1
|
Zhang F, Huang B, Xu Y, Cao G, Shen M, Liu C, Luo J. MISP Suppresses Ferroptosis via MST1/2 Kinases to Facilitate YAP Activation in Non-Small Cell Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415814. [PMID: 40019375 PMCID: PMC12021056 DOI: 10.1002/advs.202415814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Despite advances in non-small cell lung cancer (NSCLC) therapies, resistance remains a major challenge. Ferroptosis, a form of regulated cell death, plays a key role in cancer progression and treatment response. However, the mechanisms governing ferroptosis in NSCLC are not fully understood. The Hippo pathway, which regulates cell proliferation, has recently been implicated in ferroptosis regulation. In this study, we identify Mitotic Spindle Positioning (MISP) as a critical inhibitor of ferroptosis in NSCLC. MISP is upregulated in NSCLC tissues, and its loss sensitizes cells to ferroptosis, reducing cell proliferation in vitro and in vivo. Mechanistically, MISP binds to the SARAH domain of MST1/2 kinases, inhibiting their homodimerization and autophosphorylation, leading to sustained activation of YAP, a transcriptional coactivator in the Hippo pathway. YAP activation increases SLC7A11 expression, which protects cells from ferroptosis. We also identify a mutant MISP-R390/391A that disrupts MISP-MST1/2 binding, further illustrating the MST1/2-dependent inhibition of Hippo signaling. Notably, MISP is a target of YAP, creating a feedback loop that amplifies YAP signaling. Our findings suggest a novel MISP-YAP axis regulating ferroptosis, positioning MISP as a potential therapeutic target for NSCLC, especially in cases with dysregulated YAP.
Collapse
Affiliation(s)
- Fuquan Zhang
- Department of Thoracic and Cardiovascular SurgeryThe Second Affiliated Hospital of Nantong UniversityThe First People's Hospital of NantongNantong226000China
| | - Bingtao Huang
- Department of Thoracic SurgeryBinzhou Medical University HospitalBinzhou256600China
| | - Yiming Xu
- Department of Thoracic and Cardiovascular SurgeryThe Second Affiliated Hospital of Nantong UniversityThe First People's Hospital of NantongNantong226000China
| | - Guohong Cao
- Department of Thoracic SurgeryBinzhou Medical University HospitalBinzhou256600China
| | - Mingjing Shen
- Department of Cardiothoracic SurgeryThe Second Affiliated Hospital of Soochow UniversitySuzhou215004China
| | - Changmin Liu
- Department of Radiation OncologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Department of Radiation OncologyAnhui Provincial Cancer HospitalHefeiAnhui230031China
| | - Judong Luo
- Department of RadiotherapyTongji HospitalSchool of MedicineTongji UniversityShanghai200065China
| |
Collapse
|
2
|
Zheng G, Yan Z, Zou J, Zou X, Chai K, Zhang G. AR and YAP crosstalk: impacts on therapeutic strategies in prostate cancer. Front Oncol 2025; 15:1520808. [PMID: 39963114 PMCID: PMC11830605 DOI: 10.3389/fonc.2025.1520808] [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: 10/31/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Prostate cancer ranks as one of the most common types of cancer affecting men worldwide, and its progression is shaped by a diverse array of influencing factors. The AR signaling pathway plays a pivotal role in the pathogenesis of prostate cancer. While existing anti-androgen treatments show initial efficacy, they ultimately do not succeed in halting the advancement to CRPC. Recent studies have identified alterations in the Hippo-YAP signaling pathway within prostate cancer, highlighting intricate crosstalk with the AR signaling pathway. In this review, we examine the interactions and underlying mechanisms between AR and YAP, the key molecules in these two signaling pathways. AR regulates the stability and function of YAP by modulating its transcription, translation, and phosphorylation status, while YAP exerts both promotional and inhibitory regulatory effects on AR. Based on these findings, this paper investigates their significant roles in the onset, progression, and therapeutic resistance of prostate cancer, and discusses the clinical potential of YAP in prostate cancer treatment.
Collapse
Affiliation(s)
- Guansong Zheng
- First Clinical College, Gannan Medical University, Ganzhou, China
| | - Zhaojie Yan
- First Clinical College, Gannan Medical University, Ganzhou, China
| | - Junrong Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Institute of Urology, Gannan Medical University, Ganzhou, China
- Department of Jiangxi Engineering Technology Research Center of Calculi Prevention, Gannan Medical University, Ganzhou, China
| | - Xiaofeng Zou
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Keqiang Chai
- Department of Urology, Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, China
| | - Guoxi Zhang
- Department of Urology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
3
|
Han H, Huang Z, Xu C, Seo G, An J, Yang B, Liu Y, Lan T, Yan J, Ren S, Xu Y, Xiao D, Yan JK, Ahn C, Fishman DA, Meng Z, Guan KL, Qi R, Luo R, Wang W. Functional annotation of the Hippo pathway somatic mutations in human cancers. Nat Commun 2024; 15:10106. [PMID: 39572544 PMCID: PMC11582751 DOI: 10.1038/s41467-024-54480-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
The Hippo pathway is commonly altered in cancer initiation and progression; however, exactly how this pathway becomes dysregulated to promote human cancer development remains unclear. Here we analyze the Hippo somatic mutations in the human cancer genome and functionally annotate their roles in targeting the Hippo pathway. We identify a total of 85 loss-of-function (LOF) missense mutations for Hippo pathway genes and elucidate their underlying mechanisms. Interestingly, we reveal zinc-finger domain as an integral structure for MOB1 function, whose LOF mutations in head and neck cancer promote tumor growth. Moreover, the schwannoma/meningioma-derived NF2 LOF mutations not only inhibit its tumor suppressive function in the Hippo pathway, but also gain an oncogenic role for NF2 by activating the VANGL-JNK pathway. Collectively, our study not only offers a rich somatic mutation resource for investigating the Hippo pathway in human cancers, but also provides a molecular basis for Hippo-based cancer therapy.
Collapse
Affiliation(s)
- Han Han
- Department of Pathophysiology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| | - Zhen Huang
- Chemical and Materials Physics Graduate Program, University of California, Irvine, Irvine, CA, USA
| | - Congsheng Xu
- Department of Chemistry and Shenzhen Grubbs Institute, Guangdong Provincial Key Laboratory of Catalysis, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Gayoung Seo
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jeongmin An
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Bing Yang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Yuhan Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Tian Lan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jiachen Yan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Shanshan Ren
- Department of Chemistry and Shenzhen Grubbs Institute, Guangdong Provincial Key Laboratory of Catalysis, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yue Xu
- Department of Pathophysiology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Di Xiao
- Department of Pathophysiology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Jonathan K Yan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Claire Ahn
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Dmitry A Fishman
- Department of Chemistry, University of California, Irvine, Irvine, CA, USA
| | - Zhipeng Meng
- Department of Molecular and Cellular Pharmacology and Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Ruxi Qi
- Cryo-EM Center, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Ray Luo
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA.
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, Irvine, CA, USA.
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, CA, USA.
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
4
|
Singh S, Bernal Astrain G, Hincapie AM, Goudreault M, Smith MJ. Complex interplay between RAS GTPases and RASSF effectors regulates subcellular localization of YAP. EMBO Rep 2024; 25:3574-3600. [PMID: 39009833 PMCID: PMC11316025 DOI: 10.1038/s44319-024-00203-9] [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] [Received: 12/06/2023] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/17/2024] Open
Abstract
RAS GTPases bind effectors to convert upstream cues to changes in cellular function. Effectors of classical H/K/NRAS are defined by RBD/RA domains which recognize the GTP-bound conformation of these GTPases, yet the specificity of RBD/RAs for over 160 RAS superfamily proteins remains poorly explored. We have systematically mapped interactions between BRAF and four RASSF effectors, the largest family of RA-containing proteins, with all RAS, RHO and ARF small GTPases. 39 validated complexes reveal plasticity in RASSF binding, while BRAF demonstrates tight specificity for classical H/K/NRAS. Complex between RASSF5 and diverse RAS GTPases at the plasma membrane can activate Hippo signalling and sequester YAP in the cytosol. RASSF8 undergoes liquid-liquid phase separation and resides in YAP-associated membraneless condensates, which also engage several RAS and RHO GTPases. The poorly studied RASSF3 has been identified as a first potential effector of mitochondrial MIRO proteins, and its co-expression with these GTPases impacts mitochondria and peroxisome distribution. These data reveal the complex nature of GTPase-effector interactions and show their systematic elucidation can reveal completely novel and biologically relevant cellular processes.
Collapse
Affiliation(s)
- Swati Singh
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Gabriela Bernal Astrain
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Ana Maria Hincapie
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Marilyn Goudreault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Matthew J Smith
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
5
|
Rukhsana, Supty AT, Hussain M, Lee Y. STK3 higher expression association with clinical characteristics in intrinsic subtypes of breast cancer invasive ductal carcinoma patients. Breast Cancer Res Treat 2024; 206:119-129. [PMID: 38592540 DOI: 10.1007/s10549-024-07248-3] [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] [Received: 12/15/2023] [Accepted: 01/04/2024] [Indexed: 04/10/2024]
Abstract
PURPOSE STK3 has a central role in maintaining cell homeostasis, proliferation, growth, and apoptosis. Previously, we investigated the functional link between STK3/MST2, and estrogen receptor in MCF-7 breast cancer cells. To expand the investigation, this study evaluated STK3's higher expression and associated genes in breast cancer intrinsic subtypes using publicly available data. METHODS The relationship between clinical pathologic features and STK3 high expression was analyzed using descriptive and multivariate analysis. RESULTS Increased STK3 expression in breast cancer was significantly associated with higher pathological cancer stages, and a different expression level was observed in the intrinsic subtypes of breast cancer. Kaplan-Meier analysis showed that breast cancer with high STK3 had a lower survival rate in IDC patients than that with low STK3 expression (p < 0.05). The multivariate analysis unveiled a strong correlation between STK3 expression and the survival rate among IDC patients, demonstrating hazard ratios for lower expression. In the TCGA dataset, the hazard ratio was 0.56 (95% CI 0.34-0.94, p = 0.029) for patients deceased with tumor, and 0.62 (95% CI 0.42-0.92, p = 0.017) for all deceased patients. Additionally, in the METABRIC dataset, the hazard ratio was 0.76 (95% CI 0.64-0.91, p = 0.003) for those deceased with tumor. From GSEA outcomes 7 gene sets were selected based on statistical significance (FDR < 0.25 and p < 0.05). Weighted Sum model (WSM) derived top 5% genes also have higher expression in basal and lower in luminal A in association with STK3. CONCLUSION By introducing a novel bioinformatics approach that combines GSEA and WSM, the study successfully identified the top 5% of genes associated with higher expression of STK3.
