1
|
Chan EC, Ablooglu AJ, Ghosh CC, Desai A, Schaible N, Chen X, Zhao M, Olano MR, Ganesan S, Lack JB, Krishnan R, Parikh SM, Druey KM. PARP15 Is a Susceptibility Locus for Clarkson Disease (Monoclonal Gammopathy-Associated Systemic Capillary Leak Syndrome). Arterioscler Thromb Vasc Biol 2024; 44:2628-2646. [PMID: 39479769 PMCID: PMC11602389 DOI: 10.1161/atvbaha.124.321522] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/15/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Vascular leakage is a deadly complication of severe infections, ranging from bacterial sepsis to malaria. Worldwide, septicemia is among the top 10 causes of lethality because of the shock and multiorgan dysfunction that arise from the host vascular response. In the monoclonal gammopathy-associated capillary leak syndrome (MG-CLS), even otherwise mundane infections induce recurrent septic-like episodes of profound microvascular hyperpermeability and shock. There are no defined genetic risk factors for MG-CLS or effective treatments for acute crises. METHODS We characterized predicted loss-of-function mutations in PARP15 (poly[ADP-ribose] polymerase 15), a protein of unknown function that is absent in mice, in patients with MG-CLS. We analyzed barrier function in PARP15-deficient vascular endothelial cells and vascular leakage in mice engineered to express wild-type or loss-of-function variant human PARP15. RESULTS We discovered several loss-of-function PARP15 variants associated with MG-CLS. These mutations severely reduced PARP15 enzymatic function. The presence of the most frequently detected variant (G628R) correlated with clinical markers of severe vascular leakage. In human microvascular endothelial cells, PARP15 suppressed cytokine-induced barrier disruption by ADP-ribosylating the scaffold protein JIP3 (c-Jun N-terminal kinase-interacting protein 3) and inhibiting p38 MAP (mitogen-activated protein) kinase activation. Mice expressing enzymatically inactive human PARP15(G628R) were significantly more prone to inflammation-associated vascular leakage than mice expressing wild-type PARP15 in a p38-dependent fashion. CONCLUSIONS PARP15 represents a previously unrecognized genetic susceptibility factor for MG-CLS. PARP15-mediated ADP ribosylation is an essential and genetically determined mechanism of the human vascular response to inflammation.
Collapse
Affiliation(s)
- Eunice C. Chan
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Ararat J. Ablooglu
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Chandra C. Ghosh
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center; Boston, MA 02215, USA
| | - Abhishek Desai
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Niccole Schaible
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center; Boston, MA 02215, USA
| | - Xiuying Chen
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center; Boston, MA 02215, USA
| | - Ming Zhao
- Protein Chemistry Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - M. Renee Olano
- Protein Chemistry Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Sundar Ganesan
- Biological Imaging Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Justin B. Lack
- NIAID Collaborative Bioinformatics Resource, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Department of Emergency Medicine, Beth Israel Deaconess Medical Center; Boston, MA 02215, USA
| | - Samir M. Parikh
- Division of Nephrology, Departments of Internal Medicine and Pharmacology, University of Texas Southwestern Medical Center; Dallas, TX, 75225, USA
| | - Kirk M. Druey
- Lung and Vascular Inflammation Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health; Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Pittala S, Haspula D, Cui Y, Yang WM, Kim YB, Davis RJ, Wing A, Rotman Y, McGuinness OP, Inoue A, Wess J. G 12/13-mediated signaling stimulates hepatic glucose production and has a major impact on whole body glucose homeostasis. Nat Commun 2024; 15:9996. [PMID: 39557854 PMCID: PMC11574106 DOI: 10.1038/s41467-024-54299-7] [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: 01/10/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Altered hepatic glucose fluxes are critical during the pathogenesis of type 2 diabetes. G protein-coupled receptors represent important regulators of hepatic glucose production. Recent studies have shown that hepatocytes express GPCRs that can couple to G12/13, a subfamily of heterotrimeric G proteins that has attracted relatively little attention in the past. Here we show, by analyzing several mutant mouse strains, that selective activation of hepatocyte G12/13 signaling leads to pronounced hyperglycemia and that this effect involves the stimulation of the ROCK1-JNK signaling cascade. Using both mouse and human hepatocytes, we also show that activation of endogenous sphingosine-1-phosphate type 1 receptors strongly promotes glucose release in a G12/13-dependent fashion. Studies with human liver samples indicate that hepatic GNA12 (encoding Gα12) expression levels positively correlate with indices of insulin resistance and impaired glucose homeostasis, consistent with a potential pathophysiological role of enhanced hepatic G12/13 signaling.
Collapse
Affiliation(s)
- Srinivas Pittala
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA.
| | - Dhanush Haspula
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - Yinghong Cui
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA
| | - Won-Mo Yang
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Allison Wing
- Liver & Energy Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Yaron Rotman
- Liver & Energy Metabolism Section, Liver Diseases Branch, NIDDK, NIH, Bethesda, MD, USA
| | - Owen P McGuinness
- Departments of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, NIDDK, NIH, Bethesda, MD, USA.
| |
Collapse
|
3
|
Frederick MI, Hovey OFJ, Kakadia JH, Shepherd TG, Li SSC, Heinemann IU. Proteomic and Phosphoproteomic Reprogramming in Epithelial Ovarian Cancer Metastasis. Mol Cell Proteomics 2023; 22:100660. [PMID: 37820923 PMCID: PMC10652129 DOI: 10.1016/j.mcpro.2023.100660] [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: 06/23/2023] [Revised: 09/30/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is a high-risk cancer presenting with heterogeneous tumors. The high incidence of EOC metastasis from primary tumors to nearby tissues and organs is a major driver of EOC lethality. We used cellular models of spheroid formation and readherence to investigate cellular signaling dynamics in each step toward EOC metastasis. In our system, adherent cells model primary tumors, spheroid formation represents the initiation of metastatic spread, and readherent spheroid cells represent secondary tumors. Proteomic and phosphoproteomic analyses show that spheroid cells are hypoxic and show markers for cell cycle arrest. Aurora kinase B abundance and downstream substrate phosphorylation are significantly reduced in spheroids and readherent cells, explaining their cell cycle arrest phenotype. The proteome of readherent cells is most similar to spheroids, yet greater changes in the phosphoproteome show that spheroid cells stimulate Rho-associated kinase 1 (ROCK1)-mediated signaling, which controls cytoskeletal organization. In spheroids, we found significant phosphorylation of ROCK1 substrates that were reduced in both adherent and readherent cells. Application of the ROCK1-specific inhibitor Y-27632 to spheroids increased the rate of readherence and altered spheroid density. The data suggest ROCK1 inhibition increases EOC metastatic potential. We identified novel pathways controlled by Aurora kinase B and ROCK1 as major drivers of metastatic behavior in EOC cells. Our data show that phosphoproteomic reprogramming precedes proteomic changes that characterize spheroid readherence in EOC metastasis.
Collapse
Affiliation(s)
- Mallory I Frederick
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Owen F J Hovey
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jenica H Kakadia
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Trevor G Shepherd
- Department of Obstetrics & Gynaecology, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Shawn S C Li
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| | - Ilka U Heinemann
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
4
|
Li T, Zeng Z, Fan C, Xiong W. Role of stress granules in tumorigenesis and cancer therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:189006. [PMID: 37913942 DOI: 10.1016/j.bbcan.2023.189006] [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: 06/24/2023] [Revised: 09/24/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
Stress granules (SGs) are membrane-less organelles that cell forms via liquid-liquid phase separation (LLPS) under stress conditions such as oxidative stress, ER stress, heat shock and hypoxia. SG assembly is a stress-responsive mechanism by regulating gene expression and cellular signaling pathways. Cancer cells face various stress conditions in tumor microenvironment during tumorigenesis, while SGs contribute to hallmarks of cancer including proliferation, invasion, migration, avoiding apoptosis, metabolism reprogramming and immune evasion. Here, we review the connection between SGs and cancer development, the limitation of SGs on current cancer therapy and promising cancer therapeutic strategies targeting SGs in the future.
Collapse
Affiliation(s)
- Tiansheng Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Chunmei Fan
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China; Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
5
|
Yang L, Qiao P, Zhang J, Huang S, Hu A. Rho-associated kinase1 promotes laryngeal squamous cell carcinoma tumorigenesis and progression via the FAK signaling pathway. Discov Oncol 2022; 13:100. [PMID: 36197602 PMCID: PMC9535064 DOI: 10.1007/s12672-022-00561-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/08/2022] [Indexed: 11/04/2022] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is one of the most common head and neck squamous cell carcinomas (HNSCC). Rho-associated kinase1 (ROCK1) is considered to promote progression of numerous cancers, however, its role in LSCC is still unknown. Here, the expression level of ROCK1 is higher in LSCC tissues than non-tumor tissues, and the expression level of ROCK1 is positively correlated with advanced stage and poor survival prognosis. ROCK1 knockdown in TU686 and TU212 cells dramatically inhibits cellular proliferation, migration and invasion. Whereas the overexpression of ROCK1 reversed these changes. FAK signaling pathway plays an essential role in promoting LSCC progression. Inhibiting FAK activity with TAE226 observably impairs the tumor-promoting effects. In conclusion, ROCK1 promotes LSCC tumorigenesis and progression via the FAK signaling pathway, targeting the ROCK1 molecule may represent potential targets for clinical LSCC treatment.
