1
|
Wan X, Yap J, Chen J, Li Y, Faruk R, Tan NCB, Ma Y, Lim Y, Jubri KB, Hu J, Yuan J, Zhang G, Li Q, Yap YS, Lam P, Wang M, Fu NY, Hu J. Oncogenic non-V600 mutations evade the regulatory machinery of RAF including the Cdc37/Hsp90 chaperone and the 14-3-3 scaffold. Theranostics 2025; 15:2035-2051. [PMID: 39897565 PMCID: PMC11780520 DOI: 10.7150/thno.103958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/24/2024] [Indexed: 02/04/2025] Open
Abstract
The Ser/Thr kinase RAF, particularly BRAF isoform is a dominant target of oncogenic mutations and many mutations have been identified in various cancers. However, how these mutations except V600E evade the regulatory machinery of RAF protein and hence trigger its oncogenicity remains unclear. Methods: In this study, we used mutagenesis, peptide affinity assay, immunoprecipitation, immunoblot, and complementary split luciferase assay as well as mouse xenograft tumour model to investigate how the function of RAF is cooperatively regulated by Cdc37/Hsp90 chaperones and 14-3-3 scaffolds and how this regulatory machinery is evaded by prevalent non-V600 mutations. Results: We found that Cdc37/Hsp90 chaperones engaged with mature BRAF proteins promoted together with 14-3-3 scaffolds a switch of BRAF proteins from active open dimers into inactive close monomers. Most non-V600 mutations were enriched on or around the Cdc37/Hsp90-binding segments of BRAF, which impair association of CDc37/Hsp90 chaperones with BRAF and hence trap BRAF in active open conformation favouring dimerization. These BRAF mutants with high dimer propensity sustained a prolonged ERK signaling, and were effectively targeted by RAF dimer breaker plx8394 in vitro and in vivo. In contrast, CRAF and ARAF existed as immature monomers highly packaged with Cdc37/Hsp90 chaperones, which will be released upon dimerization driven by RAS-GTP binding with their N-terminus as well as 14-3-3 scaffold association with their C-terminus. Mature CRAF and ARAF dimers also sustained a prolonged ERK signaling as non-V600 BRAF mutants by virtue of absence of the C-terminal Cdc37/Hsp90-binding segment. Conclusions: Cdc37/Hsp90 chaperones and 14-3-3 scaffolds cooperatively facilitate the switch of RAF proteins from open active dimers to close inactive monomers. Non-V600 mutations disrupt this regulatory machinery, and trap RAF in dimers, which could be targeted by RAF dimer breakers.
Collapse
Affiliation(s)
- Xiaoyu Wan
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Jiajun Yap
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
- The Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore 169857
| | - Junjun Chen
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Yifan Li
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Regina Faruk
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Nazereth Chor Boon Tan
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Yiying Ma
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Yiting Lim
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Karlina Bte Jubri
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Jingyi Hu
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Jimin Yuan
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Ge Zhang
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Quan Li
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
| | - Yoon Sim Yap
- The Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore 168583
- The Oncology Academic Clinical Programme, Duke-NUS Medical School, 8 College Road, Singapore 169857
| | - Paula Lam
- The Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore 169857
- Department of Physiology, National University of Singapore, 2 Medical Drive, Singapore 117597
- Cellvec Pte. Ltd. 100 Pasir Panjang Road, Singapore 118518
| | - Mei Wang
- The Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore 169857
| | - Nai Yang Fu
- The Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore 169857
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Jiancheng Hu
- The Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore General Hospital, 30 Hospital Boulevard, Singapore 168583
- The Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore 169857
| |
Collapse
|
2
|
He Y, Guo Y, Zheng W, Yue T, Zhang H, Wang B, Feng Z, Ouzhuluobu, Cui C, Liu K, Zhou B, Zeng X, Li L, Wang T, Wang Y, Zhang C, Xu S, Qi X, Su B. Polygenic adaptation leads to a higher reproductive fitness of native Tibetans at high altitude. Curr Biol 2023; 33:4037-4051.e5. [PMID: 37643619 DOI: 10.1016/j.cub.2023.08.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/01/2023] [Accepted: 08/04/2023] [Indexed: 08/31/2023]
Abstract
The adaptation of Tibetans to high-altitude environments has been studied extensively. However, the direct assessment of evolutionary adaptation, i.e., the reproductive fitness of Tibetans and its genetic basis, remains elusive. Here, we conduct systematic phenotyping and genome-wide association analysis of 2,252 mother-newborn pairs of indigenous Tibetans, covering 12 reproductive traits and 76 maternal physiological traits. Compared with the lowland immigrants living at high altitudes, indigenous Tibetans show better reproductive outcomes, reflected by their lower abortion rate, higher birth weight, and better fetal development. The results of genome-wide association analyses indicate a polygenic adaptation of reproduction in Tibetans, attributed to the genomic backgrounds of both the mothers and the newborns. Furthermore, the EPAS1-edited mice display higher reproductive fitness under chronic hypoxia, mirroring the situation in Tibetans. Collectively, these results shed new light on the phenotypic pattern and the genetic mechanism of human reproductive fitness in extreme environments.
Collapse
Affiliation(s)
- Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| | - Yongbo Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Wangshan Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Tian Yue
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Hui Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650000, China
| | - Bin Wang
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang Hospital, Lhasa 850000, China
| | - Zhanying Feng
- CEMS, NCMIS, MDIS, Academy of Mathematics & Systems Science, Chinese Academy of Sciences, Beijing 100080, China
| | - Ouzhuluobu
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang Hospital, Lhasa 850000, China; High Altitude Medical Research Center, School of Medicine, Tibetan University, Lhasa 850000, China
| | - Chaoying Cui
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang Hospital, Lhasa 850000, China; High Altitude Medical Research Center, School of Medicine, Tibetan University, Lhasa 850000, China
| | - Kai Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Bin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xuerui Zeng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Liya Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Tianyun Wang
- Department of Medical Genetics, Center for Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yong Wang
- CEMS, NCMIS, MDIS, Academy of Mathematics & Systems Science, Chinese Academy of Sciences, Beijing 100080, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Chao Zhang
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuhua Xu
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200438, China; Human Phenome Institute, Zhangjiang Fudan International Innovation Center, and Ministry of Education Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai 201203, China
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang Hospital, Lhasa 850000, China; State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650000, China.
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China.
| |
Collapse
|
3
|
Lauinger M, Christen D, Klar RF, Roubaty C, Heilig CE, Stumpe M, Knox JJ, Radulovich N, Tamblyn L, Xie IY, Horak P, Forschner A, Bitzer M, Wittel UA, Boerries M, Ball CR, Heining C, Glimm H, Fröhlich M, Hübschmann D, Gallinger S, Fritsch R, Fröhling S, O’Kane GM, Dengjel J, Brummer T. BRAF Δβ3-αC in-frame deletion mutants differ in their dimerization propensity, HSP90 dependence, and druggability. SCIENCE ADVANCES 2023; 9:eade7486. [PMID: 37656784 PMCID: PMC11804575 DOI: 10.1126/sciadv.ade7486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 08/02/2023] [Indexed: 09/03/2023]
Abstract
In-frame BRAF exon 12 deletions are increasingly identified in various tumor types. The resultant BRAFΔβ3-αC oncoproteins usually lack five amino acids in the β3-αC helix linker and sometimes contain de novo insertions. The dimerization status of BRAFΔβ3-αC oncoproteins, their precise pathomechanism, and their direct druggability by RAF inhibitors (RAFi) has been under debate. Here, we functionally characterize BRAFΔLNVTAP>F and two novel mutants, BRAFdelinsFS and BRAFΔLNVT>F, and compare them with other BRAFΔβ3-αC oncoproteins. We show that BRAFΔβ3-αC oncoproteins not only form stable homodimers and large multiprotein complexes but also require dimerization. Nevertheless, details matter as aromatic amino acids at the deletion junction of some BRAFΔβ3-αC oncoproteins, e.g., BRAFΔLNVTAP>F, increase their stability and dimerization propensity while conferring resistance to monomer-favoring RAFi such as dabrafenib or HSP 90/CDC37 inhibition. In contrast, dimer-favoring inhibitors such as naporafenib inhibit all BRAFΔβ3-αC mutants in cell lines and patient-derived organoids, suggesting that tumors driven by such oncoproteins are vulnerable to these compounds.
Collapse
Affiliation(s)
- Manuel Lauinger
- Institute of Molecular Medicine, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Daniel Christen
- Institute of Molecular Medicine, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Rhena F. U. Klar
- Institute of Molecular Medicine, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Freeze-O Organoid Bank, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Internal Medicine I (Hematology, Oncology, and Stem Cell Transplantation), University Hospital of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine (IBSM), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carole Roubaty
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Christoph E. Heilig
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Stumpe
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Jennifer J. Knox
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Nikolina Radulovich
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Laura Tamblyn
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Irene Y. Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Peter Horak
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Andrea Forschner
- Department of Dermatology, University Hospital of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Eberhard Karls University, Tübingen, Germany
| | - Michael Bitzer
- German Cancer Consortium (DKTK), DKFZ partner site Tübingen, Eberhard Karls University, Tübingen, Germany
- Center for Personalized Medicine Tübingen, Eberhard Karls University, Tübingen, Germany
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany
| | - Uwe A. Wittel
- Department of General and Visceral Surgery, University of Freiburg Medical Center, Faculty of Medicine, 79106 Freiburg, Germany
| | - Melanie Boerries
- German Cancer Consortium (DKTK), partner site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Institute of Medical Bioinformatics and Systems Medicine (IBSM), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Claudia R. Ball
- Department for Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden–Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Technische Universität Dresden, Faculty of Biology, Technische Universität Dresden, Dresden, Germany
| | - Christoph Heining
- Department for Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden–Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
| | - Hanno Glimm
- Department for Translational Medical Oncology, National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden–Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Fröhlich
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Hübschmann
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Pattern Recognition and Digital Medicine Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Steven Gallinger
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ralph Fritsch
- Department of Internal Medicine I (Hematology, Oncology, and Stem Cell Transplantation), University Hospital of Freiburg, Freiburg, Germany
- Department of Medical Oncology and Haematology, University Hospital of Zurich, Zurich, Switzerland
| | - Stefan Fröhling
- Division of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Grainne M. O’Kane
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Tilman Brummer
- Institute of Molecular Medicine, ZBMZ, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- German Cancer Consortium (DKTK), partner site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Freeze-O Organoid Bank, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center for Biological Signalling Studies BIOSS, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
4
|
Pérez-Baena MJ, Cordero-Pérez FJ, Pérez-Losada J, Holgado-Madruga M. The Role of GAB1 in Cancer. Cancers (Basel) 2023; 15:4179. [PMID: 37627207 PMCID: PMC10453317 DOI: 10.3390/cancers15164179] [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: 07/17/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
GRB2-associated binder 1 (GAB1) is the inaugural member of the GAB/DOS family of pleckstrin homology (PH) domain-containing proteins. Upon receiving various stimuli, GAB1 transitions from the cytoplasm to the membrane where it is phosphorylated by a range of kinases. This event recruits SH2 domain-containing proteins like SHP2, PI3K's p85 subunit, CRK, and others, thereby activating distinct signaling pathways, including MAPK, PI3K/AKT, and JNK. GAB1-deficient embryos succumb in utero, presenting with developmental abnormalities in the heart, placenta, liver, skin, limb, and diaphragm myocytes. Oncogenic mutations have been identified in the context of cancer. GAB1 expression levels are disrupted in various tumors, and elevated levels in patients often portend a worse prognosis in multiple cancer types. This review focuses on GAB1's influence on cellular transformation particularly in proliferation, evasion of apoptosis, metastasis, and angiogenesis-each of these processes being a cancer hallmark. GAB1 also modulates the resistance/sensitivity to antitumor therapies, making it a promising target for future anticancer strategies.
