1
|
Qin Z, Li Y, Shao X, Li K, Bai Y, Wang B, Ma F, Shi W, Song L, Zhuang A, He F, Ding C, Yang W. HNF4A functions as a hepatocellular carcinoma oncogene or tumor suppressor depending upon the AMPK pathway activity status. Cancer Lett 2025; 623:217732. [PMID: 40254090 DOI: 10.1016/j.canlet.2025.217732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Cancer cells frequently undergo energy metabolic stress induced by the increased dynamics of nutrient supply. Hepatocyte nuclear factor 4A (HNF4A) is a master transcription factor (TF) in hepatocytes that regulates metabolism and differentiation. However, the mechanism underlying how HNF4A functions in cancer progression remains unclear due to conflicting results observed in numerous studies. To address the roles of HNF4A in hepatocellular carcinoma (HCC), we investigated the regulatory functions of HNF4A in HCC cells under different glucose supply conditions. We found that HNF4A exhibited tumor-suppressive effects on the proliferation and migration of HCC cells in glucose-sufficient conditions and tumor-promotive effects on HCC cells in glucose-insufficient conditions. Further investigation revealed that this diverse function of HNF4A was dependent upon the AMPK pathway activity. Similarly, the prognosis predicted by HNF4A was also correlated with whether the AMPKa expression levels were low or high in clinical HCC patients. Multiomics approaches consisting of proteomics and ChIP-seq revealed that key HNF4A target genes, including NEDD4 and RPS6KA2, are involved in the diverse function of HNF4A in HCC in response to the AMPK activity status. Specifically, HNF4A could bind to the promoter region of NEDD4 and RPS6KA2, and upregulating their expression. Our study has demonstrated the relationship between and synergism of AMPK and HNF4A in the progression of HCC under diverse nutrient conditions.
Collapse
Affiliation(s)
- Zhaoyu Qin
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Yan Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Xiexiang Shao
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Kai Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Yihe Bai
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Bing Wang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Fahan Ma
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Wenhao Shi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lei Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Aojia Zhuang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Fuchu He
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Chen Ding
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
2
|
Moon S, Ito Y. A simplified in vitro disease-mimicking culture system can determine the angiogenic effect of medicines on vascular diseases. Cytotechnology 2025; 77:75. [PMID: 40062227 PMCID: PMC11889311 DOI: 10.1007/s10616-025-00736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/27/2025] [Indexed: 03/21/2025] Open
Abstract
Many patients undergoing clinical regenerative treatments experience severe conditions arising from endothelial disruption. In chronic cardiac and perivascular diseases, deficiencies in vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF), and heparin, which are essential for maintaining and activating endothelial cells, can lead to angiogenic dysregulation. Endothelial disruption caused by ischemic hypoxia and a deficiency in these factors is associated with many vascular diseases. However, their pathogenic processes remain unclear at the cellular level. Therefore, the present study aimed to develop a culture system that mimics the disease environment to test the effectiveness of drug candidates in restoring damaged blood vessels in chronic vascular diseases, including coronary artery disease and peripheral vascular disease. This study focused on VEGF, IGF, and heparin and developed a pseudo-disease culture system by pre-treating human umbilical vein endothelial cells (HUVECs) with a starvation medium (EGM-2™ medium lacking VEGF, IGF, and heparin) to examine the ability of HUVECs to form a traditional 2D vascular network. The results indicated that a deficiency in these proteins results in disruptions in tube morphogenesis. Moreover, the results suggested that dysregulation of the PI3K/AKT pathway plays a key role for in vascular disruption in HUVECs. The proposed pseudo-disease starvation system provides a simple way to visualize pathological disruptions to blood vessels and assess the efficacy of drugs for vascular regeneration. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-025-00736-4.
Collapse
Affiliation(s)
- SongHo Moon
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
| | - Yuzuru Ito
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki Japan
- Life Science Development Department, CHIYODA Corporation, Yokohama, Kanagawa Japan
- National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki Japan
| |
Collapse
|
3
|
Feng Q, Yu C, Guo L, Liu X, Lin Y, Li C, Zhang W, Zong Y, Yang W, Ma Y, Wang R, Li L, Pei Y, Wang H, Liu D, Niu H, Han M, Nie L. DCBLD1 Modulates Angiogenesis by Regulation of the VEGFR-2 Endocytosis in Endothelial Cells. Arterioscler Thromb Vasc Biol 2025; 45:198-217. [PMID: 39665138 DOI: 10.1161/atvbaha.123.320443] [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: 11/17/2023] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND Unwanted angiogenesis is involved in the progression of various malignant tumors and cardiovascular diseases, and the factors that regulate angiogenesis are potential therapeutic targets. We tested the hypothesis that DCBLD1 (discoidin, CUB, and LCCL domain-containing protein 1) is a coreceptor of VEGFR-2 (vascular endothelial growth factor receptor-2) and modulates angiogenesis in endothelial cells. METHODS A carotid artery ligation model and retinal angiogenesis assay were used to study angiogenesis using globe knockout or endothelial cell-specific conditional Dcbld1 knockout mice in vivo. Immunoblotting, immunofluorescence staining, plasma membrane subfraction isolation, Coimmunoprecipitation, and mass spectrum assay were performed to clarify the molecular mechanisms. RESULTS Loss of Dcbld1 impaired VEGF (vascular endothelial growth factor) response and inhibited VEGF-induced endothelial cell proliferation and migration. Dcbld1 deletion interfered with adult and developmental angiogenesis. Mechanistically, DCBLD1 bound to VEGFR-2 and regulated the formation of VEGFR-2 complex with negative regulators: protein tyrosine phosphatases, E3 ubiquitin ligases (neuronal precursor cell-expressed developmentally downregulated gene 4, Nedd4 and c-Casitas B-lineage lymphoma, c-Cbl), and also Dcbld1 knockdown promoted lysosome-mediated VEGFR-2 degradation in endothelial cells. CONCLUSION These findings demonstrated the essential role of endothelial DCBLD1 in regulating VEGF signaling and provided evidence that DCBLD1 promotes VEGF-induced angiogenesis by limiting the dephosphorylation, ubiquitination, and lysosome degradation after VEGFR-2 endocytosis. We proposed that endothelial DCBLD1 is a potential therapeutic target for ischemic cardiovascular diseases by the modulation of angiogenesis through regulation of the VEGFR-2 endocytosis.
Collapse
Affiliation(s)
- Qi Feng
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Chao Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Lingling Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Xiaoning Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Yanling Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Chenyang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Wenjun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Yanhong Zong
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Weiwei Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Yuehua Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Runtao Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Lijing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Yunli Pei
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Huifang Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Demin Liu
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China (D.L.)
| | - Honglin Niu
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
- School of Nursing, Hebei Medical University, Shijiazhuang, Hebei Province, China (H.N.)
| | - Mei Han
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| | - Lei Nie
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Key Laboratory of Vascular Biology in Hebei Province, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., M.H., L.N.)
- Cardiovascular Medical Science Center, Hebei Medical University, Shijiazhuang, Hebei Province, China (Q.F., C.Y., L.G., X.L., Y.L., C.L., W.Z., Y.Z., W.Y., Y.M., R.W., L.L., Y.P., H.W., D.L., H.N., M.H., L.N.)
| |
Collapse
|
4
|
Wu Z, Ye Q, Zhang S, Hu L, Wang X, Yao L, Zhu L, Xiao S, Duan Z, Zhang X, Jiang S, Zhang Z, Liu D, Li D, Yang X. Vitamin K-dependent gamma-carboxyglutamic acid protein 1 promotes pancreatic ductal adenocarcinoma progression through stabilizing oncoprotein KRAS and tyrosine kinase receptor EGFR. Clin Transl Med 2025; 15:e70191. [PMID: 39843398 PMCID: PMC11753899 DOI: 10.1002/ctm2.70191] [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: 09/10/2024] [Revised: 12/13/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Vitamin K-dependent γ-glutamic acid carboxylation (Gla) proteins are calcium-binding and membrane-associated, participating in coagulation, bone turnover, and cancer biology. The molecular function of transmembrane proline-rich Gla proteins (PRRGs) remains unexplored. METHODS Analysis of pancreatic ductal adenocarcinoma (PDAC) datasets, including transcription profiles, clinical data, and tissue microarrays, was conducted to evaluate PRRG1 expression and its clinical relevance. PDAC cell lines with overexpressed, knockdown, and mutated PRRG1 were developed to study biological functions and pathways using RNA-seq, co-immunoprecipitation with mass spectrometry, Western blotting, and immunofluorescence. In vivo xenograft and orthotopic models assessed PRRG1's impact on PDAC progression, with and without warfarin treatment. RESULTS PRRG1 was significantly upregulated in PDAC compared to normal pancreas, correlating with poorer patient survival. PRRG1 knockdown reduced PDAC cell proliferation, anchorage-independent growth in vitro, and tumor growth in vivo. PRRG1 localized at the plasma membrane, interacted with the HECT E3 ligase NEDD4 via the C-terminal PPXY motif, and promoted NEDD4 self-ubiquitination, reducing its protein levels. PRRG1 knockdown elevated NEDD4, destabilizing the oncoprotein KRAS and receptor EGFR, and attenuating downstream signaling and macropinocytosis under nutrient deprivation. The vitamin K-dependent Gla modification of PRRG1 was crucial for its membrane localization and pro-tumorigenic effects, and was inhibited by low-dose warfarin, a clinical vitamin K antagonist. CONCLUSIONS This study identifies PRRG1 as a key regulator of pro-tumorigenic signaling in PDAC, suggesting the potential of repurposing the anticoagulant warfarin as a therapeutic strategy. KEY POINTS PRRG1 is identified as the transmembrane Gla protein mediating PDAC malignancy. PRRG1 recruits and induces self-ubiquitination of membrane-anchoring E3 ligase NEDD4. PRRG1 exerts a protective role toward KRAS and EGFR by inhibiting NEDD4. The anticoagulant warfarin can be utilized to inhibit PRRG1 and PDAC advancement.
Collapse
Affiliation(s)
- Zheng Wu
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Qing Ye
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Shan Zhang
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Li‐Peng Hu
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Xiao‐Qi Wang
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Lin‐Li Yao
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Lei Zhu
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Shu‐Yu Xiao
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Zong‐Hao Duan
- Department of Biliary‐Pancreatic SurgeryRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Xue‐Li Zhang
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Shu‐Heng Jiang
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Zhi‐Gang Zhang
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - De‐Jun Liu
- Department of Biliary‐Pancreatic SurgeryRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Dong‐Xue Li
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| | - Xiao‐Mei Yang
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiP.R. China
| |
Collapse
|
5
|
Gao JJ, Wu FY, Liu YJ, Li L, Lin YJ, Kang YT, Peng YM, Liu YF, Wang C, Ma ZS, Cao Y, Cao HY, Mo ZW, Li Y, Ou JS, Ou ZJ. Increase of PCSK9 expression in diabetes promotes VEGFR2 ubiquitination to inhibit endothelial function and skin wound healing. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2635-2649. [PMID: 39153050 DOI: 10.1007/s11427-023-2688-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 07/15/2024] [Indexed: 08/19/2024]
Abstract
Diabetic foot ulcers (DFUs) are a serious vascular disease. Currently, no effective methods are available for treating DFUs. Pro-protein convertase subtilisin/kexin type 9 (PCSK9) regulates lipid levels to promote atherosclerosis. However, the role of PCSK9 in DFUs remains unclear. In this study, we found that the expression of PCSK9 in endothelial cells (ECs) increased significantly under high glucose (HG) stimulation and in diabetic plasma and vessels. Specifically, PCSK9 promotes the E3 ubiquitin-protein ligase NEDD4 binding to vascular endothelial growth factor receptor 2 (VEGFR2), which led to the ubiquitination of VEGFR2, resulting in its degradation and downregulation in ECs. Furthermore, PCSK9 suppresses the expression and activation of AKT, endothelial nitric oxide synthase (eNOS), and ERK1/2, leading to decreased nitric oxide (NO) production and increased superoxide anion (O2._) generation, which impairs vascular endothelial function and angiogenesis. Importantly, using evolocumab to limit the increase in PCSK9 expression blocked the HG-induced inhibition of NO production and the increase in O2._ production, as well as inhibited the phosphorylation and expression of AKT, eNOS, and ERK1/2. Moreover, evolocumab improved vascular endothelial function and angiogenesis, and promoted wound healing in diabetes. Our findings suggest that targeting PCSK9 is a novel therapeutic approach for treating DFUs.
