]
Abstract
Human genome has ten genes that are collectedly called Ras association domain family (RASSF). RASSF is composed of two subclasses, C-RASSF and N-RASSF. Both N-RASSF and C-RASSF encode Ras association domain-containing proteins and are frequently suppressed by DNA hypermethylation in human cancers. However, C-RASSF and N-RASSF are quite different. Six C-RASSF proteins (RASSF1-6) are characterized by a C-terminal coiled-coil motif named Salvador/RASSF/Hippo domain, while four N-RASSF proteins (RASSF7-10) lack it. C-RASSF proteins interact with mammalian Ste20-like kinases-the core kinases of the tumor suppressor Hippo pathway-and cross-talk with this pathway. Some of them share the same interacting molecules such as MDM2 and exert the tumor suppressor role in similar manners. Nevertheless, each C-RASSF protein has distinct characters. In this review, we summarize our current knowledge of how C-RASSF proteins play tumor suppressor roles and discuss the similarities and differences among C-RASSF proteins.
Collapse
Affiliation(s)
- Hiroaki Iwasa
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Shakhawoat Hossain
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, 6205, Bangladesh
| | - Yutaka Hata
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan.
- Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, 113-8519, Japan.
| |
Collapse
|
39
|
Nguyen TH, Kugler JM. Ubiquitin-Dependent Regulation of the Mammalian Hippo Pathway: Therapeutic Implications for Cancer. Cancers (Basel) 2018; 10:cancers10040121. [PMID: 29673168 PMCID: PMC5923376 DOI: 10.3390/cancers10040121] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/08/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
The Hippo pathway serves as a key barrier for oncogenic transformation. It acts by limiting the activity of the proto-oncogenes YAP and TAZ. Reduced Hippo signaling and elevated YAP/TAZ activities are frequently observed in various types of tumors. Emerging evidence suggests that the ubiquitin system plays an important role in regulating Hippo pathway activity. Deregulation of ubiquitin ligases and of deubiquitinating enzymes has been implicated in increased YAP/TAZ activity in cancer. In this article, we review recent insights into the ubiquitin-mediated regulation of the mammalian Hippo pathway, its deregulation in cancer, and possibilities for targeting the Hippo pathway through the ubiquitin system.
Collapse
Affiliation(s)
- Thanh Hung Nguyen
- Institute of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| | - Jan-Michael Kugler
- Institute of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark.
| |
Collapse
|
40
|
Aharoni-Kats L, Zelinger E, Chen S, Yarden O. Altering Neurospora crassa MOB2A exposes its functions in development and affects its interaction with the NDR kinase COT1. Mol Microbiol 2018; 108:641-660. [PMID: 29600559 DOI: 10.1111/mmi.13954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2018] [Indexed: 12/30/2022]
Abstract
The Neurospora crassa Mps One Binder (MOB) proteins MOB2A and MOB2B physically interact with the Nuclear Dbf2 Related (NDR) kinase COT1 and have been shown to have overlapping functions in various aspects of asexual development. Here, we identified two N. crassa MOB2A residues, Tyr117 and Tyr119, which are potentially phosphorylated. Using phosphomimetic mob-2a mutants we have been able to establish that apart from their previously described roles, MOB2A/B are involved in additional developmental processes. Enhanced conidial germination, accompanied by conidial agglutination, in the phosphomimetic mutants indicated that MOB2A is a negative regulator of germination. Thick-section imaging of perithecia revealed slow maturation and a lack of asci alignment in the mutant strains demonstrating a role for MOB2A in sexual development. We demonstrate that even though MOB2A and MOB2B have some overlapping functions, MOB2B cannot compensate for the roles MOB2A has in conidiation and germination. Altering Tyr residues 117 and 119 impaired the physical interactions between MOB2A and COT1, most likely contributing to some of the observed effects. As cot-1 and the phosphomimetic mutants share an extragenic suppressor (gul-1), we concluded that at least some of the effects imposed by altering Tyr117 and Tyr119 are mediated by the NDR kinase.