Collapse
Affiliation(s)
- Manuel Jesús Pérez-Baena
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (M.J.P.-B.); (J.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | | | - Jesús Pérez-Losada
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC-CIC), Universidad de Salamanca/CSIC, 37007 Salamanca, Spain; (M.J.P.-B.); (J.P.-L.)
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Marina Holgado-Madruga
- Instituto de Investigación Biosanitaria de Salamanca (IBSAL), 37007 Salamanca, Spain
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, 37007 Salamanca, Spain
- Instituto de Neurociencias de Castilla y León (INCyL), 37007 Salamanca, Spain
- Virtual Institute for Good Health and Well Being (GLADE), European Campus of City Universities (EC2U), 86073 Poitiers, France
| |
Collapse
|
5
|
He L, Zhan F, Lu L, Zhang X, Wu J. Role of necroptosis and immune infiltration in preeclampsia: novel insights from bioinformatics analyses. BMC Pregnancy Childbirth 2023; 23:495. [PMID: 37403014 DOI: 10.1186/s12884-023-05821-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 06/29/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Preeclampsia (PE) is a serious pregnancy complication that can adversely affect the mother and fetus. Necroptosis is a recently discovered new form of programmed cell death involved in the pathological process of various pregnancy complications. Our study aimed to identify the necroptosis-related differentially expressed genes (NRDEGs), create a diagnosis model and related disease subtypes model based on these genes, and further investigate their relationship with immune infiltration. METHODS In this study, we identified NRDEGs by analyzing data from various databases, including Molecular Signatures, GeneCards, and Gene Expression Omnibus (GEO). Using minor absolute shrinkage and selection operator (LASSO) and logistic Cox regression analysis, we developed a novel PE diagnosis model based on NRDEGs. Furthermore, we developed PE subtype models using consensus clustering analysis based on key gene modules screened out by weighted correlation network analysis (WGCNA). Finally, we identified the difference in immune infiltration between the PE and control groups as well as between both PE subtypes by analyzing the immune cell infiltration across combined datasets and PE datasets. RESULTS Our study discovered that the necroptosis pathway was significantly enriched and active in PE samples. We identified nine NRDEGs that involved in this pathway, including BRAF, PAWR, USP22, SYNCRIP, KRT86, MERTK, BAP1, CXCL5, and STK38. Additionally, we developed a diagnostic model based on a regression model including six NRDEGs and identified two PE subtypes: Cluster1 and Cluster2, based on key module genes. Furthermore, correlation analysis showed that the abundance of immune cell infiltration was related to necroptosis genes and PE disease subtypes. CONCLUSION According to the present study, necroptosis is a phenomenon that occurs in PE and is connected to immune cell infiltration. This result suggests that necroptosis and immune-related factors may be the underlying mechanisms of PE pathophysiology. This study opens new avenues for future research into PE's pathogenesis and treatment options.
Collapse
Affiliation(s)
- Lidan He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian, 350004, China
| | - Feng Zhan
- College of Engineering, Fujian Jiangxia University, Fuzhou, 350108, China
| | - Lin Lu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian, 350004, China
| | - Xia Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian, 350004, China
| | - Jianbo Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian, 350004, China.
| |
Collapse
|
6
|
Goyal D, Limesand SW, Goyal R. Vascular Stem Cells and the Role of B-Raf Kinase in Survival, Proliferation, and Apoptosis. Int J Mol Sci 2023; 24:7483. [PMID: 37108645 PMCID: PMC10138574 DOI: 10.3390/ijms24087483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/04/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Neovascularization is an essential process in organismal development and aging. With aging, from fetal to adult life, there is a significant reduction in neovascularization potential. However, the pathways which play a role in increased neovascularization potential during fetal life are unknown. Although several studies proposed the idea of vascular stem cells (VSCs), the identification and essential survival mechanism are still not clear. In the present study, we isolated fetal VSCs from the ovine carotid artery and identified the pathways involved in their survival. We tested the hypothesis that fetal vessels contain a population of VSCs, and that B-Raf kinase is required for their survival. We conducted viability, apoptotic, and cell cycle stage assays on fetal and adult carotid arteries and isolated cells. To determine molecular mechanisms, we conducted RNAseq, PCR, and western blot experiments to characterize them and identify pathways essential for their survival. Results: A stem cell-like population was isolated from fetal carotid arteries grown in serum-free media. The isolated fetal VSCs contained markers for endothelial, smooth muscle, and adventitial cells, and formed a de novo blood vessel ex vivo. A transcriptomic analysis that compared fetal and adult arteries identified pathway enrichment for several kinases, including B-Raf kinase in fetal arteries. Furthermore, we demonstrated that B-Raf- Signal Transducer and Activator of Transcription 3 (STAT3)-Bcl2 is critical for the survival of these cells. Fetal arteries, but not adult arteries, contain VSCs, and B-Raf-STAT3-Bcl2 plays an important role in their survival and proliferation.
Collapse
Affiliation(s)
| | | | - Ravi Goyal
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ 85719, USA
| |
Collapse
|
7
|
Zerfaoui M, Tsumagari K, Toraih E, Errami Y, Ruiz E, Elaasar MSM, Krzysztof M, Sholl AB, Magdeldin S, Soudy M, Abd Elmageed ZY, Boulares AH, Kandil E. Nuclear interaction of Arp2/3 complex and BRAF V600E promotes aggressive behavior and vemurafenib resistance of thyroid cancer. Am J Cancer Res 2022; 12:3014-3033. [PMID: 35968344 PMCID: PMC9360225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/12/2022] [Indexed: 04/22/2023] Open
Abstract
The presence of mutant BRAF V600E correlates with the risk of recurrence in papillary thyroid cancer (PTC) patients. However, not all PTC patients with BRAF V600E are associated with poor prognosis. Thus, understanding the mechanisms by which certain PTC patients with nuclear BRAF V600E become aggressive and develop resistance to a selective BRAF inhibitor, PLX-4032, is urgently needed. The effect of nuclear localization of BRAFV600E using in vitro studies, xenograft mouse-model and human tissues was evaluated. PTC cells harboring a nuclear localization signal (NLS) of BRAFV600E were established and examined in nude mice implanted with TPC1-NLS-BRAFV600E cells followed by PLX-4032 treatment. Immunohistochemical (IHC) analysis was performed on 100 PTC specimens previously confirmed that they have BRAFV600E mutations. Our results demonstrate that 21 of 100 (21%) PTC tissues stained with specific BRAFV600E antibody had nuclear staining with more aggressive features compared to their cytosolic counterparts. In vitro studies show that BRAFV600E is transported between the nucleus and the cytosol through CRM1 and importin (α/β) system. Sequestration of BRAFV600E in the cytosol sensitized resistant cells to PLX-4032, whereas nuclear BRAFV600E was associated with aggressive phenotypes and developed drug resistance. Proteomic analysis revealed Arp2/3 complex members, actin-related protein 2 (ACTR2 aliases ARP2) and actin-related protein 3 (ACTR3 aliases ARP3), as the most enriched nuclear BRAFV600E partners. ACTR3 was highly correlated to lymph node stage and extrathyroidal extension and was validated with different functional assays. Our findings provide new insights into the clinical utility of the nuclear BRAFV600E as a prognostic marker for PTC aggressiveness and determine the efficacy of selective BRAFV600E inhibitor treatment which opens new avenues for future treatment options.
Collapse
Affiliation(s)
- Mourad Zerfaoui
- Department of Surgery, Tulane University School of MedicineUSA
| | - Koji Tsumagari
- Department of Surgery, Tulane University School of MedicineUSA
| | - Eman Toraih
- Department of Surgery, Tulane University School of MedicineUSA
| | - Youssef Errami
- Department of Surgery, Tulane University School of MedicineUSA
| | - Emmanuelle Ruiz
- Department of Surgery, Tulane University School of MedicineUSA
| | | | - Moroz Krzysztof
- Department of Pathology, Tulane University School of MedicineUSA
| | - Andrew B Sholl
- Department of Otolaryngology, Tulane University School of MedicineUSA
| | - Sameh Magdeldin
- Proteomics Research Program Unit, Basic Research Department, Children Cancer Hospital CairoEgypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal UniversityIsmailia 41522, Egypt
| | - Mohamed Soudy
- Proteomics Research Program Unit, Basic Research Department, Children Cancer Hospital CairoEgypt
| | - Zakaria Y Abd Elmageed
- Department of Surgery, Tulane University School of MedicineUSA
- Department of Pharmacology, Edward Via College of Osteopathic Medicine, University of LouisianaMonroe, USA
| | - A Hamid Boulares
- Department of Pharmacology, LSU Health Sciences CenterNew Orleans, LA, USA
| | - Emad Kandil
- Department of Surgery, Tulane University School of MedicineUSA
| |
Collapse
|
8
|
Connor SM, Rashid M, Ryan KJ, Patel K, Boyd JD, Smith J, Elyaman W, Bennett DA, Bradshaw EM. GW5074 Increases Microglial Phagocytic Activities: Potential Therapeutic Direction for Alzheimer's Disease. Front Cell Neurosci 2022; 16:894601. [PMID: 35677758 PMCID: PMC9169965 DOI: 10.3389/fncel.2022.894601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/21/2022] [Indexed: 11/17/2022] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), are responsible for maintaining homeostasis in the brain by clearing debris and are suggested to be inefficient in Alzheimer's Disease (AD), a progressive neurodegenerative disorder for which there is no disease-modifying drug. Besides pathological approaches, unbiased evidence from genome-wide association studies (GWAS) and gene network analysis implicate genes expressed in microglia that reduce phagocytic ability as susceptibility genes for AD. Thus, a central feature toward AD therapy is to increase the microglial phagocytic activities while maintaining synaptic integrity. Here, we developed a robust unbiased high content screening assay to identify potential therapeutics which can reduce the amyloid-beta (Aβ1-42) load by increasing microglial uptake ability. Our screen identified the small-molecule GW5074, an inhibitor of c-RAF, a serine/threonine kinase, which significantly increased the Aβ1-42 clearance activities in human monocyte-derived microglia-like (MDMi) cells, a microglia culture model that recapitulates many genetic and phenotypic aspects of human microglia. Notably, GW5074 was previously reported to be neuroprotective for cerebellar granule cells and cortical neurons. We found that GW5074 significantly increased the expression of key AD-associated microglial molecules known to modulate phagocytosis: TYROBP, SIRPβ1, and TREM2. Our results demonstrated that GW5074 is a potential therapeutic for AD, by targeting microglia.