Collapse
Affiliation(s)
- Jian-Jun Gao
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Fang-Yuan Wu
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
- Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yu-Jia Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Le Li
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yi-Jun Lin
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yue-Ting Kang
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
- Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yi-Fang Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Chen Wang
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Zhen-Sheng Ma
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Yang Cao
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Hong-Yu Cao
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
| | - Zhi-Wei Mo
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yan Li
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China.
| | - Jing-Song Ou
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, 510080, China.
- Division of Hypertension and Vascular Diseases, Department of Cardiology, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
6
|
Rotin D, Prag G. Physiological Functions of the Ubiquitin Ligases Nedd4-1 and Nedd4-2. Physiology (Bethesda) 2024; 39:18-29. [PMID: 37962894 DOI: 10.1152/physiol.00023.2023] [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: 10/05/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/15/2023] Open
Abstract
The Nedd4 family of E3 ubiquitin ligases, consisting of a C2-WW(n)-HECT domain architecture, includes the closely related Nedd4/Nedd4-1 and Nedd4L/Nedd4-2, which play critical roles in human physiology and pathophysiology.This review focuses on the regulation of enzymatic activity of these Nedd4 proteins, as well as on their roles in regulating stability and function of membrane and other signaling proteins, such as ion channels, ion transporters, and growth factor receptors. The diseases caused by impairment of such regulation are discussed, as well as opportunities and challenges for targeting these enzymes for therapy.
Collapse
Affiliation(s)
- Daniela Rotin
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Biochemistry Department, University of Toronto, Ontario, Canada
| | - Gali Prag
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Israel
- Sagol School of Neuroscience, Tel Aviv University, Israel
| |
Collapse
|
7
|
Critchley WR, Smith GA, Zachary IC, Harrison MA, Ponnambalam S. The E2 ubiquitin-conjugating enzymes UBE2D1 and UBE2D2 regulate VEGFR2 dynamics and endothelial function. J Cell Sci 2023; 136:jcs260657. [PMID: 37226882 PMCID: PMC10234107 DOI: 10.1242/jcs.260657] [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: 09/26/2022] [Accepted: 04/05/2023] [Indexed: 05/10/2023] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2, encoded by KDR) regulates endothelial function and angiogenesis. VEGFR2 undergoes ubiquitination that programs this receptor for trafficking and proteolysis, but the ubiquitin-modifying enzymes involved are ill-defined. Herein, we used a reverse genetics screen for the human E2 family of ubiquitin-conjugating enzymes to identify gene products that regulate VEGFR2 ubiquitination and proteolysis. We found that depletion of either UBE2D1 or UBE2D2 in endothelial cells caused a rise in steady-state VEGFR2 levels. This rise in plasma membrane VEGFR2 levels impacted on VEGF-A-stimulated signalling, with increased activation of canonical MAPK, phospholipase Cγ1 and Akt pathways. Analysis of biosynthetic VEGFR2 is consistent with a role for UBE2D enzymes in influencing plasma membrane VEGFR2 levels. Cell-surface-specific biotinylation and recycling studies showed an increase in VEGFR2 recycling to the plasma membrane upon reduction in UBE2D levels. Depletion of either UBE2D1 or UBE2D2 stimulated endothelial tubulogenesis, which is consistent with increased VEGFR2 plasma membrane levels promoting the cellular response to exogenous VEGF-A. Our studies identify a key role for UBE2D1 and UBE2D2 in regulating VEGFR2 function in angiogenesis.
Collapse
Affiliation(s)
- William R. Critchley
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Gina A. Smith
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Ian C. Zachary
- Centre for Cardiovascular Biology & Medicine, Rayne Building, University College London, London WC1E 6JF, UK
| | | | - Sreenivasan Ponnambalam
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
8
|
Yang Y, Yao HJ, Lin WJ, Huang SC, Li XD, He FZ. Real role of growth factor receptor-binding protein 10: Linking lipid metabolism to diabetes cardiovascular complications. World J Clin Cases 2022; 10:12875-12879. [PMID: 36569013 PMCID: PMC9782935 DOI: 10.12998/wjcc.v10.i35.12875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/13/2022] [Accepted: 11/17/2022] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular complications of patients with type 2 diabetes mellitus (T2DM) threaten the health and life of numerous individuals. Recently, growth factor receptor-binding protein 10 (GRB10) was found to play a pivotal role in vascular complications of T2DM, which participates in the regulation of lipid metabolism of T2DM patients. The genetic variation of GRB10 rs1800504 is closely related to the risk of coronary heart disease in patients with T2DM. The development of GRB10 as a key mediator in the association of lipid metabolism with cardiovascular complications in T2DM is detailed in and may provide new potential concerns for the study of cardiovascular complications in T2DM patients.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, Guangdong Province, China
| | - Hua-Jie Yao
- College of Life Science and Technology, Wuhan Polytechnic University, Wuhan 430000, Hubei Province, China
| | - Wei-Jie Lin
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, Guangdong Province, China
| | - Si-Chao Huang
- Department of Pharmacy, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, Guangdong Province, China
| | - Xiao-Dong Li
- Department of Quality Control, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, Guangdong Province, China
| | - Fa-Zhong He
- Department of Quality Control, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, Guangdong Province, China
| |
Collapse
|
9
|
Repova K, Stanko P, Baka T, Krajcirovicova K, Aziriova S, Hrenak J, Barta A, Zorad S, Reiter RJ, Adamcova M, Simko F. Lactacystin-induced kidney fibrosis: Protection by melatonin and captopril. Front Pharmacol 2022; 13:978337. [PMID: 36176443 PMCID: PMC9513205 DOI: 10.3389/fphar.2022.978337] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Lactacystin is a specific proteasome inhibitor that blocks the hydrolysis of intracellular proteins by ubiquitin/proteasome system inhibition. The administration of lactacystin to rats induced hypertension and remodeling of the left ventricle and aorta. This study tested whether lactacystin induces structural and fibrotic rebuilding of the kidneys and whether melatonin and captopril can prevent these potential changes. Six weeks of lactacystin administration to rats increased their average systolic blood pressure (SBP). In the kidneys, lactacystin reduced glomerular density, increased the glomerular tuft area, and enhanced hydroxyproline concentrations. It also elevated the intraglomerular proportion including the amounts of collagen (Col) I and Col III. Lactacystin also raised the tubulointerstitial amounts of Col I and the sum of Col I and Col III with no effect on vascular/perivascular collagen. Six weeks of captopril treatment reduced SBP, while melatonin had no effect. Both melatonin and captopril increased glomerular density, reduced the glomerular tuft area, and lowered the hydroxyproline concentration in the kidneys. Both drugs reduced the proportion and total amounts of intraglomerular and tubulointerstitial Col I and Col III. We conclude that chronic lactacystin treatment stimulated structural and fibrotic remodeling of the kidneys, and melatonin and captopril partly prevented these alterations. Considering the effect of lactacystin on both the heart and kidneys, chronic treatment with this drug may be a prospective model of cardiorenal damage suitable for testing pharmacological drugs as protective agents.
Collapse
Affiliation(s)
- Kristina Repova
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Peter Stanko
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Tomas Baka
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Kristina Krajcirovicova
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Silvia Aziriova
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | | | - Andrej Barta
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Stefan Zorad
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, United States
| | - Michaela Adamcova
- Department of Physiology, Faculty of Medicine in Hradec Kralove, Charles University, Hradec Kralove, Czechia
| | - Fedor Simko
- Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
- 3rd Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- *Correspondence: Fedor Simko,
| |
Collapse
|
10
|
Saikia Q, Reeve H, Alzahrani A, Critchley WR, Zeqiraj E, Divan A, Harrison MA, Ponnambalam S. VEGFR endocytosis: Implications for angiogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 194:109-139. [PMID: 36631189 DOI: 10.1016/bs.pmbts.2022.06.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The binding of vascular endothelial growth factor (VEGF) superfamily to VEGF receptor tyrosine kinases (VEGFRs) and co-receptors regulates vasculogenesis, angiogenesis and lymphangiogenesis. A recurring theme is that dysfunction in VEGF signaling promotes pathological angiogenesis, an important feature of cancer and pro-inflammatory disease states. Endocytosis of basal (resting) or activated VEGFRs facilitates signal attenuation and endothelial quiescence. However, increasing evidence suggest that activated VEGFRs can continue to signal from intracellular compartments such as endosomes. In this chapter, we focus on the evolving link between VEGFR endocytosis, signaling and turnover and the implications for angiogenesis. There is much interest in how such understanding of VEGFR dynamics can be harnessed therapeutically for a wide range of human disease states.
Collapse
Affiliation(s)
- Queen Saikia
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Hannah Reeve
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Areej Alzahrani
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - William R Critchley
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Elton Zeqiraj
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Aysha Divan
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Michael A Harrison
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | | |
Collapse
|
11
|
The Role of NEDD4 E3 Ubiquitin–Protein Ligases in Parkinson’s Disease. Genes (Basel) 2022; 13:genes13030513. [PMID: 35328067 PMCID: PMC8950476 DOI: 10.3390/genes13030513] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 01/25/2023] Open
Abstract
Parkinson’s disease (PD) is a debilitating neurodegenerative disease that causes a great clinical burden. However, its exact molecular pathologies are not fully understood. Whilst there are a number of avenues for research into slowing, halting, or reversing PD, one central idea is to enhance the clearance of the proposed aetiological protein, oligomeric α-synuclein. Oligomeric α-synuclein is the main constituent protein in Lewy bodies and neurites and is considered neurotoxic. Multiple E3 ubiquitin-protein ligases, including the NEDD4 (neural precursor cell expressed developmentally downregulated protein 4) family, parkin, SIAH (mammalian homologues of Drosophila seven in absentia), CHIP (carboxy-terminus of Hsc70 interacting protein), and SCFFXBL5 SCF ubiquitin ligase assembled by the S-phase kinase-associated protein (SKP1), cullin-1 (Cul1), a zinc-binding RING finger protein, and the F-box domain/Leucine-rich repeat protein 5-containing protein FBXL5), have been shown to be able to ubiquitinate α-synuclein, influencing its subsequent degradation via the proteasome or lysosome. Here, we explore the link between NEDD4 ligases and PD, which is not only via α-synuclein but further strengthened by several additional substrates and interaction partners. Some members of the NEDD4 family of ligases are thought to crosstalk even with PD-related genes and proteins found to be mutated in familial forms of PD. Mutations in NEDD4 family genes have not been observed in PD patients, most likely because of their essential survival function during development. Following further in vivo studies, it has been thought that NEDD4 ligases may be viable therapeutic targets in PD. NEDD4 family members could clear toxic proteins, enhancing cell survival and slowing disease progression, or might diminish beneficial proteins, reducing cell survival and accelerating disease progression. Here, we review studies to date on the expression and function of NEDD4 ubiquitin ligases in the brain and their possible impact on PD pathology.