Collapse
Affiliation(s)
- Liyun Yang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Peipei Qiao
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Jianwei Zhang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China
| | - Shuixian Huang
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| | - An Hu
- Department of Otolaryngology, Gongli Hospital, The Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
6
|
Zeng Y, Qin T, Flamini V, Tan C, Zhang X, Cong Y, Birkin E, Jiang WG, Yao H, Cui Y. Identification of DHX36 as a tumour suppressor through modulating the activities of the stress-associated proteins and cyclin-dependent kinases in breast cancer. Am J Cancer Res 2020; 10:4211-4233. [PMID: 33414996 PMCID: PMC7783738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/29/2020] [Indexed: 06/12/2023] Open
Abstract
The nucleic acid guanine-quadruplex structures (G4s) are involved in many aspects of cancer progression. The DEAH-box polypeptide 36 (DHX36) has been identified as a dominant nucleic acid helicase which targets and disrupts DNA and RNA G4s in an ATP-dependent manner. However, the actual role of DHX36 in breast cancer remains unknown. In this study, we observed that the gene expression of DHX36 was positively associated with patient survival in breast cancer. The abundance of DHX36 is also linked with pathologic conditions and the stage of breast cancer. By using the xenograft mouse model, we demonstrated that the stable knockdown of DHX36 via lentivirus in breast cancer cells significantly promoted tumour growth. We also found that, after the DHX36 knockdown (KD), the invasion of triple-negative breast cancer cells was enhanced. In addition, we found a significant increase in the number of cells in the S-phase and a reduction of apoptosis with the response to cisplatin. DHX36 KD also desensitized the cytotoxic cellular response to paclitaxel and cisplatin. Transcriptomic profiling analysis by RNA sequencing indicated that DHX36 altered gene expression profile through the upstream activation of TNF, IFNγ, NFκb and TGFβ1. High throughput signalling analysis showed that one cluster of stress-associated kinase proteins including p53, ROCK1 and JNK were suppressed, while the mitotic checkpoint protein-serine kinases CDK1 and CDK2 were activated, as a consequence of the DHX36 knockdown. Our study reveals that DHX36 functions as a tumour suppressor and may be considered as a potential therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Yinduo Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou 510120, China
- Cardiff China Medical Research Collaborative, Cardiff University School of MedicineHeath Park, Cardiff CF14 4XN, UK
- Breast Tumour Center, Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou 510120, China
| | - Tao Qin
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou 510120, China
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Valentina Flamini
- Cardiff China Medical Research Collaborative, Cardiff University School of MedicineHeath Park, Cardiff CF14 4XN, UK
| | - Cui Tan
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou 510120, China
- Department of Pathology, Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Xinke Zhang
- Sun Yat-sen University Cancer Centre, The State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer MedicineGuangzhou 510060, China
| | - Yizi Cong
- Cardiff China Medical Research Collaborative, Cardiff University School of MedicineHeath Park, Cardiff CF14 4XN, UK
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantai, China
| | - Emily Birkin
- Cardiff China Medical Research Collaborative, Cardiff University School of MedicineHeath Park, Cardiff CF14 4XN, UK
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of MedicineHeath Park, Cardiff CF14 4XN, UK
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumour Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou 510120, China
- Breast Tumour Center, Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou 510120, China
| | - Yuxin Cui
- Cardiff China Medical Research Collaborative, Cardiff University School of MedicineHeath Park, Cardiff CF14 4XN, UK
| |
Collapse
|
7
|
Hu J, Zhang H, Li J, Jiang X, Zhang Y, Wu Q, Shen L, Shi J, Gao N. ROCK1 activation-mediated mitochondrial translocation of Drp1 and cofilin are required for arnidiol-induced mitochondrial fission and apoptosis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:37. [PMID: 32075676 PMCID: PMC7031977 DOI: 10.1186/s13046-020-01545-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/14/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Arnidiol is a pentacyclic triterpene diol that has multiple pharmacological activities. However, the apoptotic activities of arnidiol in human cancer cells have not yet been explored, nor has the mechanism by which arnidiol induces apoptosis been examined in depth. METHODS MDA-MB-231 cells and xenografted mice were treated with arnidiol. Mitochondrial fission and apoptosis were determined by immunofluorescence, flow cytometry and related molecular biological techniques. The interaction and colocalization of cofilin and Drp1 was determined by immunoprecipitation and immunofluorescence assays. RESULTS Arnidiol induces mitochondrial fission and apoptosis through mitochondrial translocation of Drp1 and cofilin. Importantly, the interaction of Drp1 and cofilin in mitochondria is involved in arnidiol-induced mitochondrial fission and apoptosis. Knockdown of either Drp1 or cofilin abrogated arnidiol-induced mitochondrial translocation, interaction of Drp1 and cofilin, mitochondrial fission and apoptosis. Only dephosphorylated Drp1 (Ser637) and cofilin (Ser3) were translocated to the mitochondria. Mutants of Drp1 S637A and cofilin S3A, which mimic the dephosphorylated forms, enhanced mitochondrial fission and apoptosis induced by arnidiol, whereas mutants of Drp1 S637D and cofilin S3E, which mimic the phosphorylated forms, suppressed mitochondrial fission and apoptosis induced by arnidiol. A mechanistic study revealed that ROCK1 activation plays an important role in the arnidiol-mediated Drp1 and cofilin dephosphorylation and mitochondrial translocation, mitochondrial fission, and apoptosis. CONCLUSIONS Our data reveal a novel role of both Drp1 and cofilin in the regulation of mitochondrial fission and apoptosis and suggest that arnidiol could be developed as a potential agent for the treatment of human cancer.
Collapse
Affiliation(s)
- Jinjiao Hu
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Hongwei Zhang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jie Li
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiuxing Jiang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yanhao Zhang
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Qin Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Liwen Shen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jingshan Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| | - Ning Gao
- College of Pharmacy, Army Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China. .,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
8
|
Shah P, Zhao B, Qiang L, He YY. Phosphorylation of xeroderma pigmentosum group C regulates ultraviolet-induced DNA damage repair. Nucleic Acids Res 2019; 46:5050-5060. [PMID: 29660033 PMCID: PMC6007576 DOI: 10.1093/nar/gky239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/21/2018] [Indexed: 12/18/2022] Open
Abstract
Nucleotide excision repair (NER) is the most versatile DNA repair system that removes bulky DNA damage induced by various endogenous and exogenous factors, including UV radiation. Defects in NER can lead to the xeroderma pigmentosum (XP) syndrome, mainly characterized by increased carcinogenesis in the skin. The function of NER factors, including xeroderma pigmentosum group C (XPC), can be regulated by post-translational modifications such as ubiquitination. However, the role of phosphorylation in XPC function remains unknown. Here, we show that phosphorylation of XPC acts as a novel post-translational regulatory mechanism of the NER pathway. We show that XPC is phosphorylated at serine 94. Moreover, after UVB irradiation, XPC phosphorylation regulates recruitment of ubiquitinated XPC and its downstream NER factors to the chromatin. In addition, upon evaluating the predicted kinases for XPC phosphorylation, we found that casein kinase II (CK2) promotes NER. Furthermore, CK2 kinase mediates XPC phosphorylation at serine 94, and also promotes recruitment of ubiquitinated XPC to the chromatin after UVB irradiation. Our findings have identified XPC phosphorylation as a new mechanism for regulating NER following UV-induced DNA damage.
Collapse
Affiliation(s)
- Palak Shah
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637, USA.,Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Baozhong Zhao
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637, USA
| | - Lei Qiang
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637, USA.,School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210008, China
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, IL 60637, USA.,Committee on Molecular Pathogenesis and Molecular Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
9
|
Abstract
The study aimed to measure the presence of rho-associated protein kinase 1 (ROCK1) mRNA in serum samples collected from glioma and investigate its diagnostic significance in glioma.The presence of ROCK1 mRNA was examined by quantitative real-time polymerase chain reaction (qRT-PCR). The relationship between ROCK1 mRNA and clinical characteristics was analyzed via Chi-square test. The criteria of diagnosis evaluation, including sensitivity, specificity, optimal cutoff point, and area under the curve (AUC) were determined through the receiver operating characteristic (ROC) curve analysis.ROCK1 mRNA was significantly increased in serum samples collected from glioma patients compared to the controls (P <.05). Besides, high ROCK1 mRNA expression was tightly related with Karnofsky Performance Status (KPS) score (P = .024) and World Health Organization (WHO) grade (P = .029). However, there was no association between ROCK1 expression and gender, neurological disorders, family history and cigarette smoking (all, P >.05). In addition, the optimal cutoff point was 3.025, with the sensitivity and specificity of 88.89% and 79.25%, respectively. The AUC was 0.881, indicating that ROCK1 was a diagnostic biomarker for glioma patients (P <.0001, 95% CI = 0.829-0.933).Serum ROCK1 mRNA is significantly up-regulated in glioma cases compared to healthy controls. ROCK1 may be a potential diagnostic biomarker in glioma.