Collapse
Affiliation(s)
- Sarah M. Connor
- Columbia University Irving Medical Center, New York, NY, United States
| | - Mamunur Rashid
- Columbia University Irving Medical Center, New York, NY, United States
| | - Katie J. Ryan
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
| | - Kruti Patel
- Ann Romney Center for Neurologic Diseases, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
| | - Justin D. Boyd
- Laboratory for Drug Discovery in Neurodegeneration at the Harvard NeuroDiscovery Center, Harvard Medical School, Boston, MA, United States
| | - Jennifer Smith
- The Institute of Chemistry and Cell Biology (ICCB)-Longwood Screening Facility, Harvard Medical School, Boston, MA, United States
| | - Wassim Elyaman
- Columbia University Irving Medical Center, New York, NY, United States
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY, United States
| | - David A. Bennett
- Alzheimer Disease Center, Rush University Medical Center, Chicago, IL, United States
| | - Elizabeth M. Bradshaw
- Columbia University Irving Medical Center, New York, NY, United States
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY, United States
| |
Collapse
|
9
|
Venkatanarayan A, Liang J, Yen I, Shanahan F, Haley B, Phu L, Verschueren E, Hinkle TB, Kan D, Segal E, Long JE, Lima T, Liau NPD, Sudhamsu J, Li J, Klijn C, Piskol R, Junttila MR, Shaw AS, Merchant M, Chang MT, Kirkpatrick DS, Malek S. CRAF dimerization with ARAF regulates KRAS-driven tumor growth. Cell Rep 2022; 38:110351. [PMID: 35139374 DOI: 10.1016/j.celrep.2022.110351] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 09/21/2021] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
KRAS, which is mutated in ∼30% of all cancers, activates the RAF-MEK-ERK signaling cascade. CRAF is required for growth of KRAS mutant lung tumors, but the requirement for CRAF kinase activity is unknown. Here, we show that subsets of KRAS mutant tumors are dependent on CRAF for growth. Kinase-dead but not dimer-defective CRAF rescues growth inhibition, suggesting that dimerization but not kinase activity is required. Quantitative proteomics demonstrates increased levels of CRAF:ARAF dimers in KRAS mutant cells, and depletion of both CRAF and ARAF rescues the CRAF-loss phenotype. Mechanistically, CRAF depletion causes sustained ERK activation and induction of cell-cycle arrest, while treatment with low-dose MEK or ERK inhibitor rescues the CRAF-loss phenotype. Our studies highlight the role of CRAF in regulating MAPK signal intensity to promote tumorigenesis downstream of mutant KRAS and suggest that disrupting CRAF dimerization or degrading CRAF may have therapeutic benefit.
Collapse
Affiliation(s)
| | - Jason Liang
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ivana Yen
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Frances Shanahan
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Benjamin Haley
- Department of Molecular Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lilian Phu
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Erik Verschueren
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Trent B Hinkle
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - David Kan
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ehud Segal
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason E Long
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Tony Lima
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Nicholas P D Liau
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jawahar Sudhamsu
- Department of Structural Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jason Li
- Department of Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Christiaan Klijn
- Department of Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Robert Piskol
- Department of Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Melissa R Junttila
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Andrey S Shaw
- Department of Research Biology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Mark Merchant
- Department of Translational Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Matthew T Chang
- Department of Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Donald S Kirkpatrick
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Shiva Malek
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
10
|
Cucu I, Nicolescu MI. A Synopsis of Signaling Crosstalk of Pericytes and Endothelial Cells in Salivary Gland. Dent J (Basel) 2021; 9:dj9120144. [PMID: 34940041 PMCID: PMC8700478 DOI: 10.3390/dj9120144] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
The salivary gland (SG) microvasculature constitutes a dynamic cellular organization instrumental to preserving tissue stability and homeostasis. The interplay between pericytes (PCs) and endothelial cells (ECs) culminates as a key ingredient that coordinates the development, maturation, and integrity of vessel building blocks. PCs, as a variety of mesenchymal stem cells, enthrall in the field of regenerative medicine, supporting the notion of regeneration and repair. PC-EC interconnections are pivotal in the kinetic and intricate process of angiogenesis during both embryological and post-natal development. The disruption of this complex interlinkage corresponds to SG pathogenesis, including inflammation, autoimmune disorders (Sjögren’s syndrome), and tumorigenesis. Here, we provided a global portrayal of major signaling pathways between PCs and ECs that cooperate to enhance vascular steadiness through the synergistic interchange. Additionally, we delineated how the crosstalk among molecular networks affiliate to contribute to a malignant context. Additionally, within SG microarchitecture, telocytes and myoepithelial cells assemble a labyrinthine companionship, which together with PCs appear to synchronize the regenerative potential of parenchymal constituents. By underscoring the intricacy of signaling cascades within cellular latticework, this review sketched a perceptive basis for target-selective drugs to safeguard SG function.
Collapse
Affiliation(s)
- Ioana Cucu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mihnea Ioan Nicolescu
- Division of Histology, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Laboratory of Radiobiology, “Victor Babeș” National Institute of Pathology, 050096 Bucharest, Romania
- Correspondence:
| |
Collapse
|
11
|
Yap J, Deepak RNVK, Tian Z, Ng WH, Goh KC, Foo A, Tee ZH, Mohanam MP, Sim YRM, Degirmenci U, Lam P, Chen Z, Fan H, Hu J. The stability of R-spine defines RAF inhibitor resistance: A comprehensive analysis of oncogenic BRAF mutants with in-frame insertion of αC-β4 loop. SCIENCE ADVANCES 2021; 7:7/24/eabg0390. [PMID: 34108213 PMCID: PMC8189578 DOI: 10.1126/sciadv.abg0390] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 04/15/2021] [Indexed: 06/12/2023]
Abstract
Although targeting BRAF mutants with RAF inhibitors has achieved promising outcomes in cancer therapy, drug resistance remains a remarkable challenge, and underlying molecular mechanisms are not fully understood. Here, we characterized a previously unknown group of oncogenic BRAF mutants with in-frame insertions (LLRins506 or VLRins506) of αC-β4 loop. Using structure modeling and molecular dynamics simulation, we found that these insertions formed a large hydrophobic network that stabilizes R-spine and thus triggers the catalytic activity of BRAF. Furthermore, these insertions disrupted BRAF dimer interface and impaired dimerization. Unlike BRAF(V600E), these BRAF mutants with low dimer affinity were strongly resistant to all RAF inhibitors in clinic or clinical trials, which arises from their stabilized R-spines. As predicted by molecular docking, the stabilized R-spines in other BRAF mutants also conferred drug resistance. Together, our data indicated that the stability of R-spine but not dimer affinity determines the RAF inhibitor resistance of oncogenic BRAF mutants.
Collapse
Affiliation(s)
- Jiajun Yap
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| | - R N V Krishna Deepak
- Bioinformatics Institute, A*STAR, 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Zizi Tian
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Wan Hwa Ng
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| | - Kah Chun Goh
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| | - Alicia Foo
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| | - Zi Heng Tee
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| | - Manju Payini Mohanam
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| | - Yuen Rong M Sim
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| | - Ufuk Degirmenci
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| | - Paula Lam
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive, MD9, Singapore 117593, Singapore
- Cellvec Pte. Ltd., 100 Pasir Panjang Road, #04-02, Singapore 118518, Singapore
| | - Zhongzhou Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hao Fan
- Bioinformatics Institute, A*STAR, 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore.
| | - Jiancheng Hu
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore.
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Crescen, Singapore 169610, Singapore
| |
Collapse
|
12
|
Pang C, Lim CS, Brookes J, Tsui J, Hamilton G. Emerging importance of molecular pathogenesis of vascular malformations in clinical practice and classifications. Vasc Med 2020; 25:364-377. [PMID: 32568624 DOI: 10.1177/1358863x20918941] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Vascular malformations occur during early vascular development resulting in abnormally formed vessels that can manifest as arterial, venous, capillary or lymphatic lesions, or in combination, and include local tissue overdevelopment. Vascular malformations are largely caused by sporadic somatic gene mutations. This article aims to review and discuss current molecular signaling pathways and therapeutic targets for vascular malformations and to classify vascular malformations according to the molecular pathways involved. A literature review was performed using Embase and Medline. Different MeSH terms were combined for the search strategy, with the aim of encompassing all studies describing the classification, pathogenesis, and treatment of vascular malformations. Major pathways involved in the pathogenesis of vascular malformations are vascular endothelial growth factor (VEGF), Ras/Raf/MEK/ERK, angiopoietin-TIE2, transforming growth factor beta (TGF-β), and PI3K/AKT/mTOR. These pathways are involved in controlling cellular growth, apoptosis, differentiation, and proliferation, and play a central role in endothelial cell signaling and angiogenesis. Many vascular malformations share similar aberrant molecular signaling pathways with cancers and inflammatory disorders. Therefore, selective anticancer agents and immunosuppressants may be beneficial in treating vascular malformations of specific mutations. The current classification systems of vascular malformations, including the International Society of the Study of Vascular Anomalies (ISSVA) classification, are primarily observational and clinical, and are not based on the molecular pathways involved in the pathogenesis of the condition. Several molecular pathways with potential therapeutic targets have been demonstrated to contribute to the development of various vascular anomalies. Classifying vascular malformations based on their molecular pathogenesis may improve treatment by determining the underlying nature of the condition and their potential therapeutic target.
Collapse
Affiliation(s)
- Calver Pang
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Surgical Biotechnology, Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, United Kingdom
| | - Chung Sim Lim
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Surgical Biotechnology, Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, United Kingdom.,NIHR, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Jocelyn Brookes
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Interventional Radiology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Janice Tsui
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Surgical Biotechnology, Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, United Kingdom.,NIHR, University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - George Hamilton
- Department of Vascular Surgery, Royal Free London NHS Foundation Trust, London, United Kingdom.,Department of Surgical Biotechnology, Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, United Kingdom
| |
Collapse
|
13
|
Drosten M, Barbacid M. Targeting the MAPK Pathway in KRAS-Driven Tumors. Cancer Cell 2020; 37:543-550. [PMID: 32289276 DOI: 10.1016/j.ccell.2020.03.013] [Citation(s) in RCA: 320] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/26/2020] [Accepted: 03/16/2020] [Indexed: 12/19/2022]
Abstract
KRAS mutations occur in a quarter of all of human cancers, yet no selective drug has been approved to treat these tumors. Despite the recent development of drugs that block KRASG12C, the majority of KRAS oncoproteins remain undruggable. Here, we review recent efforts to validate individual components of the mitogen-activated protein kinase (MAPK) pathway as targets to treat KRAS-mutant cancers by comparing genetic information derived from experimental mouse models of KRAS-driven lung and pancreatic tumors with the outcome of selective MAPK inhibitors in clinical trials. We also review the potential of RAF1 as a key target to block KRAS-mutant cancers.
Collapse
Affiliation(s)
- Matthias Drosten
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| | - Mariano Barbacid
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro 3, 28029 Madrid, Spain.
| |
Collapse
|
14
|
|
15
|
Outhwaite JE, Patel J, Simmons DG. Secondary Placental Defects in Cxadr Mutant Mice. Front Physiol 2019; 10:622. [PMID: 31338035 PMCID: PMC6628872 DOI: 10.3389/fphys.2019.00622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/02/2019] [Indexed: 12/20/2022] Open
Abstract
The Coxsackie virus and adenovirus receptor (CXADR) is an adhesion molecule known for its role in virus-cell interactions, epithelial integrity, and organogenesis. Loss of Cxadr causes numerous embryonic defects in mice, notably abnormal development of the cardiovascular system, and embryonic lethality. While CXADR expression has been reported in the placenta, the precise cellular localization and function within this tissue are unknown. Since impairments in placental development and function can cause secondary cardiovascular abnormalities, a phenomenon referred to as the placenta-heart axis, it is possible placental phenotypes in Cxadr mutant embryos may underlie the reported cardiovascular defects and embryonic lethality. In the current study, we determine the cellular localization of placental Cxadr expression and whether there are placental abnormalities in the absence of Cxadr. In the placenta, CXADR is expressed specifically by trophoblast labyrinth progenitors as well as cells of the visceral yolk sac (YS). In the absence of Cxadr, we observed altered expression of angiogenic factors coupled with poor expansion of trophoblast and fetal endothelial cell subpopulations, plus diminished placental transport. Unexpectedly, preserving endogenous trophoblast Cxadr expression revealed the placental defects to be secondary to primary embryonic and/or YS phenotypes. Moreover, further tissue-restricted deletions of Cxadr suggest that the secondary placental defects are likely influenced by embryonic lineages such as the fetal endothelium or those within the extraembryonic YS vascular plexus.
