1
|
Fang Y, Wan JP, Wang Z, Song SY, Zhang CX, Yang L, Zhang QY, Yan CY, Wu FY, Lu SY, Sun F, Han B, Zhao SX, Dong M, Song HD. Deficiency of the HGF/Met pathway leads to thyroid dysgenesis by impeding late thyroid expansion. Nat Commun 2024; 15:3165. [PMID: 38605010 PMCID: PMC11009301 DOI: 10.1038/s41467-024-47363-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 03/28/2024] [Indexed: 04/13/2024] Open
Abstract
The mechanisms of bifurcation, a key step in thyroid development, are largely unknown. Here we find three zebrafish lines from a forward genetic screening with similar thyroid dysgenesis phenotypes and identify a stop-gain mutation in hgfa and two missense mutations in met by positional cloning from these zebrafish lines. The elongation of the thyroid primordium along the pharyngeal midline was dramatically disrupted in these zebrafish lines carrying a mutation in hgfa or met. Further studies show that MAPK inhibitor U0126 could mimic thyroid dysgenesis in zebrafish, and the phenotypes are rescued by overexpression of constitutively active MEK or Snail, downstream molecules of the HGF/Met pathway, in thyrocytes. Moreover, HGF promotes thyrocyte migration, which is probably mediated by downregulation of E-cadherin expression. The delayed bifurcation of the thyroid primordium is also observed in thyroid-specific Met knockout mice. Together, our findings reveal that HGF/Met is indispensable for the bifurcation of the thyroid primordium during thyroid development mediated by downregulation of E-cadherin in thyrocytes via MAPK-snail pathway.
Collapse
Affiliation(s)
- Ya Fang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Jia-Ping Wan
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Endocrinology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zheng Wang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shi-Yang Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cao-Xu Zhang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Liu Yang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qian-Yue Zhang
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chen-Yan Yan
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Feng-Yao Wu
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Sang-Yu Lu
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Feng Sun
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bing Han
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Shuang-Xia Zhao
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Mei Dong
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Huai-Dong Song
- Department of Molecular Diagnostics & Endocrinology, The Core Laboratory in Medical Center of Clinical Research, Shanghai Ninth People's Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
2
|
Schumacher JA, Wright ZA, Rufin Florat D, Anand SK, Dasyani M, Batta SPR, Laverde V, Ferrari K, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E and ABL signaling regulate blood vessel size. PLoS Genet 2024; 20:e1010851. [PMID: 38190417 PMCID: PMC10798624 DOI: 10.1371/journal.pgen.1010851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/19/2024] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Blood vessels in different vascular beds vary in size, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vessel size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow, eventually leading to the DA collapse. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA size in she mutants correlated with an increased endothelial expression of claudin 5a (cldn5a), which encodes a protein enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates vessel and lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surya Prakash Rao Batta
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Valentina Laverde
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Kaitlin Ferrari
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| |
Collapse
|
3
|
Schumacher JA, Wright ZA, Florat DR, Anand SK, Dasyani M, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E (SHE) and ABL signaling regulate blood vessel size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547455. [PMID: 37461480 PMCID: PMC10349984 DOI: 10.1101/2023.07.03.547455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Blood vessels in different vascular beds vary in lumen diameter, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vascular lumen size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA lumen, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA lumen in she mutants correlated with an increased endothelial expression of claudin 5a and 5b (cldn5a / cldn5b), which encode proteins enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA lumen size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH 45267, USA
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH 45267, USA
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| |
Collapse
|
4
|
Escudero-Flórez M, Torres-Hoyos D, Miranda-Brand Y, Boudreau RL, Gallego-Gómez JC, Vicente-Manzanares M. Dengue Virus Infection Alters Inter-Endothelial Junctions and Promotes Endothelial-Mesenchymal-Transition-Like Changes in Human Microvascular Endothelial Cells. Viruses 2023; 15:1437. [PMID: 37515125 PMCID: PMC10386726 DOI: 10.3390/v15071437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Dengue virus (DENV) is a pathogenic arbovirus that causes human disease. The most severe stage of the disease (severe dengue) is characterized by vascular leakage, hypovolemic shock, and organ failure. Endothelial dysfunction underlies these phenomena, but the causal mechanisms of endothelial dysfunction are poorly characterized. This study investigated the role of c-ABL kinase in DENV-induced endothelial dysfunction. Silencing c-ABL with artificial miRNA or targeting its catalytic activity with imatinib revealed that c-ABL is required for the early steps of DENV infection. DENV-2 infection and conditioned media from DENV-infected cells increased endothelial expression of c-ABL and CRKII phosphorylation, promoted expression of mesenchymal markers, e.g., vimentin and N-cadherin, and decreased the levels of endothelial-specific proteins, e.g., VE-cadherin and ZO-1. These effects were reverted by silencing or inhibiting c-ABL. As part of the acquisition of a mesenchymal phenotype, DENV infection and treatment with conditioned media from DENV-infected cells increased endothelial cell motility in a c-ABL-dependent manner. In conclusion, DENV infection promotes a c-ABL-dependent endothelial phenotypic change that leads to the loss of intercellular junctions and acquisition of motility.