Collapse
Affiliation(s)
- Yunyang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital
- Department of Neurosurgery, Tianjin First Central Hospital
| | - Jianjun Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital
- Department of Neurosurgery, Tianjin 4th Center Hospital, Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, Tianjin 4th Center Hospital, Tianjin, China
| | - Xinyu Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital
| |
Collapse
|
10
|
Hoque A, Williamson NA, Ameen SS, Ciccotosto GD, Hossain MI, Oakhill JS, Ng DCH, Ang CS, Cheng HC. Quantitative proteomic analyses of dynamic signalling events in cortical neurons undergoing excitotoxic cell death. Cell Death Dis 2019; 10:213. [PMID: 30824683 PMCID: PMC6397184 DOI: 10.1038/s41419-019-1445-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/20/2019] [Accepted: 02/01/2019] [Indexed: 12/23/2022]
Abstract
Excitotoxicity, caused by overstimulation or dysregulation of ionotropic glutamate receptors (iGluRs), is a pathological process directing neuronal death in many neurological disorders. The aberrantly stimulated iGluRs direct massive influx of calcium ions into the affected neurons, leading to changes in expression and phosphorylation of specific proteins to modulate their functions and direct their participation in the signalling pathways that induce excitotoxic neuronal death. To define these pathways, we used quantitative proteomic approaches to identify these neuronal proteins (referred to as the changed proteins) and determine how their expression and/or phosphorylation dynamically changed in association with excitotoxic cell death. Our data, available in ProteomeXchange with identifier PXD008353, identified over 100 changed proteins exhibiting significant alterations in abundance and/or phosphorylation levels at different time points (5–240 min) in neurons after glutamate overstimulation. Bioinformatic analyses predicted that many of them are components of signalling networks directing defective neuronal morphology and functions. Among them, the well-known neuronal survival regulators including mitogen-activated protein kinases Erk1/2, glycogen synthase kinase 3 (GSK3) and microtubule-associated protein (Tau), were selected for validation by biochemical approaches, which confirmed the findings of the proteomic analysis. Bioinformatic analysis predicted Protein Kinase B (Akt), c-Jun kinase (JNK), cyclin-dependent protein kinase 5 (Cdk5), MAP kinase kinase (MEK), Casein kinase 2 (CK2), Rho-activated protein kinase (Rock) and Serum/glucocorticoid-regulated kinase 1 (SGK1) as the potential upstream kinases phosphorylating some of the changed proteins. Further biochemical investigation confirmed the predictions of sustained changes of the activation states of neuronal Akt and CK2 in excitotoxicity. Thus, future investigation to define the signalling pathways directing the dynamic alterations in abundance and phosphorylation of the identified changed neuronal proteins will help elucidate the molecular mechanism of neuronal death in excitotoxicity.
Collapse
Affiliation(s)
- Ashfaqul Hoque
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.,Metabolic Signalling Laboratory, St. Vincent's Institute for Medical Research, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Nicholas A Williamson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - S Sadia Ameen
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Giuseppe D Ciccotosto
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - M Iqbal Hossain
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute for Medical Research, University of Melbourne, Fitzroy, VIC, 3065, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, 3000, Australia
| | - Dominic C H Ng
- School of Biomedical Sciences, University of Queensland, St. Lucia, QLD, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia. .,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
11
|
Haun F, Neumann S, Peintner L, Wieland K, Habicht J, Schwan C, Østevold K, Koczorowska MM, Biniossek M, Kist M, Busch H, Boerries M, Davis RJ, Maurer U, Schilling O, Aktories K, Borner C. Identification of a novel anoikis signalling pathway using the fungal virulence factor gliotoxin. Nat Commun 2018; 9:3524. [PMID: 30166526 PMCID: PMC6117259 DOI: 10.1038/s41467-018-05850-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 07/25/2018] [Indexed: 01/02/2023] Open
Abstract
Anoikis is a form of apoptosis induced by cell detachment. Integrin inactivation plays a major role in the process but the exact signalling pathway is ill-defined. Here we identify an anoikis pathway using gliotoxin (GT), a virulence factor of the fungus Aspergillus fumigatus, which causes invasive aspergillosis in humans. GT prevents integrin binding to RGD-containing extracellular matrix components by covalently modifying cysteines in the binding pocket. As a consequence, focal adhesion kinase (FAK) is inhibited resulting in dephosphorylation of p190RhoGAP, allowing activation of RhoA. Sequential activation of ROCK, MKK4/MKK7 and JNK then triggers pro-apoptotic phosphorylation of Bim. Cells in suspension or lacking integrin surface expression are insensitive to GT but are sensitised to ROCK-MKK4/MKK7-JNK-dependent anoikis upon attachment to fibronectin or integrin upregulation. The same signalling pathway is triggered by FAK inhibition or inhibiting integrin αV/β3 with Cilengitide. Thus, GT can target integrins to induce anoikis on lung epithelial cells.
Collapse
Affiliation(s)
- Florian Haun
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany.,Faculty of Biology, Albert Ludwigs University Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Albertstrasse 19a, 79104, Freiburg, Germany
| | - Simon Neumann
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Lukas Peintner
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Katrin Wieland
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Jüri Habicht
- Institute of Immunology, University Hospital Heidelberg, Im Neuenheimer Feld 305, 69120, Heidelberg, Germany
| | - Carsten Schwan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwigs University Freiburg, Albertstrasse 25, 79102, Freiburg, Germany
| | - Kristine Østevold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwigs University Freiburg, Albertstrasse 25, 79102, Freiburg, Germany
| | - Maria Magdalena Koczorowska
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Martin Biniossek
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Matthias Kist
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany
| | - Hauke Busch
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany.,Institute of Experimental Dermatology and Institute of Cardiogenetics, University of Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Melanie Boerries
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), INF 280, 69120, Heidelberg, Germany
| | - Roger J Davis
- Howard Hughes Medical Institute & Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Ulrich Maurer
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Albertstrasse 19a, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Schänzlestrasse 14, 79104, Freiburg, Germany
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Albertstrasse 19a, 79104, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Schänzlestrasse 14, 79104, Freiburg, Germany
| | - Klaus Aktories
- Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Albertstrasse 19a, 79104, Freiburg, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Albert Ludwigs University Freiburg, Albertstrasse 25, 79102, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, Schänzlestrasse 14, 79104, Freiburg, Germany
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104, Freiburg, Germany. .,Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Albertstrasse 19a, 79104, Freiburg, Germany. .,BIOSS Centre for Biological Signalling Studies, Schänzlestrasse 14, 79104, Freiburg, Germany.
| |
Collapse
|
12
|
Wu HT, Xie CR, Lv J, Qi HQ, Wang F, Zhang S, Fang QL, Wang FQ, Lu YY, Yin ZY. The tumor suppressor DLC1 inhibits cancer progression and oncogenic autophagy in hepatocellular carcinoma. J Transl Med 2018; 98:1014-1024. [PMID: 29785050 DOI: 10.1038/s41374-018-0062-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/23/2018] [Accepted: 03/04/2018] [Indexed: 12/26/2022] Open
Abstract
Downregulation of deleted in liver cancer 1 (DLC1) is associated with poor prognosis of various cancers, but its functional mechanisms in hepatocellular carcinoma (HCC) remains unclear. In the present study, we investigated the roles of DLC1 in tumor progression and autophagy of HCC. We found that DLC1 was frequently downregulated in HCC tissues. Underexpression of DLC1 correlated with AFP level, vascular invasion, poor differentiation, and poor prognosis. In vitro assays revealed that DLC1 not only suppressed the proliferation, migration, and invasion of HCC cells, but also inhibited autophagy of HCC cells. Mechanistic investigation revealed that DLC1 decreased TCF4 expression and the interaction between β-catenin and TCF4, then inactivated Wnt/β-catenin signaling. Additionally, DLC1 suppressed the ROCK1 activity and the dissociation of the Beclin1-Bcl2 complex, thereby inhibiting autophagy of HCC cells. In conclusion, our findings imply that loss of DLC1 contributes to the progression and oncogenic autophagy of HCC.