Collapse
Affiliation(s)
- Jennifer E Outhwaite
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Jatin Patel
- Translational Research Institute, UQ Diamantina Institute, The University of Queensland, Brisbane, QLD, Australia
| | - David G Simmons
- Faculty of Medicine, School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
16
|
Declercq M, Treps L. BRAF, A gatekeeper controlling endothelial permeability. FEBS J 2019; 286:2273-2276. [PMID: 31081213 DOI: 10.1111/febs.14861] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/23/2019] [Indexed: 12/18/2022]
Abstract
The RAF/MEK/ERK signal transduction pathway is commonly deregulated in cancer and is activated by various stimuli regulating a variety of cell responses. In wild-type endothelial cells, upon permeability stimuli, ROKα, RAF1, BRAF, and RAP1 become activated, inducing a cascade of reactions resulting in F-actin remodeling and increased cell permeability. Here, Dorard et al. showed that BRAF ablated cells had more RAF1/ROKα dimerization and relocalization to VE-cadherin occurred, ultimately leading to less F-actin content and reduced vessel permeability.
Collapse
Affiliation(s)
- Mathias Declercq
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lucas Treps
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, Leuven, Belgium.,Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
| |
Collapse
|
17
|
A Premature Stop Codon in RAF1 Is the Priority Candidate Causative Mutation of the Inherited Chicken Wingless-2 Developmental Syndrome. Genes (Basel) 2019; 10:genes10050353. [PMID: 31075853 PMCID: PMC6562611 DOI: 10.3390/genes10050353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/23/2019] [Accepted: 04/26/2019] [Indexed: 11/17/2022] Open
Abstract
The chicken wingless-2 (wg-2) mutation is inherited in an autosomal recessive fashion, and the resulting phenotype in mutant (wg-2/wg-2) individuals is a developmental syndrome characterized by absent wings, truncated legs, craniofacial as well as skin and feather defects, and kidney malformations. Mapping and genotyping established that the mutation resides within 227 kilobases (kb) of chromosome 12 in a wg-2 congenic inbred line. A capture array was designed to target and sequence the candidate region along with flanking DNA in 24 birds from the line. Many point mutations and insertions or deletions were identified, and analysis of the linked variants indicated a point mutation predicted to cause a premature stop codon in the RAF1 gene. Expression studies were conducted inclusive of all genes in the candidate region. Interestingly, RAF1 transcription was elevated, yet the protein was absent in the mutants relative to normal individuals. RAF1 encodes a protein integral to the Ras/Raf/MAPK signaling pathway controlling cellular proliferation, and notably, human RASopathies are developmental syndromes caused by germline mutations in genes of this pathway. Our work indicates RAF1 as the priority candidate causative gene for wg-2 and provides a new animal model to study an important signaling pathway implicated in limb development, as well as RASopathies.
Collapse
|
18
|
Kurtzeborn K, Kwon HN, Kuure S. MAPK/ERK Signaling in Regulation of Renal Differentiation. Int J Mol Sci 2019; 20:E1779. [PMID: 30974877 PMCID: PMC6479953 DOI: 10.3390/ijms20071779] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/20/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects derived from abnormalities in renal differentiation during embryogenesis. CAKUT is the major cause of end-stage renal disease and chronic kidney diseases in children, but its genetic causes remain largely unresolved. Here we discuss advances in the understanding of how mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) activity contributes to the regulation of ureteric bud branching morphogenesis, which dictates the final size, shape, and nephron number of the kidney. Recent studies also demonstrate that the MAPK/ERK pathway is directly involved in nephrogenesis, regulating both the maintenance and differentiation of the nephrogenic mesenchyme. Interestingly, aberrant MAPK/ERK signaling is linked to many cancers, and recent studies suggest it also plays a role in the most common pediatric renal cancer, Wilms' tumor.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
| | - Hyuk Nam Kwon
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland.
- GM-unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland.
| |
Collapse
|
19
|
B-Raf deficiency impairs tumor initiation and progression in a murine breast cancer model. Oncogene 2019; 38:1324-1339. [DOI: 10.1038/s41388-018-0663-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 11/22/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023]
|
20
|
Prediction of sorafenib treatment–related gene expression for hepatocellular carcinoma: preoperative MRI and histopathological correlation. Eur Radiol 2018; 29:2272-2282. [DOI: 10.1007/s00330-018-5882-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/19/2018] [Accepted: 11/09/2018] [Indexed: 01/15/2023]
|
21
|
Glenfield C, McLysaght A. Pseudogenes Provide Evolutionary Evidence for the Competitive Endogenous RNA Hypothesis. Mol Biol Evol 2018; 35:2886-2899. [PMID: 30252115 PMCID: PMC6278865 DOI: 10.1093/molbev/msy183] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The competitive endogenous RNA (ceRNA) hypothesis is an attractively simple model to explain the biological role of many putatively functionless noncoding RNAs. Under this model, there exist transcripts in the cell whose role is to titrate out microRNAs such that the expression level of another target sequence is altered. That it is logistically possible for expression of one microRNA recognition element (MRE)-containing transcript to affect another is seen in the multiple examples of pathogenic effects of inappropriate expression of MRE-containing RNAs. However, the role, if any, of ceRNAs in normal biological processes and at physiological levels is disputed. By comparison of parent genes and pseudogenes we show, both for a specific example and genome-wide, that the pseudo-3' untranslated regions (3'UTRs) of expressed pseudogenes are frequently retained and are under selective constraint in mammalian genomes. We found that the pseudo-3'UTR of BRAFP1, a previously described oncogenic ceRNA, has reduced substitutions relative to its pseudo-coding sequence, and we show sequence constraint on MREs shared between the parent gene, BRAF, and the pseudogene. Investigation of RNA-seq data reveals expression of BRAFP1 in normal somatic tissues in human and in other primates, consistent with biological ceRNA functionality of this pseudogene in nonpathogenic cellular contexts. Furthermore, we find that on a genome-wide scale pseudo-3'UTRs of mammalian pseudogenes (n = 1,629) are under stronger selective constraint than their pseudo-coding sequence counterparts, and are more often retained and expressed. Our results suggest that many human pseudogenes, often considered nonfunctional, may have an evolutionarily constrained role, consistent with the ceRNA hypothesis.
Collapse
Affiliation(s)
- Cian Glenfield
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Aoife McLysaght
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
22
|
Yuan J, Ng WH, Tian Z, Yap J, Baccarini M, Chen Z, Hu J. Activating mutations in MEK1 enhance homodimerization and promote tumorigenesis. Sci Signal 2018; 11:eaar6795. [PMID: 30377225 DOI: 10.1126/scisignal.aar6795] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
RAS-RAF-MEK-ERK signaling has a well-defined role in cancer biology. Although aberrant pathway activation occurs mostly upstream of the kinase MEK, mutations in MEK are prevalent in some cancer subsets. Here, we found that cancer-related, activating mutations in MEK can be classified into two groups: those that relieve inhibitory interactions with the helix A region and those that are in-frame deletions of the β3-αC loop, which enhance MEK1 homodimerization. The former, helix A-associated mutants, are inhibited by traditional MEK inhibitors. However, we found that the increased homodimerization associated with the loop-deletion mutants promoted intradimer cross-phosphorylation of the activation loop and conferred differential resistance to MEK inhibitors both in vitro and in vivo. MEK1 dimerization was required both for its activation by the kinase RAF and for its catalytic activity toward the kinase ERK. Our findings not only identify a previously unknown group of MEK mutants and provide insight into some key steps in RAF-MEK-ERK activation but also have implications for the design of therapies targeting RAS-ERK signaling in cancers.
Collapse
Affiliation(s)
- Jimin Yuan
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Drive, 169610 Singapore, Singapore
| | - Wan Hwa Ng
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Drive, 169610 Singapore, Singapore
| | - Zizi Tian
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiajun Yap
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Drive, 169610 Singapore, Singapore
| | - Manuela Baccarini
- Max F. Perutz Laboratories, University of Vienna, Doktor-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Zhongzhou Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Jiancheng Hu
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, 11 Hospital Drive, 169610 Singapore, Singapore.
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, 169857 Singapore, Singapore
| |
Collapse
|
23
|
Yuan J, Ng WH, Lam PYP, Wang Y, Xia H, Yap J, Guan SP, Lee ASG, Wang M, Baccarini M, Hu J. The dimer-dependent catalytic activity of RAF family kinases is revealed through characterizing their oncogenic mutants. Oncogene 2018; 37:5719-5734. [PMID: 29930381 PMCID: PMC6202329 DOI: 10.1038/s41388-018-0365-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 05/18/2018] [Accepted: 05/21/2018] [Indexed: 12/19/2022]
Abstract
Although extensively studied for three decades, the molecular mechanisms that regulate the RAF/MEK/ERK kinase cascade remain ambiguous. Recent studies identified the dimerization of RAF as a key event in the activation of this cascade. Here, we show that in-frame deletions in the β3-αC loop activate ARAF as well as BRAF and other oncogenic kinases by enforcing homodimerization. By characterizing these RAF mutants, we find that ARAF has less allosteric and catalytic activity than the other two RAF isoforms, which arises from its non-canonical APE motif. Further, these RAF mutants exhibit a strong oncogenic potential, and a differential inhibitor resistance that correlates with their dimer affinity. Using these unique mutants, we demonstrate that active RAFs, including the BRAF(V600E) mutant, phosphorylate MEK in a dimer-dependent manner. This study characterizes a special category of oncogenic kinase mutations, and elucidates the molecular basis that underlies the differential ability of RAF isoforms to stimulate MEK-ERK pathway. Further, this study reveals a unique catalytic feature of RAF family kinases that can be exploited to control their activities for cancer therapies.
Collapse
Affiliation(s)
- Jimin Yuan
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Wan Hwa Ng
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Paula Y P Lam
- Division of Cellular and Molecular Research, Singapore, Singapore
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore
| | - Yu Wang
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Hongping Xia
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Jiajun Yap
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Shou Ping Guan
- Division of Cellular and Molecular Research, Singapore, Singapore
| | - Ann S G Lee
- Division of Medical Sciences, National Cancer Centre Singapore, 11 Hospital Drive, Singapore, 169610, Singapore
- Office of Clinical & Academic Faculty Affairs, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
- Department of Physiology, National University of Singapore, 2 Medical Drive, 117597, Singapore, Singapore
| | - Mei Wang
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore
| | - Manuela Baccarini
- Max F. Perutz Laboratories, University of Vienna, Doktor-Bohr-Gasse 9, 1030, Vienna, Austria
| | - Jiancheng Hu
- Division of Cellular and Molecular Research, Singapore, Singapore.