Collapse
|
12
|
βIV-spectrin as a stalk cell-intrinsic regulator of VEGF signaling. Nat Commun 2022; 13:1326. [PMID: 35288568 PMCID: PMC8921520 DOI: 10.1038/s41467-022-28933-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 02/14/2022] [Indexed: 11/25/2022] Open
Abstract
Defective angiogenesis underlies over 50 malignant, ischemic and inflammatory disorders yet long-term therapeutic applications inevitably fail, thus highlighting the need for greater understanding of the vast crosstalk and compensatory mechanisms. Based on proteomic profiling of angiogenic endothelial components, here we report βIV-spectrin, a non-erythrocytic cytoskeletal protein, as a critical regulator of sprouting angiogenesis. Early loss of endothelial-specific βIV-spectrin promotes embryonic lethality in mice due to hypervascularization and hemorrhagic defects whereas neonatal depletion yields higher vascular density and tip cell populations in developing retina. During sprouting, βIV-spectrin expresses in stalk cells to inhibit their tip cell potential by enhancing VEGFR2 turnover in a manner independent of most cell-fate determining mechanisms. Rather, βIV-spectrin recruits CaMKII to the plasma membrane to directly phosphorylate VEGFR2 at Ser984, a previously undefined phosphoregulatory site that strongly induces VEGFR2 internalization and degradation. These findings support a distinct spectrin-based mechanism of tip-stalk cell specification during vascular development. Defective angiogenesis remains a high source of morbidity in multiple disorders. Here they show that βIV-spectrin, a membrane-associated cytoskeletal protein, is essential for regulation of endothelial tip cell populations and VEGF signaling during sprouting angiogenesis.
Collapse
|
13
|
Shi Y, Li X, Yang J. Cx43 upregulation in HUVECs under stretch via TGF-β1 and cytoskeletal network. Open Med (Wars) 2022; 17:463-474. [PMID: 35350835 PMCID: PMC8919824 DOI: 10.1515/med-2022-0432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Many physiological and pathophysiological processes in cells or tissues are involved in mechanical stretch, which induces the gap junction gene expression and cytokine TGF beta changes. However, the underlying mechanisms of the gap junction gene expression remain unknown. Here, we showed that the mRNA and protein levels of Cx43 in human umbilical vein endothelial cells (HUVECs) were significantly increased after 24 h stretch stimulation, and TGF beta1 (not TGF beta2) expression was also upregulated. Administration of TGF beta1 into HUVECs without stretch also induced upregulation of Cx43 expression. However, SB431542, a specific inhibitor of the TGF beta1 receptor, blocked the Cx43 protein upregulation caused by TGF beta1. Further, the increase of Cx43 protein expression under the stretch condition was partially blocked by SB431542; it was also partially blocked by simultaneous administration of anti-TGF beta1 monoclonal neutralization antibody. Importantly, the upregulation of Cx43 induced by stretch was blocked by the administration of actin and microtubule inhibitors, while NEDD4, a key element in mediating Cx43 protein ubiquitination and degradation, was not changed under the stretch condition. In conclusion, upregulation of Cx43 expression under the 24 h stretch condition is mediated via TGF beta1 receptor signaling pathway, and it also involves the actin and microtubule cytoskeletal network.
Collapse
Affiliation(s)
- Yumeng Shi
- Department of Ophthalmology and Visual Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai 200031, China
| | - Xinbo Li
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Oregon, USA
| | - Jin Yang
- Department of Ophthalmology and Visual Science, Eye Ear Nose and Throat Hospital of Fudan University, Shanghai 200031, China
| |
Collapse
|
14
|
The NEDD4 ubiquitin E3 ligase: a snapshot view of its functional activity and regulation. Biochem Soc Trans 2022; 50:473-485. [PMID: 35129615 DOI: 10.1042/bst20210731] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/17/2022]
Abstract
Due to its fundamental role in all eukaryotic cells, a deeper understanding of the molecular mechanisms underlying ubiquitination is of central importance. Being responsible for chain specificity and substrate recognition, E3 ligases are the selective elements of the ubiquitination process. In this review, we discuss different cellular pathways regulated by one of the first identified E3 ligase, NEDD4, focusing on its pathophysiological role, its known targets and modulators. In addition, we highlight small molecule inhibitors that act on NEDD4 and discuss new strategies to effectively target this E3 enzyme.
Collapse
|
15
|
Yang Y, Qiu W, Meng Q, Liu M, Lin W, Yang H, Wang R, Dong J, Yuan N, Zhou Z, He F. GRB10 rs1800504 Polymorphism Is Associated With the Risk of Coronary Heart Disease in Patients With Type 2 Diabetes Mellitus. Front Cardiovasc Med 2021; 8:728976. [PMID: 34651026 PMCID: PMC8505721 DOI: 10.3389/fcvm.2021.728976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic vascular complications are one of the main causes of death and disability. Previous studies have reported that genetic variation is associated with diabetic vascular complications. In this study, we aimed to investigate the association between GRB10 polymorphisms and susceptibility to type 2 diabetes mellitus (T2DM) vascular complications. Eight single nucleotide polymorphisms (SNPs) in the GRB10 gene were genotyped by MassARRAY system and 934 patients with type 2 diabetes mellitus (T2DM) were included for investigation. We found that GRB10 rs1800504 CC+CT genotypes were significantly associated with increased risk of coronary heart disease (CHD) compared with TT genotype (OR = 2.24; 95%CI: 1.36-3.70, p = 0.002). Consistently, levels of cholesterol (CHOL) (CC+CT vs. TT, 4.44 ± 1.25 vs. 4.10 ± 1.00 mmol/L; p = 0.009) and low density lipoprotein cholesterin (LDL-CH) (CC+CT vs. TT, 2.81 ± 1.07 vs. 2.53 ± 0.82 mmol/L; p = 0.01) in T2DM patients with TT genotype were significant lower than those of CC+CT genotypes. We further validated in MIHA cell that the total cholesterol (TC) level in GRB10-Mut was significantly reduced compared with GRB10-WT; p = 0.0005. Likewise, the reversed palmitic acid (PA) induced lipid droplet formation in GRB10-Mut was more effective than in GRB10-WT. These results suggest that rs1800504 of GRB10 variant may be associated with the blood lipids and then may also related to the risk of CHD in patients with T2DM.
Collapse
Affiliation(s)
- Yang Yang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Wentao Qiu
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China.,College of Pharmacy, Jinan University, Guangzhou, China
| | - Qian Meng
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Mouze Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Weijie Lin
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Haikui Yang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Ruiqi Wang
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Jiamei Dong
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Ningning Yuan
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Zhiling Zhou
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| | - Fazhong He
- Department of Pharmacy, Zhuhai People's Hospital (Zhuhai Hospital Affiliated With Jinan University), Zhuhai, China
| |
Collapse
|
16
|
Zhang J, Yuan HK, Chen S, Zhang ZR. Detrimental or beneficial: Role of endothelial ENaC in vascular function. J Cell Physiol 2021; 237:29-48. [PMID: 34279047 DOI: 10.1002/jcp.30505] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/19/2022]
Abstract
In the past, it was believed that the expression of the epithelial sodium channel (ENaC) was restricted to epithelial tissues, such as the distal nephron, airway, sweat glands, and colon, where it is critical for sodium homeostasis. Over the past two decades, this paradigm has shifted due to the finding that ENaC is also expressed in various nonepithelial tissues, notably in vascular endothelial cells. In this review, the recent findings of the expression, regulation, and function of the endothelial ENaC (EnNaC) are discussed. The expression of EnNaC subunits is reported in a variety of endothelial cell lines and vasculatures, but this is controversial across different species and vessels and is not a universal finding in all vascular beds. The expression density of EnNaC is very faint compared to ENaC in the epithelium. To date, little is known about the regulatory mechanism of EnNaC. Through it can be regulated by aldosterone, the detailed downstream signaling remains elusive. EnNaC responds to increased extracellular sodium with the feedforward activation mechanism, which is quite different from the Na+ self-inhibition mechanism of ENaC. Functionally, EnNaC was shown to be a determinant of cellular mechanics and vascular tone as it can sense shear stress, and its activation or insertion into plasma membrane causes endothelial stiffness and reduced nitric oxide production. However, in some blood vessels, EnNaC is essential for maintaining the integrity of endothelial barrier function. In this context, we discuss the possible reasons for the distinct role of EnNaC in vasculatures.
Collapse
Affiliation(s)
- Jun Zhang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Kai Yuan
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Chen
- Department of Biopharmaceutical Sciences, School of Pharmacy, Harbin Medical University (Daqing), Daqing, China
| | - Zhi-Ren Zhang
- Departments of Pharmacy and Cardiology, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder & Cancer Related Cardiovascular Diseases, NHC Key Laboratory of Cell Transplantation, Harbin Medical University & Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China
| |
Collapse
|
17
|
Wang H, Wang L, Pan H, Wang Y, Shi M, Yu H, Wang C, Pan X, Chen Z. Exosomes Derived From Macrophages Enhance Aerobic Glycolysis and Chemoresistance in Lung Cancer by Stabilizing c-Myc via the Inhibition of NEDD4L. Front Cell Dev Biol 2021; 8:620603. [PMID: 33748098 PMCID: PMC7969980 DOI: 10.3389/fcell.2020.620603] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/29/2020] [Indexed: 01/20/2023] Open
Abstract
As one of the most common and lethal cancer, lung cancer severely threatens the health of human. It has been reported that tumor-associated macrophages promote initiation, progression, as well as chemoresistance in human cancers. However, the underneath molecular mechanism that drives chemoresistance in lung cancer is yet not fully characterized. In this article, we demonstrated that M2 macrophage-derived exosomes (MDE) is the key factor to promote cisplatin-resistance in lung cancer. MDE exhibited high expression level of several miRNA including miR-3679-5p. Mechanistically, miR-3679-5p was delivered to lung cancer cells by MDE, downregulating the expression of a known E3 ligase, NEDD4L, which has been identified as a key regulator controlling the stability of c-Myc. Such decreased NEDD4L expression level resulted in the stabilization of c-Myc and elevated glycolysis. The enhanced glycolysis drives the chemoresistance in lung cancer. Taken together, our findings not only show that M2 macrophage induce chemoresistance in lung cancer through MDE mediated miR-3679-5R/NEDD4L/c-Myc signaling cascade, but also shed the light on the mechanism of the cross-talk between M2 macrophage and lung cancers. By pinpointing a potential novel survival signaling pathway, our data could provide a new potential therapeutic target for lung cancer treatment and management.