Collapse
Affiliation(s)
- Hui-Ta Wu
- Department of Oncology, Zhongshan Hospital, Xiamen University, Fujian, 361004, Xiamen, China
| | - Cheng-Rong Xie
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Fujian, 361004, Xiamen, China
| | - Jie Lv
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Fujian, 361004, Xiamen, China
| | - He-Qiang Qi
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Fujian, 361004, Xiamen, China
| | - Fei Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of, Fujian, 350025, Fuzhou, China
| | - Sheng Zhang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Fujian, 361004, Xiamen, China
| | - Qin-Liang Fang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Fujian, 361004, Xiamen, China
| | - Fu-Qiang Wang
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Fujian, 361004, Xiamen, China
| | - Yu-Yan Lu
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Fujian, 361004, Xiamen, China
| | - Zhen-Yu Yin
- Department of Hepatobiliary Surgery, Zhongshan Hospital, Xiamen University, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Fujian, 361004, Xiamen, China.
| |
Collapse
|
13
|
Ma Q, Liu Y, Chen L. JIP3 deficiency attenuates cardiac hypertrophy by suppression of JNK pathway. Biochem Biophys Res Commun 2018; 503:1-7. [PMID: 29604277 DOI: 10.1016/j.bbrc.2018.03.208] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 03/27/2018] [Indexed: 01/22/2023]
Abstract
Pathological cardiac hypertrophy is a leading cause of morbidity and mortality worldwide; however, our understanding of the molecular mechanisms revealing the disease is still unclear. In the present study, we suggested that c-Jun N-terminal kinase (JNK)-interacting protein 3 (JIP3), involved in various cellular processes, played an essential role in regulating pathological cardiac hypertrophy through in vivo and in vitro studies. JIP3 was highly expressed in human hearts with hypertrophic cardiomyopathy (HCM), and in mouse hypertrophic hearts. Following, the wild type (WT) and JIP3-knockout (KO) mice subjected to aortic banding (AB) challenge were used as animal models with cardiac hypertrophy. The results showed that JIP3-KO mice after AB operation exhibited attenuated cardiac function, reduced fibrosis levels and decreased hypertrophic marker proteins, including atrial natriuretic peptides (Anp) and brain/B-type natriuretic peptides (Bnp) and β-myosin heavy chain (β-Mhc). Loss of JIP3 also ameliorated oxidative stress, inflammatory response, apoptosis and endoplasmic reticulum (ER) stress in hearts of mice after AB surgery. Consistently, the expressions of ER stress-related molecules, such as phosphorylated-α-subunit of the eukaryotic initiation factor-2 (eIF2α), glucose-regulated protein (GRP) 78 and C/-EBP homologous protein (CHOP), were markedly decreased by JIP3-deficiency in hearts of AB-operated mice. JNK and its down-streaming signal of p90rsk was highly activated by AB operation in WT mice, while being significantly reversed by JIP3-ablation. Intriguingly, the in vitro results showed that promoting JNK activation by using its activator of anisomycin enhanced AngII-stimulated ER stress, oxidative stress, apoptosis and inflammatory response in cardiomyocytes isolated from WT mice. However, JIP3-KO-attenuated these pathologies was rescued by anisomycin treatment in AngII-incubated cardiomyocytes. Together, the findings indicated that blockage of JIP3 could alleviate cardiac hypertrophy via inactivating JNK pathway, and thus might be a promising strategy to prevent pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Qinghua Ma
- Department of Cardiology, Linyi Central Hospital of Shandong Province, Linyi 276400, China
| | - Yuxiu Liu
- Department of Geriatric Medicine, Linyi Central Hospital of Shandong Province, Linyi 276400, China
| | - Lianghua Chen
- Department of Cardiology, Shandong Provincial Hospital, Jinan 250021, China.
| |
Collapse
|
14
|
van der Velden JL, Wagner DE, Lahue KG, Abdalla ST, Lam YW, Weiss DJ, Janssen-Heininger YMW. TGF-β1-induced deposition of provisional extracellular matrix by tracheal basal cells promotes epithelial-to-mesenchymal transition in a c-Jun NH 2-terminal kinase-1-dependent manner. Am J Physiol Lung Cell Mol Physiol 2018; 314:L984-L997. [PMID: 29469614 PMCID: PMC6032072 DOI: 10.1152/ajplung.00053.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 02/14/2018] [Accepted: 02/15/2018] [Indexed: 12/16/2022] Open
Abstract
Epithelial cells have been suggested as potential drivers of lung fibrosis, although the epithelial-dependent pathways that promote fibrogenesis remain unknown. Extracellular matrix is increasingly recognized as an environment that can drive cellular responses in various pulmonary diseases. In this study, we demonstrate that transforming growth factor-β1 (TGF-β1)-stimulated mouse tracheal basal (MTB) cells produce provisional matrix proteins in vitro, which initiate mesenchymal changes in subsequently freshly plated MTB cells via Rho kinase- and c-Jun NH2-terminal kinase (JNK1)-dependent processes. Repopulation of decellularized lung scaffolds, derived from mice with bleomycin-induced fibrosis or from patients with idiopathic pulmonary fibrosis, with wild-type MTB cells resulted in a loss of epithelial gene expression and augmentation of mesenchymal gene expression compared with cells seeded into decellularized normal lungs. In contrast, Jnk1-/- basal cells seeded into fibrotic lung scaffolds retained a robust epithelial expression profile, failed to induce mesenchymal genes, and differentiated into club cell secretory protein-expressing cells. This new paradigm wherein TGF-β1-induced extracellular matrix derived from MTB cells activates a JNK1-dependent mesenchymal program, which impedes subsequent normal epithelial cell homeostasis, provides a plausible scenario of chronic aberrant epithelial repair, thought to be critical in lung fibrogenesis. This study identifies JNK1 as a possible target for inhibition in settings wherein reepithelialization is desired.
Collapse
Affiliation(s)
- Jos L van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Darcy E Wagner
- Department of Medicine, University of Vermont , Burlington, Vermont
- Department of Experimental Medical Sciences, Lung Bioengineering, and Regeneration, Lund University , Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University , Lund, Sweden
| | - Karolyn G Lahue
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Sarah T Abdalla
- Department of Pathology and Laboratory Medicine, University of Vermont , Burlington, Vermont
| | - Ying-Wai Lam
- Department of Biology, University of Vermont , Burlington, Vermont
- Vermont Genetics Networks Proteomics Facility, University of Vermont , Burlington, Vermont
| | - Daniel J Weiss
- Department of Medicine, University of Vermont , Burlington, Vermont
| | | |
Collapse
|
15
|
Mining for Candidate Genes Related to Pancreatic Cancer Using Protein-Protein Interactions and a Shortest Path Approach. BIOMED RESEARCH INTERNATIONAL 2015; 2015:623121. [PMID: 26613085 PMCID: PMC4647023 DOI: 10.1155/2015/623121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 10/15/2015] [Indexed: 12/13/2022]
Abstract
Pancreatic cancer (PC) is a highly malignant tumor derived from pancreas tissue and is one of the leading causes of death from cancer. Its molecular mechanism has been partially revealed by validating its oncogenes and tumor suppressor genes; however, the available data remain insufficient for medical workers to design effective treatments. Large-scale identification of PC-related genes can promote studies on PC. In this study, we propose a computational method for mining new candidate PC-related genes. A large network was constructed using protein-protein interaction information, and a shortest path approach was applied to mine new candidate genes based on validated PC-related genes. In addition, a permutation test was adopted to further select key candidate genes. Finally, for all discovered candidate genes, the likelihood that the genes are novel PC-related genes is discussed based on their currently known functions.
Collapse
|
16
|
Wang Z, Chen Y, Lü Y, Chen X, Cheng L, Mi X, Xu X, Deng W, Zhang Y, Wang N, Li J, Li Y, Wang X. Effects of JIP3 on epileptic seizures: Evidence from temporal lobe epilepsy patients, kainic-induced acute seizures and pentylenetetrazole-induced kindled seizures. Neuroscience 2015; 300:314-24. [PMID: 26002316 DOI: 10.1016/j.neuroscience.2015.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 05/01/2015] [Accepted: 05/05/2015] [Indexed: 11/26/2022]
Abstract
JNK-interacting protein 3 (JIP3), also known as JNK stress-activated protein kinase-associated protein 1 (JSAP1), is a scaffold protein mainly involved in the regulation of the pro-apoptotic signaling cascade mediated by c-Jun N-terminal kinase (JNK). Overexpression of JIP3 in neurons in vitro has been reported to lead to accelerated activation of JNK and enhanced apoptosis response to cellular stress. However, the occurrence and the functional significance of stress-induced modulations of JIP3 levels in vivo remain elusive. In this study, we investigated the expression of JIP3 in temporal lobe epilepsy (TLE) and in a kainic acid (KA)-induced mouse model of epileptic seizures, and determined whether down-regulation of JIP3 can decrease susceptibility to seizures and neuron damage induced by KA. We found that JIP3 was markedly increased in TLE patients and a mouse model of epileptic seizures; mice underexpressing JIP3 through lentivirus bearing LV-Letm1-RNAi showed decreased susceptibility, delayed first seizure and decreased seizure duration response to the epileptogenic properties of KA. Subsequently, a decreased activation of JNK following seizure induction was observed in mice underexpressing JIP3, which also exhibited less neuronal apoptosis in the CA3 region of the hippocampus, as assessed three days after KA administration. We also found that mice underexpressing JIP3 exhibited a delayed pentylenetetrazole (PTZ)-induced kindling seizure process.