- Cancer and Stem Cell Program, Duke-NUS Medical School, 8 College Road, 169857, Singapore, Singapore.
| |
Collapse
|
24
|
Camm EJ, Botting KJ, Sferruzzi-Perri AN. Near to One's Heart: The Intimate Relationship Between the Placenta and Fetal Heart. Front Physiol 2018; 9:629. [PMID: 29997513 PMCID: PMC6029139 DOI: 10.3389/fphys.2018.00629] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 05/09/2018] [Indexed: 01/19/2023] Open
Abstract
The development of the fetal heart is exquisitely controlled by a multitude of factors, ranging from humoral to mechanical forces. The gatekeeper regulating many of these factors is the placenta, an external fetal organ. As such, resistance within the placental vascular bed has a direct influence on the fetal circulation and therefore, the developing heart. In addition, the placenta serves as the interface between the mother and fetus, controlling substrate exchange and release of hormones into both circulations. The intricate relationship between the placenta and fetal heart is appreciated in instances of clinical placental pathology. Abnormal umbilical cord insertion is associated with congenital heart defects. Likewise, twin-to-twin transfusion syndrome, where monochorionic twins have unequal sharing of their placenta due to inter-twin vascular anastomoses, can result in cardiac remodeling and dysfunction in both fetuses. Moreover, epidemiological studies have suggested a link between placental phenotypic traits and increased risk of cardiovascular disease in adult life. To date, the mechanistic basis of the relationships between the placenta, fetal heart development and later risk of cardiac dysfunction have not been fully elucidated. However, studies using environmental exposures and gene manipulations in experimental animals are providing insights into the pathways involved. Likewise, surgical instrumentation of the maternal and fetal circulations in large animal species has enabled the manipulation of specific humoral and mechanical factors to investigate their roles in fetal cardiac development. This review will focus on such studies and what is known to date about the link between the placenta and heart development.
Collapse
Affiliation(s)
- Emily J Camm
- Department of Physiology, Development and Neuroscience and Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Kimberley J Botting
- Department of Physiology, Development and Neuroscience and Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience and Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
25
|
Dorard C, Vucak G, Baccarini M. Deciphering the RAS/ERK pathway in vivo. Biochem Soc Trans 2017; 45:27-36. [PMID: 28202657 DOI: 10.1042/bst20160135] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 12/19/2022]
Abstract
The RAS/ERK pathway has been intensely studied for about three decades, not least because of its role in human pathologies. ERK activation is observed in the majority of human cancers; in about one-third of them, it is driven by mutational activation of pathway components. The pathway is arguably one of the best targets for molecule-based pharmacological intervention, and several small-molecule inhibitors are in clinical use. Genetically engineered mouse models have greatly contributed to our understanding of signaling pathways in development, tissue homeostasis, and disease. In the specific case of the RAS/ERK pathway, they have revealed unique biological roles of structurally and functionally similar proteins, new kinase-independent effectors, and unsuspected relationships with other cascades. This short review summarizes the contribution of mouse models to our current understanding of the pathway.
Collapse
Affiliation(s)
- Coralie Dorard
- Max F. Perutz Laboratories, Center for Molecular Biology, University of Vienna, Vienna 1030, Austria
| | - Georg Vucak
- Max F. Perutz Laboratories, Center for Molecular Biology, University of Vienna, Vienna 1030, Austria
| | - Manuela Baccarini
- Max F. Perutz Laboratories, Center for Molecular Biology, University of Vienna, Vienna 1030, Austria
| |
Collapse
|
26
|
Abstract
This is a report of a 36 week male infant who suffered abdominal distension and difficulty opening bowels within first few days of life and showed a pattern of hypoventilation and apnea associated with sleep. His diagnostic studies confirmed the diagnosis of congenital central hypoventilation syndrome CCHS (PHOX2B mutation) and Hirschsprung's disease and later found a further mutation of BRAF oncogene. This describes a novel association between these mutations and the shared qualities of tumorigenesis between BRAF and PHOX2B.
Collapse
|
27
|
Hernandez MA, Patel B, Hey F, Giblett S, Davis H, Pritchard C. Regulation of BRAF protein stability by a negative feedback loop involving the MEK-ERK pathway but not the FBXW7 tumour suppressor. Cell Signal 2016; 28:561-71. [PMID: 26898828 PMCID: PMC6399479 DOI: 10.1016/j.cellsig.2016.02.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/08/2016] [Accepted: 02/15/2016] [Indexed: 01/06/2023]
Abstract
The V600EBRAF oncogenic mutation is detected in a wide range of human cancers and induces hyperactivation of the downstream MEK–ERK signalling cascade. Although output of the BRAF–MEK–ERK pathway is regulated by feed-forward RAF activity, feedback control also plays an important role. One such feedback pathway has been identified in Caenorhabditis elegans and involves ERK-mediated phosphorylation of BRAF within a CDC4 phosphodegron (CPD), targeting BRAF for degradation via CDC4 (also known as FBXW7), a component of the SKP1/CUL1/F-box (SCF) E3 ubiquitin ligase complex. Here we investigate this pathway in mammalian cells. Short-term expression of autochthonous V600EBRAF in mouse embryonic fibroblasts (MEFs) leads to down-regulation of BRAF protein levels in a proteasome-dependent manner and V600EBRAF has a reduced half-life compared to WTBRAF in HEK293T cells. These effects were reversed by treatment with the MEK inhibitor PD184352. We have identified the equivalent CPD at residues 400–405 in human BRAF and have found that mutation of ERK phosphorylation sites at residues T401 and S405 in V600EBRAF increases the half-life of the protein. While BRAF and FBXW7 co-immunoprecipitated, the overexpression of FBXW7 did not influence the half-life of either WTBRAF or V600EBRAF. Furthermore, disruption of the substrate-binding site of mouse FBXW7 using the R482Q mutation did not affect the interaction with BRAF and the expression levels of WTBRAF and V600EBRAF were not altered in MEFs derived from mice with the homozygous knockin R482QFBXW7 mutation. Overall these data confirm the existence of a negative feedback pathway by which BRAF protein stability is regulated by ERK. However, unlike the situation in C. elegans, FBXW7 does not play a unique role in mediating subsequent BRAF degradation. Expression of oncogenic V600EBRAF down-regulates BRAF expression at the protein level. V600EBRAF has a shorter half-life than WTBRAF. BRAF protein stability is subjected to feedback control by the MEK/ERK pathway. This feedback pathway is associated with the oncogene-induced senescence phenotype. ERK phosphorylation sites at T401 and S405 within a conserved CDC4 (FBXW7) phosphodegron of BRAF are involved in the feedback control pathway. The FBXW7 substrate recognition component of the SKP1/CUL1/F-box (SCF) complex binds to BRAF but is not uniquely involved in the regulation of its protein turnover.
Collapse
Affiliation(s)
- Maria Aguilar Hernandez
- Department of Molecular Cell Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Bipin Patel
- Department of Molecular Cell Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Fiona Hey
- Department of Molecular Cell Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Susan Giblett
- Department of Molecular Cell Biology, University of Leicester, University Road, Leicester LE1 7RH, UK
| | - Hayley Davis
- Gastrointestinal Stem Cell Biology Laboratory, Wellcome Trust Centre for Human Genetics, Oxford University, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Catrin Pritchard
- Department of Molecular Cell Biology, University of Leicester, University Road, Leicester LE1 7RH, UK; Department of Cancer Studies, University of Leicester, University Road, Leicester LE1 7RH, UK.
| |
Collapse
|
28
|
Dambach DM, Misner D, Brock M, Fullerton A, Proctor W, Maher J, Lee D, Ford K, Diaz D. Safety Lead Optimization and Candidate Identification: Integrating New Technologies into Decision-Making. Chem Res Toxicol 2015; 29:452-72. [DOI: 10.1021/acs.chemrestox.5b00396] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Donna M. Dambach
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dinah Misner
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Mathew Brock
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Aaron Fullerton
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - William Proctor
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Jonathan Maher
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dong Lee
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Kevin Ford
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| | - Dolores Diaz
- Department of Safety Assessment, Genentech, Inc., 1 DNA
Way, South San Francisco, California 94080, United States
| |
Collapse
|
29
|
Köhler M, Röring M, Schorch B, Heilmann K, Stickel N, Fiala GJ, Schmitt LC, Braun S, Ehrenfeld S, Uhl FM, Kaltenbacher T, Weinberg F, Herzog S, Zeiser R, Schamel WW, Jumaa H, Brummer T. Activation loop phosphorylation regulates B-Raf in vivo and transformation by B-Raf mutants. EMBO J 2015; 35:143-61. [PMID: 26657898 DOI: 10.15252/embj.201592097] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/28/2015] [Indexed: 12/19/2022] Open
Abstract
Despite being mutated in cancer and RASopathies, the role of the activation segment (AS) has not been addressed for B-Raf signaling in vivo. Here, we generated a conditional knock-in mouse allowing the expression of the B-Raf(AVKA) mutant in which the AS phosphoacceptor sites T599 and S602 are replaced by alanine residues. Surprisingly, despite producing a kinase-impaired protein, the Braf(AVKA) allele does not phenocopy the lethality of Braf-knockout or paradoxically acting knock-in alleles. However, Braf(AVKA) mice display abnormalities in the hematopoietic system, a distinct facial morphology, reduced ERK pathway activity in the brain, and an abnormal gait. This phenotype suggests that maximum B-Raf activity is required for the proper development, function, and maintenance of certain cell populations. By establishing conditional murine embryonic fibroblast cultures, we further show that MEK/ERK phosphorylation and the immediate early gene response toward growth factors are impaired in the presence of B-Raf(AVKA). Importantly, alanine substitution of T599/S602 impairs the transformation potential of oncogenic non-V600E B-Raf mutants and a fusion protein, suggesting that blocking their phosphorylation could represent an alternative strategy to ATP-competitive inhibitors.
Collapse
Affiliation(s)
- Martin Köhler
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Michael Röring
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Björn Schorch
- Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Katharina Heilmann
- Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Natalie Stickel
- Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany Department of Hematology and Oncology, University Medical Center ALU, Freiburg, Germany
| | - Gina J Fiala
- Spemann Graduate School for Biology and Medicine, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Lisa C Schmitt
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Sandra Braun
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany
| | - Sophia Ehrenfeld
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Franziska M Uhl
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Thorsten Kaltenbacher
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Florian Weinberg
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany
| | - Sebastian Herzog
- Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University Medical Center ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Comprehensive Cancer Centre, Freiburg, Germany German Consortium for Translational Cancer Research DKTK, Standort Freiburg, Germany
| | - Wolfgang W Schamel
- Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany Center for Chronic Immunodeficiency CCI, University Medical Center, Freiburg, Germany
| | - Hassan Jumaa
- Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Department of Molecular Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany Institute of Immunology, University Hospital Ulm, Ulm, Germany
| | - Tilman Brummer
- Faculty of Medicine, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University (ALU), Freiburg, Germany Centre for Biological Systems Analysis ZBSA, ALU, Freiburg, Germany Faculty of Biology, ALU, Freiburg, Germany Centre for Biological Signalling Studies BIOSS, ALU, Freiburg, Germany Comprehensive Cancer Centre, Freiburg, Germany German Consortium for Translational Cancer Research DKTK, Standort Freiburg, Germany
| |
Collapse
|
30
|
Zandvakili I, Davis AK, Hu G, Zheng Y. Loss of RhoA Exacerbates, Rather Than Dampens, Oncogenic K-Ras Induced Lung Adenoma Formation in Mice. PLoS One 2015; 10:e0127923. [PMID: 26030593 PMCID: PMC4452309 DOI: 10.1371/journal.pone.0127923] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/20/2015] [Indexed: 12/25/2022] Open
Abstract
Numerous cellular studies have indicated that RhoA signaling is required for oncogenic Ras-induced transformation, suggesting that RhoA is a useful target in Ras induced neoplasia. However, to date very limited data exist to genetically attribute RhoA function to Ras-mediated tumorigenesis in mammalian models. In order to assess whether RhoA is required for K-Ras-induced lung cancer initiation, we utilized the K-RasG12D Lox-Stop-Lox murine lung cancer model in combination with a conditional RhoAflox/flox and RhoC-/- knockout mouse models. Deletion of the floxed Rhoa gene and expression of K-RasG12D was achieved by either CCSP-Cre or adenoviral Cre, resulting in simultaneous expression of K-RasG12D and deletion of RhoA from the murine lung. We found that deletion of RhoA, RhoC or both did not adversely affect normal lung development. Moreover, we found that deletion of either RhoA or RhoC alone did not suppress K-RasG12D induced lung adenoma initiation. Rather, deletion of RhoA alone exacerbated lung adenoma formation, whereas dual deletion of RhoA and RhoC together significantly reduced K-RasG12D induced adenoma formation. Deletion of RhoA appears to induce a compensatory mechanism that exacerbates adenoma formation. The compensatory mechanism is at least partly mediated by RhoC. This study suggests that targeting of RhoA alone may allow for compensation and a paradoxical exacerbation of neoplasia, while simultaneous targeting of both RhoA and RhoC is likely to lead to more favorable outcomes.