Collapse
Affiliation(s)
- Huan Wang
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| | - Lie Wang
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| | - Haiyan Pan
- Department of Internal Medicine, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| | - Yaona Wang
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| | - Miao Shi
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| | - Hang Yu
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| | - Chaoye Wang
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| | - Xinfu Pan
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| | - Zhijun Chen
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Zhejiang University, Zhoushan, China
| |
Collapse
|
18
|
Hao J, Ci X, Wang Y, Choi SYC, Sullivan SE, Xue H, Wu R, Dong X, Haegert AM, Collins CC, Lin D, Wang Y. GRB10 sustains AR activity by interacting with PP2A in prostate cancer cells. Int J Cancer 2020; 148:469-480. [PMID: 33038264 DOI: 10.1002/ijc.33335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/06/2020] [Accepted: 09/30/2020] [Indexed: 01/22/2023]
Abstract
Prostate cancer (PCa) progression is driven by androgen receptor (AR) signaling. Unfortunately, androgen-deprivation therapy and the use of even more potent AR pathway inhibitors (ARPIs) cannot bring about a cure. ARPI resistance (ie, castration-resistant PCa, CRPC) will inevitably develop. Previously, we demonstrated that GRB10 is an AR transcriptionally repressed gene that functionally contributes to CRPC development and ARPI resistance. GRB10 expression is elevated prior to CRPC development in our patient-derived xenograft models and is significantly upregulated in clinical CRPC samples. Here, we analyzed transcriptomic data from GRB10 knockdown in PCa cells and found that AR signaling is downregulated. While the mRNA expression of AR target genes decreased upon GRB10 knockdown, AR expression was not affected at the mRNA or protein level. We further found that phosphorylation of AR serine 81 (S81), which is critical for AR transcriptional activity, is decreased by GRB10 knockdown and increased by its overexpression. Luciferase assay using GRB10-knockdown cells also indicate reduced AR activity. Immunoprecipitation coupled with mass spectrometry revealed an interaction between GRB10 and the PP2A complex, which is a known phosphatase of AR. Further validations and analyses showed that GRB10 binds to the PP2Ac catalytic subunit with its PH domain. Mechanistically, GRB10 knockdown increased PP2Ac protein stability, which in turn decreased AR S81 phosphorylation and reduced AR activity. Our findings indicate a reciprocal feedback between GRB10 and AR signaling, implying the importance of GRB10 in PCa progression.
Collapse
Affiliation(s)
- Jun Hao
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Xinpei Ci
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Yong Wang
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Stephen Yiu Chuen Choi
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Sarah E Sullivan
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Kinesiology and Physical Education, McGill University, Quebec, Canada
| | - Hui Xue
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Rebecca Wu
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Xin Dong
- Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Anne M Haegert
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colin C Collins
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dong Lin
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Experimental Therapeutics, BC Cancer Agency, Vancouver, British Columbia, Canada
| |
Collapse
|
19
|
VEGF/VEGFR-2 system exerts neuroprotection against Phoneutria nigriventer spider envenomation through PI3K-AKT-dependent pathway. Toxicon 2020; 185:76-90. [PMID: 32649934 DOI: 10.1016/j.toxicon.2020.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 01/19/2023]
Abstract
This study was undertaken to elucidate why VEGF/VEGFR-2 is elevated in the hippocampus of rats injected with Phoneutria nigriventer spider venom (PNV). PNV delays Na+ channels inactivation; blocks Ca2+ and K+ channels, increases glutamate release, causes blood-brain breakdown (BBBb), brain edema and severe excitotoxicity. Analytical FT-IR spectroscopy showed profound alteration in molecular biochemical state, with evidences for VEGFR-2 (KDR/Flk-1) signaling mediation. By blocking VEGF/VEGFR-2 binding via pre-treatment with itraconazole we demonstrated that animals' condition was deteriorated soon at 1-2 h post-PNV exposure concurrently with decreased expression of VEGF, BBB-associated proteins, ZO-1, β-catenin, laminin, P-gp (P-glycoprotein), Neu-N (neuron's viability marker) and MAPKphosphorylated-p38, while phosphorylated-ERK and Src pathways were increased. At 5 h and coinciding with incipient signs of animals' recuperation, the proteins associated with protection (HIF-1α, VEGF, VEGFR-1, VEGFR-2, Neu-N, occludin, β-catenin, laminin, P-gp efflux protein, phosphorylated-p38) increased thus indicating p38 pathway activation together with paracellular route strengthening. However, the BBB transcellular trafficking and caspase-3 increased (pro-apoptotic pathway activation). At 24 h, the transcellular route reestablished physiological state but the pro-survival pathway PI3K/(p-Akt) dropped in animals underwent VEGF/VEGFR-2 binding inhibition, whereas it was significantly activated at matched interval in PNV group without prior itraconazole; these results demonstrate impaired VEGF' survival effects at 24 h. The inhibition of VEGF/VEGFR-2 binding identified 5 h as turning point at which multi-level dynamic interplay was elicited to reverse hippocampal damage. Collectively, the data confirmed VEGFR-2 signaling via serine-threonine kinase Akt as neuroprotective pathway against PNV-induced damage. Further studies are needed to elucidate mechanisms underlying PNV effects.
Collapse
|
20
|
Wang ZW, Hu X, Ye M, Lin M, Chu M, Shen X. NEDD4 E3 ligase: Functions and mechanism in human cancer. Semin Cancer Biol 2020; 67:92-101. [PMID: 32171886 DOI: 10.1016/j.semcancer.2020.03.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/03/2020] [Accepted: 03/07/2020] [Indexed: 12/11/2022]
Abstract
A growing amount of evidence indicates that the neuronally expressed developmentally downregulated 4 (NEDD4, also known as NEDD4-1) E3 ligase plays a critical role in a variety of cellular processes via the ubiquitination-mediated degradation of multiple substrates. The abnormal regulation of NEDD4 protein has been implicated in cancer development and progression. In this review article, we briefly delineate the downstream substrates and upstream regulators of NEDD4, which are involved in carcinogenesis. Moreover, we succinctly elucidate the functions of NEDD4 protein in tumorigenesis and progression, including cell proliferation, apoptosis, cell cycle, migration, invasion, epithelial mesenchymal transition (EMT), cancer stem cells, and drug resistance. The findings regarding NEDD4 functions are further supported by knockout mouse models and human tumor tissue studies. This review could provide a promising and optimum anticancer therapeutic strategy via targeting the NEDD4 protein.
Collapse
Affiliation(s)
- Zhi-Wei Wang
- Center of Scientific Research, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Xiaoli Hu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Miaomiao Ye
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Min Lin
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Man Chu
- Center of Scientific Research, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Xian Shen
- Department of Gastrointestinal Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China.
| |
Collapse
|
21
|
Yamak A, Hu D, Mittal N, Buikema JW, Ditta S, Lutz PG, Moog-Lutz C, Ellinor PT, Domian IJ. Loss of Asb2 Impairs Cardiomyocyte Differentiation and Leads to Congenital Double Outlet Right Ventricle. iScience 2020; 23:100959. [PMID: 32179481 PMCID: PMC7078385 DOI: 10.1016/j.isci.2020.100959] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/17/2019] [Accepted: 02/26/2020] [Indexed: 11/21/2022] Open
Abstract
Defining the pathways that control cardiac development facilitates understanding the pathogenesis of congenital heart disease. Herein, we identify enrichment of a Cullin5 Ub ligase key subunit, Asb2, in myocardial progenitors and differentiated cardiomyocytes. Using two conditional murine knockouts, Nkx+/Cre.Asb2fl/fl and AHF-Cre.Asb2fl/fl, and tissue clarifying technique, we reveal Asb2 requirement for embryonic survival and complete heart looping. Deletion of Asb2 results in upregulation of its target Filamin A (Flna), and concurrent Flna deletion partially rescues embryonic lethality. Conditional AHF-Cre.Asb2 knockouts harboring one Flna allele have double outlet right ventricle (DORV), which is rescued by biallelic Flna excision. Transcriptomic and immunofluorescence analyses identify Tgfβ/Smad as downstream targets of Asb2/Flna. Finally, using CRISPR/Cas9 genome editing, we demonstrate Asb2 requirement for human cardiomyocyte differentiation suggesting a conserved mechanism between mice and humans. Collectively, our study provides deeper mechanistic understanding of the role of the ubiquitin proteasome system in cardiac development and suggests a previously unidentified murine model for DORV. Flna removal partially rescues embryonic lethality of Asb2-heart-specific knockout AHF-Asb2 knockouts harboring one Flna allele have double outlet right ventricle Asb2-Flna regulate TGFβ-Smad2 signaling in the heart Conserved role of Asb2 in heart morphogenesis between mice and humans
Collapse
Affiliation(s)
- Abir Yamak
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Dongjian Hu
- Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Nikhil Mittal
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA
| | - Jan W Buikema
- Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; University Medical Center Utrecht, 3584 CX Utrecht, Netherlands
| | - Sheraz Ditta
- Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Department of Pharmaceutical Sciences, Utrecht University, 3512 JE Utrecht, Netherlands
| | - Pierre G Lutz
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Christel Moog-Lutz
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Patrick T Ellinor
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ibrahim J Domian
- Harvard Medical School, Boston, MA 02115, USA; Cardiovascular Research Center, Massachusetts General Hospital, 185 Cambridge Street, CPZN3200, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| |
Collapse
|
22
|
Novellasdemunt L, Kucharska A, Jamieson C, Prange‐Barczynska M, Baulies A, Antas P, van der Vaart J, Gehart H, Maurice MM, Li VSW. NEDD4 and NEDD4L regulate Wnt signalling and intestinal stem cell priming by degrading LGR5 receptor. EMBO J 2020; 39:e102771. [PMID: 31867777 PMCID: PMC6996568 DOI: 10.15252/embj.2019102771] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/12/2022] Open
Abstract
The intestinal stem cell (ISC) marker LGR5 is a receptor for R-spondin (RSPO) that functions to potentiate Wnt signalling in the proliferating crypt. It has been recently shown that Wnt plays a priming role for ISC self-renewal by inducing RSPO receptor LGR5 expression. Despite its pivotal role in homeostasis, regeneration and cancer, little is known about the post-translational regulation of LGR5. Here, we show that the HECT-domain E3 ligases NEDD4 and NEDD4L are expressed in the crypt stem cell regions and regulate ISC priming by degrading LGR receptors. Loss of Nedd4 and Nedd4l enhances ISC proliferation, increases sensitivity to RSPO stimulation and accelerates tumour development in Apcmin mice with increased numbers of high-grade adenomas. Mechanistically, we find that both NEDD4 and NEDD4L negatively regulate Wnt/β-catenin signalling by targeting LGR5 receptor and DVL2 for proteasomal and lysosomal degradation. Our findings unveil the previously unreported post-translational control of LGR receptors via NEDD4/NEDD4L to regulate ISC priming. Inactivation of NEDD4 and NEDD4L increases Wnt activation and ISC numbers, which subsequently enhances tumour predisposition and progression.