Collapse
MESH Headings
- Aged
- Female
- Humans
- Middle Aged
- Biomarkers, Tumor/genetics
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/mortality
- Breast Neoplasms/metabolism
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Kaplan-Meier Estimate
- Neoplasm Staging
- Prognosis
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Serine-Threonine Kinase 3/analysis
- Serine-Threonine Kinase 3/genetics
Collapse
Affiliation(s)
- Rukhsana
- Department of Integrative Bioscience and Biotechnology, College of Life Science, Sejong University, 209-Neungdong-ro, Gwangjin-gu, 05006, Seoul, Korea
- College of Science and Engineering, University of Derby, Kedleston Road, Derby, DE22 1GB, UK
| | - Afia Tasnim Supty
- Department of Integrative Bioscience and Biotechnology, College of Life Science, Sejong University, 209-Neungdong-ro, Gwangjin-gu, 05006, Seoul, Korea
| | - Maqbool Hussain
- College of Science and Engineering, University of Derby, Kedleston Road, Derby, DE22 1GB, UK.
| | - YoungJoo Lee
- Department of Integrative Bioscience and Biotechnology, College of Life Science, Sejong University, 209-Neungdong-ro, Gwangjin-gu, 05006, Seoul, Korea.
| |
Collapse
|
6
|
Gu Y, Ding C, Yu T, Liu B, Tang W, Wang Z, Tang X, Liang G, Peng J, Zhang X, Li Z. SIRT7 promotes Hippo/YAP activation and cancer cell proliferation in hepatocellular carcinoma via suppressing MST1. Cancer Sci 2024; 115:1209-1223. [PMID: 38288904 PMCID: PMC11006999 DOI: 10.1111/cas.16091] [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] [Received: 10/19/2023] [Revised: 12/22/2023] [Accepted: 01/14/2024] [Indexed: 04/12/2024] Open
Abstract
Abnormal activation of the oncogene YAP in the Hippo pathway is a major feature in liver cancer and inactivation of MST1/2 has been shown to be responsible for the overactivation of YAP that led to tumorigenesis. However, mechanisms underlying MST1/2 dysregulation remain poorly understood. RNA-seq analysis and genome (KEGG) pathway enrichment analysis were used to identify genes and pathways that were regulated by SIRT7. qRT-PCR, ChIP, and luciferase assay were used to investigate transcriptional regulation. Mass spectrometry, co-immunoprecipitation and immunoprecipitation were used to exam protein-protein interaction and post-transcriptional modification. A xenograft mouse model was used to confirm the effect of SIRT7 and SIRT7 inhibitors on hepatocellular carcinoma (HCC) proliferation in vivo. We found that SIRT7 suppresses MST1 by both transcriptional regulation and post-transcriptional modification, which in turn promotes YAP nuclear localization and transcriptional activation in liver cancer. Mechanistically, we revealed that SIRT7 suppresses MST1 transcription by binding to the MST1 promoter and inducing H3K18 deacetylation in its promoter region. In addition, SIRT7 directly binds to and deacetylates MST1, which primes acetylation-dependent MST1 ubiquitination and protein degradation. In clinical samples, we confirmed a negative correlation between SIRT7 and MST1 protein levels, and high SIRT7 expression correlated with elevated YAP expression and nuclear localization. In addition, SIRT7 specific inhibitor 2800Z sufficiently inhibited HCC growth by disrupting the SIRT7/MST1/YAP axis. Our data thus revealed the previously undescribed function of SIRT7 in regulating the Hippo pathway in HCC and further proved that targeting SIRT7 might provide novel therapeutic options for the treatment of liver cancer.
Collapse
Affiliation(s)
- Yiying Gu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Cong Ding
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Tingzi Yu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Bohao Liu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Wenbin Tang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Zhiqiang Wang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Xiaohui Tang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Gaoshuang Liang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Jinying Peng
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Xiangwen Zhang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| | - Zhuan Li
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and The Key Laboratory of Model Animals and Stem Cell Biology of Hunan ProvinceHunan Normal University School of MedicineChangshaHunanChina
| |
Collapse
|
7
|
Li T, Wen Y, Lu Q, Hua S, Hou Y, Du X, Zheng Y, Sun S. MST1/2 in inflammation and immunity. Cell Adh Migr 2023; 17:1-15. [PMID: 37909712 PMCID: PMC10761064 DOI: 10.1080/19336918.2023.2276616] [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: 05/04/2023] [Accepted: 10/09/2023] [Indexed: 11/03/2023] Open
Abstract
The mammalian Sterile 20-like kinase 1/2 (MST1/2) belongs to the serine/threonine (GC) protein kinase superfamily. Collective studies confirm the vital role MST1/2 in inflammation and immunity. MST1/2 is closely related to the progress of inflammation. Generally, MST1/2 aggravates the inflammatory injury through MST1-JNK, MST1-mROS, MST1-Foxo3, and NF-κB pathways, as well as several regulatory factors such as tumor necrosis factor-α (TNF-α), mitochondrial extension factor 1 (MIEF1), and lipopolysaccharide (LPS). Moreover, MST1/2 is also involved in the regulation of immunity to balance immune activation and tolerance by regulating MST1/2-Rac, MST1-Akt1/c-myc, MST1-Foxos, MST1-STAT, Btk pathways, and lymphocyte function-related antigen 1 (LFA-1), which subsequently prevents immunodeficiency syndrome and autoimmune diseases. This article reviews the effects of MST1/2 on inflammation and immunity.
Collapse
Affiliation(s)
- Tongfen Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yiqiong Wen
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Qiongfen Lu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shu Hua
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yunjiao Hou
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaohua Du
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yuanyuan Zheng
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| |
Collapse
|
8
|
Sun Y, Jin X, Meng J, Guo F, Chen T, Zhao X, Wu H, Ren D. MST2 methylation by PRMT5 inhibits Hippo signaling and promotes pancreatic cancer progression. EMBO J 2023; 42:e114558. [PMID: 37905571 PMCID: PMC10690468 DOI: 10.15252/embj.2023114558] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 11/02/2023] Open
Abstract
The Hippo signaling axis is a tumor suppressor pathway that is activated by various extra-pathway factors to regulate cell differentiation and organ development. Recent studies have reported that autophosphorylation of the core kinase cassette stimulates activation of the Hippo signaling cascade. Here, we demonstrate that protein arginine methyltransferase 5 (PRMT5) contributes to inactivation of the Hippo signaling pathway in pancreatic cancer. We show that the Hippo pathway initiator serine/threonine kinase 3 (STK3, also known as MST2) of Hippo signaling pathway can be symmetrically di-methylated by PRMT5 at arginine-461 (R461) and arginine-467 (R467) in its SARAH domain. Methylation suppresses MST2 autophosphorylation and kinase activity by blocking its homodimerization, thereby inactivating Hippo signaling pathway in pancreatic cancer. Moreover, we also show that the specific PRMT5 inhibitor GSK3326595 re-activates the dysregulated Hippo signaling pathway and inhibits the growth of human pancreatic cancer xenografts in immunodeficient mice, thus suggesting potential clinical application of PRMT5 inhibitors in pancreatic cancer.
Collapse
Affiliation(s)
- Yan Sun
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xin Jin
- Department of Urology, The Second Xiangya HospitalCentral South UniversityChangshaChina
- Uro‐Oncology Institute of Central South UniversityChangshaChina
| | - Junpeng Meng
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of General SurgeryThe Second Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Feng Guo
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Taoyu Chen
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaoyan Zhao
- Department of Hematology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dianyun Ren
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Sino‐German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
9
|
Smith MJ. Defining bone fide effectors of RAS GTPases. Bioessays 2023; 45:e2300088. [PMID: 37401638 DOI: 10.1002/bies.202300088] [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: 05/24/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
RAS GTPases play essential roles in normal development and are direct drivers of human cancers. Three decades of study have failed to wholly characterize pathways stimulated by activated RAS, driven by engagement with 'effector' proteins that have RAS binding domains (RBDs). Bone fide effectors must bind directly to RAS GTPases in a nucleotide-dependent manner, and this interaction must impart a clear change in effector activity. Despite this, for most proteins currently deemed effectors there is little mechanistic understanding of how binding to the GTPase alters protein function. There has also been limited effort to comprehensively resolve the specificity of effector binding to the full array of RAS superfamily GTPase proteins. This review will summarize what is known about RAS-driven activation for an array of potential effector proteins, focusing on structural and mechanistic effects and highlighting how little is still known regarding this key paradigm of cellular signal transduction.
Collapse
Affiliation(s)
- Matthew J Smith
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Québec, Canada
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
10
|
Weingartner KA, Tran T, Tripp KW, Kavran JM. Dimerization and autophosphorylation of the MST family of kinases are controlled by the same set of residues. Biochem J 2023; 480:1165-1182. [PMID: 37459121 PMCID: PMC10500444 DOI: 10.1042/bcj20230067] [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] [Received: 03/08/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/28/2023]
Abstract
The Hippo pathway controls tissue growth and regulates stem cell fate through the activities of core kinase cassette that begins with the Sterile 20-like kinase MST1/2. Activation of MST1/2 relies on trans-autophosphorylation but the details of the mechanisms regulating that reaction are not fully elucidated. Proposals include dimerization as a first step and include multiple models for potential kinase-domain dimers. Efforts to verify and link these dimers to trans-autophosphorylation were unsuccessful. We explored the link between dimerization and trans-autophosphorylation for MST2 and the entire family of MST kinases. We analyzed crystal lattice contacts of structures of MST kinases and identified an ensemble of kinase-domain dimers compatible with trans-autophosphorylation. These dimers share a common dimerization interface comprised of the activation loop and αG-helix while the arrangements of the kinase-domains within the dimer varied depending on their activation state. We then verified the dimerization interface and determined its function using MST2. Variants bearing alanine substitutions of the αG-helix prevented dimerization of the MST2 kinase domain both in solution and in cells. These substitutions also blocked autophosphorylation of full-length MST2 and its Drosophila homolog Hippo in cells. These variants retain the same secondary structure as wild-type and capacity to phosphorylate a protein substrate, indicating the loss of MST2 activation can be directly attributed to a loss of dimerization rather than loss of either fold or catalytic function. Together this data functionally links dimerization and autophosphorylation for MST2 and suggests this activation mechanism is conserved across both species and the entire MST family.
Collapse
Affiliation(s)
- Kyler A. Weingartner
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Thao Tran
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Katherine W. Tripp
- The T.C. Jenkins Department of Biophysics, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Jennifer M. Kavran
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
- Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
11
|
Liu YX, Wan S, Yang XQ, Wang Y, Gan WJ, Ye WL, He XS, Chen JJ, Yang Y, Yang XM, Guo X, Gao XJ, Lu YT, Deng ZY, Hu G, Wu H. TRIM21 is a druggable target for the treatment of metastatic colorectal cancer through ubiquitination and activation of MST2. Cell Chem Biol 2023:S2451-9456(23)00152-6. [PMID: 37354905 DOI: 10.1016/j.chembiol.2023.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 04/09/2023] [Accepted: 05/25/2023] [Indexed: 06/26/2023]
Abstract
Metastatic colorectal cancer (mCRC) is characterized by poorer prognosis of patients and limited therapeutic approach, partly due to the lack of effective target. Using mouse models and tumor organoids, this study reported a tripartite motif 21 (TRIM21) protein, exerting potential inhibitory effects on the invasion and metastasis of CRC. Mechanistically, TRIM21 directly interacted with and ubiquitinated MST2 at lysine 473 (K473) via K63-linkage. This ubiquitination enabled the formation of MST2 homodimer and enhanced its kinase activity, ultimately resulting in the functional inactivation of yes-associated protein (YAP) and inhibition of an epithelial-mesenchymal transition (EMT) feature. We identified that vilazodone, an antidepressant, directly bound to TRIM21 to exert effective anti-metastatic action both in vitro and in vivo. Collectively, these findings revealed a previously unrecognized interplay between TRIM21 and the Hippo-YAP signaling. These results suggested that vilazodone could be repositioned as an anti-tumor drug to inhibit CRC metastasis by targeting TRIM21.