Collapse
Affiliation(s)
- Z Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China; Department of Neurology, Fuling Central Hospital, Chongqing 408000, China
| | - Y Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Y Lü
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - X Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - L Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - X Mi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - X Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - W Deng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Y Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - N Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - J Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Y Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - X Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China.
| |
Collapse
|
17
|
Julian L, Olson MF. Rho-associated coiled-coil containing kinases (ROCK): structure, regulation, and functions. Small GTPases 2014; 5:e29846. [PMID: 25010901 PMCID: PMC4114931 DOI: 10.4161/sgtp.29846] [Citation(s) in RCA: 362] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 07/02/2014] [Accepted: 07/07/2014] [Indexed: 12/29/2022] Open
Abstract
Rho-associated coiled-coil containing kinases (ROCK) were originally identified as effectors of the RhoA small GTPase. (1)(-) (5) They belong to the AGC family of serine/threonine kinases (6) and play vital roles in facilitating actomyosin cytoskeleton contractility downstream of RhoA and RhoC activation. Since their discovery, ROCK kinases have been extensively studied, unveiling their manifold functions in processes including cell contraction, migration, apoptosis, survival, and proliferation. Two mammalian ROCK homologs have been identified, ROCK1 (also called ROCK I, ROKβ, Rho-kinase β, or p160ROCK) and ROCK2 (also known as ROCK II, ROKα, or Rho kinase), hereafter collectively referred to as ROCK. In this review, we will focus on the structure, regulation, and functions of ROCK.
Collapse
Affiliation(s)
- Linda Julian
- Beatson Institute for Cancer Research; Glasgow, UK
| | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Rho kinases (ROCKs) are involved in regulating a variety of physiologic functions including cytoskeletal reorganization, migration, adhesion, survival and proliferation. They do so via activating several different downstream substrates such as myosin light chain phosphatase, LIM kinase and ezrin/radixin/moesin proteins. To date, most of the conclusions with regard to the function of ROCKs have involved the use of cell line models, pharmacologic inhibitors and dominant negative approaches. Importantly, the role of ROCK in hematopoiesis or leukemogenesis in the context of whole organism remains poorly understood. RECENT FINDINGS Recent studies utilizing mice deficient in the expression of ROCK1 have begun to shed some light into the physiologic role(s) of ROCK in both normal and abnormal hematopoiesis. Findings, thus far, suggest that ROCK plays an essential role in regulating growth and survival in different hematopoietic lineages via distinct mechanisms, in part, by utilizing distinct downstream substrates including maintaining the activation of tumor-suppressor genes. SUMMARY In blood cells, emerging data suggest that ROCK plays an essential role in negatively regulating inflammatory and erythropoietic stress and positively regulates the growth and survival of leukemic cells.
Collapse
Affiliation(s)
- Raghuveer Singh Mali
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Department of Pediatrics, Indianapolis, Indiana, USA
| | | | | |
Collapse
|
19
|
Abstract
Rho kinase (ROCK) is a major downstream effector of the small GTPase RhoA. ROCK family, consisting of ROCK1 and ROCK2, plays central roles in the organization of actin cytoskeleton and is involved in a wide range of fundamental cellular functions, such as contraction, adhesion, migration, proliferation, and apoptosis. Due to the discovery of effective inhibitors, such as fasudil and Y27632, the biological roles of ROCK have been extensively explored with particular attention on the cardiovascular system. In many preclinical models of cardiovascular diseases, including vasospasm, arteriosclerosis, hypertension, pulmonary hypertension, stroke, ischemia-reperfusion injury, and heart failure, ROCK inhibitors have shown a remarkable efficacy in reducing vascular smooth muscle cell hypercontraction, endothelial dysfunction, inflammatory cell recruitment, vascular remodeling, and cardiac remodeling. Moreover, fasudil has been used in the clinical trials of several cardiovascular diseases. The continuing utilization of available pharmacological inhibitors and the development of more potent or isoform-selective inhibitors in ROCK signaling research and in treating human diseases are escalating. In this review, we discuss the recent molecular, cellular, animal, and clinical studies with a focus on the current understanding of ROCK signaling in cardiovascular physiology and diseases. We particularly note that emerging evidence suggests that selective targeting ROCK isoform based on the disease pathophysiology may represent a novel therapeutic approach for the disease treatment including cardiovascular diseases.
Collapse
|
20
|
Enkhtuya R, Sato T, Wakasugi M, Tuvshintugs B, Miyata H, Sakurai T, Matsunaga T, Yoshioka K. The scaffold protein JLP plays a key role in regulating ultraviolet B-induced apoptosis in mice. Genes Cells 2014; 19:350-8. [PMID: 24520900 DOI: 10.1111/gtc.12135] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/23/2013] [Indexed: 11/27/2022]
Abstract
The ultraviolet B (UVB) component of sunlight can cause severe damage to skin cells and even induce skin cancer. Growing evidence indicates that the UVB-induced signaling network is complex and involves diverse cellular processes. In this study, we investigated the role of c-Jun NH2 -terminal kinase-associated leucine zipper protein (JLP), a scaffold protein for mitogen-activated protein kinase (MAPK) signaling cascades, in UVB-induced apoptosis. We found that UVB-induced skin epidermal apoptosis was prevented in Jlp knockout (KO) as well as in keratinocyte-specific Jlp KO mice. Analysis of the repair of UVB-induced DNA damage over time showed no evidence for the involvement of JLP in this process. In contrast, UVB-stimulated p38 MAPK activation in the skin was impaired in both Jlp KO and keratinocyte-specific Jlp KO mice. Moreover, topical treatment of UVB-irradiated mouse skin with a p38 inhibitor significantly suppressed the epidermal apoptosis in wild-type mice, but not in Jlp KO mice. Our findings suggest that JLP in skin basal keratinocytes plays an important role in UVB-induced apoptosis by modulating p38 MAPK signaling pathways. This is the first study to show a critical role for JLP in an in vivo response to environmental stimulation.
Collapse
Affiliation(s)
- Radnaa Enkhtuya
- Division of Molecular Cell Signaling, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Gurkar AU, Chu K, Raj L, Bouley R, Lee SH, Kim YB, Dunn SE, Mandinova A, Lee SW. Identification of ROCK1 kinase as a critical regulator of Beclin1-mediated autophagy during metabolic stress. Nat Commun 2014; 4:2189. [PMID: 23877263 PMCID: PMC3740589 DOI: 10.1038/ncomms3189] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 06/26/2013] [Indexed: 01/19/2023] Open
Abstract
The Ser/Thr Rho kinase 1 (ROCK1) is known to play major roles in a wide range of cellular activities, including those involved in tumor metastasis and apoptosis. Here we identify an indispensable function of ROCK1 in metabolic stress-induced autophagy. Applying a proteomics approach, we characterize Beclin1, a proximal component of the PI(3)kinase class III lipid-kinase complex that induces autophagy, as an interacting partner of ROCK1. Upon nutrient deprivation, activated ROCK1 promotes autophagy by binding and phosphorylating Beclin1 at Thr119. This results in the specific dissociation of the Beclin1-Bcl-2 complex, without affecting the Beclin1-UVRAG interaction. Conversely, inhibition of ROCK1 activity increases Beclin1-Bcl-2 association, thus reducing nutritional stress-mediated autophagy. Genetic knockout of ROCK1 function in mice also leads to impaired autophagy as evidenced by reduced autophagosome formation. These results show that ROCK1 acts as a prominent upstream regulator of Beclin1-mediated autophagy and maintains a homeostatic balance between apoptosis and autophagy.
Collapse
Affiliation(s)
- Aditi U Gurkar
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Zhang W, Zhao J, Lee JF, Gartung A, Jawadi H, Lambiv WL, Honn KV, Lee MJ. ETS-1-mediated transcriptional up-regulation of CD44 is required for sphingosine-1-phosphate receptor subtype 3-stimulated chemotaxis. J Biol Chem 2013; 288:32126-32137. [PMID: 24064218 PMCID: PMC3820853 DOI: 10.1074/jbc.m113.495218] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/18/2013] [Indexed: 12/17/2022] Open
Abstract
Sphingosine-1-phosphate (S1P)-regulated chemotaxis plays critical roles in various physiological and pathophysiological conditions. S1P-regulated chemotaxis is mediated by the S1P family of G-protein-coupled receptors. However, molecular details of the S1P-regulated chemotaxis are incompletely understood. Cultured human lung adenocarcinoma cell lines abundantly express S1P receptor subtype 3 (S1P3), thus providing a tractable in vitro system to characterize molecular mechanism(s) underlying the S1P3 receptor-regulated chemotactic response. S1P treatment enhances CD44 expression and induces membrane localization of CD44 polypeptides via the S1P3/Rho kinase (ROCK) signaling pathway. Knockdown of CD44 completely diminishes the S1P-stimulated chemotaxis. Promoter analysis suggests that the CD44 promoter contains binding sites of the ETS-1 (v-ets erythroblastosis virus E26 oncogene homolog 1) transcriptional factor. ChIP assay confirms that S1P treatment stimulates the binding of ETS-1 to the CD44 promoter region. Moreover, S1P induces the expression and nuclear translocation of ETS-1. Knockdown of S1P3 or inhibition of ROCK abrogates the S1P-induced ETS-1 expression. Furthermore, knockdown of ETS-1 inhibits the S1P-induced CD44 expression and cell migration. In addition, we showed that S1P3/ROCK signaling up-regulates ETS-1 via the activity of JNK. Collectively, we characterized a novel signaling axis, i.e., ROCK-JNK-ETS-1-CD44 pathway, which plays an essential role in the S1P3-regulated chemotactic response.