Collapse
Affiliation(s)
| | - Anna Kucharska
- Stem Cell and Cancer Biology LaboratoryThe Francis Crick InstituteLondonUK
| | - Cara Jamieson
- Oncode Institute and Department of Cell BiologyCenter for Molecular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Anna Baulies
- Stem Cell and Cancer Biology LaboratoryThe Francis Crick InstituteLondonUK
| | - Pedro Antas
- Stem Cell and Cancer Biology LaboratoryThe Francis Crick InstituteLondonUK
| | - Jelte van der Vaart
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) UtrechtUtrechtThe Netherlands
| | - Helmuth Gehart
- Hubrecht InstituteRoyal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Centre (UMC) UtrechtUtrechtThe Netherlands
| | - Madelon M Maurice
- Oncode Institute and Department of Cell BiologyCenter for Molecular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Vivian SW Li
- Stem Cell and Cancer Biology LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
23
|
Wang Y, Hoeppner LH, Angom RS, Wang E, Dutta S, Doeppler HR, Wang F, Shen T, Scarisbrick IA, Guha S, Storz P, Bhattacharya R, Mukhopadhyay D. Protein kinase D up-regulates transcription of VEGF receptor-2 in endothelial cells by suppressing nuclear localization of the transcription factor AP2β. J Biol Chem 2019; 294:15759-15767. [PMID: 31492751 PMCID: PMC6816101 DOI: 10.1074/jbc.ra119.010152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/19/2019] [Indexed: 01/29/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF) signals primarily through its cognate receptor VEGF receptor-2 (VEGFR-2) to control vasculogenesis and angiogenesis, key physiological processes in cardiovascular disease and cancer. In human umbilical vein endothelial cells (HUVECs), knockdown of protein kinase D-1 (PKD1) or PKD2 down-regulates VEGFR-2 expression and inhibits VEGF-induced cell proliferation and migration. However, how PKD regulates VEGF signaling is unclear. Previous bioinformatics analyses have identified binding sites for the transcription factor activating enhancer-binding protein 2 (AP2) in the VEGFR-2 promoter. Using ChIP analyses, here we found that PKD knockdown in HUVECs increases binding of AP2β to the VEGFR-2 promoter. Luciferase reporter assays with serial deletions of AP2-binding sites within the VEGFR-2 promoter revealed that its transcriptional activity negatively correlates with the number of these sites. Next we demonstrated that AP2β up-regulation decreases VEGFR-2 expression and that loss of AP2β enhances VEGFR-2 expression in HUVECs. In vivo experiments confirmed increased VEGFR-2 immunostaining in the spinal cord of AP2β knockout mouse embryos. Mechanistically, we observed that PKD phosphorylates AP2β at Ser258 and Ser277 and suppresses its nuclear accumulation. Inhibition of PKD activity with a pan-PKD inhibitor increased AP2β nuclear localization, and overexpression of both WT and constitutively active PKD1 or PKD2 reduced AP2β nuclear localization through a Ser258- and Ser277-dependent mechanism. Furthermore, substitution of Ser277 in AP2β increased its binding to the VEGFR-2 promoter. Our findings uncover evidence of a molecular pathway that regulates VEGFR-2 expression, insights that may shed light on the etiology of diseases associated with aberrant VEGF/VEGFR signaling.
Collapse
Affiliation(s)
- Ying Wang
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
| | - Luke H Hoeppner
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Rochester, Minnesota 55905
| | - Ramcharan Singh Angom
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
| | - Shamit Dutta
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
| | - Heike R Doeppler
- Department of Cancer Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
| | - Fei Wang
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
- Department of Neurosurgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010050, China
| | - Tao Shen
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
- Department of Colorectal Surgery, Third Affiliated Hospital of Kunming Medical University, Kunming 650221, China
| | - Isobel A Scarisbrick
- Department of Physical Medicine and Rehabilitation, College of Medicine and Science, Mayo Clinic, Rochester, Minnesota 55905
| | - Sushovan Guha
- University of Arizona College of Medicine, Phoenix, Arizona 85004
| | - Peter Storz
- Department of Cancer Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
| | - Resham Bhattacharya
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Rochester, Minnesota 55905
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, College of Medicine and Science, Mayo Clinic, Jacksonville, Florida 32224
| |
Collapse
|
24
|
Xu J, Sheng Z, Li F, Wang S, Yuan Y, Wang M, Yu Z. NEDD4 protects vascular endothelial cells against Angiotensin II-induced cell death via enhancement of XPO1-mediated nuclear export. Exp Cell Res 2019; 383:111505. [PMID: 31326389 DOI: 10.1016/j.yexcr.2019.111505] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 07/14/2019] [Accepted: 07/18/2019] [Indexed: 01/11/2023]
Abstract
NEDD4 is an E3 ubiquitin ligase containing the HECT domain, which regulates various cellular processes, but its role in vascular endothelial cells is unknown. In the present study, we found that NEDD4 bound directly to XPO1 by co-immunoprecipitation screening. In HUVECs (human umbilical vein endothelial cells), overexpression of NEDD4 reduced Ang II-induced ROS level and cell apoptosis. Ang II stimulation led to nuclear accumulation of cargoes, while overexpression of NEDD4 enhanced the XPO1-dependent nuclear export of its cargoes. KPT185, an inhibitor of XPO1, can abolished the protective effect of NEDD4 under Ang II treatment. In addition, NEDD4 could promote the interaction between XPO1 and RanBP3 via K63-linked ubiquitination of XPO1. These results suggested that NEDD4 played a protective role in vascular endothelial cell injury through regulating XPO1-mediated nuclear export.
Collapse
Affiliation(s)
- Jianning Xu
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zhiyong Sheng
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Fuxin Li
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Shu Wang
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Ying Yuan
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Meng Wang
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zhihong Yu
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
25
|
Schacher NM, Raaz-Schrauder D, Pasutto F, Stumpfe FM, Tauchi M, Dietel B, Achenbach S, Urschel K. Impact of single nucleotide polymorphisms in the VEGFR2 gene on endothelial cell activation under non‑uniform shear stress. Int J Mol Med 2019; 44:1366-1376. [PMID: 31432097 PMCID: PMC6713417 DOI: 10.3892/ijmm.2019.4301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/26/2019] [Indexed: 12/20/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) in vascular endothelial growth factor receptor 2 (VEGFR2) are associated with coronary artery disease, hypertension and myocardial infarction. However, their association with atherosclerosis remains to be fully elucidated. The purpose of the present study was to determine whether SNPs are involved in atherogenesis, by analyzing their impact on human umbilical vein endothelial cells (HUVECs) under laminar and non‑uniform shear stress in a well‑established in vitro model that simulates shear stress‑induced proatherogenic processes at vessel bifurcations. All experiments were performed using freshly isolated HUVECs. Three SNPs in the VEGFR2 gene (rs1870377 T>A, rs2071559 A>G and rs2305948 C>T) were genotyped and the expression levels of VEGFR2 were semi‑quantitatively determined using western blotting. Subsequently, the HUVECs were seeded in bifurcating flow‑through cell culture slides and flow (9.6 ml/min) was applied for 19 h, including tumor necrosis factor‑α stimulation during the final 2 h of flow. The protein expression levels of VCAM‑1, E‑selectin and VEGFR2 and the adhesion of THP‑1 cells were analyzed in laminar and non‑uniform shear stress regions. Data were analyzed for associations with the respective SNPs. The total expression of VEGFR2 was significantly lower under non‑uniform shear stress than under laminar shear stress conditions, independent of the genotype. The expression of VEGFR2 between the different shear stress patterns was not significantly altered by the different SNPs. The expression levels of VCAM‑1 and E‑selectin were lower in the A/A genotype compared with those in other genotypes in rs1870377 T>A and rs2071559 A>G. In conclusion, the results suggested that SNPs within the VEGFR2 gene have a significant impact on shear stress‑related endothelial activation.
Collapse
Affiliation(s)
- Nora M Schacher
- Department of Medicine 2‑Cardiology and Angiology, Erlangen University Hospital, Friedrich‑Alexander University Erlangen‑Nürnberg, D‑91054 Erlangen, Germany
| | - Dorette Raaz-Schrauder
- Department of Medicine 2‑Cardiology and Angiology, Erlangen University Hospital, Friedrich‑Alexander University Erlangen‑Nürnberg, D‑91054 Erlangen, Germany
| | - Francesca Pasutto
- Institute of Human Genetics, Friedrich‑Alexander University Erlangen‑Nürnberg, D‑91051 Erlangen, Germany
| | - Florian M Stumpfe
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen‑EMN, Friedrich‑Alexander University Erlangen‑Nürnberg, D‑91054 Erlangen, Germany
| | - Miyuki Tauchi
- Department of Medicine 2‑Cardiology and Angiology, Erlangen University Hospital, Friedrich‑Alexander University Erlangen‑Nürnberg, D‑91054 Erlangen, Germany
| | - Barbara Dietel
- Department of Medicine 2‑Cardiology and Angiology, Erlangen University Hospital, Friedrich‑Alexander University Erlangen‑Nürnberg, D‑91054 Erlangen, Germany
| | - Stephan Achenbach
- Department of Medicine 2‑Cardiology and Angiology, Erlangen University Hospital, Friedrich‑Alexander University Erlangen‑Nürnberg, D‑91054 Erlangen, Germany
| | - Katharina Urschel
- Department of Medicine 2‑Cardiology and Angiology, Erlangen University Hospital, Friedrich‑Alexander University Erlangen‑Nürnberg, D‑91054 Erlangen, Germany
| |
Collapse
|
26
|
Smith JR, David LL, Appukuttan B, Wilmarth PA. Angiogenic and Immunologic Proteins Identified by Deep Proteomic Profiling of Human Retinal and Choroidal Vascular Endothelial Cells: Potential Targets for New Biologic Drugs. Am J Ophthalmol 2018; 193:197-229. [PMID: 29559410 PMCID: PMC6109601 DOI: 10.1016/j.ajo.2018.03.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 03/06/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE Diseases that involve retinal or choroidal vascular endothelial cells are leading causes of vision loss: age-related macular degeneration, retinal ischemic vasculopathies, and noninfectious posterior uveitis. Proteins differentially expressed by these endothelial cell populations are potential drug targets. We used deep proteomic profiling to define the molecular phenotype of human retinal and choroidal endothelial cells at the protein level. METHODS Retinal and choroidal vascular endothelial cells were separately isolated from 5 human eye pairs by selection on CD31. Total protein was extracted and digested, and peptide fractions were analyzed by reverse-phase liquid chromatography tandem mass spectrometry. Peptide sequences were assigned to fragment ion spectra, and proteins were inferred from openly accessible protein databases. Protein abundance was determined by spectral counting. Publicly available software packages were used to identify proteins that were differentially expressed between human retinal and choroidal endothelial cells, and to classify proteins that were highly abundant in each endothelial cell population. RESULTS Human retinal and/or choroidal vascular endothelial cells expressed 5042 nonredundant proteins. Setting the differential expression false discovery rate at 0.05, 498 proteins of 3454 quantifiable proteins (14.4%) with minimum mean spectral counts of 2.5 were differentially abundant in the 2 cell populations. Retinal and choroidal endothelial cells were enriched in angiogenic proteins, and retinal endothelial cells were also enriched in immunologic proteins. CONCLUSIONS This work describes the different protein expression profiles of human retinal and choroidal vascular endothelial cells, and provides multiple candidates for further study as novel treatments or drug targets for posterior eye diseases. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
Collapse
Affiliation(s)
- Justine R Smith
- Flinders University, Adelaide, Australia; Oregon Health & Science University, Portland, Oregon, USA.
| | - Larry L David
- Flinders University, Adelaide, Australia; Oregon Health & Science University, Portland, Oregon, USA
| | - Binoy Appukuttan
- Flinders University, Adelaide, Australia; Oregon Health & Science University, Portland, Oregon, USA
| | - Phillip A Wilmarth
- Flinders University, Adelaide, Australia; Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
27
|
Luo L, Jiang W, Liu H, Bu J, Tang P, Du C, Xu Z, Luo H, Liu B, Xiao B, Zhou Z, Liu F. De-silencing Grb10 contributes to acute ER stress-induced steatosis in mouse liver. J Mol Endocrinol 2018; 60:285-297. [PMID: 29555819 DOI: 10.1530/jme-18-0018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022]
Abstract
The growth factor receptor bound protein GRB10 is an imprinted gene product and a key negative regulator of the insulin, IGF1 and mTORC1 signaling pathways. GRB10 is highly expressed in mouse fetal liver but almost completely silenced in adult mice, suggesting a potential detrimental role of this protein in adult liver function. Here we show that the Grb10 gene could be reactivated in adult mouse liver by acute endoplasmic reticulum stress (ER stress) such as tunicamycin or a short-term high-fat diet (HFD) challenge, concurrently with increased unfolded protein response (UPR) and hepatosteatosis. Lipogenic gene expression and acute ER stress-induced hepatosteatosis were significantly suppressed in the liver of the liver-specific GRB10 knockout mice, uncovering a key role of Grb10 reactivation in acute ER stress-induced hepatic lipid dysregulation. Mechanically, acute ER stress induces Grb10 reactivation via an ATF4-mediated increase in Grb10 gene transcription. Our study demonstrates for the first time that the silenced Grb10 gene can be reactivated by acute ER stress and its reactivation plays an important role in the early development of hepatic steatosis.