Collapse
Affiliation(s)
- Liran Aharoni-Kats
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610000, Israel
| | - Einat Zelinger
- Centre for Scientific Imaging, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610000, Israel
| | - She Chen
- Proteomics Centre, The National Institute of Biological Sciences, Beijing 102206, China
| | - Oded Yarden
- Department of Plant Pathology and Microbiology, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610000, Israel
| |
Collapse
|
41
|
Byun YS, Kim EK, Araki K, Yamamura KI, Lee K, Yoon WK, Won YS, Kim HC, Choi KC, Nam KH. Fryl deficiency is associated with defective kidney development and function in mice. Exp Biol Med (Maywood) 2018; 243:408-417. [PMID: 29409347 DOI: 10.1177/1535370218758249] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
FRY like transcription coactivator ( Fryl) gene located on chromosome 5 is a paralog of FRY microtubule binding protein ( Fry) in vertebrates. It encodes a protein with unknown functions. Fryl gene is conserved in various species ranging from eukaryotes to human. Although there are several reports on functions of Fry gene, functions of Fryl gene remain unclear. A mouse line containing null mutation in Fryl gene by gene trapping was produced in this study for the first time. The survival and growth of Fryl-/- mice were observed. Fryl gene expression levels in mouse tissues were determined and histopathologic analyses were conducted. Most Fryl-/- mice died soon after birth. Rare Fryl-/- survivors showed growth retardation with significantly lower body weight compared to their littermate controls. Although they could breed, more than half of Fryl-/- survivors died of hydronephrosis before age 1. No abnormal histopathologic lesion was apparent in full-term embryo or adult tissues except the kidney. Abnormal lining cell layer detachments from walls of collecting and convoluted tubules in kidneys were apparent in Fryl-/- neonates and full-term embryos. Fryl gene was expressed in renal tubular tissues including the glomeruli and convoluted and collecting tubules. This indicates that defects in tubular systems are associated with Fryl functions and death of Fryl-/- neonates. Fryl protein is required for normal development and functional maintenance of kidney in mice. This is the first report of in vivo Fryl gene functions. Impact statement FRY like transcription coactivator ( Fryl) gene is conserved in various species ranging from eukaryotes to human. It expresses a protein with unknown function. We generated a Fryl gene mutant mouse line and found that most homozygous mice died soon after their birth. Rare Fryl-/- survivors showed growth retardation with significantly lower body weight compared to their littermate controls. Although they could breed, more than half of Fryl-/- survivors died of hydronephrosis before age 1. Full-term mutant embryos showed abnormal collecting and convoluted tubules in kidneys where Fryl gene was expressed. Collectively, these results indicate that Fryl protein is required for normal development and functional maintenance of kidney in mice. To the best of our knowledge, this is the first report on in vivo Fryl gene functions.
Collapse
Affiliation(s)
- Yong-Sub Byun
- 1 Laboratory Animal Resource Center, 204180 Korea Research Institute of Bioscience and Biotechnology , Chungbuk 28116, Korea
- 2 Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Chungbuk 28644, Korea
| | - Eun-Kyoung Kim
- 1 Laboratory Animal Resource Center, 204180 Korea Research Institute of Bioscience and Biotechnology , Chungbuk 28116, Korea
| | - Kimi Araki
- 3 Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Ken-Ichi Yamamura
- 3 Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Kihoon Lee
- 1 Laboratory Animal Resource Center, 204180 Korea Research Institute of Bioscience and Biotechnology , Chungbuk 28116, Korea
| | - Won-Kee Yoon
- 1 Laboratory Animal Resource Center, 204180 Korea Research Institute of Bioscience and Biotechnology , Chungbuk 28116, Korea
| | - Young-Suk Won
- 1 Laboratory Animal Resource Center, 204180 Korea Research Institute of Bioscience and Biotechnology , Chungbuk 28116, Korea
| | - Hyoung-Chin Kim
- 1 Laboratory Animal Resource Center, 204180 Korea Research Institute of Bioscience and Biotechnology , Chungbuk 28116, Korea
| | - Kyung-Chul Choi
- 2 Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Chungbuk 28644, Korea
| | - Ki-Hoan Nam
- 1 Laboratory Animal Resource Center, 204180 Korea Research Institute of Bioscience and Biotechnology , Chungbuk 28116, Korea
| |
Collapse
|
42
|