Collapse
Affiliation(s)
- Yu-Xuan Liu
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Shan Wan
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Xiao-Qin Yang
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Yi Wang
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Juan Gan
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Long Ye
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Xiao-Shun He
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Jun-Jie Chen
- Analysis and Measurement Centre, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Yun Yang
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Xue-Mei Yang
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Xin Guo
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Xiao-Jiao Gao
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China
| | - Yi-Tan Lu
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Zhi-Yong Deng
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China.
| | - Guang Hu
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China.
| | - Hua Wu
- Department of Pathology, Medical Center of Soochow University & Suzhou Medical College of Soochow University & Institute for Excellence in Clinical Medicine of Kunshan First People's Hospital and Soochow University, Soochow University, Suzhou 215123, China.
| |
Collapse
|
12
|
Yin Y, Tan M, Han L, Zhang L, Zhang Y, Zhang J, Pan W, Bai J, Jiang T, Li H. The hippo kinases MST1/2 in cardiovascular and metabolic diseases: A promising therapeutic target option for pharmacotherapy. Acta Pharm Sin B 2023; 13:1956-1975. [PMID: 37250161 PMCID: PMC10213817 DOI: 10.1016/j.apsb.2023.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/09/2022] [Accepted: 11/18/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) and metabolic disorders are major components of noncommunicable diseases, causing an enormous health and economic burden worldwide. There are common risk factors and developmental mechanisms among them, indicating the far-reaching significance in exploring the corresponding therapeutic targets. MST1/2 kinases are well-established proapoptotic effectors that also bidirectionally regulate autophagic activity. Recent studies have demonstrated that MST1/2 influence the outcome of cardiovascular and metabolic diseases by regulating immune inflammation. In addition, drug development against them is in full swing. In this review, we mainly describe the roles and mechanisms of MST1/2 in apoptosis and autophagy in cardiovascular and metabolic events as well as emphasis on the existing evidence for their involvement in immune inflammation. Moreover, we summarize the latest progress of pharmacotherapy targeting MST1/2 and propose a new mode of drug combination therapy, which may be beneficial to seek more effective strategies to prevent and treat CVDs and metabolic disorders.
Collapse
Affiliation(s)
- Yunfei Yin
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Mingyue Tan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lianhua Han
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lei Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yue Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jun Zhang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Wanqian Pan
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jiaxiang Bai
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Department of Orthopedics, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tingbo Jiang
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Hongxia Li
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
13
|
Bonello TT, Cai D, Fletcher GC, Wiengartner K, Pengilly V, Lange KS, Liu Z, Lippincott‐Schwartz J, Kavran JM, Thompson BJ. Phase separation of Hippo signalling complexes. EMBO J 2023; 42:e112863. [PMID: 36807601 PMCID: PMC10015380 DOI: 10.15252/embj.2022112863] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/12/2023] [Accepted: 01/23/2023] [Indexed: 02/22/2023] Open
Abstract
The Hippo pathway was originally discovered to control tissue growth in Drosophila and includes the Hippo kinase (Hpo; MST1/2 in mammals), scaffold protein Salvador (Sav; SAV1 in mammals) and the Warts kinase (Wts; LATS1/2 in mammals). The Hpo kinase is activated by binding to Crumbs-Expanded (Crb-Ex) and/or Merlin-Kibra (Mer-Kib) proteins at the apical domain of epithelial cells. Here we show that activation of Hpo also involves the formation of supramolecular complexes with properties of a biomolecular condensate, including concentration dependence and sensitivity to starvation, macromolecular crowding, or 1,6-hexanediol treatment. Overexpressing Ex or Kib induces formation of micron-scale Hpo condensates in the cytoplasm, rather than at the apical membrane. Several Hippo pathway components contain unstructured low-complexity domains and purified Hpo-Sav complexes undergo phase separation in vitro. Formation of Hpo condensates is conserved in human cells. We propose that apical Hpo kinase activation occurs in phase separated "signalosomes" induced by clustering of upstream pathway components.
Collapse
Affiliation(s)
- Teresa T Bonello
- EMBL Australia, John Curtin School of Medical ResearchAustralian National UniversityCanberraACTAustralia
| | - Danfeng Cai
- HHMI Janelia Research CampusAshburnVAUSA
- Department of Biochemistry and Molecular BiologyBloomberg School of Public HealthBaltimoreMDUSA
| | | | - Kyler Wiengartner
- Department of Biochemistry and Molecular BiologyBloomberg School of Public HealthBaltimoreMDUSA
| | - Victoria Pengilly
- EMBL Australia, John Curtin School of Medical ResearchAustralian National UniversityCanberraACTAustralia
| | - Kimberly S Lange
- Department of Biochemistry and Molecular BiologyBloomberg School of Public HealthBaltimoreMDUSA
| | - Zhe Liu
- HHMI Janelia Research CampusAshburnVAUSA
| | | | - Jennifer M Kavran
- Department of Biochemistry and Molecular BiologyBloomberg School of Public HealthBaltimoreMDUSA
- Department of Biophysics and Biophysical Chemistry, and Department of OncologyJohns Hopkins School of MedicineBaltimoreMDUSA
| | - Barry J Thompson
- EMBL Australia, John Curtin School of Medical ResearchAustralian National UniversityCanberraACTAustralia
- Epithelial Biology LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
14
|
Weingartner KA, Tran T, Tripp KW, Kavran JM. Dimerization and autophosphorylation of the MST family of kinases are controlled by the same set of residues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531926. [PMID: 36945437 PMCID: PMC10028985 DOI: 10.1101/2023.03.09.531926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
The Hippo pathway controls tissue growth and regulates stem cell fate through the activities of core kinase cassette that begins with the Sterile 20-like kinase MST1/2. Activation of MST1/2 relies on trans -autophosphorylation but the details of the mechanisms regulating that reaction are not fully elucidated. Proposals include dimerization as a first step and include multiple models for potential kinase-domain dimers. Efforts to verify and link these dimers to trans -autophosphorylation were unsuccessful. We explored the link between dimerization and trans -autophosphorylation for MST2 and the entire family of MST kinases. We analyzed crystal lattice contacts of structures of MST kinases and identified an ensemble of kinase-domain dimers compatible with trans -autophosphorylation. These dimers share a common dimerization interface comprised of the activation loop and αG-helix while the arrangements of the kinase-domains within the dimer varied depending on their activation state. We then verified the dimerization interface and determined its function using MST2. Variants bearing alanine substitutions of the αG-helix prevented dimerization of the MST2 kinase domain both in solution and in cells. These substitutions also blocked autophosphorylation of full-length MST2 and its Drosophila homolog Hippo in cells. These variants retain the same secondary structure as wild-type and capacity to phosphorylate a protein substrate, indicating the loss of MST2 activation can be directly attributed to a loss of dimerization rather than loss of either fold or catalytic function. Together this data functionally links dimerization and autophosphorylation for MST2 and suggests this activation mechanism is conserved across both species and the entire MST family.
Collapse
|
15
|
Koehler TJ, Tran T, Weingartner KA, Kavran JM. Kinetic Regulation of the Mammalian Sterile 20-like Kinase 2 (MST2). Biochemistry 2022; 61:1683-1693. [PMID: 35895874 PMCID: PMC10167949 DOI: 10.1021/acs.biochem.2c00022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Canonically, MST1/2 functions as a core kinase of the Hippo pathway and noncanonically during both apoptotic signaling and with RASSFs in T-cells. Faithful signal transduction by MST1/2 relies on both appropriate activation and regulated substrate phosphorylation by the activated kinase. Considerable progress has been made in understanding the molecular mechanisms regulating the activation of MST1/2 and identifying downstream signaling events. Here, we investigated the ability of MST2 to phosphorylate a peptide substrate and how that activity is regulated. Using a steady-state kinetic system, we parse the contribution of different factors to substrate phosphorylation, including the domains of MST2, phosphorylation, caspase cleavage, and complex formation. We found that in the unphosphorylated state, the SARAH domain stabilizes interactions with a peptide substrate and promotes turnover. Phosphorylation drives the activity of MST2, and once activated, MST2 is not further regulated by complex formation with other Hippo pathway components (SAV1, MOB1A, and RASSF5). We also show that the phosphorylated, caspase-cleaved MST2 is as active as the full-length one, suggesting that caspase-stimulated activity arises through noncatalytic mechanisms. The kinetic analysis presented here establishes a framework for interpreting how signaling events and post-translational modifications contribute to the signaling of MST2 in vivo.
Collapse
Affiliation(s)
- Thomas J Koehler
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Thao Tran
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Kyler A Weingartner
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Jennifer M Kavran
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205, United States.,Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States.,Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, United States
| |
Collapse
|
16
|
O-GlcNAcylation: An Emerging Protein Modification Regulating the Hippo Pathway. Cancers (Basel) 2022; 14:cancers14123013. [PMID: 35740678 PMCID: PMC9221189 DOI: 10.3390/cancers14123013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/13/2022] [Accepted: 06/16/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The contact point between the Hippo pathway, which serves as a central hub for various external environments, and O-GlcNAcylation, which is a non-canonical glycosylation process acting as a dynamic regulator in various signal transduction pathways, has recently been identified. This review aims to summarize the function of O-GlcNAcylation as an intrinsic and extrinsic regulator of the Hippo pathway. Abstract The balance between cellular proliferation and apoptosis and the regulation of cell differentiation must be established to maintain tissue homeostasis. These cellular responses involve the kinase cascade-mediated Hippo pathway as a crucial regulator. Hence, Hippo pathway dysregulation is implicated in diverse diseases, including cancer. O-GlcNAcylation is a non-canonical glycosylation that affects multiple signaling pathways through its interplay with phosphorylation in the nucleus and cytoplasm. An abnormal increase in the O-GlcNAcylation levels in various cancer cells is a potent factor in Hippo pathway dysregulation. Intriguingly, Hippo pathway dysregulation also disrupts O-GlcNAc homeostasis, leading to a persistent elevation of O-GlcNAcylation levels, which is potentially pathogenic in several diseases. Therefore, O-GlcNAcylation is gaining attention as a protein modification that regulates the Hippo pathway. This review presents a framework on how O-GlcNAcylation regulates the Hippo pathway and forms a self-perpetuating cycle with it. The pathological significance of this self-perpetuating cycle and clinical strategies for targeting O-GlcNAcylation that causes Hippo pathway dysregulation are also discussed.