Collapse
Affiliation(s)
- Liping Luo
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wanxiang Jiang
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Liu
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jicheng Bu
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ping Tang
- The State Key Laboratory of BiotherapyWest China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chongyangzi Du
- The State Key Laboratory of BiotherapyWest China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhipeng Xu
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hairong Luo
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bilian Liu
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bo Xiao
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- The State Key Laboratory of BiotherapyWest China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Liu
- Department of Metabolism and Endocrinology and the Metabolic Syndrome Research Center of Central South UniversityThe Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of PharmacologyUniversity of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
28
|
Critchley WR, Pellet-Many C, Ringham-Terry B, Harrison MA, Zachary IC, Ponnambalam S. Receptor Tyrosine Kinase Ubiquitination and De-Ubiquitination in Signal Transduction and Receptor Trafficking. Cells 2018; 7:E22. [PMID: 29543760 PMCID: PMC5870354 DOI: 10.3390/cells7030022] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/13/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) are membrane-based sensors that enable rapid communication between cells and their environment. Evidence is now emerging that interdependent regulatory mechanisms, such as membrane trafficking, ubiquitination, proteolysis and gene expression, have substantial effects on RTK signal transduction and cellular responses. Different RTKs exhibit both basal and ligand-stimulated ubiquitination, linked to trafficking through different intracellular compartments including the secretory pathway, plasma membrane, endosomes and lysosomes. The ubiquitin ligase superfamily comprising the E1, E2 and E3 enzymes are increasingly implicated in this post-translational modification by adding mono- and polyubiquitin tags to RTKs. Conversely, removal of these ubiquitin tags by proteases called de-ubiquitinases (DUBs) enables RTK recycling for another round of ligand sensing and signal transduction. The endocytosis of basal and activated RTKs from the plasma membrane is closely linked to controlled proteolysis after trafficking and delivery to late endosomes and lysosomes. Proteolytic RTK fragments can also have the capacity to move to compartments such as the nucleus and regulate gene expression. Such mechanistic diversity now provides new opportunities for modulating RTK-regulated cellular responses in health and disease states.
Collapse
Affiliation(s)
- William R Critchley
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Caroline Pellet-Many
- Centre for Cardiovascular Biology & Medicine, Rayne Building, University College London, London WC1E 6PT, UK.
| | - Benjamin Ringham-Terry
- Centre for Cardiovascular Biology & Medicine, Rayne Building, University College London, London WC1E 6PT, UK.
| | | | - Ian C Zachary
- Centre for Cardiovascular Biology & Medicine, Rayne Building, University College London, London WC1E 6PT, UK.
| | - Sreenivasan Ponnambalam
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
29
|
Smith GA, Fearnley GW, Abdul-Zani I, Wheatcroft SB, Tomlinson DC, Harrison MA, Ponnambalam S. Ubiquitination of basal VEGFR2 regulates signal transduction and endothelial function. Biol Open 2017; 6:1404-1415. [PMID: 28798148 PMCID: PMC5665470 DOI: 10.1242/bio.027896] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/04/2017] [Indexed: 01/19/2023] Open
Abstract
Cell surface receptors can undergo recycling or proteolysis but the cellular decision-making events that sort between these pathways remain poorly defined. Vascular endothelial growth factor A (VEGF-A) and vascular endothelial growth factor receptor 2 (VEGFR2) regulate signal transduction and angiogenesis, but how signaling and proteolysis is regulated is not well understood. Here, we provide evidence that a pathway requiring the E1 ubiquitin-activating enzyme UBA1 controls basal VEGFR2 levels, hence metering plasma membrane receptor availability for the VEGF-A-regulated endothelial cell response. VEGFR2 undergoes VEGF-A-independent constitutive degradation via a UBA1-dependent ubiquitin-linked pathway. Depletion of UBA1 increased VEGFR2 recycling from endosome-to-plasma membrane and decreased proteolysis. Increased membrane receptor availability after UBA1 depletion elevated VEGF-A-stimulated activation of key signaling enzymes such as PLCγ1 and ERK1/2. Although UBA1 depletion caused an overall decrease in endothelial cell proliferation, surviving cells showed greater VEGF-A-stimulated responses such as cell migration and tubulogenesis. Our study now suggests that a ubiquitin-linked pathway regulates the balance between receptor recycling and degradation which in turn impacts on the intensity and duration of VEGF-A-stimulated signal transduction and the endothelial response.
Collapse
Affiliation(s)
- Gina A Smith
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Gareth W Fearnley
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Izma Abdul-Zani
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular & Metabolic Medicine, Faculty of Medicine & Health, University of Leeds, Leeds LS2 9JT, UK
| | - Darren C Tomlinson
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | | | - Sreenivasan Ponnambalam
- Endothelial Cell Biology Unit, School of Molecular & Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
30
|
Yamana S, Tokiyama A, Fujita H, Terao Y, Horibe S, Sasaki N, Satomi-Kobayashi S, Hirata KI, Rikitake Y. Necl-4 enhances the PLCγ–c-Raf–MEK–ERK pathway without affecting internalization of VEGFR2. Biochem Biophys Res Commun 2017; 490:169-175. [DOI: 10.1016/j.bbrc.2017.05.185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 05/29/2017] [Indexed: 02/03/2023]
|
31
|
Yu B, Liu Z, Fu Y, Wang Y, Zhang L, Cai Z, Yu F, Wang X, Zhou J, Kong W. CYLD Deubiquitinates Nicotinamide Adenine Dinucleotide Phosphate Oxidase 4 Contributing to Adventitial Remodeling. Arterioscler Thromb Vasc Biol 2017; 37:1698-1709. [PMID: 28751569 DOI: 10.1161/atvbaha.117.309859] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Transdifferentiation of adventitial fibroblasts (AFs) into myofibroblasts plays a critical role during the vascular remodeling that occurs during atherosclerosis, restenosis, and aortic aneurysm. The ubiquitination/deubiquitination regulatory system is essential for the quality control of proteins. The involvement of ubiquitination/deubiquitination during AF transdifferentiation remains largely unknown. In this study, we determined the role of cylindromatosis (CYLD), a deubiquitinase, in the process of AF differentiation and activation in vitro and in vivo. APPROACH AND RESULTS Transforming growth factor-β1 and homocysteine, 2 known inducers of AF transdifferentiation, greatly upregulated CYLD expression in a time- and dose-dependent manner. The silencing of CYLD significantly inhibited AF transdifferentiation and activation as evidenced by the expression of contractile proteins, the production of the proinflammatory cytokines MCP-1 (monocyte chemotactic protein 1) and IL-6 (interleukin-6), the deposition of extracellular matrix, and cell migration. We further asked whether CYLD mediates AF activation via the regulation of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) as it is an essential factor during AF transdifferentiation. Indeed, the silencing of CYLD repressed transforming growth factor-β1-induced and homocysteine-induced Nox4 upregulation and reactive oxygen species production, whereas Nox4 overexpression greatly rescued the inhibitory effect on AF activation by CYLD silencing. Most interestingly, transforming growth factor-β1 and homocysteine repressed Nox4 ubiquitination and prolonged the half-life of Nox4. Moreover, Nox4 was deubiquitinated via a direct interaction with the ubiquitin-specific protease domain of CYLD. In accordance, hyperhomocysteinemia significantly increased adventitial CYLD and Nox4 expression, promoted AF transdifferentiation, and aggravated CaPO4-induced abdominal aortic aneurysm in mice. These effects were abolished in CYLD-/- mice. CONCLUSIONS CYLD contributes to the transdifferentiation of AFs via deubiquitinating Nox4 and may play a role in vascular remodeling.
Collapse
Affiliation(s)
- Bing Yu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Ziyi Liu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Yi Fu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Yingbao Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Lu Zhang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Zeyu Cai
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Fang Yu
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Xian Wang
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.)
| | - Jun Zhou
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.).
| | - Wei Kong
- From the Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (B.Y., Z.L., Y.F., Y.W., L.Z., Z.C., F.Y., X.W., W.K.); and State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, P. R. China (J.Z.).
| |
Collapse
|
32
|
Thrombospondins: A Role in Cardiovascular Disease. Int J Mol Sci 2017; 18:ijms18071540. [PMID: 28714932 PMCID: PMC5536028 DOI: 10.3390/ijms18071540] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 07/05/2017] [Accepted: 07/13/2017] [Indexed: 12/16/2022] Open
Abstract
Thrombospondins (TSPs) represent extracellular matrix (ECM) proteins belonging to the TSP family that comprises five members. All TSPs have a complex multidomain structure that permits the interaction with various partners including other ECM proteins, cytokines, receptors, growth factors, etc. Among TSPs, TSP1, TSP2, and TSP4 are the most studied and functionally tested. TSP1 possesses anti-angiogenic activity and is able to activate transforming growth factor (TGF)-β, a potent profibrotic and anti-inflammatory factor. Both TSP2 and TSP4 are implicated in the control of ECM composition in hypertrophic hearts. TSP1, TSP2, and TSP4 also influence cardiac remodeling by affecting collagen production, activity of matrix metalloproteinases and TGF-β signaling, myofibroblast differentiation, cardiomyocyte apoptosis, and stretch-mediated enhancement of myocardial contraction. The development and evaluation of TSP-deficient animal models provided an option to assess the contribution of TSPs to cardiovascular pathology such as (myocardial infarction) MI, cardiac hypertrophy, heart failure, atherosclerosis, and aortic valve stenosis. Targeting of TSPs has a significant therapeutic value for treatment of cardiovascular disease. The activation of cardiac TSP signaling in stress and pressure overload may be therefore beneficial.
Collapse
|
33
|
Wang L, Zhu B, Wang S, Wu Y, Zhan W, Xie S, Shi H, Yu R. Regulation of glioma migration and invasion via modification of Rap2a activity by the ubiquitin ligase Nedd4-1. Oncol Rep 2017; 37:2565-2574. [PMID: 28405688 PMCID: PMC5428538 DOI: 10.3892/or.2017.5572] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 10/18/2016] [Indexed: 12/26/2022] Open
Abstract
Νeuronal precursor cell expressed and developmentally downregulated protein (Nedd4-1) is an E3 ubiquitin ligase with critical roles in the pathogenesis of cancer. Herein, we demonstrated that Nedd4-1 protein was upregulated in glioma tissues vs. that in non-cancerous tissues by western blotting and immunohistochemistry. Scratch migration and Transwell chamber assays indicated that downregulation of Nedd4-1 significantly reduced the migration and invasion of the glioma cell lines U251 and U87. Conversely, overexpression of Nedd4-1 obviously enhanced the migratory and invasive capacities in both cell lines. To investigate the role of Nedd4-1 and the intracellular pathways involved, we performed pull-down and co-immunoprecipitation assays, and recognized that Nedd4-1, TNIK and Rap2a formed a complex. Moreover, Nedd4-1 selectively ubiquitinated its specific substrates, the wild-type Rap2a (WT-Rap2a) and dominant-active Rap2a (DA-Rap2a) rather than the dominant-negative Rap2a (DN-Rap2a) in the U251 cells. Subsequently, we demonstrated that Rap2a was robustly ubiquitinated by Nedd4-1 along with the K63-linked, but not the K48-linked ubiquitin chain, which significantly inhibited GTP-Rap2a activity by GST-RalGDS pull-down assay. To further verify whether the ubiquitination of Rap2a by Nedd4-1 regulated the migration and invasion of glioma cells, Nedd4-1, HA-tagged ubiquitin and its mutants as well as WT-Rap2a were co-transfected in the U251 and U87 cell lines. The results confirmed that Nedd4-1 inhibited GTP-Rap2a activity, and promoted the migration and invasion of glioma cells. In brief, our findings demonstrated the important role of Nedd4-1 in regulating the migration and invasion of glioma cells via the Nedd4-1/Rap2a pathway, which may qualify Nedd4-1 as a viable therapeutic target for glioma.