Collapse
Affiliation(s)
- Wenliang Zhang
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Jiawei Zhao
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Jen-Fu Lee
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | - Allison Gartung
- From the Department of Pathology,; the Bioactive Lipid Research Program
| | | | | | - Kenneth V Honn
- From the Department of Pathology,; the Bioactive Lipid Research Program,; the Karmanos Cancer Institute
| | - Menq-Jer Lee
- From the Department of Pathology,; the Bioactive Lipid Research Program,; the Karmanos Cancer Institute; the Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan 48201.
| |
Collapse
|
23
|
Kedersha N, Ivanov P, Anderson P. Stress granules and cell signaling: more than just a passing phase? Trends Biochem Sci 2013; 38:494-506. [PMID: 24029419 DOI: 10.1016/j.tibs.2013.07.004] [Citation(s) in RCA: 467] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/23/2013] [Accepted: 07/29/2013] [Indexed: 12/27/2022]
Abstract
Stress granules (SGs) contain translationally-stalled mRNAs, associated preinitiation factors, and specific RNA-binding proteins. In addition, many signaling proteins are recruited to SGs and/or influence their assembly, which is transient, lasting only until the cells adapt to stress or die. Beyond their role as mRNA triage centers, we posit that SGs constitute RNA-centric signaling hubs analogous to classical multiprotein signaling domains such as transmembrane receptor complexes. As signaling centers, SG formation communicates a 'state of emergency', and their transient existence alters multiple signaling pathways by intercepting and sequestering signaling components. SG assembly and downstream signaling functions may require a cytosolic phase transition facilitated by intrinsically disordered, aggregation-prone protein regions shared by RNA-binding and signaling proteins.
Collapse
Affiliation(s)
- Nancy Kedersha
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Smith 652, One Jimmy Fund Way, Boston, MA 02115, USA
| | | | | |
Collapse
|
24
|
Schofield AV, Bernard O. Rho-associated coiled-coil kinase (ROCK) signaling and disease. Crit Rev Biochem Mol Biol 2013; 48:301-16. [PMID: 23601011 DOI: 10.3109/10409238.2013.786671] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The small Rho GTPase family of proteins, encompassing the three major G-protein classes Rho, Rac and cell division control protein 42, are key mitogenic signaling molecules that regulate multiple cancer-associated cellular phenotypes including cell proliferation and motility. These proteins are known for their role in the regulation of actin cytoskeletal dynamics, which is achieved through modulating the activity of their downstream effector molecules. The Rho-associated coiled-coil kinase 1 and 2 (ROCK1 and ROCK2) proteins were the first discovered Rho effectors that were primarily established as players in RhoA-mediated stress fiber formation and focal adhesion assembly. It has since been discovered that the ROCK kinases actively phosphorylate a large cohort of actin-binding proteins and intermediate filament proteins to modulate their functions. It is well established that global cellular morphology, as modulated by the three cytoskeletal networks: actin filaments, intermediate filaments and microtubules, is regulated by a variety of accessory proteins whose activities are dependent on their phosphorylation by the Rho-kinases. As a consequence, they regulate many key cellular functions associated with malignancy, including cell proliferation, motility and viability. In this current review, we focus on the role of the ROCK-signaling pathways in disease including cancer.
Collapse
Affiliation(s)
- Alice V Schofield
- St Vincent's Institute of Medical Research, Cytoskeleton and Cancer Unit and Department of Medicine, St Vincent's Hospital, University of Melbourne, Victoria 3065, Australia
| | | |
Collapse
|
25
|
Arf6-GEF BRAG1 regulates JNK-mediated synaptic removal of GluA1-containing AMPA receptors: a new mechanism for nonsyndromic X-linked mental disorder. J Neurosci 2012; 32:11716-26. [PMID: 22915114 DOI: 10.1523/jneurosci.1942-12.2012] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Activity-dependent modifications of excitatory synapses contribute to synaptic maturation and plasticity, and are critical for learning and memory. Consequently, impairments in synapse formation or synaptic transmission are thought to be responsible for several types of mental disabilities. BRAG1 is a guanine nucleotide exchange factor for the small GTP-binding protein Arf6 that localizes to the postsynaptic density of excitatory synapses. Mutations in BRAG1 have been identified in families with X-linked intellectual disability (XLID). These mutations mapped to either the catalytic domain or an IQ-like motif; however, the pathophysiological basis of these mutations remains unknown. Here, we show that the BRAG1 IQ motif binds apo-calmodulin (CaM), and that calcium-induced CaM release triggers a reversible conformational change in human BRAG1. We demonstrate that BRAG1 activity, stimulated by activation of NMDA-sensitive glutamate receptors, depresses AMPA receptor (AMPA-R)-mediated transmission via JNK-mediated synaptic removal of GluA1-containing AMPA-Rs in rat hippocampal neurons. Importantly, a BRAG1 mutant that fails to activate Arf6 also fails to depress AMPA-R signaling, indicating that Arf6 activity is necessary for this process. Conversely, a mutation in the BRAG1 IQ-like motif that impairs CaM binding results in hyperactivation of Arf6 signaling and constitutive depression of AMPA transmission. Our findings reveal a role for BRAG1 in response to neuronal activity with possible clinical relevance to nonsyndromic XLID.
Collapse
|
26
|
|
27
|
ROCK1 functions as a critical regulator of stress erythropoiesis and survival by regulating p53. Blood 2012; 120:2868-78. [PMID: 22889758 DOI: 10.1182/blood-2011-10-384172] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Erythropoiesis is a dynamic, multistep process whereby hematopoietic stem cells differentiate toward a progressively committed erythroid lineage through intermediate progenitors. Although several downstream signaling molecules have been identified that regulate steady-state erythropoiesis, the major regulators under conditions of stress remain poorly defined. Rho kinases (ROCKs) belong to a family of serine/threonine kinases. Using gene-targeted ROCK1-deficient mice, we show that lack of ROCK1 in phenylhydrazine-induced oxidative stress model results in enhanced recovery from hemolytic anemia as well as enhanced splenic stress erythropoiesis compared with control mice. Deficiency of ROCK1 also results in enhanced survival, whereas wild-type mice die rapidly in response to stress. Enhanced survivability of ROCK1-deficient mice is associated with reduced level of reactive oxygen species. BM transplantation studies revealed that enhanced stress erythropoiesis in ROCK1-deficient mice is stem cell autonomous. We show that ROCK1 binds to p53 and regulates its stability and expression. In the absence of ROCK1, p53 phosphorylation and expression is significantly reduced. Our findings reveal that ROCK1 functions as a physiologic regulator of p53 under conditions of erythroid stress. These findings are expected to offer new perspectives on stress erythropoiesis and may provide a potential therapeutic target in human disease characterized by anemia.
Collapse
|
28
|
Wang W, Wang Y, Long J, Wang J, Haudek SB, Overbeek P, Chang BH, Schumacker PT, Danesh FR. Mitochondrial fission triggered by hyperglycemia is mediated by ROCK1 activation in podocytes and endothelial cells. Cell Metab 2012; 15:186-200. [PMID: 22326220 PMCID: PMC3278719 DOI: 10.1016/j.cmet.2012.01.009] [Citation(s) in RCA: 373] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 10/16/2011] [Accepted: 01/10/2012] [Indexed: 11/18/2022]
Abstract
Several lines of evidence suggest that mitochondrial dysfunction plays a critical role in the pathogenesis of microvascular complications of diabetes, including diabetic nephropathy. However, the signaling pathways by which hyperglycemia leads to mitochondrial dysfunction are not fully understood. Here we examined the role of Rho-associated coiled coil-containing protein kinase 1 (ROCK1) on mitochondrial dynamics by generating two diabetic mouse models with targeted deletions of ROCK1 and an inducible podocyte-specific knockin mouse expressing a constitutively active (cA) mutant of ROCK1. Our findings suggest that ROCK1 mediates hyperglycemia-induced mitochondrial fission by promoting dynamin-related protein-1 (Drp1) recruitment to the mitochondria. Deletion of ROCK1 in diabetic mice prevented mitochondrial fission, whereas podocyte-specific cA-ROCK1 mice exhibited increased mitochondrial fission. Importantly, we found that ROCK1 triggers mitochondrial fission by phosphorylating Drp1 at serine 600 residue. These findings provide insights into the unexpected role of ROCK1 in a signaling cascade that regulates mitochondrial dynamics.