Collapse
|
17
|
Guo CL. Self-Sustained Regulation or Self-Perpetuating Dysregulation: ROS-dependent HIF-YAP-Notch Signaling as a Double-Edged Sword on Stem Cell Physiology and Tumorigenesis. Front Cell Dev Biol 2022; 10:862791. [PMID: 35774228 PMCID: PMC9237464 DOI: 10.3389/fcell.2022.862791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/29/2022] [Indexed: 12/19/2022] Open
Abstract
Organ development, homeostasis, and repair often rely on bidirectional, self-organized cell-niche interactions, through which cells select cell fate, such as stem cell self-renewal and differentiation. The niche contains multiplexed chemical and mechanical factors. How cells interpret niche structural information such as the 3D topology of organs and integrate with multiplexed mechano-chemical signals is an open and active research field. Among all the niche factors, reactive oxygen species (ROS) have recently gained growing interest. Once considered harmful, ROS are now recognized as an important niche factor in the regulation of tissue mechanics and topology through, for example, the HIF-YAP-Notch signaling pathways. These pathways are not only involved in the regulation of stem cell physiology but also associated with inflammation, neurological disorder, aging, tumorigenesis, and the regulation of the immune checkpoint molecule PD-L1. Positive feedback circuits have been identified in the interplay of ROS and HIF-YAP-Notch signaling, leading to the possibility that under aberrant conditions, self-organized, ROS-dependent physiological regulations can be switched to self-perpetuating dysregulation, making ROS a double-edged sword at the interface of stem cell physiology and tumorigenesis. In this review, we discuss the recent findings on how ROS and tissue mechanics affect YAP-HIF-Notch-PD-L1 signaling, hoping that the knowledge can be used to design strategies for stem cell-based and ROS-targeting therapy and tissue engineering.
Collapse
Affiliation(s)
- Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
18
|
Kilanowska A, Ziółkowska A. Apoptosis in Type 2 Diabetes: Can It Be Prevented? Hippo Pathway Prospects. Int J Mol Sci 2022; 23:636. [PMID: 35054822 PMCID: PMC8775644 DOI: 10.3390/ijms23020636] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/20/2021] [Accepted: 01/05/2022] [Indexed: 02/04/2023] Open
Abstract
Diabetes mellitus is a heterogeneous disease of complex etiology and pathogenesis. Hyperglycemia leads to many serious complications, but also directly initiates the process of β cell apoptosis. A potential strategy for the preservation of pancreatic β cells in diabetes may be to inhibit the implementation of pro-apoptotic pathways or to enhance the action of pancreatic protective factors. The Hippo signaling pathway is proposed and selected as a target to manipulate the activity of its core proteins in therapy-basic research. MST1 and LATS2, as major upstream signaling kinases of the Hippo pathway, are considered as target candidates for pharmacologically induced tissue regeneration and inhibition of apoptosis. Manipulating the activity of components of the Hippo pathway offers a wide range of possibilities, and thus is a potential tool in the treatment of diabetes and the regeneration of β cells. Therefore, it is important to fully understand the processes involved in apoptosis in diabetic states and completely characterize the role of this pathway in diabetes. Therapy consisting of slowing down or stopping the mechanisms of apoptosis may be an important direction of diabetes treatment in the future.
Collapse
Affiliation(s)
- Agnieszka Kilanowska
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zyty 28, 65-001 Zielona Gora, Poland;
| | | |
Collapse
|
19
|
Singh A, Erijman A, Noronha A, Kumar H, Peleg Y, Yarden Y, Shifman JM. Engineered variants of the Ras effector protein RASSF5 (NORE1A) promote anticancer activities in lung adenocarcinoma. J Biol Chem 2021; 297:101353. [PMID: 34717958 PMCID: PMC8605244 DOI: 10.1016/j.jbc.2021.101353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/12/2021] [Accepted: 10/25/2021] [Indexed: 11/24/2022] Open
Abstract
Within the superfamily of small GTPases, Ras appears to be the master regulator of such processes as cell cycle progression, cell division, and apoptosis. Several oncogenic Ras mutations at amino acid positions 12, 13, and 61 have been identified that lose their ability to hydrolyze GTP, giving rise to constitutive signaling and eventually development of cancer. While disruption of the Ras/effector interface is an attractive strategy for drug design to prevent this constitutive activity, inhibition of this interaction using small molecules is impractical due to the absence of a cavity to which such molecules could bind. However, proteins and especially natural Ras effectors that bind to the Ras/effector interface with high affinity could disrupt Ras/effector interactions and abolish procancer pathways initiated by Ras oncogene. Using a combination of computational design and in vitro evolution, we engineered high-affinity Ras-binding proteins starting from a natural Ras effector, RASSF5 (NORE1A), which is encoded by a tumor suppressor gene. Unlike previously reported Ras oncogene inhibitors, the proteins we designed not only inhibit Ras-regulated procancer pathways, but also stimulate anticancer pathways initiated by RASSF5. We show that upon introduction into A549 lung carcinoma cells, the engineered RASSF5 mutants decreased cell viability and mobility to a significantly greater extent than WT RASSF5. In addition, these mutant proteins induce cellular senescence by increasing acetylation and decreasing phosphorylation of p53. In conclusion, engineered RASSF5 variants provide an attractive therapeutic strategy able to oppose cancer development by means of inhibiting of procancer pathways and stimulating anticancer processes.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ariel Erijman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ashish Noronha
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Hemant Kumar
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yoav Peleg
- Life Sciences Core Facilities (LSCF) Structural Proteomics Unit (SPU), Weizmann Institute of Science, Rehovot, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Julia M Shifman
- Department of Biological Chemistry, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
20
|
RAS GTPase signalling to alternative effector pathways. Biochem Soc Trans 2021; 48:2241-2252. [PMID: 33125484 DOI: 10.1042/bst20200506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
Abstract
RAS GTPases are fundamental regulators of development and drivers of an extraordinary number of human cancers. RAS oncoproteins constitutively signal through downstream effector proteins, triggering cancer initiation, progression and metastasis. In the absence of targeted therapeutics to mutant RAS itself, inhibitors of downstream pathways controlled by the effector kinases RAF and PI3K have become tools in the treatment of RAS-driven tumours. Unfortunately, the efficacy of this approach has been greatly minimized by the prevalence of acquired drug resistance. Decades of research have established that RAS signalling is highly complex, and in addition to RAF and PI3K these small GTPase proteins can interact with an array of alternative effectors that feature RAS binding domains. The consequence of RAS binding to these effectors remains relatively unexplored, but these pathways may provide targets for combinatorial therapeutics. We discuss here three candidate alternative effectors: RALGEFs, RASSF5 and AFDN, detailing their interaction with RAS GTPases and their biological significance. The metastatic nature of RAS-driven cancers suggests more attention should be granted to these alternate pathways, as they are highly implicated in the regulation of cell adhesion, polarity, cell size and cytoskeletal architecture.
Collapse
|
21
|
Huang Y, Ren Q. A newly identified Hippo homologue from the oriental river prawn Macrobrachium nipponense is involved in the antimicrobial immune response. Vet Res 2021; 52:76. [PMID: 34078461 PMCID: PMC8170997 DOI: 10.1186/s13567-021-00945-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/27/2021] [Indexed: 01/02/2023] Open
Abstract
The Hippo signalling pathway plays a vital role in organ size control, cell proliferation, apoptosis, and immune regulation. In this study, a Hippo homologue with three isoforms (named MnHippo-a, MnHippo-b, and MnHippo-c) was isolated and characterized for the first time from the freshwater prawn Macrobrachium nipponense. The deduced amino acid sequences of MnHippo-a (698 aa), MnHippo-b (688 aa), and MnHippo-c (656 aa) were highly similar, and they all contained an N-terminal S_TKc (serine/threonine protein kinase catalytic) domain and a C-terminal Mst1_SARAH (Sav/Rassf/Hpo) domain. MnHippo-a and MnHippo-c were derived from alternative splicing. Phylogenetic analysis was performed, and the results revealed that MnHippo was a member of the clade containing STPK4 and Hippo of Penaeus vannamei. The expression distribution showed that MnHippo was constitutively expressed in various tissues of uninfected prawns and highly expressed in the hepatopancreas and intestine. In prawns challenged with Vibrio parahaemolyticus and Staphylococcus aureus, the expression of MnHippo in haemocytes was significantly upregulated. Furthermore, in MnHippo-knockdown prawns injected with V. parahaemolyticus or S. aureus, the transcription levels of five antimicrobial peptides were downregulated. MnHippo silencing weakened the clearance of V. parahaemolyticus and S. aureus in prawns. The survival rate of the MnHippo-dsRNA group was obviously decreased from 2 to 6 days post-injection with V. parahaemolyticus or S. aureus. Hence, MnHippo might be involved in the antibacterial immune defence of M. nipponense.
Collapse
Affiliation(s)
- Ying Huang
- College of Oceanography, Hohai University, 1 Xikang Road, Nanjing, 210098, Jiangsu, China
| | - Qian Ren
- College of Marine Science and Engineering, Nanjing Normal University, 1 Wenyuan Road, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
22
|
Regulation of MST complexes and activity via SARAH domain modifications. Biochem Soc Trans 2021; 49:675-683. [PMID: 33860801 DOI: 10.1042/bst20200559] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 11/17/2022]
Abstract
Three elements of the Hippo tumor suppressor pathway - MST1/2, SAV1, and RASSF1-6 - share in common a C-terminal interaction motif termed the SARAH domain. Proteins containing this domain are capable of self-association as homodimers and also of trans-association with other SARAH domain containing proteins as well as selected additional proteins that lack this domain. Recently, the association of MST1/2 with itself or with other proteins has been shown to be regulated by phosphorylation at sites near or within the SARAH domain. In this review, we focus on recent findings regarding the regulation of such MST1/2 interactions, with an emphasis on the effects of these events on Hippo pathway activity.
Collapse
|
23
|
Cryo-EM structure of the Hippo signaling integrator human STRIPAK. Nat Struct Mol Biol 2021; 28:290-299. [PMID: 33633399 PMCID: PMC8315899 DOI: 10.1038/s41594-021-00564-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 01/20/2021] [Indexed: 01/31/2023]
Abstract
The striatin-interacting phosphatase and kinase (STRIPAK) complex is a large, multisubunit protein phosphatase 2A (PP2A) assembly that integrates diverse cellular signals in the Hippo pathway to regulate cell proliferation and survival. The architecture and assembly mechanism of this critical complex are poorly understood. Using cryo-EM, we determine the structure of the human STRIPAK core comprising PP2AA, PP2AC, STRN3, STRIP1, and MOB4 at 3.2-Å resolution. Unlike the canonical trimeric PP2A holoenzyme, STRIPAK contains four copies of STRN3 and one copy of each the PP2AA-C heterodimer, STRIP1, and MOB4. The STRN3 coiled-coil domains form an elongated homotetrameric scaffold that links the complex together. An inositol hexakisphosphate (IP6) is identified as a structural cofactor of STRIP1. Mutations of key residues at subunit interfaces disrupt the integrity of STRIPAK, causing aberrant Hippo pathway activation. Thus, STRIPAK is established as a noncanonical PP2A complex with four copies of regulatory STRN3 for enhanced signal integration.