Collapse
Affiliation(s)
- Inuk Zandvakili
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Medical-Scientist Training Program, College of Medicine, The University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Ashley Kuenzi Davis
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Guodong Hu
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Yi Zheng
- Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Medical-Scientist Training Program, College of Medicine, The University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
31
|
de Iriarte Rodríguez R, Magariños M, Pfeiffer V, Rapp UR, Varela-Nieto I. C-Raf deficiency leads to hearing loss and increased noise susceptibility. Cell Mol Life Sci 2015; 72:3983-98. [PMID: 25975225 PMCID: PMC4575698 DOI: 10.1007/s00018-015-1919-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 04/21/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022]
Abstract
The family of RAF kinases transduces extracellular information to the nucleus, and their activation is crucial for cellular regulation on many levels, ranging from embryonic development to carcinogenesis. B-RAF and C-RAF modulate neurogenesis and neuritogenesis during chicken inner ear development. C-RAF deficiency in humans is associated with deafness in the rare genetic insulin-like growth factor 1 (IGF-1), Noonan and Leopard syndromes. In this study, we show that RAF kinases are expressed in the developing inner ear and in adult mouse cochlea. A homozygous C-Raf deletion in mice caused profound deafness with no evident cellular aberrations except for a remarkable reduction of the K+ channel Kir4.1 expression, a trait that suffices as a cause of deafness. To explore the role of C-Raf in cellular protection and repair, heterozygous C-Raf+/− mice were exposed to noise. A reduced C-RAF level negatively affected hearing preservation in response to noise through mechanisms involving the activation of JNK and an exacerbated apoptotic response. Taken together, these results strongly support a role for C-RAF in hearing protection.
Collapse
Affiliation(s)
- Rocío de Iriarte Rodríguez
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain.,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Marta Magariños
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain. .,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain. .,Departamento de Biología, Universidad Autónoma de Madrid, Darwin 2, 28049, Madrid, Spain.
| | - Verena Pfeiffer
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Versbacher Strasse 5, 97078, Würzburg, Germany.,Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstraße 6, 97070, Würzburg, Germany
| | - Ulf R Rapp
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Versbacher Strasse 5, 97078, Würzburg, Germany.,Molecular Mechanisms of Lung Cancer, Max Planck Institute for Heart and Lung Research, Parkstr. 1, 61231, Bad Nauheim, Germany
| | - Isabel Varela-Nieto
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Arturo Duperier 4, 28029, Madrid, Spain.,Centre for Biomedical Network Research (CIBERER), Institute of Health Carlos III (ISCIII), Madrid, Spain.,Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| |
Collapse
|
32
|
Kang MC, Park SJ, Kim HJ, Lee J, Yu DH, Bae KB, Ji YR, Park SJ, Jeong J, Jang WY, Kim JH, Choi MS, Lee DS, Lee HS, Lee S, Kim SH, Kim MO, Park G, Choo YS, Cho JY, Ryoo ZY. Gestational loss and growth restriction by angiogenic defects in placental growth factor transgenic mice. Arterioscler Thromb Vasc Biol 2014; 34:2276-82. [PMID: 25147341 DOI: 10.1161/atvbaha.114.303693] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Angiogenesis is an important biological process during development, reproduction, and in immune responses. Placental growth factor (PlGF) is a member of vascular endothelial growth factor that is critical for angiogenesis and vasculogenesis. We generated transgenic mice overexpressing PlGF in specifically T cells using the human CD2-promoter to investigate the effects of PlGF overexpression. APPROACH AND RESULTS Transgenic mice were difficult to obtain owing to high lethality; for this reason, we investigated why gestational loss occurred in these transgenic mice. Here, we report that placenta detachment and inhibition of angiogenesis occurred in PlGF transgenic mice during the gestational period. Moreover, even when transgenic mice were born, their growth was restricted. CONCLUSIONS Conclusively, PlGF overexpression prevents angiogenesis by inhibiting Braf, extracellular signal-regulated kinase activation, and downregulation of HIF-1α in the mouse placenta. Furthermore, it affected regulatory T cells, which are important for maintenance of pregnancy.
Collapse
Affiliation(s)
- Min-Cheol Kang
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Seo Jin Park
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Hei Jung Kim
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Jinhee Lee
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Dong Hoon Yu
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Ki Beom Bae
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Young Rae Ji
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Si Jun Park
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Jain Jeong
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Woo Young Jang
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Jung-Hak Kim
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Myung-Sook Choi
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Dong-Seok Lee
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Hyun-Shik Lee
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Sanggyu Lee
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Sung Hyun Kim
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Myoung Ok Kim
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Gyeongsin Park
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Yeon Sik Choo
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Je-Yoel Cho
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.)
| | - Zae Young Ryoo
- From the School of Life Sciences and Biotechnology (M.K., S.J.P., H.J.K., J.L., D.H.Y., K.B.B., Y.R.J., S.J.P., J.J., W.Y.J., J.-H.K., D.-S.L., H.-S.L., S.L., S.H.K., M.O.K., Z.Y.R.), Department of Food Science and Nutrition (M.S.C.), and School of Biology (Y.S.C.), Kyungpook National University, Daegu, Korea; Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea (G.P.); and Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Korea (J.-Y.C.).
| |
Collapse
|
33
|
Nadeau V, Charron J. Essential role of the ERK/MAPK pathway in blood-placental barrier formation. Development 2014; 141:2825-37. [PMID: 24948605 DOI: 10.1242/dev.107409] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mammalian genome contains two ERK/MAP kinase kinase genes, Map2k1 and Map2k2, which encode dual-specificity kinases responsible for ERK activation. Loss of Map2k1 function in mouse causes embryonic lethality due to placental defects, whereas Map2k2 mutants have a normal lifespan. The majority of Map2k1(+/-) Map2k2(+/-) embryos die during gestation from the underdevelopment of the placenta labyrinth, demonstrating that both kinases are involved in placenta formation. Map2k1(+/-) Map2k2(+/-) mutants show reduced vascularization of the labyrinth and defective formation of syncytiotrophoblast layer II (SynT-II) leading to the accumulation of multinucleated trophoblast giant cells (MTGs). To define the cell type-specific contribution of the ERK/MAPK pathway to placenta development, we performed deletions of Map2k1 function in different Map2k1 Map2k2 allelic backgrounds. Loss of MAP kinase kinase activity in pericytes or in allantois-derived tissues worsens the MTG phenotype. These results define the contribution of the ERK/MAPK pathway in specific embryonic and extraembryonic cell populations for normal placentation. Our data also indicate that MTGs could result from the aberrant fusion of SynT-I and -II. Using mouse genetics, we demonstrate that the normal development of SynT-I into a thin layer of multinucleated cells depends on the presence of SynT-II. Lastly, the combined mutations of Map2k1 and Map2k2 alter the expression of several genes involved in cell fate specification, cell fusion and cell polarity. Thus, appropriate ERK/MAPK signaling in defined cell types is required for the proper growth, differentiation and morphogenesis of the placenta.
Collapse
Affiliation(s)
- Valérie Nadeau
- Centre de recherche sur le cancer de l'Université Laval, Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, Canada G1R 2J6
| | - Jean Charron
- Centre de recherche sur le cancer de l'Université Laval, Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec, QC, Canada G1R 2J6
| |
Collapse
|
34
|
"RAF" neighborhood: protein-protein interaction in the Raf/Mek/Erk pathway. FEBS Lett 2014; 588:2398-406. [PMID: 24937142 PMCID: PMC4099524 DOI: 10.1016/j.febslet.2014.06.025] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 06/05/2014] [Accepted: 06/06/2014] [Indexed: 12/19/2022]
Abstract
The Raf/Mek/Erk signaling pathway, activated downstream of Ras primarily to promote proliferation, represents the best studied of the evolutionary conserved MAPK cascades. The investigation of the pathway has continued unabated since its discovery roughly 30 years ago. In the last decade, however, the identification of unexpected in vivo functions of pathway components, as well as the discovery of Raf mutations in human cancer, the ensuing quest for inhibitors, and the efforts to understand their mechanism of action, have boosted interest tremendously. From this large body of work, protein-protein interaction has emerged as a recurrent, crucial theme. This review focuses on the role of protein complexes in the regulation of the Raf/Mek/Erk pathway and in its cross-talk with other signaling cascades. Mapping these interactions and finding a way of exploiting them for therapeutic purposes is one of the challenges of future molecule-targeted therapy.
Collapse
|
35
|
Watanabe-Takano H, Takano K, Sakamoto A, Matsumoto K, Tokuhisa T, Endo T, Hatano M. DA-Raf-dependent inhibition of the Ras-ERK signaling pathway in type 2 alveolar epithelial cells controls alveolar formation. Proc Natl Acad Sci U S A 2014; 111:E2291-300. [PMID: 24843139 PMCID: PMC4050578 DOI: 10.1073/pnas.1321574111] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Alveolar formation is coupled to the spatiotemporally regulated differentiation of alveolar myofibroblasts (AMYFs), which contribute to the morphological changes of interalveolar walls. Although the Ras-ERK signaling pathway is one of the key regulators for alveolar formation in developing lungs, the intrinsic molecular and cellular mechanisms underlying its role remain largely unknown. By analyzing the Ras-ERK signaling pathway during postnatal development of lungs, we have identified a critical role of DA-Raf1 (DA-Raf)-a dominant-negative antagonist for the Ras-ERK signaling pathway-in alveolar formation. DA-Raf-deficient mice displayed alveolar dysgenesis as a result of the blockade of AMYF differentiation. DA-Raf is predominantly expressed in type 2 alveolar epithelial cells (AEC2s) in developing lungs, and DA-Raf-dependent MEK1/2 inhibition in AEC2s suppresses expression of tissue inhibitor of matalloprotienase 4 (TIMP4), which prevents a subsequent proteolytic cascade matrix metalloproteinase (MMP)14-MMP2. Furthermore, MMP14-MMP2 proteolytic cascade regulates AMYF differentiation and alveolar formation. Therefore, DA-Raf-dependent inhibition of the Ras-ERK signaling pathway in AEC2s is required for alveolar formation via triggering MMP2 activation followed by AMYF differentiation. These findings reveal a pivotal role of the Ras-ERK signaling pathway in the dynamic regulation of alveolar development.