Collapse
Affiliation(s)
- Wenjian Wang
- Department of Medicine-Nephrology, Baylor College of Medicine, Houston, TX
| | - Yin Wang
- Department of Medicine-Nephrology, Baylor College of Medicine, Houston, TX
| | - Jianyin Long
- Department of Medicine-Nephrology, Baylor College of Medicine, Houston, TX
| | - Jinrong Wang
- Department of Medicine-Nephrology, Baylor College of Medicine, Houston, TX
| | - Sandra B. Haudek
- Department of Medicine-Division of Cardiovascular Sciences, Baylor College of Medicine, Houston, TX
| | - Paul Overbeek
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Benny H.J. Chang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Paul T. Schumacker
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Farhad R. Danesh
- Department of Medicine-Nephrology, Baylor College of Medicine, Houston, TX
- Department of Pharmacology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
29
|
Reipschläger S, Kubatzky K, Taromi S, Burger M, Orth J, Aktories K, Schmidt G. Toxin-induced RhoA activity mediates CCL1-triggered signal transducers and activators of transcription protein signaling. J Biol Chem 2012; 287:11183-94. [PMID: 22311973 DOI: 10.1074/jbc.m111.313395] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RhoA is reportedly involved in signal transducers and activators of transcription (STAT)-dependent transcription. However, the pathway connecting the GTPase and STAT signaling has not been characterized. Here, we made use of bacterial toxins, which directly activate Rho GTPases to analyze this pathway. Cytotoxic necrotizing factors (CNFs) are produced by pathogenic Escherichia coli strains and by Yersinia pseudotuberculosis. They activate small GTPases of the Rho family by deamidation of a glutamine, which is crucial for GTP hydrolysis. We show that RhoA activation leads to phosphorylation and activation of STAT3 and identify signal proteins involved in this pathway. RhoA-dependent STAT3 stimulation requires ROCK and Jun kinase activation as well as AP1-induced protein synthesis. The secretion of one or more factors activates the JAK-STAT pathway in an auto/paracrine manner. We identify CCL1/I-309 as an essential cytokine, which is produced and secreted upon RhoA activation and which is able to activate STAT3-dependent signaling pathways.
Collapse
Affiliation(s)
- Simone Reipschläger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Albert-Ludwigs-University of Freiburg, Albert-Str. 25, 79104 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Street CA, Bryan BA. Rho kinase proteins--pleiotropic modulators of cell survival and apoptosis. Anticancer Res 2011; 31:3645-3657. [PMID: 22110183 PMCID: PMC3226732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Rho kinase (ROCK) proteins are Rho-GTPase activated serine/threonine kinases that function as modulators of actin-myosin cytoskeletal dynamics via regulation of Lin11, Isl-1 & Mec-3 domain (LIM) kinase, myosin light chain (MLC), and MLC phosphatase. A strong correlation between cytoskeletal rearrangements and tumor cell invasion, metastasis, and deregulated microenvironment interaction has been reported in the literature, and the utilization of pharmacological inhibitors of ROCK signaling for the treatment of cancer is actively being pursued by a number of pharmaceutical companies. Indeed, in many preclinical models ROCK inhibitors have shown remarkable efficacy in reducing tumor growth and metastasis. Interestingly, ROCK signaling has been shown to be either pro-apoptotic or pro-survival in a cell type and context dependent manner, though the molecular mechanisms controlling ROCK-mediated cell fate decisions are unknown. This review summarizes the many pleiotropic roles of ROCK signaling in survival and apoptosis, and suggests that controlled modulation of ROCK activity in tumor cells has the potential to significantly affect tumor survival and patient outcome.
Collapse
Affiliation(s)
- Catharine A. Street
- Ghosh Science and Technology Center, Department of Biology, Worcester State University, Worcester, MA, 01602, U.S.A
| | - Brad A. Bryan
- Center of Excellence in Cancer Research, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, U.S.A
| |
Collapse
|
31
|
Abstract
This review addresses the roles of Rho/Rho-kinase (ROCK) pathway in the pathogenesis of diabetes complications. Diabetes can cause many serious complications and can result in physical disability or even increased mortality. However, there are not many effective ways to treat these complications. The small guanosine-5'-triphosphate-binding protein Rho and its downstream target Rho-kinase mediate important cellular functions, such as cell morphology, motility, secretion, proliferation, and gene expression. Recently, the Rho/Rho-kinase pathway has attracted a great deal of attention in diabetes-related research. These studies have provided evidence that the activity and gene expression of Rho-kinase are upregulated in some tissues in animal models of type 1 or type 2 diabetes and in cell lines cultured with high concentrations of glucose. Inhibitors of Rho-kinase could prevent or ameliorate the pathological changes in diabetic complications. The inhibitory effects of statins on the Rho/Rho-kinase signalling pathway may also play a role in the prevention of diabetic complications. However, the precise molecular mechanism by which the Rho/Roh-kinase pathway participates in the development or progression of diabetic complications has not been extensively investigated. This article evaluates the relationship between Rho/Roh-kinase activation and diabetic complications, as well as the roles of Roh-kinase inhibitors and statins in the complications of diabetes, with the objective of providing a novel target for the treatment of long-term diabetic complications.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Endocrinology, The Second Hospital of He Bei Medical University, Shijiazhuang, China
| | | |
Collapse
|
32
|
Kim HG, Hwang SY, Aaronson SA, Mandinova A, Lee SW. DDR1 receptor tyrosine kinase promotes prosurvival pathway through Notch1 activation. J Biol Chem 2011; 286:17672-81. [PMID: 21398698 DOI: 10.1074/jbc.m111.236612] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
DDR1 (discoidin domain receptor tyrosine kinase 1) kinase s highly expressed in a variety of human cancers and occasionally mutated in lung cancer and leukemia. It is now clear that aberrant signaling through the DDR1 receptor is closely associated with various steps of tumorigenesis, although little is known about the molecular mechanism(s) underlying the role of DDR1 in cancer. Besides the role of DDR1 in tumorigenesis, we previously identified DDR1 kinase as a transcriptional target of tumor suppressor p53. DDR1 is functionally activated as determined by its tyrosine phosphorylation, in response to p53-dependent DNA damage. In this study, we report the characterization of the Notch1 protein as an interacting partner of DDR1 receptor, as determined by tandem affinity protein purification. Upon ligand-mediated DDR1 kinase activation, Notch1 was activated, bound to DDR1, and activated canonical Notch1 targets, including Hes1 and Hey2. Moreover, DDR1 ligand (collagen I) treatment significantly increased the active form of Notch1 receptor in the nuclear fraction, whereas DDR1 knockdown cells show little or no increase of the active form of Notch1 in the nuclear fraction, suggesting a novel intracellular mechanism underlying autocrine activation of wild-type Notch signaling through DDR1. DDR1 activation suppressed genotoxic-mediated cell death, whereas Notch1 inhibition by a γ-secretase inhibitor, DAPT, enhanced cell death in response to stress. Moreover, the DDR1 knockdown cancer cells showed the reduced transformed phenotypes in vitro and in vivo xenograft studies. The results suggest that DDR1 exerts prosurvival effect, at least in part, through the functional interaction with Notch1.
Collapse
Affiliation(s)
- Hyung-Gu Kim
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | |
Collapse
|
33
|
Xu B, Zhou Y, O K, Choy PC, Pierce GN, Siow YL. Regulation of stress-associated scaffold proteins JIP1 and JIP3 on the c-Jun NH2-terminal kinase in ischemia-reperfusion. Can J Physiol Pharmacol 2011; 88:1084-92. [PMID: 21076496 DOI: 10.1139/y10-088] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ischemia-reperfusion (IR)-induced cell apoptosis involves the activation of c-Jun NH2-terminal kinase (JNK). The activation of JNK requires the presence of scaffold proteins called JNK-interacting proteins (JIP), which bind several members of a signaling cascade for proper signaling specificity. In this study, the expression of scaffold proteins JIP1 and JIP3 and their roles in the regulation of JNK activity were investigated in simulated IR in a cell model (H9c2). JIP1 protein expression was significantly decreased, whereas JIP3 protein expression was increased in IR H9c2 cells. Adenovirus-induced overexpression of JIP1 reduced IR-induced JNK activity and apoptosis. Conversely, overexpression of JIP3 increased JNK activity and apoptosis following IR. Depletion of endogenous JIP1 by siRNA treatment increased the IR-induced JNK activity, whereas siRNA-mediated depletion of endogenous JIP3 inhibited JNK activity. These results suggest that JIP1 and JIP3 play important roles in the activation of JNK during simulated IR challenge in H9c2 cells.