Zhang W, Shen J, Gu F, Zhang Y, Wu W, Weng J, Liao Y, Deng Z, Yuan Q, Zheng L, Zhang Y, Shen W. Monopolar spindle-one-binder protein 2 regulates the activity of large tumor suppressor/yes-associated protein to inhibit the motility of SMMC-7721 hepatocellular carcinoma cells. Oncol Lett 2018; 15:5375-5383. [PMID: 29552181 DOI: 10.3892/ol.2018.7952] [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: 07/02/2017] [Accepted: 01/03/2018] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence implicates monopolar spindle-one-binder protein (MOB)2 as an inhibitor of nuclear-Dbf2-related kinase (NDR) by competing with MOB1 for interaction with NDR1/2. NDR/large tumor suppressor (LATS) kinases may function similarly to yes-associated protein (YAP) kinases and be considered as members of the Hippo core cassette. MOB2 appears to serve roles in cell survival, cell cycle progression, responses to DNA damage and cell motility. However, the underlying mechanisms involved remain unclarified. In the present study, it was demonstrated that the knockout of MOB2 by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 promoted migration and invasion, induced phosphorylation of NDR1/2 and decreased phosphorylation of YAP in SMMC-7721 cells when compared with the blank vector-transduced cells. By contrast, the overexpression of MOB2 resulted in the opposite results. Mechanistically, MOB2 regulated the alternative interaction of MOB1 with NDR1/2 and LATS1, which resulted in increased phosphorylation of LATS1 and MOB1 and thereby led to the inactivation of YAP and consequently inhibition of cell motility. The results of the present study provide evidence of MOB2 serving a positive role in LATS/YAP activation by activating the Hippo signaling pathway.
Collapse
Affiliation(s)
- Weicheng Zhang
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jingyuan Shen
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Fengming Gu
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Ying Zhang
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Wenjuan Wu
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China.,Department of Medical Oncology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Jiachun Weng
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Yuexia Liao
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Zijing Deng
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Qing Yuan
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Lu Zheng
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China
| | - Yu Zhang
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, P.R. China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| | - Weigan Shen
- Department of Cell Biology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225001, P.R. China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, P.R. China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225001, P.R. China
| |
Collapse
|
43
|
Bettoun A, Joffre C, Zago G, Surdez D, Vallerand D, Gundogdu R, Sharif AAD, Gomez M, Cascone I, Meunier B, White MA, Codogno P, Parrini MC, Camonis JH, Hergovich A. Mitochondrial clearance by the STK38 kinase supports oncogenic Ras-induced cell transformation. Oncotarget 2018; 7:44142-44160. [PMID: 27283898 PMCID: PMC5190085 DOI: 10.18632/oncotarget.9875] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/12/2016] [Indexed: 01/12/2023] Open
Abstract
Oncogenic Ras signalling occurs frequently in many human cancers. However, no effective targeted therapies are currently available to treat patients suffering from Ras-driven tumours. Therefore, it is imperative to identify downstream effectors of Ras signalling that potentially represent promising new therapeutic options. Particularly, considering that autophagy inhibition can impair the survival of Ras-transformed cells in tissue culture and mouse models, an understanding of factors regulating the balance between autophagy and apoptosis in Ras-transformed human cells is needed. Here, we report critical roles of the STK38 protein kinase in oncogenic Ras transformation. STK38 knockdown impaired anoikis resistance, anchorage-independent soft agar growth, and in vivo xenograft growth of Ras-transformed human cells. Mechanistically, STK38 supports Ras-driven transformation through promoting detachment-induced autophagy. Even more importantly, upon cell detachment STK38 is required to sustain the removal of damaged mitochondria by mitophagy, a selective autophagic process, to prevent excessive mitochondrial reactive oxygen species production that can negatively affect cancer cell survival. Significantly, knockdown of PINK1 or Parkin, two positive regulators of mitophagy, also impaired anoikis resistance and anchorage-independent growth of Ras-transformed human cells, while knockdown of USP30, a negative regulator of PINK1/Parkin-mediated mitophagy, restored anchorage-independent growth of STK38-depleted Ras-transformed human cells. Therefore, our findings collectively reveal novel molecular players that determine whether Ras-transformed human cells die or survive upon cell detachment, which potentially could be exploited for the development of novel strategies to target Ras-transformed cells.