Collapse
Affiliation(s)
- Lei Wang
- Insitute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Bingxin Zhu
- Insitute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Shiquan Wang
- Insitute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Yuxuan Wu
- Insitute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Wenjian Zhan
- Insitute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Shao Xie
- Insitute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Hengliang Shi
- Insitute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| | - Rutong Yu
- Insitute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, P.R. China
| |
Collapse
|
34
|
Gilda JE, Gomes AV. Proteasome dysfunction in cardiomyopathies. J Physiol 2017; 595:4051-4071. [PMID: 28181243 DOI: 10.1113/jp273607] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) plays a critical role in removing unwanted intracellular proteins and is involved in protein quality control, signalling and cell death. Because the heart is subject to continuous metabolic and mechanical stress, the proteasome plays a particularly important role in the heart, and proteasome dysfunction has been suggested as a causative factor in cardiac dysfunction. Proteasome impairment has been detected in cardiomyopathies, heart failure, myocardial ischaemia, and hypertrophy. Proteasome inhibition is also sufficient to cause cardiac dysfunction in healthy pigs, and patients using a proteasome inhibitor for cancer therapy have a higher incidence of heart failure. In this Topical Review we discuss the experimental data which suggest UPS dysfunction is a common feature of cardiomyopathies, with an emphasis on hypertrophic cardiomyopathy caused by sarcomeric mutations. We also propose potential mechanisms by which cardiomyopathy-causing mutations may lead to proteasome impairment, such as altered calcium handling and increased oxidative stress due to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jennifer E Gilda
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA
| | - Aldrin V Gomes
- Department of Neurobiology, Physiology, and Behaviour, University of California, Davis, CA, 95616, USA.,Department of Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| |
Collapse
|
35
|
Abstract
Newly synthesized transmembrane proteins undergo a series of steps to ensure that only the required amount of correctly folded protein is localized to the membrane. The regulation of protein quality and its abundance at the membrane are often controlled by ubiquitination, a multistep enzymatic process that results in the attachment of ubiquitin, or chains of ubiquitin to the target protein. Protein ubiquitination acts as a signal for sorting, trafficking, and the removal of membrane proteins via endocytosis, a process through which multiple ubiquitin ligases are known to specifically regulate the functions of a number of ion channels, transporters, and signaling receptors. Endocytic removal of these proteins through ubiquitin-dependent endocytosis provides a way to rapidly downregulate the physiological outcomes, and defects in such controls are directly linked to human pathologies. Recent evidence suggests that ubiquitination is also involved in the shedding of membranes and associated proteins as extracellular vesicles, thereby not only controlling the cell surface levels of some membrane proteins, but also their potential transport to neighboring cells. In this review, we summarize the mechanisms and functions of ubiquitination of membrane proteins and provide specific examples of ubiquitin-dependent regulation of membrane proteins.
Collapse
Affiliation(s)
- Natalie Foot
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Tanya Henshall
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| |
Collapse
|
36
|
Hrgovic I, Doll M, Pinter A, Kaufmann R, Kippenberger S, Meissner M. Histone deacetylase inhibitors interfere with angiogenesis by decreasing endothelial VEGFR-2 protein half-life in part via a VE-cadherin-dependent mechanism. Exp Dermatol 2017; 26:194-201. [DOI: 10.1111/exd.13159] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Igor Hrgovic
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Monika Doll
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Andreas Pinter
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Roland Kaufmann
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Stefan Kippenberger
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| | - Markus Meissner
- Department of Dermatology, Venereology and Allergology; Johann Wolfgang Goethe-University; Frankfurt/Main Germany
| |
Collapse
|
37
|
Tubeimoside-1 suppresses tumor angiogenesis by stimulation of proteasomal VEGFR2 and Tie2 degradation in a non-small cell lung cancer xenograft model. Oncotarget 2017; 7:5258-72. [PMID: 26701724 PMCID: PMC4868684 DOI: 10.18632/oncotarget.6676] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/07/2015] [Indexed: 12/29/2022] Open
Abstract
Tubeimoside-1 (TBMS1) is a potent anti-tumor phytochemical. Its functional and molecular mode of action, however, remains elusive so far. Since angiogenesis is essential for tumor progression and metastasis, we herein investigated the anti-angiogenic effects of the compound. In a non-small cell lung cancer (NSCLC) xenograft model we found that treatment of CD1 nu/nu mice with TBMS1 (5 mg/kg) significantly suppressed the growth and vascularization of NCI-H460 flank tumors. Moreover, TBMS1 dose-dependently reduced vascular sprouting in a rat aortic ring assay. In vitro, TBMS1 induced endothelial cell apoptosis without decreasing the viability of NSCLC tumor cells and inhibited the migration of endothelial cells by disturbing their actin filament organization. TBMS1 further stimulated the proteasomal degradation of vascular endothelial growth factor receptor-2 (VEGFR2) and Tie2 in endothelial cells, which down-regulated AKT/mTOR signaling. These findings indicate that TBMS1 represents a novel phytochemical for anti-angiogenic treatment of cancer and other angiogenesis-related diseases.
Collapse
|
38
|
Girnita L, Takahashi SI, Crudden C, Fukushima T, Worrall C, Furuta H, Yoshihara H, Hakuno F, Girnita A. Chapter Seven - When Phosphorylation Encounters Ubiquitination: A Balanced Perspective on IGF-1R Signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:277-311. [PMID: 27378760 DOI: 10.1016/bs.pmbts.2016.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cell-surface receptors govern the critical information passage from outside to inside the cell and hence control important cellular decisions such as survival, growth, and differentiation. These receptors, structurally grouped into different families, utilize common intracellular signaling-proteins and pathways, yet promote divergent biological consequences. In rapid processing of extracellular signals to biological outcomes, posttranslational modifications offer a repertoire of protein processing options. Protein ubiquitination was originally identified as a signal for protein degradation through the proteasome system. It is now becoming increasingly recognized that both ubiquitin and ubiquitin-like proteins, all evolved from a common ubiquitin structural superfold, are used extensively by the cell and encompass signal tags for many different cellular fates. In this chapter we examine the current understanding of the ubiquitin regulation surrounding the insulin-like growth factor and insulin signaling systems, major members of the larger family of receptor tyrosine kinases (RTKs) and key regulators of fundamental physiological and pathological states.
Collapse
Affiliation(s)
- L Girnita
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
| | - S-I Takahashi
- Departments of Animal Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - C Crudden
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - T Fukushima
- Departments of Animal Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan; Department of Biological Sciences, Faculty of Bioscience and Biotechnology, Tokyo Institute of Technology, Kanagawa, Japan
| | - C Worrall
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - H Furuta
- Departments of Animal Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - H Yoshihara
- Departments of Animal Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - F Hakuno
- Departments of Animal Sciences and Applied Biological Chemistry, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - A Girnita
- Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden; Dermatology Department, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
39
|
Intestinal knockout of Nedd4 enhances growth of Apc min tumors. Oncogene 2016; 35:5839-5849. [PMID: 27086928 DOI: 10.1038/onc.2016.125] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 03/07/2016] [Accepted: 03/11/2016] [Indexed: 12/24/2022]
Abstract
Nedd4 (Nedd4-1) is an E3 ubiquitin ligase that belongs to the HECT family and comprises a C2-WW(n)-HECT domain architecture. Although it has been reported to regulate growth factor receptors and cellular signaling, its role in cancer development has been controversial, with some studies proposing that it promotes cancer while others suggest it inhibits tumor growth. Here, we tested the effect of Nedd4 on intestinal tumor formation and growth using Nedd4-knockout mice (Nedd4 floxed (fl) mice crossed to villin-Cre mice). Although we find that knockout of Nedd4 on its own does not cause tumor growth, its knockout in the context of Apc+/min-derived colorectal tumors leads to augmentation of tumor growth, suggesting that Nedd4 normally suppresses intestinal WNT signaling and growth of colonic tumors. WNT signaling microarray, immunoblotting and immunohistochemistry analyses of tumors derived from the Villin-Cre;Nedd4fl/fl;Apc+/min colons demonstrated elevated expression of the WNT upstream effectors LEF1 (full length) and YY1 in these tumors relative to control (Apc+/min alone) tumors. Together, these results suggest that Nedd4 suppresses colonic WNT signaling and tumor growth, at least in part, by suppressing the transcription factors LEF1 and YY1.
Collapse
|
40
|
Age-dependent expression of VEGFR2 in deep brain arteries in small vessel disease, CADASIL, and healthy brains. Neurobiol Aging 2016; 42:110-5. [PMID: 27143427 DOI: 10.1016/j.neurobiolaging.2016.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/16/2016] [Accepted: 03/02/2016] [Indexed: 01/07/2023]
Abstract
Vascular myocytes are central to brain aging. Small vessel disease (SVD; arteriolosclerosis) is a widespread cause of lacunar stroke and vascular dementia and is characterized by fibrosis and depletion of vascular myocytes in small penetrating arteries. Vascular endothelial growth factor (VEGF) is associated with brain aging, and Immunolabeling for vascular endothelial growth factor receptor 2 (VEGFR2) is a potent determinant of cell fate. Here, we tested whether VEGFR2 in vascular myocytes is associated with older age and SVD in human brain. Immunolabeling for VEGFR2 in deep gray matter was assessed in older people with or without moderate-severe SVD or in younger people without brain pathology or with a monogenic form of SVD (Cerebral Autosomal-Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy). All cases were without Alzheimer's disease pathology. Myocyte VEGFR2 was associated with increasing age (p = 0.0026) but not with SVD pathology or with sclerotic index or blood vessel density. We conclude that VEGFR2 is consistently expressed in small artery myocytes of older people and may mediate effects of VEGF on brain vascular aging.
Collapse
|
41
|
Smith G, Tomlinson D, Harrison M, Ponnambalam S. Chapter Eight - Ubiquitin-Mediated Regulation of Cellular Responses to Vascular Endothelial Growth Factors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:313-38. [DOI: 10.1016/bs.pmbts.2016.02.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
42
|
Mukhopadhyay A, Ravikumar G, Dwarkanath P, Meraaj H, Thomas A, Crasta J, Thomas T, Kurpad A, Sridhar T. Placental expression of the insulin receptor binding protein GRB10: Relation to human fetoplacental growth and fetal gender. Placenta 2015; 36:1225-30. [DOI: 10.1016/j.placenta.2015.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 08/13/2015] [Accepted: 09/08/2015] [Indexed: 11/27/2022]
|
43
|
The cellular response to vascular endothelial growth factors requires co-ordinated signal transduction, trafficking and proteolysis. Biosci Rep 2015; 35:BSR20150171. [PMID: 26285805 PMCID: PMC4613718 DOI: 10.1042/bsr20150171] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/18/2015] [Indexed: 01/18/2023] Open
Abstract
VEGFs (vascular endothelial growth factors) are a family of conserved disulfide-linked soluble secretory glycoproteins found in higher eukaryotes. VEGFs mediate a wide range of responses in different tissues including metabolic homoeostasis, cell proliferation, migration and tubulogenesis. Such responses are initiated by VEGF binding to soluble and membrane-bound VEGFRs (VEGF receptor tyrosine kinases) and co-receptors. VEGF and receptor splice isoform diversity further enhances complexity of membrane protein assembly and function in signal transduction pathways that control multiple cellular responses. Different signal transduction pathways are simultaneously activated by VEGFR-VEGF complexes with membrane trafficking along the endosome-lysosome network further modulating signal output from multiple enzymatic events associated with such pathways. Balancing VEGFR-VEGF signal transduction with trafficking and proteolysis is essential in controlling the intensity and duration of different intracellular signalling events. Dysfunction in VEGF-regulated signal transduction is important in chronic disease states including cancer, atherosclerosis and blindness. This family of growth factors and receptors is an important model system for understanding human disease pathology and developing new therapeutics for treating such ailments.