Collapse
|
24
|
MST2 silencing induces apoptosis and inhibits tumor growth for estrogen receptor alpha-positive MCF-7 breast cancer. Toxicol Appl Pharmacol 2020; 408:115257. [PMID: 33007383 DOI: 10.1016/j.taap.2020.115257] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/17/2020] [Accepted: 09/27/2020] [Indexed: 12/14/2022]
Abstract
Mammalian sterile 20-like kinase 1/2 (MST1/2) plays an important role in cell growth and apoptosis and functions as a tumor suppressor. Previously, we showed that MST2 overexpression activates Estrogen receptor alpha (ERα) in human breast cancer MCF-7 cells in the absence of a ligand. Here, we examined the role of MST2 in the growth of ER-positive MCF-7 cells. Cell cycle, apoptosis, and mammosphere formation assay method were implemented to detect the biological effects of MST2 ablation on the growth of MCF-7 cells in vitro. The effect of MST2-siRNA on MCF-7 cells tumor growth in vivo was studied in tumor-bearing mouse model. Kaplan-Meier plotter analysis was used to determine the effect of MST2 on overall survival in breast cancer patients. MST2 overexpression increased cell viability marginally. The ablation of MST2 using siRNA dramatically suppressed the viability of the MCF-7 cells, but not ER-negative MDA-MB-231 breast cancer cells. Furthermore, MST2 knockdown increased caspase-dependent apoptosis and led to decreased mammosphere formation. Treatment of MCF-7 tumor-bearing mice with MST2 siRNA significantly inhibited tumor growth. The tumor weight was reduced further when tamoxifen was added. Patients with ER-positive breast cancer with low MST2 expression had better overall survival than did those with high MST2 expression in Kaplan-Meier survival analyses using public datasets. Our results provide new insight into the role of MST2, a key component of the Hippo signaling pathway, in mediating breast cancer progression.
Collapse
|
25
|
Tran T, Mitra J, Ha T, Kavran JM. Increasing kinase domain proximity promotes MST2 autophosphorylation during Hippo signaling. J Biol Chem 2020; 295:16166-16179. [PMID: 32994222 DOI: 10.1074/jbc.ra120.015723] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/22/2020] [Indexed: 01/20/2023] Open
Abstract
The Hippo pathway plays an important role in developmental biology, mediating organ size by controlling cell proliferation through the activity of a core kinase cassette. Multiple upstream events activate the pathway, but how each controls this core kinase cassette is not fully understood. Activation of the core kinase cassette begins with phosphorylation of the kinase MST1/2 (also known as STK3/4). Here, using a combination of in vitro biochemistry and cell-based assays, including chemically induced dimerization and single-molecule pulldown, we revealed that increasing the proximity of adjacent kinase domains, rather than formation of a specific protein assembly, is sufficient to trigger autophosphorylation. We validate this mechanism in cells and demonstrate that multiple events associated with the active pathway, including SARAH domain-mediated homodimerization, membrane recruitment, and complex formation with the effector protein SAV1, each increase the kinase domain proximity and autophosphorylation of MST2. Together, our results reveal that multiple and distinct upstream signals each utilize the same common molecular mechanism to stimulate MST2 autophosphorylation. This mechanism is likely conserved among MST2 homologs. Our work also highlights potential differences in Hippo signal propagation between each activating event owing to differences in the dynamics and regulation of each protein ensemble that triggers MST2 autophosphorylation and possible redundancy in activation.
Collapse
Affiliation(s)
- Thao Tran
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jaba Mitra
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA; Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Taekjip Ha
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA; Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; T. C. Jenkins Department of Biophysics, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Howard Hughes Medical Institute, Baltimore, Maryland, USA
| | - Jennifer M Kavran
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA; Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
26
|
Hossain MS, Roy AS, Islam MS. In silico analysis predicting effects of deleterious SNPs of human RASSF5 gene on its structure and functions. Sci Rep 2020; 10:14542. [PMID: 32884013 PMCID: PMC7471297 DOI: 10.1038/s41598-020-71457-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 07/17/2020] [Indexed: 11/09/2022] Open
Abstract
Ras association domain-containing protein 5 (RASSF5), one of the prospective biomarkers for tumors, generally plays a crucial role as a tumor suppressor. As deleterious effects can result from functional differences through SNPs, we sought to analyze the most deleterious SNPs of RASSF5 as well as predict the structural changes associated with the mutants that hamper the normal protein-protein interactions. We adopted both sequence and structure based approaches to analyze the SNPs of RASSF5 protein. We also analyzed the putative post translational modification sites as well as the altered protein-protein interactions that encompass various cascades of signals. Out of all the SNPs obtained from the NCBI database, only 25 were considered as highly deleterious by six in silico SNP prediction tools. Among them, upon analyzing the effect of these nsSNPs on the stability of the protein, we found 17 SNPs that decrease the stability. Significant deviation in the energy minimization score was observed in P350R, F321L, and R277W. Besides this, docking analysis confirmed that P350R, A319V, F321L, and R277W reduce the binding affinity of the protein with H-Ras, where P350R shows the most remarkable deviation. Protein-protein interaction analysis revealed that RASSF5 acts as a hub connecting two clusters consisting of 18 proteins and alteration in the RASSF5 may lead to disassociation of several signal cascades. Thus, based on these analyses, our study suggests that the reported functional SNPs may serve as potential targets for different proteomic studies, diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Md Shahadat Hossain
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Arpita Singha Roy
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Sajedul Islam
- Department of Biochemistry and Biotechnology, University of Barishal, Barishal, Bangladesh.
| |
Collapse
|
27
|
Ooki T, Hatakeyama M. Hyaluronan Degradation Promotes Cancer via Hippo-YAP Signaling: An Intervention Point for Cancer Therapy. Bioessays 2020; 42:e2000005. [PMID: 32449813 DOI: 10.1002/bies.202000005] [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] [Received: 01/15/2020] [Revised: 03/16/2020] [Indexed: 12/14/2022]
Abstract
High-molecular-weight hyaluronan acts as a ligand of the tumor-suppressive Hippo signal, whereas degradation of hyaluronan from a high-molecular-weight form to a low-molecular-weight forms by hyaluronidase 2 inhibits Hippo signal activation and thereby activates the pro-oncogenic transcriptional coactivator yes-associated protein (YAP), which creates a cancer-predisposing microenvironment and drives neoplastic transformation of cells through both cell-autonomous and non-cell-autonomous mechanisms. In fact, accumulation of low-molecular-weight hyaluronan in tissue stroma is observed in many types of cancers. Since inhibition of YAP activity suppresses tumor growth in vivo, pharmacological intervention of the Hippo-YAP signal is an attractive approach for future drug development. In this review, pharmacological intervention of excessive hyaluronan degradation as a novel approach for inhibition of the Hippo-YAP signal is also discussed. Development of hyaluronidase inhibitors may provide novel therapeutic strategies for human malignant tumors.
Collapse
Affiliation(s)
- Takuya Ooki
- Division of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-0033, Japan
| | - Masanori Hatakeyama
- Division of Microbiology, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
28
|
Bae SJ, Ni L, Luo X. STK25 suppresses Hippo signaling by regulating SAV1-STRIPAK antagonism. eLife 2020; 9:e54863. [PMID: 32292165 PMCID: PMC7182433 DOI: 10.7554/elife.54863] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/10/2020] [Indexed: 12/11/2022] Open
Abstract
The MST-LATS kinase cascade is central to the Hippo pathway that controls tissue homeostasis, development, and organ size. The PP2A complex STRIPAKSLMAP blocks MST1/2 activation. The GCKIII family kinases associate with STRIPAK, but the functions of these phosphatase-associated kinases remain elusive. We previously showed that the scaffolding protein SAV1 promotes Hippo signaling by counteracting STRIPAK (Bae et al., 2017). Here, we show that the GCKIII kinase STK25 promotes STRIPAK-mediated inhibition of MST2 in human cells. Depletion of STK25 enhances MST2 activation without affecting the integrity of STRIPAKSLMAP. STK25 directly phosphorylates SAV1 and diminishes the ability of SAV1 to inhibit STRIPAK. Thus, STK25 as the kinase component of STRIPAK can inhibit the function of the STRIPAK inhibitor SAV1. This mutual antagonism between STRIPAK and SAV1 controls the initiation of Hippo signaling.
Collapse
Affiliation(s)
- Sung Jun Bae
- Department of Pharmacology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Lisheng Ni
- Department of Pharmacology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Xuelian Luo
- Department of Pharmacology, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Biophysics University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
29
|
Cairns L, Patterson A, Weingartner KA, Koehler TJ, DeAngelis DR, Tripp KW, Bothner B, Kavran JM. Biophysical characterization of SARAH domain-mediated multimerization of Hippo pathway complexes in Drosophila. J Biol Chem 2020; 295:6202-6213. [PMID: 32213597 DOI: 10.1074/jbc.ra120.012679] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/23/2020] [Indexed: 11/06/2022] Open
Abstract
Hippo pathway signaling limits cell growth and proliferation and maintains the stem-cell niche. These cellular events result from the coordinated activity of a core kinase cassette that is regulated, in part, by interactions involving Hippo, Salvador, and dRassF. These interactions are mediated by a conserved coiled-coil domain, termed SARAH, in each of these proteins. SARAH domain-mediated homodimerization of Hippo kinase leads to autophosphorylation and activation. Paradoxically, SARAH domain-mediated heterodimerization between Hippo and Salvador enhances Hippo kinase activity in cells, whereas complex formation with dRassF inhibits it. To better understand the mechanism by which each complex distinctly modulates Hippo kinase and pathway activity, here we biophysically characterized the entire suite of SARAH domain-mediated complexes. We purified the three SARAH domains from Drosophila melanogaster and performed an unbiased pulldown assay to identify all possible interactions, revealing that isolated SARAH domains are sufficient to recapitulate the cellular assemblies and that Hippo is a universal binding partner. Additionally, we found that the Salvador SARAH domain homodimerizes and demonstrate that this interaction is conserved in Salvador's mammalian homolog. Using native MS, we show that each of these complexes is dimeric in solution. We also measured the stability of each SARAH domain complex, finding that despite similarities at both the sequence and structural levels, SARAH domain complexes differ in stability. The identity, stoichiometry, and stability of these interactions characterized here comprehensively reveal the nature of SARAH domain-mediated complex formation and provide mechanistic insights into how SARAH domain-mediated interactions influence Hippo pathway activity.