Collapse
Affiliation(s)
- Audrey Bettoun
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France
| | - Carine Joffre
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France.,Present address: Cancer Research Center of Toulouse, UMR1037, Toulouse, 31100, France
| | - Giulia Zago
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France
| | - Didier Surdez
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France
| | - David Vallerand
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France
| | - Ramazan Gundogdu
- University College London, Cancer Institute, London, WC1E 6BT, United Kingdom
| | - Ahmad A D Sharif
- University College London, Cancer Institute, London, WC1E 6BT, United Kingdom
| | - Marta Gomez
- University College London, Cancer Institute, London, WC1E 6BT, United Kingdom
| | - Ilaria Cascone
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France
| | - Brigitte Meunier
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France
| | - Michael A White
- University of Texas Southwestern Medical Center, Dallas, Texas, 75390, USA
| | - Patrice Codogno
- Inserm U1151-CNRS UMR 8253, Institut Necker Enfants-Malades, Paris, 75993, France
| | - Maria Carla Parrini
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France
| | - Jacques H Camonis
- Institut Curie, Inserm U830, Paris Sciences et Lettres University Paris, 75248, France
| | - Alexander Hergovich
- University College London, Cancer Institute, London, WC1E 6BT, United Kingdom
| |
Collapse
|
44
|
Abstract
The mammalian STE20-like (MST) protein kinases are composed of MST1, MST2, MST3, MST4 and YSK1. They play crucial roles in cell growth, migration, polarity and apoptosis. Dysfunction of these kinases often leads to diseases. MST kinases are extensively involved in development and function of immune system. Here, we review recent progresses on the regulatory function of MST kinases in innate immune signaling.
Collapse
Affiliation(s)
- Zhubing Shi
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhaocai Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
45
|
Wang SP, Wang LH. Disease implication of hyper-Hippo signalling. Open Biol 2017; 6:rsob.160119. [PMID: 27805903 PMCID: PMC5090056 DOI: 10.1098/rsob.160119] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022] Open
Abstract
The Hippo signalling pathway regulates cellular proliferation, apoptosis and differentiation, thus exerting profound effects on cellular homeostasis. Inhibition of Hippo signalling has been frequently implicated in human cancers, indicating a well-known tumour suppressor function of the Hippo pathway. However, it is less certain whether and how hyperactivation of the Hippo pathway affects biological outcome in living cells. This review describes current knowledge of the regulatory mechanisms of the Hippo pathway, mainly focusing on hyperactivation of the Hippo signalling nexus. The disease implications of hyperactivated Hippo signalling have also been discussed, including arrhythmogenic cardiomyopathy, Sveinsson's chorioretinal atrophy, Alzheimer's disease, amyotrophic lateral sclerosis and diabetes. By highlighting the significance of disease-relevant Hippo signalling activation, this review can offer exciting prospects to address the onset and potential reversal of Hippo-related disorders.
Collapse
Affiliation(s)
- Shu-Ping Wang
- Laboratory of Biochemistry and Molecular Biology, The Rockefeller University, New York, NY 10065, USA
| | - Lan-Hsin Wang
- Graduate Institute of Life Sciences, National Defense Medical Center, 161, Sec. 6, Minquan E. Rd., Neihu Dist, Taipei City 114, Taiwan
| |
Collapse
|
46
|
Li D, Ni H, Rui Q, Gao R, Chen G. Deletion of Mst1 attenuates neuronal loss and improves neurological impairment in a rat model of traumatic brain injury. Brain Res 2017; 1688:15-21. [PMID: 29054447 DOI: 10.1016/j.brainres.2017.10.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/10/2017] [Accepted: 10/17/2017] [Indexed: 10/18/2022]
Abstract
Neuronal cell death following traumatic brain injury (TBI) is a considerable contributor to neurological deficits. In our work, we explored the functions of Mammalian STE20-like kinase-1 (Mst1), a apoptosis-promoting kinase and also a pivotal bridgebuilder of apoptotic signaling, in the etiopathogenesis of an experimental rat model of TBI. We found that the phosphorylation level of Mst1 in injured area was significantly increased after TBI. Furthermore, we discovered that inhibition of Mst1 phosphorylation can effectively reduce neuronal cell death by inhibiting the activation of caspase 3 and suppressing the damage of DNA during TBI. In addition, the decreased of Mst1 phosphorylation level, not only reduced brain edema and blood-brain barrier (BBB) damage in injured region but also weakened the impairment of neurologic behavior during TBI. In conclusion, our work demonstrates that Mst1 plays an important role in TBI-induced neuronal cell death, suggesting that Mst1 is expected to be a potential therapeutic target for TBI.