Collapse
Affiliation(s)
- Bing Xu
- Centre for Research and Treatment of Atherosclerosis, University of Manitoba, Winnipeg, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Shi J, Zhang L, Wei L. Rho-kinase in development and heart failure: insights from genetic models. Pediatr Cardiol 2011; 32:297-304. [PMID: 21327630 PMCID: PMC3085170 DOI: 10.1007/s00246-011-9920-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 02/04/2011] [Indexed: 11/26/2022]
Abstract
Rho-kinase (ROCK) belongs to the AGC (protein kinase A/protein kinase G/protein kinase C, PKA/PKG/PKC) family of serine/threonine kinases and is a major downstream effector of small GTPase RhoA. Rho-kinase is involved in a wide range of fundamental cellular functions such as contraction, adhesion, migration, and proliferation. Two ROCK isoforms, ROCK1 and ROCK2, are assumed to be functionally redundant, based largely on the major common activators, the high degree of homology within the kinase domain, and studies from overexpression with kinase constructs and chemical inhibitors (e.g., Y27632 and fasudil), which inhibit both ROCK1 and ROCK2. Gene targeting and RNA interference approaches allow further dissection of distinct cellular, physiologic, and pathophysiologic functions of the two ROCK isoforms. This review focuses on the current understanding of ROCK isoform biology, with a particular emphasis on their functions in mouse development and the pathogenesis of heart failure.
Collapse
Affiliation(s)
- Jianjian Shi
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202-5225, USA
| | | | | |
Collapse
|
35
|
Walker A, Su H, Conti MA, Harb N, Adelstein RS, Sato N. Non-muscle myosin II regulates survival threshold of pluripotent stem cells. Nat Commun 2010; 1:71. [PMID: 20842192 PMCID: PMC3430968 DOI: 10.1038/ncomms1074] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 08/11/2010] [Indexed: 12/22/2022] Open
Abstract
Human pluripotent stem (hPS) cells such as human embryonic stem (hES) and induced pluripotent stem (hiPS) cells are vulnerable under single cell conditions, which hampers practical applications; yet, the mechanisms underlying this cell death remain elusive. In this paper, we demonstrate that treatment with a specific inhibitor of non-muscle myosin II (NMII), blebbistatin, enhances the survival of hPS cells under clonal density and suspension conditions, and, in combination with a synthetic matrix, supports a fully defined environment for self-renewal. Consistent with this, genetically engineered mouse embryonic stem cells lacking an isoform of NMII heavy chain (NMHCII), or hES cells expressing a short hairpin RNA to knock down NMHCII, show greater viability than controls. Moreover, NMII inhibition increases the expression of self-renewal regulators Oct3/4 and Nanog, suggesting a mechanistic connection between NMII and self-renewal. These results underscore the importance of the molecular motor, NMII, as a novel target for chemically engineering the survival and self-renewal of hPS cells.
Collapse
Affiliation(s)
- Andrea Walker
- Department of Biochemistry, University of California, Riverside, Riverside, California 29521, USA
| | | | | | | | | | | |
Collapse
|
36
|
Hahmann C, Schroeter T. Rho-kinase inhibitors as therapeutics: from pan inhibition to isoform selectivity. Cell Mol Life Sci 2010; 67:171-7. [PMID: 19907920 PMCID: PMC11115778 DOI: 10.1007/s00018-009-0189-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 09/08/2009] [Accepted: 09/17/2009] [Indexed: 01/02/2023]
Abstract
The emerging critical implications of Rho/Rho-kinase (ROCK) signaling in neurodegenerative diseases, glaucoma, renoprotection, diabetes and cancer have sparked growing interest in the pharmacological potential of ROCK inhibitors beyond their current application in cardiovascular disease. This article discusses the therapeutic benefits of novel ROCK inhibitors in development, and highlights the recent advances in the current understanding of disease-dependent and isoform-specific functions of ROCK and their potential impact on future therapeutic strategies.
Collapse
Affiliation(s)
- C. Hahmann
- Discovery Biology, Translational Research Institute, The Scripps Research Institute, Scripps Florida, 130 Scripps Way, Jupiter, FL 33458 USA
| | - T. Schroeter
- Discovery Biology, Translational Research Institute, The Scripps Research Institute, Scripps Florida, 130 Scripps Way, Jupiter, FL 33458 USA
| |
Collapse
|
37
|
Tsai NP, Wei LN. RhoA/ROCK1 signaling regulates stress granule formation and apoptosis. Cell Signal 2009; 22:668-75. [PMID: 20004716 DOI: 10.1016/j.cellsig.2009.12.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 11/30/2009] [Accepted: 12/03/2009] [Indexed: 01/31/2023]
Abstract
Cells form stress granules (SGs), in response to unfavorable environments, to avoid apoptosis, but it is unclear whether and how SG formation and cellular apoptosis are coordinately regulated. In this study we detected the small GTPase, Ras homolog gene family member A (RhoA), and its downstream kinase, Rho-associated, coiled-coil containing protein kinase 1 (ROCK1), in SG, and found that their stress-induced activities were important for SG formation and subsequent global translational repression. Importantly, only activated RhoA and ROCK1 were sequestered into SG. Sequestration of activated ROCK1 into SG prevented ROCK1 from interacting with JNK-interacting protein 3 (JIP-3) and its activation of c-Jun N-terminal kinase (JNK), a pathway triggering apoptosis, thereby protecting cells from apoptosis. This study identifies a specific signaling pathway, mediated by RhoA and ROCK1, which determines cell fate by promoting SG formation or initiating apoptosis during stress.
Collapse
Affiliation(s)
- Nien-Pei Tsai
- Department of Pharmacology, University of Minnesota Medical School, 6-120 Jackson Hall, 321 Church St. SE, Minneapolis, MN 55455, USA
| | | |
Collapse
|
38
|
Cao C, Wan Y. Parameters of protection against ultraviolet radiation-induced skin cell damage. J Cell Physiol 2009; 220:277-84. [PMID: 19360745 DOI: 10.1002/jcp.21780] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epidemiological and experimental evidence has supported the notion that solar ultraviolet (UV) radiation is the leading cause of skin cell damage and skin cancer. Non-melanoma skin cancer, one of the malignancies with the most rapidly increasing incidence, is suggested to be directly related to the total exposure to solar UV light. Over the past few years, the mechanisms of cellular responses to UV radiation have received unprecedented attention. Understanding how skin cells respond to UV radiation will undoubtedly help decipher what goes wrong in a variety of clinical skin disorders including skin cancer and will facilitate the development of novel therapeutic strategies. In the past decade, studies have established that UV radiation induces multifarious signal transduction pathways, some of which lead to apoptotic cell death, while others protect against this process. In this review, we summarize some of the most recent progresses regarding the involvement of multiple signal pathways in UV radiation-induced apoptosis in skin cells, especially in keratinocytes. These pathways include pro-apoptosis components such as MAPK, AMPK, and p53 as well as pro-survival components, namely, AKT and mTORC complexes.
Collapse
Affiliation(s)
- Cong Cao
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| | | |
Collapse
|
39
|
Lee SW, Ongusaha PP, VanHook AM. Science Signaling
Podcast: 02 December 2008. Sci Signal 2008. [DOI: 10.1126/scisignal.148pc13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Sam Lee and Pat Ongusaha discuss their research on the mechanisms by which ultraviolet B radiation induces cell death.
Collapse
Affiliation(s)
- Sam W. Lee
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, 13th Street, Building 149, Charlestown, MA 02129, USA
| | - Pat P. Ongusaha
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, 13th Street, Building 149, Charlestown, MA 02129, USA
| | - Annalisa M. VanHook
- Science Signaling, American Association for the Advancement of Science, 1200 New York Avenue, N.W., Washington, DC 20005, USA
| |
Collapse
|
40
|
Ongusaha PP, Qi HH, Raj L, Kim YB, Aaronson SA, Davis RJ, Shi Y, Liao JK, Lee SW. Identification of ROCK1 as an upstream activator of the JIP-3 to JNK signaling axis in response to UVB damage. Sci Signal 2008; 1:ra14. [PMID: 19036714 PMCID: PMC2649725 DOI: 10.1126/scisignal.1161938] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Although apoptosis triggered by ultraviolet B (UVB)-mediated activation of the c-Jun N-terminal kinase (JNK) pathway is mediated by both intrinsic and extrinsic pathways, the mechanism of initiation of JNK activation remains obscure. Here, we report the characterization of the JNK-interacting protein 3 (JIP-3) scaffolding protein as an interacting partner of Rho-associated kinase 1 (ROCK1), as determined by tandem affinity protein purification. Upon UVB-induced stress in keratinocytes, ROCK1 was activated, bound to JIP-3, and activated the JNK pathway. Moreover, phosphorylation of JIP-3 by ROCK1 was crucial for the recruitment of JNK. Inhibition of the activity of ROCK1 in keratinocytes resulted in decreased activation of the JNK pathway and thus a reduction in apoptosis. ROCK1(+/-) mice exhibited decreased UVB-mediated activation of JNK and apoptosis relative to wild-type mice. Our findings present a new molecular mechanism by which ROCK1 functions as a UVB sensor that regulates apoptosis, an important event in the prevention of skin cancer.
Collapse
Affiliation(s)
- Pat P Ongusaha
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|