Collapse
Affiliation(s)
- Leah Cairns
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 20215
| | - Angela Patterson
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, 59717
| | - Kyler A Weingartner
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 20215
| | - T J Koehler
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 20215
| | - Daniel R DeAngelis
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 20215
| | - Katherine W Tripp
- The T. C. Jenkins Department of Biophysics, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, Maryland, 201218
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, 59717
| | - Jennifer M Kavran
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 20215; Department of Biophysics and Biophysical Chemistry, School of Medicine, Johns Hopkins University, Baltimore, Maryland 20215; Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 20215.
| |
Collapse
|
30
|
Duhart JC, Raftery LA. Mob Family Proteins: Regulatory Partners in Hippo and Hippo-Like Intracellular Signaling Pathways. Front Cell Dev Biol 2020; 8:161. [PMID: 32266255 DOI: 10.3389/fcell.2020.00161/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/28/2020] [Indexed: 05/26/2023] Open
Abstract
Studies in yeast first delineated the function of Mob proteins in kinase pathways that regulate cell division and shape; in multicellular eukaryotes Mobs regulate tissue growth and morphogenesis. In animals, Mobs are adaptors in Hippo signaling, an intracellular signal-transduction pathway that restricts growth, impacting the development and homeostasis of animal organs. Central to Hippo signaling are the Nuclear Dbf2-Related (NDR) kinases, Warts and LATS1 and LATS2, in flies and mammals, respectively. A second Hippo-like signaling pathway has been uncovered in animals, which regulates cell and tissue morphogenesis. Central to this emergent pathway are the NDR kinases, Tricornered, STK38, and STK38L. In Hippo signaling, NDR kinase activation is controlled by three activating interactions with a conserved set of proteins. This review focuses on one co-activator family, the highly conserved, non-catalytic Mps1-binder-related (Mob) proteins. In this context, Mobs are allosteric activators of NDR kinases and adaptors that contribute to assembly of multiprotein NDR kinase activation complexes. In multicellular eukaryotes, the Mob family has expanded relative to model unicellular yeasts; accumulating evidence points to Mob functional diversification. A striking example comes from the most sequence-divergent class of Mobs, which are components of the highly conserved Striatin Interacting Phosphatase and Kinase (STRIPAK) complex, that antagonizes Hippo signaling. Mobs stand out for their potential to modulate the output from Hippo and Hippo-like kinases, through their roles both in activating NDR kinases and in antagonizing upstream Hippo or Hippo-like kinase activity. These opposing Mob functions suggest that they coordinate the relative activities of the Tricornered/STK38/STK38L and Warts/LATS kinases, and thus have potential to assemble nodes for pathway signaling output. We survey the different facets of Mob-dependent regulation of Hippo and Hippo-like signaling and highlight open questions that hinge on unresolved aspects of Mob functions.
Collapse
Affiliation(s)
- Juan Carlos Duhart
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Laurel A Raftery
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| |
Collapse
|
31
|
Duhart JC, Raftery LA. Mob Family Proteins: Regulatory Partners in Hippo and Hippo-Like Intracellular Signaling Pathways. Front Cell Dev Biol 2020; 8:161. [PMID: 32266255 PMCID: PMC7096357 DOI: 10.3389/fcell.2020.00161] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/28/2020] [Indexed: 12/16/2022] Open
Abstract
Studies in yeast first delineated the function of Mob proteins in kinase pathways that regulate cell division and shape; in multicellular eukaryotes Mobs regulate tissue growth and morphogenesis. In animals, Mobs are adaptors in Hippo signaling, an intracellular signal-transduction pathway that restricts growth, impacting the development and homeostasis of animal organs. Central to Hippo signaling are the Nuclear Dbf2-Related (NDR) kinases, Warts and LATS1 and LATS2, in flies and mammals, respectively. A second Hippo-like signaling pathway has been uncovered in animals, which regulates cell and tissue morphogenesis. Central to this emergent pathway are the NDR kinases, Tricornered, STK38, and STK38L. In Hippo signaling, NDR kinase activation is controlled by three activating interactions with a conserved set of proteins. This review focuses on one co-activator family, the highly conserved, non-catalytic Mps1-binder-related (Mob) proteins. In this context, Mobs are allosteric activators of NDR kinases and adaptors that contribute to assembly of multiprotein NDR kinase activation complexes. In multicellular eukaryotes, the Mob family has expanded relative to model unicellular yeasts; accumulating evidence points to Mob functional diversification. A striking example comes from the most sequence-divergent class of Mobs, which are components of the highly conserved Striatin Interacting Phosphatase and Kinase (STRIPAK) complex, that antagonizes Hippo signaling. Mobs stand out for their potential to modulate the output from Hippo and Hippo-like kinases, through their roles both in activating NDR kinases and in antagonizing upstream Hippo or Hippo-like kinase activity. These opposing Mob functions suggest that they coordinate the relative activities of the Tricornered/STK38/STK38L and Warts/LATS kinases, and thus have potential to assemble nodes for pathway signaling output. We survey the different facets of Mob-dependent regulation of Hippo and Hippo-like signaling and highlight open questions that hinge on unresolved aspects of Mob functions.
Collapse
Affiliation(s)
| | - Laurel A. Raftery
- School of Life Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| |
Collapse
|
32
|
Chen Y, Han H, Seo G, Vargas RE, Yang B, Chuc K, Zhao H, Wang W. Systematic analysis of the Hippo pathway organization and oncogenic alteration in evolution. Sci Rep 2020; 10:3173. [PMID: 32081887 PMCID: PMC7035326 DOI: 10.1038/s41598-020-60120-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 02/06/2020] [Indexed: 02/08/2023] Open
Abstract
The Hippo pathway is a central regulator of organ size and a key tumor suppressor via coordinating cell proliferation and death. Initially discovered in Drosophila, the Hippo pathway has been implicated as an evolutionarily conserved pathway in mammals; however, how this pathway was evolved to be functional from its origin is still largely unknown. In this study, we traced the Hippo pathway in premetazoan species, characterized the intrinsic functions of its ancestor components, and unveiled the evolutionary history of this key signaling pathway from its unicellular origin. In addition, we elucidated the paralogous gene history for the mammalian Hippo pathway components and characterized their cancer-derived somatic mutations from an evolutionary perspective. Taken together, our findings not only traced the conserved function of the Hippo pathway to its unicellular ancestor components, but also provided novel evolutionary insights into the Hippo pathway organization and oncogenic alteration.
Collapse
Affiliation(s)
- Yuxuan Chen
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA.,Department of Ecology, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Han Han
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Gayoung Seo
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Rebecca Elizabeth Vargas
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Bing Yang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Kimberly Chuc
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA
| | - Huabin Zhao
- Department of Ecology, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
33
|
Shrestha BK, Skytte Rasmussen M, Abudu YP, Bruun JA, Larsen KB, Alemu EA, Sjøttem E, Lamark T, Johansen T. NIMA-related kinase 9–mediated phosphorylation of the microtubule-associated LC3B protein at Thr-50 suppresses selective autophagy of p62/sequestosome 1. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49883-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
34
|
Shrestha BK, Skytte Rasmussen M, Abudu YP, Bruun JA, Larsen KB, Alemu EA, Sjøttem E, Lamark T, Johansen T. NIMA-related kinase 9–mediated phosphorylation of the microtubule-associated LC3B protein at Thr-50 suppresses selective autophagy of p62/sequestosome 1. J Biol Chem 2020; 295:1240-1260. [DOI: https:/doi.org/10.1016/s0021-9258(17)49883-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
|
35
|
Shrestha BK, Skytte Rasmussen M, Abudu YP, Bruun JA, Larsen KB, Alemu EA, Sjøttem E, Lamark T, Johansen T. NIMA-related kinase 9-mediated phosphorylation of the microtubule-associated LC3B protein at Thr-50 suppresses selective autophagy of p62/sequestosome 1. J Biol Chem 2019; 295:1240-1260. [PMID: 31857374 DOI: 10.1074/jbc.ra119.010068] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/11/2019] [Indexed: 12/21/2022] Open
Abstract
Human ATG8 family proteins (ATG8s) are active in all steps of the macroautophagy pathway, and their lipidation is essential for autophagosome formation. Lipidated ATG8s anchored to the outer surface of the phagophore serve as scaffolds for binding of other core autophagy proteins and various effector proteins involved in trafficking or fusion events, whereas those at the inner surface are needed for assembly of selective autophagy substrates. Their scaffolding role depends on specific interactions between the LC3-interacting region (LIR) docking site (LDS) in ATG8s and LIR motifs in various interaction partners. LC3B is phosphorylated at Thr-50 within the LDS by serine/threonine kinase (STK) 3 and STK4. Here, we identified LIR motifs in STK3 and atypical protein kinase Cζ (PKCζ) and never in mitosis A (NIMA)-related kinase 9 (NEK9). All three kinases phosphorylated LC3B Thr-50 in vitro A phospho-mimicking substitution of Thr-50 impaired binding of several LIR-containing proteins, such as ATG4B, FYVE, and coiled-coil domain-containing 1 (FYCO1), and autophagy cargo receptors p62/sequestosome 1 (SQSTM1) and neighbor of BRCA1 gene (NBR1). NEK9 knockdown or knockout enhanced degradation of the autophagy receptor and substrate p62. Of note, the suppression of p62 degradation was mediated by NEK9-mediated phosphorylation of LC3B Thr-50. Consistently, reconstitution of LC3B-KO cells with the phospho-mimicking T50E variant inhibited autophagic p62 degradation. PKCζ knockdown did not affect autophagic p62 degradation, whereas STK3/4 knockouts inhibited autophagic p62 degradation independently of LC3B Thr-50 phosphorylation. Our findings suggest that NEK9 suppresses LC3B-mediated autophagy of p62 by phosphorylating Thr-50 within the LDS of LC3B.
Collapse
Affiliation(s)
- Birendra Kumar Shrestha
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Mads Skytte Rasmussen
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Yakubu Princely Abudu
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Jack-Ansgar Bruun
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Kenneth Bowitz Larsen
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Endalkachew A Alemu
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Eva Sjøttem
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Trond Lamark
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
| |
Collapse
|
36
|
Flinn MA, Link BA, O'Meara CC. Upstream regulation of the Hippo-Yap pathway in cardiomyocyte regeneration. Semin Cell Dev Biol 2019; 100:11-19. [PMID: 31606277 DOI: 10.1016/j.semcdb.2019.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/27/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022]
Abstract
The response of the adult mammalian heart to injury such as myocardial infarction has long been described as primarily fibrotic scarring and adverse remodeling with little to no regeneration of cardiomyocytes. Emerging studies have challenged this paradigm by demonstrating that, indeed, adult mammalian cardiomyocytes are capable of completing cytokinesis albeit at levels vastly insufficient to compensate for the loss of functional cardiomyocytes following ischemic injury. Thus, there is great interest in identifying mechanisms to guide adult cardiomyocyte cell cycle re-entry and facilitate endogenous heart regeneration. The Hippo signaling pathway is a core kinase cascade that functions to suppress the transcriptional co-activators Yap and Taz by phosphorylation and therefore cytoplasmic retention or phospho-degradation. This pathway has recently sparked interest in the field of cardiac regeneration as inhibition of Hippo kinase signaling or overdriving the transcriptional co-activator, Yap, significantly promotes proliferation of terminally differentiated adult mammalian cardiomyocytes and can restore function in failing mouse hearts. Thus, the Hippo pathway is an attractive therapeutic target for promoting cardiomyocyte renewal and cardiac regeneration. Although the core kinases and transcriptional activators of the Hippo pathway have been studied extensively over the last twenty years, the regulatory inputs of this pathway, particularly in vertebrates, are poorly understood. Recent studies have elucidated several upstream regulatory inputs to the Hippo pathway in adult mammalian cardiomyocytes that influence cell proliferation and heart regeneration. Considering upstream inputs to the Hippo pathway are thought to be context and cell type specific, targeting these various components could serve as a therapeutic approach for refining Hippo-Yap signaling in the heart. Here, we provide an overview of the emerging regulatory inputs to the Hippo pathway as they relate to mammalian cardiomyocytes and heart regeneration.