Collapse
Affiliation(s)
- Di Li
- Department of Neurosurgery and Translational Medicine Center, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Haibo Ni
- Department of Neurosurgery, The First People 's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Qin Rui
- Clinical Laboratory, The First People's Hospital of Zhangjiagang, Soochow University, Suzhou, China
| | - Rong Gao
- Department of Neurosurgery, The First People 's Hospital of Zhangjiagang, Soochow University, Suzhou, China.
| | - Gang Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
47
|
Kulaberoglu Y, Lin K, Holder M, Gai Z, Gomez M, Assefa Shifa B, Mavis M, Hoa L, Sharif AAD, Lujan C, Smith ESJ, Bjedov I, Tapon N, Wu G, Hergovich A. Stable MOB1 interaction with Hippo/MST is not essential for development and tissue growth control. Nat Commun 2017; 8:695. [PMID: 28947795 PMCID: PMC5612953 DOI: 10.1038/s41467-017-00795-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/28/2017] [Indexed: 01/07/2023] Open
Abstract
The Hippo tumor suppressor pathway is essential for development and tissue growth control, encompassing a core cassette consisting of the Hippo (MST1/2), Warts (LATS1/2), and Tricornered (NDR1/2) kinases together with MOB1 as an important signaling adaptor. However, it remains unclear which regulatory interactions between MOB1 and the different Hippo core kinases coordinate development, tissue growth, and tumor suppression. Here, we report the crystal structure of the MOB1/NDR2 complex and define key MOB1 residues mediating MOB1's differential binding to Hippo core kinases, thereby establishing MOB1 variants with selective loss-of-interaction. By studying these variants in human cancer cells and Drosophila, we uncovered that MOB1/Warts binding is essential for tumor suppression, tissue growth control, and development, while stable MOB1/Hippo binding is dispensable and MOB1/Trc binding alone is insufficient. Collectively, we decrypt molecularly, cell biologically, and genetically the importance of the diverse interactions of Hippo core kinases with the pivotal MOB1 signal transducer.The Hippo tumor suppressor pathway is essential for development and tissue growth control. Here the authors employ a multi-disciplinary approach to characterize the interactions of the three Hippo kinases with the signaling adaptor MOB1 and show how they differently affect development, tissue growth and tumor suppression.
Collapse
Affiliation(s)
- Yavuz Kulaberoglu
- Tumour Suppressor Signalling Network Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Kui Lin
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Maxine Holder
- Apoptosis and Proliferation Control Laboratory, Francis Crick Institute, London, NW1 1BF, UK
| | - Zhongchao Gai
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Marta Gomez
- Tumour Suppressor Signalling Network Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Belul Assefa Shifa
- Tumour Suppressor Signalling Network Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Merdiye Mavis
- Tumour Suppressor Signalling Network Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Lily Hoa
- Tumour Suppressor Signalling Network Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Ahmad A D Sharif
- Tumour Suppressor Signalling Network Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Celia Lujan
- Molecular Biology of Cancer laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Ewan St John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK
| | - Ivana Bjedov
- Molecular Biology of Cancer laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK
| | - Nicolas Tapon
- Apoptosis and Proliferation Control Laboratory, Francis Crick Institute, London, NW1 1BF, UK
| | - Geng Wu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Alexander Hergovich
- Tumour Suppressor Signalling Network Laboratory, UCL Cancer Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
48
|
STK38L kinase ablation promotes loss of cell viability in a subset of KRAS-dependent pancreatic cancer cell lines. Oncotarget 2017; 8:78556-78572. [PMID: 29108249 PMCID: PMC5667982 DOI: 10.18632/oncotarget.20833] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/27/2017] [Indexed: 01/07/2023] Open
Abstract
Pancreatic ductal adenocarcinomas (PDACs) are highly aggressive malignancies, associated with poor clinical prognosis and limited therapeutic options. Oncogenic KRAS mutations are found in over 90% of PDACs, playing a central role in tumor progression. Global gene expression profiling of PDAC reveals 3-4 major molecular subtypes with distinct phenotypic traits and pharmacological vulnerabilities, including variations in oncogenic KRAS pathway dependencies. PDAC cell lines of the aberrantly differentiated endocrine exocrine (ADEX) subtype are robustly KRAS-dependent for survival. The KRAS gene is located on chromosome 12p11-12p12, a region amplified in 5-10% of primary PDACs. Within this amplicon, we identified co-amplification of KRAS with the STK38L gene in a subset of primary human PDACs and PDAC cell lines. Therefore, we determined whether PDAC cell lines are dependent on STK38L expression for proliferation and viability. STK38L encodes a serine/threonine kinase, which shares homology with Hippo pathway kinases LATS1/2. We show that STK38L expression is elevated in a subset of primary PDACs and PDAC cell lines displaying ADEX subtype characteristics, including overexpression of mutant KRAS. RNAi-mediated depletion of STK38L in a subset of ADEX subtype cell lines inhibits cellular proliferation and induces apoptosis. Concomitant with these effects, STK38L depletion causes increased expression of the LATS2 kinase and the cell cycle regulator p21. LATS2 depletion partially rescues the cytostatic and cytotoxic effects of STK38L depletion. Lastly, high STK38L mRNA expression is associated with decreased overall patient survival in PDACs. Collectively, our findings implicate STK38L as a candidate targetable vulnerability in a subset of molecularly-defined PDACs.