Collapse
|
44
|
Ye X, Wang L, Shang B, Wang Z, Wei W. NEDD4: a promising target for cancer therapy. Curr Cancer Drug Targets 2015; 14:549-56. [PMID: 25088038 DOI: 10.2174/1568009614666140725092430] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 05/11/2014] [Accepted: 07/11/2014] [Indexed: 11/22/2022]
Abstract
The Neuronally expressed developmentally downregulated 4 (NEDD4), functioning largely as an E3 ubiquitin ligase, has been demonstrated to play a critical role in the development and progression of human cancers. In this review, to understand the regulatory mechanism(s) of NEDD4 as well as the signaling pathways controlled by NEDD4, we briefly describe the NEDD4 upstream regulators and its downstream ubiquitin substrates. Moreover, we further discuss its oncogenic roles in human malignancies. Therefore, targeting NEDD4 could be a potential therapeutic strategy for treatment of human cancers.
Collapse
Affiliation(s)
| | | | | | | | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, USA.
| |
Collapse
|
45
|
Liu J, Wan L, Liu P, Inuzuka H, Liu J, Wang Z, Wei W. SCF(β-TRCP)-mediated degradation of NEDD4 inhibits tumorigenesis through modulating the PTEN/Akt signaling pathway. Oncotarget 2015; 5:1026-37. [PMID: 24657926 PMCID: PMC4011580 DOI: 10.18632/oncotarget.1675] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The HECT domain-containing ubiquitin E3 ligase NEDD4 is widely expressed in mammalian tissues and plays a crucial role in governing a wide spectrum of cellular processes including cell growth, tissue development and homeostasis. Recent reports have indicated that NEDD4 might facilitate tumorigenesis through targeted degradation of multiple tumor suppressor proteins including PTEN. However, the molecular mechanism by which NEDD4 stability is regulated has not been fully elucidated. Here we report that SCF(β-TRCP) governs NEDD4 protein stability by targeting it for ubiquitination and subsequent degradation in a Casein Kinase-I (CKI) phosphorylation-dependent manner. Specifically, depletion of β-TRCP, or inactivation of CKI, stabilized NEDD4, leading to down-regulation of its ubiquitin target PTEN and subsequent activation of the mTOR/Akt oncogenic pathway. Furthermore, we found that CKIδ-mediated phosphorylation of Ser347 and Ser348 on NEDD4 promoted its interaction with SCF(β-TRCP) for subsequent ubiquitination and degradation. As a result, compared to ectopic expression of wild-type NEDD4, introducing a non-degradable NEDD4 (S347A/S348A-NEDD4) promoted cancer cell growth and migration. Hence, our findings revealed the CKI/SCF(β-TRCP) signaling axis as the upstream negative regulator of NEDD4, and further suggested that enhancing NEDD4 degradation, presumably with CKI or SCF(β-TRCP) agonists, could be a promising strategy for treating human cancers.
Collapse
Affiliation(s)
- Jia Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Life Science, FIST, Xi'an Jiaotong University, Xi'an, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Latham AM, Kankanala J, Fishwick CWG, Ponnambalam S. Identification of Receptor Tyrosine Kinase Inhibitors Using Cell Surface Biotinylation and Affinity Isolation. Methods Mol Biol 2015; 1332:121-31. [PMID: 26285749 DOI: 10.1007/978-1-4939-2917-7_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The mammalian vascular endothelial growth factor receptor tyrosine kinases (VEGFRs) bind circulating growth factors and regulate the process of angiogenesis. The discovery of new small molecules that target the enzymatic activity of the VEGFR family as potential antiangiogenic drugs is of much commercial interest in the pharmaceutical sector. Here, we describe the use of a combined cell surface biotinylation and affinity isolation procedure to monitor ligand-stimulated VEGFR trafficking in endothelial cells, in which novel VEGFR inhibitors from chemical libraries can be identified by their ability to inhibit receptor internalization. Unlike a traditional cell-free enzyme activity assay, such a cell-based approach provides a physiologically relevant readout of inhibitor activity. In this example, we use the VEGF-A-VEGFR-2 axis and the well-characterized tyrosine kinase inhibitor sunitinib as a working model; however this technique is highly applicable for the identification of inhibitors to other receptor tyrosine kinases.
Collapse
Affiliation(s)
- Antony M Latham
- Endothelial Cell Biology Unit, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | | | | | | |
Collapse
|
47
|
Boase NA, Kumar S. NEDD4: The founding member of a family of ubiquitin-protein ligases. Gene 2014; 557:113-22. [PMID: 25527121 DOI: 10.1016/j.gene.2014.12.020] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/01/2014] [Accepted: 12/10/2014] [Indexed: 01/31/2023]
Abstract
Ubiquitination plays a crucial role in regulating proteins post-translationally. The focus of this review is on NEDD4, the founding member of the NEDD4 family of ubiquitin ligases that is evolutionarily conserved in eukaryotes. Many potential substrates of NEDD4 have been identified and NEDD4 has been shown to play a critical role in the regulation of a number of membrane receptors, endocytic machinery components and the tumour suppressor PTEN. In this review we will discuss the diverse pathways in which NEDD4 is involved, and the patho-physiological significance of this important ubiquitin ligase.
Collapse
Affiliation(s)
- Natasha Anne Boase
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5001, Australia.
| |
Collapse
|
48
|
Liu J, Shaik S, Dai X, Wu Q, Zhou X, Wang Z, Wei W. Targeting the ubiquitin pathway for cancer treatment. Biochim Biophys Acta Rev Cancer 2014; 1855:50-60. [PMID: 25481052 DOI: 10.1016/j.bbcan.2014.11.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 12/15/2022]
Abstract
Proteasome-mediated degradation is a common mechanism by which cells renew their intracellular proteins and maintain protein homeostasis. In this process, the E3 ubiquitin ligases are responsible for targeting specific substrates (proteins) for ubiquitin-mediated degradation. However, in cancer cells, the stability and the balance between oncoproteins and tumor suppressor proteins are disturbed in part due to deregulated proteasome-mediated degradation. This ultimately leads to either stabilization of oncoprotein(s) or increased degradation of tumor suppressor(s), contributing to tumorigenesis and cancer progression. Therefore, E3 ubiquitin ligases including the SCF types of ubiquitin ligases have recently evolved as promising therapeutic targets for the development of novel anti-cancer drugs. In this review, we highlighted the critical components along the ubiquitin pathway including E1, E2, various E3 enzymes and DUBs that could serve as potential drug targets and also described the available bioactive compounds that target the ubiquitin pathway to control various cancers.
Collapse
Affiliation(s)
- Jia Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shavali Shaik
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Xiangpeng Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Qiong Wu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, China
| | - Xiuxia Zhou
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou 215123, China
| | - Zhiwei Wang
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou 215123, China.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
49
|
Grb10 is involved in BCR-ABL-positive leukemia in mice. Leukemia 2014; 29:858-68. [DOI: 10.1038/leu.2014.283] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 08/14/2014] [Accepted: 09/05/2014] [Indexed: 11/08/2022]
|
50
|
Cipriani P, Di Benedetto P, Capece D, Zazzeroni F, Liakouli V, Ruscitti P, Pantano I, Berardicurti O, Carubbi F, Alesse E, Giacomelli R. Impaired Cav-1 expression in SSc mesenchymal cells upregulates VEGF signaling: a link between vascular involvement and fibrosis. FIBROGENESIS & TISSUE REPAIR 2014; 7:13. [PMID: 25237397 PMCID: PMC4166421 DOI: 10.1186/1755-1536-7-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/27/2014] [Indexed: 11/12/2022]
Abstract
BACKGROUND Systemic sclerosis (SSc) is characterized by vascular alteration and fibrosis, the former probably leading to fibrosis via the ability of both endothelial cells and pericytes to differentiate toward myofibroblast. It is well known that vascular endothelial growth factor A (VEGF-A, hereafter referred to as VEGF) may induce a profibrotic phenotype on perivascular cells. Caveolin-1 (Cav-1) is involved in the regulation of VEGF signaling, playing a role in the transport of internalized VEGF receptor 2 (VEGFR2) toward degradation, thus decreasing VEGF signaling. In this work, we assessed the levels of Cav-1 in SSc bone marrow mesenchymal stem cells (SSc-MSCs), a pericyte surrogate, and correlate these results with VEGF signaling, focusing onpotential pathogenic pathways leading to fibrosis. RESULTS WE EXPLORED THE VEGF SIGNALING ASSESSING: (1) Cav-1 expression; (2) its co-localization with VEGFR2; (3) the activity of VEGFR2, by IF, immunoprecipitation, and western blot. In SSc-MSCs, Cav-1 levels were lower when compared to healthy controls (HC)-MSCs. Furthermore, the Cav-1/VEGFR2 co-localization and the ubiquitination of VEGFR2 were impaired in SSc-MSCs, suggesting a decreased degradation of the receptor and, as a consequence, the tyrosine phosphorylation of VEGFR2 and the PI3-kinase-Akt pathways were significantly increased when compared to HC. Furthermore, an increased connective tissue growth factor (CTGF) expression was observed in SSc-MSCs. Taken together, these data suggested the upregulation of VEGF signaling in SSc-MSCs. Furthermore, after silencing Cav-1 expression in HC-MSCs, an increased CTGF expression in HC-MSCs was observed, mirroring the results obtained in SSc-MSCs, and confirming the potential role that the lack of Cav-1 may play in the persistent VEGF signaling . CONCLUSIONS During SSc, the lower levels of Cav-1 may contribute to the pathogenesis of fibrosis via an upregulation of the VEGF signaling in perivascular cells which are shifted to a profibrotic phenotype.
Collapse
Affiliation(s)
- Paola Cipriani
- Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Paola Di Benedetto
- Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Daria Capece
- Department of Applied Clinical Sciences and Biotechnology, University of L’Aquila, Coppito 2, 67100 L’Aquila, Italy
| | - Francesca Zazzeroni
- Department of Applied Clinical Sciences and Biotechnology, University of L’Aquila, Coppito 2, 67100 L’Aquila, Italy
| | - Vasiliki Liakouli
- Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Piero Ruscitti
- Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Ilenia Pantano
- Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Onorina Berardicurti
- Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Francesco Carubbi
- Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| | - Edoardo Alesse
- Department of Applied Clinical Sciences and Biotechnology, University of L’Aquila, Coppito 2, 67100 L’Aquila, Italy
| | - Roberto Giacomelli
- Department of Applied Clinical Sciences and Biotechnology, Rheumatology Unit, School of Medicine, University of L’Aquila, Delta 6 Building, Via dell’Ospedale, 67100 L’Aquila, Italy
| |
Collapse
|