Collapse
Affiliation(s)
- Michael A Flinn
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian A Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Caitlin C O'Meara
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
37
|
Zheng Y, Pan D. The Hippo Signaling Pathway in Development and Disease. Dev Cell 2019; 50:264-282. [PMID: 31386861 PMCID: PMC6748048 DOI: 10.1016/j.devcel.2019.06.003] [Citation(s) in RCA: 598] [Impact Index Per Article: 99.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 05/23/2019] [Accepted: 06/09/2019] [Indexed: 12/13/2022]
Abstract
The Hippo signaling pathway regulates diverse physiological processes, and its dysfunction has been implicated in an increasing number of human diseases, including cancer. Here, we provide an updated review of the Hippo pathway; discuss its roles in development, homeostasis, regeneration, and diseases; and highlight outstanding questions for future investigation and opportunities for Hippo-targeted therapies.
Collapse
Affiliation(s)
- Yonggang Zheng
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9040, USA.
| |
Collapse
|
38
|
Szimler T, Gráczer É, Györffy D, Végh B, Szilágyi A, Hajdú I, Závodszky P, Vas M. New type of interaction between the SARAH domain of the tumour suppressor RASSF1A and its mitotic kinase Aurora A. Sci Rep 2019; 9:5550. [PMID: 30944388 PMCID: PMC6447619 DOI: 10.1038/s41598-019-41972-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/08/2019] [Indexed: 12/11/2022] Open
Abstract
The tumour suppressor protein RASSF1A is phosphorylated by Aurora A kinase, thereby impairing its tumour suppressor function. Consequently, inhibiting the interaction between Aurora A and RASSF1A may be used for anti-tumour therapy. We used recombinant variants of RASSF1A to map the sites of interaction with Aurora A. The phosphorylation kinetics of three truncated RASSF1A variants has been analysed. Compared to the RASSF1A form lacking the 120 residue long N-terminal part, the Km value of the phosphorylation is increased from 10 to 45 μM upon additional deletion of the C-terminal SARAH domain. On the other hand, deletion of the flexible loop (Δ177-197) that precedes the phosphorylation site/s (T202/S203) results in a reduction of the kcat value from about 40 to 7 min-1. Direct physical interaction between the isolated SARAH domain and Aurora A was revealed by SPR. These data demonstrate that the SARAH domain of RASSF1A is involved in the binding to Aurora A kinase. Structural modelling confirms that a novel complex is feasible between the SARAH domain and the kinase domain of Aurora A. In addition, a regulatory role of the loop in the catalytic phosphorylation reaction has been demonstrated both experimentally and by structural modelling.
Collapse
Affiliation(s)
- T Szimler
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., H-1117, Budapest, Hungary
| | - É Gráczer
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., H-1117, Budapest, Hungary
| | - D Györffy
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., H-1117, Budapest, Hungary
| | - B Végh
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., H-1117, Budapest, Hungary
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, Eötvös Loránd University, Pázmány Péter sétány 1/C, H-1117, Budapest, Hungary
| | - A Szilágyi
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., H-1117, Budapest, Hungary
| | - I Hajdú
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., H-1117, Budapest, Hungary.
| | - P Závodszky
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., H-1117, Budapest, Hungary
| | - M Vas
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok krt. 2., H-1117, Budapest, Hungary.
| |
Collapse
|
39
|
Abstract
The Hippo pathway controls organ size and maintains tissue homeostasis through a central MST-LATS kinase cascade. When Hippo signaling is on, activated MST1/2 partner with SAV1 to phosphorylate and activate the LATS1/2-MOB1 complexes, which in turn phosphorylate and inactivate YAP/TAZ transcription co-activators. This process halts the expression of Hippo-responsive genes, thereby inhibiting cell proliferation and promoting apoptosis. Our studies have shown that two core adaptor proteins MOB1 and SAV1 use distinctive mechanisms to enhance Hippo signaling. MOB1 promotes MST-dependent LATS activation through dynamic scaffolding and allosteric regulation. SAV1 promotes MST activation by antagonizing the PP2A phosphatase activity. Here we describe the detailed methods for the purification and crystallization of the MST2-SAV1 and pMOB1-LATS1 complexes, for assaying the SAV1-dependent inhibition of PP2A, and for analyzing LATS1 kinase activation using in vitro reconstitution.
Collapse
Affiliation(s)
- Lisheng Ni
- Departments of Pharmacology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuelian Luo
- Departments of Pharmacology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
40
|
A Network Pharmacology-Based Approach to Investigate the Novel TCM Formula against Huntington's Disease and Validated by Support Vector Machine Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:6020197. [PMID: 30643534 PMCID: PMC6311282 DOI: 10.1155/2018/6020197] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/05/2018] [Indexed: 12/16/2022]
Abstract
Several pathways are crucial in Huntington's disease (HD). Based on the concept of multitargets, network pharmacology-based analysis was employed to find out related proteins in disease network. The network target method aims to find out related mechanism of efficacy substances in rational design way. Traditional Chinese medicine prescriptions would be used for research and development against HD. Virtual screening was performed to obtain drug molecules with high binding capacity from traditional Chinese medicine (TCM) database@Taiwan. Quantitative structure-activity relationship (QSAR) models were conducted by MLR, SVM, CoMFA, and CoMSIA, constructed to predict the bioactivities of candidates. The compounds with high-dock score were further analyzed compared with control. Traditional Chinese medicine reported in the literature could be the training set provided for constructing novel formula by SVM model. We tried to find a novel formula that can bind well with these targets at the same time, which indicates our design could be highly related to the HD. Additionally, the candidates would validate by a long-term molecular dynamics (MD) simulation, 5 microseconds. Thus, we suggested the herbs Brucea javanica, Holarrhena antidysenterica, Dichroa febrifuga, Erythrophleum guineense, etc. which contained active compounds might be a novel medicine formula toward Huntington's disease.
Collapse
|
41
|
Matsui Y, Zhang Y, Paulson RF, Lai ZC. Dual Role of a C-Terminally Truncated Isoform of Large Tumor Suppressor Kinase 1 in the Regulation of Hippo Signaling and Tissue Growth. DNA Cell Biol 2018; 38:91-106. [PMID: 30461308 DOI: 10.1089/dna.2018.4340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The considerable amount of experimental evidence has defined the Hippo pathway as a tumor suppressive pathway and increased expression and/or activity of its oncogenic effectors is frequently observed in cancer. However, clinical studies have failed to attribute cancer development and progression to mutations in the pathway. In explaining this conundrum, we investigated the expression and functions of a C-terminally truncated isoform of large tumor suppressor kinase 1 (LATS1) called short LATS1 (sLATS1) in human cell lines and Drosophila. Intriguingly, through overexpression of sLATS1, we demonstrated that sLATS1 either activates or suppresses the activity of Yes-associated protein (YAP), one of the effectors of the Hippo pathway, in a cell type-specific manner. The activation is mediated through inhibition of full-length LATS1, whereas suppression of YAP is accomplished through sLATS1-YAP interaction. In HEK293T cells, the former mechanism may affect the cellular response more dominantly, whereas in U2OS cells and developing tissues in Drosophila, the latter mechanism may be solely carried out. Finally, to find the clinical relevance of this molecule, we examined the expression of sLATS1 in breast cancer patients. The transcriptome analysis showed that the ratio of sLATS1 to LATS1 was increased in tumor tissues comparing to their adjacent normal tissues.
Collapse
Affiliation(s)
- Yurika Matsui
- 1 Intercollege Graduate Degree Program in Molecular, Cellular, and Integrative Biosciences, The Pennsylvania State University, University Park , Pennsylvania
| | - Yifan Zhang
- 2 Department of Biology, The Pennsylvania State University , University Park, Pennsylvania
| | - Robert F Paulson
- 3 Department of Veterinary and Biomedical Sciences, and The Pennsylvania State University , University Park, Pennsylvania
| | - Zhi-Chun Lai
- 1 Intercollege Graduate Degree Program in Molecular, Cellular, and Integrative Biosciences, The Pennsylvania State University, University Park , Pennsylvania.,2 Department of Biology, The Pennsylvania State University , University Park, Pennsylvania.,4 Department of Biochemistry and Molecular Biology, The Pennsylvania State University , University Park, Pennsylvania
| |
Collapse
|
42
|
Creixell P, Pandey JP, Palmeri A, Bhattacharyya M, Creixell M, Ranganathan R, Pincus D, Yaffe MB. Hierarchical Organization Endows the Kinase Domain with Regulatory Plasticity. Cell Syst 2018; 7:371-383.e4. [PMID: 30243563 DOI: 10.1016/j.cels.2018.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/24/2018] [Accepted: 08/13/2018] [Indexed: 02/08/2023]
Abstract
The functional diversity of kinases enables specificity in cellular signal transduction. Yet how more than 500 members of the human kinome specifically receive regulatory inputs and convey information to appropriate substrates-all while using the common signaling output of phosphorylation-remains enigmatic. Here, we perform statistical co-evolution analysis, mutational scanning, and quantitative live-cell assays to reveal a hierarchical organization of the kinase domain that facilitates the orthogonal evolution of regulatory inputs and substrate outputs while maintaining catalytic function. We find that three quasi-independent "sectors"-groups of evolutionarily coupled residues-represent functional units in the kinase domain that encode for catalytic activity, substrate specificity, and regulation. Sector positions impact both disease and pharmacology: the catalytic sector is significantly enriched for somatic cancer mutations, and residues in the regulatory sector interact with allosteric kinase inhibitors. We propose that this functional architecture endows the kinase domain with inherent regulatory plasticity.