Collapse
|
49
|
Erdman VV, Karimov DD, Nasibullin TR, Timasheva IR, Tuktarova IA, Mustafina OE. The role of Alu polymorphism of PLAT, PKHD1L1, STK38L, and TEAD1 genes in development of a longevity trait. ADVANCES IN GERONTOLOGY 2017. [DOI: 10.1134/s2079057017020059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
50
|
Liu J, Kong X, Lee YM, Zhang MK, Guo LY, Lin Y, Lim TK, Lin Q, Xu XQ. Stk38 Modulates Rbm24 Protein Stability to Regulate Sarcomere Assembly in Cardiomyocytes. Sci Rep 2017; 7:44870. [PMID: 28322254 PMCID: PMC5359592 DOI: 10.1038/srep44870] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/13/2017] [Indexed: 01/12/2023] Open
Abstract
RNA-binding protein Rbm24 is a key regulator of heart development and required for sarcomere assembly and heart contractility. Yet, its underlying mechanism remains unclear. Here, we link serine/threonine kinase 38 (Stk38) signaling to the regulation of Rbm24 by showing that Rbm24 phosphorylation and its function could be modulated by Stk38. Using co-immunoprecipitation coupled with mass spectrometry technique, we identified Stk38 as an endogenous binding partner of Rbm24. Stk38 knockdown resulted in decreased Rbm24 protein level in cardiomyocytes. Further studies using Stk38 kinase inhibitor or activator showed that Rbm24 protein stability was regulated in a kinase activity-dependent manner. Deficiency of Stk38 caused reduction of sarcomere proteins and disarrangement of sarcomere, suggesting that Stk38 is essential for Rbm24 to regulate sarcomere assembly. Our results revealed that Stk38 kinase catalyzes the phosphorylation of Rbm24 during sarcomerogensis and this orchestrates accurate sarcomere alignment. This furthers our understanding of the regulatory mechanism of cardiac sarcomere assembly in both physiologic and pathologic contexts, and uncovers a potential novel pathway to cardiomyopathy through modulating the Stk38/Rbm24 protein activity.
Collapse
Affiliation(s)
- Jing Liu
- The Institute of Stem Cell and Regenerative Medicine, Medical College, Xiamen University, 361100, P.R. China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Medical College, Xiamen University, 361100, P.R. China
| | - Xu Kong
- The Institute of Stem Cell and Regenerative Medicine, Medical College, Xiamen University, 361100, P.R. China
| | - Yew Mun Lee
- Department of Biological Sciences, National University of Singapore, 117543, Singapore
| | - Meng Kai Zhang
- The Institute of Stem Cell and Regenerative Medicine, Medical College, Xiamen University, 361100, P.R. China
| | - Li Yan Guo
- The Institute of Stem Cell and Regenerative Medicine, Medical College, Xiamen University, 361100, P.R. China
| | - Yu Lin
- The Institute of Stem Cell and Regenerative Medicine, Medical College, Xiamen University, 361100, P.R. China
| | - Teck Kwang Lim
- Department of Biological Sciences, National University of Singapore, 117543, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, 117543, Singapore
| | - Xiu Qin Xu
- The Institute of Stem Cell and Regenerative Medicine, Medical College, Xiamen University, 361100, P.R. China.,Fujian Key Laboratory of Organ and Tissue Regeneration, Medical College, Xiamen University, 361100, P.R. China
| |
Collapse
|