Collapse
Affiliation(s)
- Haruko Watanabe-Takano
- Departments of Biomedical Science andDepartment of Biology, Graduate School of Science andJapan Society for the Promotion of Science, Chiyoda-ku, Tokyo 102-0083, Japan; and
| | - Kazunori Takano
- Department of Biology, Graduate School of Science andGraduate School of Advanced Integration Science, Chiba University, Yayoicho, Inage-ku, Chiba 263-8522, Japan
| | - Akemi Sakamoto
- Developmental Genetics, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kenji Matsumoto
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
| | - Takeshi Tokuhisa
- Developmental Genetics, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Takeshi Endo
- Department of Biology, Graduate School of Science andGraduate School of Advanced Integration Science, Chiba University, Yayoicho, Inage-ku, Chiba 263-8522, Japan;
| | | |
Collapse
|
36
|
Han KY, Fahd DC, Tshionyi M, Allemann N, Jain S, Chang JH, Azar DT. MT1-MMP modulates bFGF-induced VEGF-A expression in corneal fibroblasts. Protein Pept Lett 2013; 19:1334-9. [PMID: 22670674 DOI: 10.2174/092986612803521639] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 05/10/2012] [Accepted: 05/11/2012] [Indexed: 11/22/2022]
Abstract
The cornea is physiologically avascular. Following a corneal injury, wound healing often proceeds without neovascularization (NV); however, corneal NV may be induced during wound healing in certain inflammatory, infectious, degenerative, and traumatic states. Such states disrupt the physiologic balance between pro-angiogenic and antiangiogenic mediators, favoring angiogenesis. Contributors to such states are matrix metalloproteinases (MMPs), which are key factors in both extracellular matrix remodeling and angiogenesis. Similarly, vascular endothelial growth factor A (VEGF-A) and basic fibroblast growth factor (bFGF) exert pro-angiogenic effects. Here, we elaborate on the facilitative role of MMPs-specifically Membrane Type 1 MMP (MT1-MMP, MMP14)-in corneal NV. Additionally, we provide new insight into the signaling relating to MT1-MMP, Ras, and ERK in the bFGF-induced VEGF-A expression pathways within the corneal fibroblasts.
Collapse
Affiliation(s)
- Kyu-Yeon Han
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Pfeiffer V, Götz R, Xiang C, Camarero G, Braun A, Zhang Y, Blum R, Heinsen H, Nieswandt B, Rapp UR. Ablation of BRaf impairs neuronal differentiation in the postnatal hippocampus and cerebellum. PLoS One 2013; 8:e58259. [PMID: 23505473 PMCID: PMC3591433 DOI: 10.1371/journal.pone.0058259] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 02/01/2013] [Indexed: 12/30/2022] Open
Abstract
This study focuses on the role of the kinase BRaf in postnatal brain development. Mice expressing truncated, non-functional BRaf in neural stem cell-derived brain tissue demonstrate alterations in the cerebellum, with decreased sizes and fuzzy borders of the glomeruli in the granule cell layer. In addition we observed reduced numbers and misplaced ectopic Purkinje cells that showed an altered structure of their dendritic arborizations in the hippocampus, while the overall cornus ammonis architecture appeared to be unchanged. In male mice lacking BRaf in the hippocampus the size of the granule cell layer was normal at postnatal day 12 (P12) but diminished at P21, as compared to control littermates. This defect was caused by a reduced ability of dentate gyrus progenitor cells to differentiate into NeuN positive granule cell neurons. In vitro cell culture of P0/P1 hippocampal cells revealed that BRaf deficient cells were impaired in their ability to form microtubule-associated protein 2 positive neurons. Together with the alterations in behaviour, such as autoaggression and loss of balance fitness, these observations indicate that in the absence of BRaf all neuronal cellular structures develop, but neuronal circuits in the cerebellum and hippocampus are partially disturbed besides impaired neuronal generation in both structures.
Collapse
Affiliation(s)
- Verena Pfeiffer
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
| | - Rudolf Götz
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
- Institute for Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Chaomei Xiang
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
| | - Guadelupe Camarero
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
| | - Attila Braun
- Rudolf Virchow Centre, DFG Research Centre for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Yina Zhang
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
| | - Robert Blum
- Institute for Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Helmut Heinsen
- Department of Psychiatry, Morphological Brain Research Unit, University of Würzburg, Würzburg, Germany
| | - Bernhard Nieswandt
- Rudolf Virchow Centre, DFG Research Centre for Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Ulf R. Rapp
- Institute for Medical Radiation and Cell Research (MSZ), University of Würzburg, Würzburg, Germany
- Department of Molecular Mechanisms in Lung Cancer, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail:
| |
Collapse
|
38
|
Choi HJ, Sanders TA, Tormos KV, Ameri K, Tsai JD, Park AM, Gonzalez J, Rajah AM, Liu X, Quinonez DM, Rinaudo PF, Maltepe E. ECM-dependent HIF induction directs trophoblast stem cell fate via LIMK1-mediated cytoskeletal rearrangement. PLoS One 2013; 8:e56949. [PMID: 23437279 PMCID: PMC3578927 DOI: 10.1371/journal.pone.0056949] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 01/16/2013] [Indexed: 01/24/2023] Open
Abstract
The Hypoxia-inducible Factor (HIF) family of transcriptional regulators coordinates the expression of dozens of genes in response to oxygen deprivation. Mammalian development occurs in a hypoxic environment and HIF-null mice therefore die in utero due to multiple embryonic and placental defects. Mouse embryonic stem cells do not differentiate into placental cells; therefore, trophoblast stem cells (TSCs) are used to study mouse placental development. Consistent with a requirement for HIF activity during placental development in utero, TSCs derived from HIF-null mice exhibit severe differentiation defects and fail to form trophoblast giant cells (TGCs) in vitro. Interestingly, differentiating TSCs induce HIF activity independent of oxygen tension via unclear mechanisms. Here, we show that altering the extracellular matrix (ECM) composition upon which TSCs are cultured changes their differentiation potential from TGCs to multinucleated syncytiotropholasts (SynTs) and blocks oxygen-independent HIF induction. We further find that modulation of Mitogen Activated Protein Kinase Kinase-1/2 (MAP2K1/2, MEK-1/2) signaling by ECM composition is responsible for this effect. In the absence of ECM-dependent cues, hypoxia-signaling pathways activate this MAPK cascade to drive HIF induction and redirect TSC fate along the TGC lineage. In addition, we show that integrity of the microtubule and actin cytoskeleton is critical for TGC fate determination. HIF-2α ensures TSC cytoskeletal integrity and promotes invasive TGC formation by interacting with c-MYC to induce non-canonical expression of Lim domain kinase 1-an enzyme that regulates microtubule and actin stability, as well as cell invasion. Thus, we find that HIF can integrate positional and metabolic cues from within the TSC niche to regulate placental development by modulating the cellular cytoskeleton via non-canonical gene expression.
Collapse
Affiliation(s)
- Hwa J. Choi
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
| | - Timothy A. Sanders
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
| | - Kathryn V. Tormos
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Kurosh Ameri
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
| | - Justin D. Tsai
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
| | - Angela M. Park
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
| | - Julissa Gonzalez
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Anthony M. Rajah
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
| | - Xiaowei Liu
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Diana M. Quinonez
- Department of Biology, San Francisco State University, San Francisco, California, United States of America
| | - Paolo F. Rinaudo
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| | - Emin Maltepe
- Department of Pediatrics, University of California San Francisco, San Francisco, California, United States of America
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, California, United States of America
- Developmental and Stem Cell Biology Program, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
39
|
Zeng L, Ehrenreiter K, Menon J, Menard R, Kern F, Nakazawa Y, Bevilacqua E, Imamoto A, Baccarini M, Rosner MR. RKIP regulates MAP kinase signaling in cells with defective B-Raf activity. Cell Signal 2013; 25:1156-65. [PMID: 23416466 DOI: 10.1016/j.cellsig.2013.02.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 02/08/2013] [Indexed: 01/05/2023]
Abstract
MAP kinase (MAPK) signaling results from activation of Raf kinases in response to external or internal stimuli. Here, we demonstrate that Raf kinase inhibitory protein (RKIP) regulates the activation of MAPK when B-Raf signaling is defective. We used multiple models including mouse embryonic fibroblasts (MEFs) and primary keratinocytes from RKIP- or Raf-deficient mice as well as allografts in mice to investigate the mechanism. Loss of B-Raf protein or activity significantly reduces MAPK activation in these cells. We show that RKIP depletion can rescue the compromised ERK activation and promote proliferation, and this rescue occurs through a Raf-1 dependent mechanism. These results provide formal evidence that RKIP is a bona fide regulator of Raf-1. We propose a new model in which RKIP plays a key role in regulating the ability of cells to signal through Raf-1 to ERK in B-Raf compromised cells.
Collapse
Affiliation(s)
- Lingchun Zeng
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Maymó JL, Pérez Pérez A, Maskin B, Dueñas JL, Calvo JC, Sánchez Margalet V, Varone CL. The alternative Epac/cAMP pathway and the MAPK pathway mediate hCG induction of leptin in placental cells. PLoS One 2012; 7:e46216. [PMID: 23056265 PMCID: PMC3462743 DOI: 10.1371/journal.pone.0046216] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 08/29/2012] [Indexed: 12/21/2022] Open
Abstract
Pleiotropic effects of leptin have been identified in reproduction and pregnancy, particularly in the placenta, where it works as an autocrine hormone. In this work, we demonstrated that human chorionic gonadotropin (hCG) added to JEG-3 cell line or to placental explants induces endogenous leptin expression. We also found that hCG increased cAMP intracellular levels in BeWo cells in a dose-dependent manner, stimulated cAMP response element (CRE) activity and the cotransfection with an expression plasmid of a dominant negative mutant of CREB caused a significant inhibition of hCG stimulation of leptin promoter activity. These results demonstrate that hCG indeed activates cAMP/PKA pathway, and that this pathway is involved in leptin expression. Nevertheless, we found leptin induction by hCG is dependent on cAMP levels. Treatment with (Bu)2cAMP in combination with low and non stimulatory hCG concentrations led to an increase in leptin expression, whereas stimulatory concentrations showed the opposite effect. We found that specific PKA inhibition by H89 caused a significant increase of hCG leptin induction, suggesting that probably high cAMP levels might inhibit hCG effect. It was found that hCG enhancement of leptin mRNA expression involved the MAPK pathway. In this work, we demonstrated that hCG leptin induction through the MAPK signaling pathway is inhibited by PKA. We observed that ERK1/2 phosphorylation increased when hCG treatment was combined with H89. In view of these results, the involvement of the alternative cAMP/Epac signaling pathway was studied. We observed that a cAMP analogue that specifically activates Epac (CPT-OMe) stimulated leptin expression by hCG. In addition, the overexpression of Epac and Rap1 proteins increased leptin promoter activity and enhanced hCG. In conclusion, we provide evidence suggesting that hCG induction of leptin gene expression in placenta is mediated not only by activation of the MAPK signaling pathway but also by the alternative cAMP/Epac signaling pathway.