Collapse
Affiliation(s)
- Pau Creixell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Departments of Biology and Bioengineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jai P Pandey
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | | - Moitrayee Bhattacharyya
- Department of Molecular and Cell Biology and California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marc Creixell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Departments of Biology and Bioengineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rama Ranganathan
- Center for Physics of Evolving Systems, Department of Biochemistry and Molecular Biology, Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - David Pincus
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Michael B Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Departments of Biology and Bioengineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Divisions of Acute Care Surgery, Trauma, and Critical Care and Surgical Oncology, Harvard Medical School, Boston 02215, USA.
| |
Collapse
|
43
|
Activation mechanisms of the Hippo kinase signaling cascade. Biosci Rep 2018; 38:BSR20171469. [PMID: 30038061 PMCID: PMC6131212 DOI: 10.1042/bsr20171469] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 11/21/2022] Open
Abstract
First discovered two decades ago through genetic screens in Drosophila, the Hippo pathway has been shown to be conserved in metazoans and controls organ size and tissue homeostasis through regulating the balance between cell proliferation and apoptosis. Dysregulation of the Hippo pathway leads to aberrant tissue growth and tumorigenesis. Extensive studies in Drosophila and mammals have identified the core components of Hippo signaling, which form a central kinase cascade to ultimately control gene expression. Here, we review recent structural, biochemical, and cellular studies that have revealed intricate phosphorylation-dependent mechanisms in regulating the formation and activation of the core kinase complex in the Hippo pathway. These studies have established the dimerization-mediated activation of the Hippo kinase (mammalian Ste20-like 1 and 2 (MST1/2) in mammals), the dynamic scaffolding and allosteric roles of adaptor proteins in downstream kinase activation, and the importance of multisite linker autophosphorylation by Hippo and MST1/2 in fine-tuning the signaling strength and robustness of the Hippo pathway. We highlight the gaps in our knowledge in this field that will require further mechanistic studies.
Collapse
|
44
|
Pefani DE, Tognoli ML, Pirincci Ercan D, Gorgoulis V, O'Neill E. MST2 kinase suppresses rDNA transcription in response to DNA damage by phosphorylating nucleolar histone H2B. EMBO J 2018; 37:e98760. [PMID: 29789391 PMCID: PMC6068430 DOI: 10.15252/embj.201798760] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/18/2018] [Accepted: 04/27/2018] [Indexed: 12/15/2022] Open
Abstract
The heavily transcribed rDNA repeats that give rise to the ribosomal RNA are clustered in a unique chromatin structure, the nucleolus. Due to its highly repetitive nature and transcriptional activity, the nucleolus is considered a hotspot of genomic instability. Breaks in rDNA induce a transient transcriptional shut down to conserve energy and promote rDNA repair; however, how nucleolar chromatin is modified and impacts on rDNA repair is unknown. Here, we uncover that phosphorylation of serine 14 on histone H2B marks transcriptionally inactive nucleolar chromatin in response to DNA damage. We identified that the MST2 kinase localises at the nucleoli and targets phosphorylation of H2BS14p in an ATM-dependent manner. We show that establishment of H2BS14p is necessary for damage-induced rDNA transcriptional shut down and maintenance of genomic integrity. Ablation of MST2 kinase, or upstream activators, results in defective establishment of nucleolar H2BS14p, perturbed DNA damage repair, sensitisation to rDNA damage and increased cell lethality. We highlight the impact of chromatin regulation in the rDNA damage response and targeting of the nucleolus as an emerging cancer therapeutic approach.
Collapse
Affiliation(s)
- Dafni Eleftheria Pefani
- CRUK/MRC Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Maria Laura Tognoli
- CRUK/MRC Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Deniz Pirincci Ercan
- CRUK/MRC Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
- Radboud University, Nijmegen, The Netherlands
| | - Vassilis Gorgoulis
- Laboratory of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Faculty of Biology, Medicine and Health, Manchester Academic Health Centre, University of Manchester, Manchester, UK
| | - Eric O'Neill
- CRUK/MRC Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
- Systems Biology Ireland, University College Dublin, Dublin 4, Ireland
| |
Collapse
|
45
|
Homeostatic Control of Hpo/MST Kinase Activity through Autophosphorylation-Dependent Recruitment of the STRIPAK PP2A Phosphatase Complex. Cell Rep 2018; 21:3612-3623. [PMID: 29262338 DOI: 10.1016/j.celrep.2017.11.076] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/25/2017] [Accepted: 11/21/2017] [Indexed: 12/22/2022] Open
Abstract
The Hippo pathway controls organ size and tissue homeostasis through a kinase cascade leading from the Ste20-like kinase Hpo (MST1/2 in mammals) to the transcriptional coactivator Yki (YAP/TAZ in mammals). Whereas previous studies have uncovered positive and negative regulators of Hpo/MST, how they are integrated to maintain signaling homeostasis remains poorly understood. Here, we identify a self-restricting mechanism whereby autophosphorylation of an unstructured linker in Hpo/MST creates docking sites for the STRIPAK PP2A phosphatase complex to inactivate Hpo/MST. Mutation of the phospho-dependent docking sites in Hpo/MST or deletion of Slmap, the STRIPAK subunit recognizing these docking sites, results in constitutive activation of Hpo/MST in both Drosophila and mammalian cells. In contrast, autophosphorylation of the Hpo/MST linker at distinct sites is known to recruit Mats/MOB1 to facilitate Hippo signaling. Thus, multisite autophosphorylation of Hpo/MST linker provides an evolutionarily conserved built-in molecular platform to maintain signaling homeostasis by coupling antagonistic signaling activities.
Collapse
|
46
|
Abstract
Ras oncoproteins can promote or suppress cellular apoptosis, but the mechanisms underlying these varied responses remain incompletely understood. Ras is linked to the Hippo tumor suppressor pathway, a highly conserved signaling cassette that regulates organ size in animals ranging from flies to humans. The proximal members of this pathway, Mammalian Ste20-like kinases (Msts) -1 and -2, self-associate in homodimers and also form heterodimers with other proteins. Formation of such complexes is known to regulate Mst kinase activity and thus, the Hippo pathway. In a manuscript that recently appeared in Current Biology, we showed that activated Hras promotes the formation of Mst1/Mst2 heterodimers, that activation of Erk was required for this event, and that these heterodimers were much less active than Mst1/Mst1 or Mst2/Mst2 homodimers. Interestingly, the formation of such heterodimers was required to deactivate the Hippo pathway and to enable transformation by Hras. In this Commentary, we discuss the background for this study and surprising implications thereof.
Collapse
|
47
|
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: 4.0] [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
|
48
|
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: 2.4] [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
|
49
|
Cairns L, Tran T, Fowl BH, Patterson A, Kim YJ, Bothner B, Kavran JM. Salvador has an extended SARAH domain that mediates binding to Hippo kinase. J Biol Chem 2018. [PMID: 29519817 DOI: 10.1074/jbc.ra117.000923] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Hippo pathway controls cell proliferation and differentiation through the precisely tuned activity of a core kinase cassette. The activity of Hippo kinase is modulated by interactions between its C-terminal coiled-coil, termed the SARAH domain, and the SARAH domains of either dRassF or Salvador. Here, we wanted to understand the molecular basis of SARAH domain-mediated interactions and their influence on Hippo kinase activity. We focused on Salvador, a positive effector of Hippo activity and the least well-characterized SARAH domain-containing protein. We determined the crystal structure of a complex between Salvador and Hippo SARAH domains from Drosophila This structure provided insight into the organization of the Salvador SARAH domain including a folded N-terminal extension that expands the binding interface with Hippo SARAH domain. We also found that this extension improves the solubility of the Salvador SARAH domain, enhances binding to Hippo, and is unique to Salvador. We therefore suggest expanding the definition of the Salvador SARAH domain to include this extended region. The heterodimeric assembly observed in the crystal was confirmed by cross-linked MS and provided a structural basis for the mutually exclusive interactions of Hippo with either dRassF or Salvador. Of note, Salvador influenced the kinase activity of Mst2, the mammalian Hippo homolog. In co-transfected HEK293T cells, human Salvador increased the levels of Mst2 autophosphorylation and Mst2-mediated phosphorylation of select substrates, whereas Salvador SARAH domain inhibited Mst2 autophosphorylation in vitro These results suggest Salvador enhances the effects of Hippo kinase activity at multiple points in the Hippo pathway.
Collapse
Affiliation(s)
- Leah Cairns
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Thao Tran
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Brendan H Fowl
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Angela Patterson
- the Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, and
| | - Yoo Jin Kim
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215
| | - Brian Bothner
- the Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, and
| | - Jennifer M Kavran
- From the Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 20215, .,the Departments of Biophysics and Biophysical Chemistry, School of Medicine, and.,Oncology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 20215
| |
Collapse
|
50
|
Wong KF, Liu AM, Hong W, Xu Z, Luk JM. Integrin α2β1 inhibits MST1 kinase phosphorylation and activates Yes-associated protein oncogenic signaling in hepatocellular carcinoma. Oncotarget 2018; 7:77683-77695. [PMID: 27765911 PMCID: PMC5363613 DOI: 10.18632/oncotarget.12760] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/25/2016] [Indexed: 12/22/2022] Open
Abstract
The Hippo pathway regulates the down-stream target Yes-associated protein (YAP) to maintain organ homeostasis, which is commonly inactivated in many types of cancers. However, how cell adhesion dysregulates the Hippo pathway activating YAP oncogene in hepatocellular carcinoma (HCC) remains unclear. Our findings demonstrate that α2β1 integrin (but not other β1 integrins) expressed in HCC cells, after binding to collagen extracellular matrix, could inhibit MST1 kinase phosphorylation and activate YAP pro-oncogenic activities. Knockdown of integrin α2 gene (ITGA2) suppressed YAP targeted gene expression in vitro. α2β1 and collagen binding resulted in suppressing Hippo signaling of mammalian sterile 20-like kinase 1 (MST1) and Large tumor suppressor homolog 1 (LATS1) with concomitant activation of YAP-mediated connective tissue growth factor (CTGF) gene expression. In vitro kinase assay showed that MST1 is an immediate downstream target of integrin α2 with S1180 residue as the critical phosphorylation site. Clinical correlational analysis using a gene expression dataset of 228 HCC tumors revealed that ITGA2 expression was significantly associated with tumor progression, and co-expression with YAP targeted genes (AXL receptor tyrosine kinase, CTGF, cyclin D1, glypican 3, insulin like growth factor 1 receptor, and SRY-box 4) correlated with survivals of HCC patients. In conclusion, α2β1 integrin activation through cellular adhesion impacts the Hippo pathway in solid tumors and modulates MST1-YAP signaling cascade. Targeting integrin α2 holds promises for treating YAP-positive HCC.
Collapse
Affiliation(s)
- Kwong-Fai Wong
- Department of Pharmacology, National University Health System, Singapore.,Department of Surgery, National University Health System, Singapore
| | - Angela M Liu
- Department of Pharmacology, National University Health System, Singapore.,Department of Surgery, National University Health System, Singapore
| | - Wanjin Hong
- Institute of Molecular and Cell Biology, Biopolis, Singapore
| | - Zhi Xu
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - John M Luk
- Department of Pharmacology, National University Health System, Singapore.,Department of Surgery, National University Health System, Singapore.,Department of Pathology, University of Hong Kong, Queen Mary Hospital, Pokfulam, Hong Kong.,Department of Translational and Clinical Medicine, Arbele Limited, Hong Kong Science Park, Shatin, Hong Kong
| |
Collapse
|