Collapse
Affiliation(s)
- Julieta Lorena Maymó
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Antonio Pérez Pérez
- Departamento de Bioquímica Médica y Biología Molecular. Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Bernardo Maskin
- Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - José Luis Dueñas
- Servicio de Ginecología y Obstetricia, Hospital Universitario Virgen Macarena, Sevilla, España
| | - Juan Carlos Calvo
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental (IBYME), Buenos Aires, Argentina
| | - Víctor Sánchez Margalet
- Departamento de Bioquímica Médica y Biología Molecular. Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Cecilia Laura Varone
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
41
|
Kleiman DA, Buitrago D, Crowley MJ, Beninato T, Veach AJ, Zanzonico PB, Jin M, Fahey TJ, Zarnegar R. Thyroid stimulating hormone increases iodine uptake by thyroid cancer cells during BRAF silencing. J Surg Res 2012; 182:85-93. [PMID: 22998776 DOI: 10.1016/j.jss.2012.08.053] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 07/21/2012] [Accepted: 08/24/2012] [Indexed: 12/25/2022]
Abstract
BACKGROUND The BRAF(V600E) mutation is present in 62% of radioactive iodine-resistant thyroid tumors and is associated with downregulation of the sodium-iodide symporter (NIS) and thyroid stimulating hormone receptor (TSHr). We sought to evaluate the combined effect of BRAF inhibition and TSH supplementation on (131)I uptake of BRAF(V600E)-mutant human thyroid cancer cells. MATERIALS AND METHODS WRO cells (a BRAF(V600E)-mutant follicular-derived papillary thyroid carcinoma cell line) were transfected with small interfering RNA targeting BRAF for 72 h in a physiological TSH environment. NIS and TSHr expression were then evaluated at three levels: gene expression, protein levels, and (131)I uptake. These three main outcomes were then reassessed in TSH-depleted media and media supplemented with supratherapeutic concentrations of TSH. RESULTS NIS gene expression increased 5.5-fold 36 h after transfection (P = 0.01), and TSHr gene expression increased 2.8-fold at 24 h (P = 0.02). NIS and TSHr protein levels were similarly increased 48 and 24 h after transfection, respectively. Seventy-two hours after BRAF inhibition, (131)I uptake was unchanged in TSH-depleted media, increased by 7.5-fold (P < 0.01) in physiological TSH media, and increased by 9.1-fold (P < 0.01) in supratherapeutic TSH media. CONCLUSIONS The combined strategy of BRAF inhibition and TSH supplementation results in greater (131)I uptake than when either technique is used alone. This represents a simple and feasible approach that may improve outcomes in patients with radioactive iodine-resistant thyroid carcinomas for which current treatment algorithms are ineffective.
Collapse
Affiliation(s)
- David A Kleiman
- Division of Endocrine and Minimally Invasive Surgery, Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Ras-driven tumorigenesis is assumed to depend on Raf for ERK activation and proliferation; yet, an in vivo requirement for Raf as MEK/ERK activator in this setting has not been demonstrated to date. Here, we show that epidermis-restricted B-Raf ablation restrains the onset and stops the progression of established Ras-driven tumors by limiting MEK/ERK activation and proliferation. Concomitant elimination of B-Raf and Raf-1 enforces the abrupt regression of established tumors owing to the decrease in ERK activation and proliferation caused by B-Raf ablation combined with the ERK-independent increase in Rho-dependent kinase (Rok) signaling and differentiation triggered by Raf-1 inactivation. Thus, B-Raf and Raf-1 have non-redundant functions in Ras-driven tumorigenesis. Of note, Raf kinase inhibitors achieve impressive results in melanomas harboring oncogenic BRAF, but are ineffective against Ras-driven tumors; moreover, therapy-related skin tumors driven by a paradox ERK activation as well as primary and acquired resistance have been reported. Our results suggest that therapies targeting both Raf kinase-dependent and -independent pathways may be effective against a broader range of malignancies and reduce the risks of adverse effects and/or resistance.
Collapse
|
43
|
Nadeau V, Bissonauth V, Charron J. [Mek1 and Mek2 functions in the formation of the blood placental barrier]. Med Sci (Paris) 2012; 28:409-15. [PMID: 22549869 DOI: 10.1051/medsci/2012284019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The ERK/MAPK signaling pathway is involved in several cellular functions. Inactivation in mice of genes encoding members of this pathway is often associated with embryonic death resulting from abnormal placental development. The placenta is essential for nutritional and gaseous exchanges between maternal and embryonic circulations, as well as for the removal of metabolic wastes. These exchanges take place without direct contact between the two circulations. In mice, the hematoplacental barrier consists in a triple layer of trophoblast cells and endothelial cells of the embryo. MEK1 and MEK2 are double specificity serine-threonine/tyrosine kinases responsible for the activation of ERK1 and ERK2. Mek1 inactivation results in placental anomalies due to trophoblast cell proliferation and differentiation defects leading to severe delays in the development of placenta and causing the death of the embryo. Although Mek2(-/-) mutant mice survived without any apparent phenotype, double heterozygous Mek1(+/-)Mek2(+/-) mutants die during gestation from placental malformations. Together, these data emphasize the crucial role of the ERK/MAPK cascade in the formation of extraembryonic structures.
Collapse
Affiliation(s)
- Valérie Nadeau
- Centre de recherche en cancérologie de l'université Laval, centre de recherche du centre hospitalier universitaire de Québec, Québec G1R 2J6, Canada.
| | | | | |
Collapse
|
44
|
Manousaridis I, Mavridou S, Goerdt S, Leverkus M, Utikal J. Cutaneous side effects of inhibitors of the RAS/RAF/MEK/ERK signalling pathway and their management. J Eur Acad Dermatol Venereol 2012; 27:11-8. [PMID: 22540151 DOI: 10.1111/j.1468-3083.2012.04546.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mutations in genes encoding for proteins along the RAS-RAF-MEK-ERK pathway have been detected in a variety of tumor entities, including malignant melanoma, thyroid, colonic and ovarian carcinomas, and some sarcomas. Thus, a number of inhibitors of this pathway have been developed, whose antitumor potential is currently being assessed in different clinical trials. Up to now one drug of this category (vemurafenib) has been approved by the FDA and the European Commission for late-stage melanoma. Although these new targeted anticancer therapies are generally considered to be safe and well tolerated, certain toxicities have been attributed to them, with cutaneous side effects being perhaps the most frequent amongst them. Based on results of clinical trials and on case series, a distinct profile of cutaneous toxicity has been observed, which is similar to that of EGFR and multikinase inhibitors. As exanthema, palmar-plantar erythrodysesthesia syndrome, hyperkeratosis, xerosis, pruritus, photosensitivity, and paronychia, can be controlled in most cases with common conservative modalities, special attention should be given to the early detection of epithelial skin tumors (mainly keratoakanthomas) that can be induced during therapy with these agents. This report reviews all current published data on cutaneous side effects of RAS-RAF-MEK-ERK pathway inhibitors, and attempts to provide the clinician with clear hints for their management.
Collapse
Affiliation(s)
- I Manousaridis
- Department of Dermatology, Venereology and Allergology, University Medical Centre Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | | | | | | | | |
Collapse
|
45
|
Charron J, Bissonauth V, Nadeau V. Implication of MEK1 and MEK2 in the establishment of the blood-placenta barrier during placentogenesis in mouse. Reprod Biomed Online 2012; 25:58-67. [PMID: 22561024 DOI: 10.1016/j.rbmo.2012.02.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 02/02/2012] [Accepted: 02/02/2012] [Indexed: 02/07/2023]
Abstract
The ERK/MAPK signalling cascade is involved in many cellular functions. In mice, the targeted ablation of genes coding for members of this pathway is often associated with embryonic death due to the abnormal development of the placenta. The placenta is essential for nutritional and gaseous exchanges between maternal and embryonic circulations, as well as for the elimination of metabolic waste. These exchanges occur without direct contact between the two circulations. In mice, the blood-placenta barrier consists of a triple layer of trophoblast cells adjacent to endothelial cells from the embryo. In the ERK/MAPK cascade, MEK1 and MEK2 are dual-specificity kinases responsible for the activation of the ERK1 and ERK2 kinases. Inactivation of Mek1 causes placental malformations resulting from defective proliferation and differentiation of the labyrinthine trophoblast cells and leading to a severe delay in the development and the vascularization of the placenta, which explains the embryonic death. Although Mek2(-/-) mutants survive without any apparent phenotype, a large proportion of Mek1(+/-)Mek2(+/-) double heterozygous mutants die during gestation from placenta anomalies affecting the establishment of the blood-placenta barrier. Together, these data reveal how crucial is the role of the ERK/MAPK pathway during the formation of the placenta.
Collapse
Affiliation(s)
- Jean Charron
- Centre de recherche en cancérologie de l'Université Laval, Centre Hospitalier Universitaire de Québec, L'Hôtel-Dieu de Québec, Canada.
| | | | | |
Collapse
|
46
|
Yu S, Graf WD. BRAF gene deletion broadens the clinical spectrum neuro-cardio-facial-cutaneous syndromes. J Child Neurol 2011; 26:1593-6. [PMID: 21862832 DOI: 10.1177/0883073811413830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Point mutations in the human BRAF gene are associated with a group of heterogeneous autosomal dominant neuro-cardio-facial-cutaneous syndromes. We identified a novel 93 kb intragenic deletion of the BRAF gene in a boy with severe developmental encephalopathy using microarray-based comparative genomic hybridization. The unique genotype and phenotype in this patient expands the spectrum of BRAF-related neurodevelopmental disorders. We propose a BRAF gene loss-of-function mechanism to best explain the biological basis of this severe developmental encephalopathy with postnatal growth deficiency.
Collapse
Affiliation(s)
- Shihui Yu
- Department of Pathology, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri, USA
| | | |
Collapse
|
47
|
|
48
|
Sun T, Cao H, Xu L, Zhu B, Gu Q, Xu X. Insulin-Like Growth Factor Binding Protein-Related Protein 1 Mediates VEGF-Induced Proliferation, Migration and Tube Formation of Retinal Endothelial Cells. Curr Eye Res 2011; 36:341-9. [DOI: 10.3109/02713683.2010.545498] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
49
|
Matallanas D, Birtwistle M, Romano D, Zebisch A, Rauch J, von Kriegsheim A, Kolch W. Raf family kinases: old dogs have learned new tricks. Genes Cancer 2011; 2:232-60. [PMID: 21779496 PMCID: PMC3128629 DOI: 10.1177/1947601911407323] [Citation(s) in RCA: 281] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
First identified in the early 1980s as retroviral oncogenes, the Raf proteins have been the objects of intense research. The discoveries 10 years later that the Raf family members (Raf-1, B-Raf, and A-Raf) are bona fide Ras effectors and upstream activators of the ubiquitous ERK pathway increased the interest in these proteins primarily because of the central role that this cascade plays in cancer development. The important role of Raf in cancer was corroborated in 2002 with the discovery of B-Raf genetic mutations in a large number of tumors. This led to intensified drug development efforts to target Raf signaling in cancer. This work yielded not only recent clinical successes but also surprising insights into the regulation of Raf proteins by homodimerization and heterodimerization. Surprising insights also came from the hunt for new Raf targets. Although MEK remains the only widely accepted Raf substrate, new kinase-independent roles for Raf proteins have emerged. These include the regulation of apoptosis by suppressing the activity of the proapoptotic kinases, ASK1 and MST2, and the regulation of cell motility and differentiation by controlling the activity of Rok-α. In this review, we discuss the regulation of Raf proteins and their role in cancer, with special focus on the interacting proteins that modulate Raf signaling. We also describe the new pathways controlled by Raf proteins and summarize the successes and failures in the development of efficient anticancer therapies targeting Raf. Finally, we also argue for the necessity of more systemic approaches to obtain a better understanding of how the Ras-Raf signaling network generates biological specificity.
Collapse
Affiliation(s)
- David Matallanas
- Systems Biology Ireland, University College Dublin, Dublin, Ireland
| | | | | | | | | | | | | |
Collapse
|
50
|
Davis GE, Stratman AN, Sacharidou A. Molecular Control of Vascular Tube Morphogenesis and Stabilization: Regulation by Extracellular Matrix, Matrix Metalloproteinases, and Endothelial Cell–Pericyte Interactions. BIOPHYSICAL REGULATION OF VASCULAR DIFFERENTIATION AND ASSEMBLY 2011. [DOI: 10.1007/978-1-4419-7835-6_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|