Collapse
Affiliation(s)
- Manuela Escudero-Flórez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia; (M.E.-F.); (D.T.-H.); (Y.M.-B.)
| | - David Torres-Hoyos
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia; (M.E.-F.); (D.T.-H.); (Y.M.-B.)
| | - Yaneth Miranda-Brand
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia; (M.E.-F.); (D.T.-H.); (Y.M.-B.)
| | - Ryan L. Boudreau
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA;
| | - Juan Carlos Gallego-Gómez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia; (M.E.-F.); (D.T.-H.); (Y.M.-B.)
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
5
|
IQGAP1 Is a Phosphotyrosine-Regulated Scaffold for SH2-Containing Proteins. Cells 2023; 12:cells12030483. [PMID: 36766826 PMCID: PMC9913818 DOI: 10.3390/cells12030483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/07/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The scaffold protein IQGAP1 associates with over 150 interactors to influence multiple biological processes. The molecular mechanisms that underly spatial and temporal regulation of these interactions, which are crucial for proper cell functions, remain poorly understood. The receptor tyrosine kinase MET phosphorylates IQGAP1 on Tyr1510. Separately, Src homology 2 (SH2) domains mediate protein-protein interactions by binding specific phosphotyrosine residues. Here, we investigate whether MET-catalyzed phosphorylation of Tyr1510 of IQGAP1 regulates the docking of SH2-containing proteins. Using a peptide array, we identified SH2 domains from several proteins, including the non-receptor tyrosine kinases Abl1 and Abl2, that bind to the Tyr1510 of IQGAP1 in a phosphorylation-dependent manner. Using pure proteins, we validated that full-length Abl1 and Abl2 bind directly to phosphorylated Tyr1510 of IQGAP1. In cells, MET inhibition decreases endogenous IQGAP1 phosphorylation and interaction with endogenous Abl1 and Abl2, indicating that binding is regulated by MET-catalyzed phosphorylation of IQGAP1. Functionally, IQGAP1 modulates basal and HGF-stimulated Abl signaling. Moreover, IQGAP1 binds directly to MET, inhibiting its activation and signaling. Collectively, our study demonstrates that IQGAP1 is a phosphotyrosine-regulated scaffold for SH2-containing proteins, thereby uncovering a previously unidentified mechanism by which IQGAP1 coordinates intracellular signaling.
Collapse
|
6
|
Lan F, Chen X, Xiong Z, Cao Z, Lu L, Zhong Y, Zhan X, Yang Y, Shao Y, Li M, Han Z, Zhu X. Comprehensive transcriptomic and co-expression analysis of ABL1 gene and molecularly targeted drugs in hepatocellular carcinoma based on multi-database mining. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:146. [PMID: 35834027 DOI: 10.1007/s12032-022-01730-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. Consequently, it is essential to identify biomarkers for treatment response and the prognosis prediction. We investigated whether ABL1 can function as a biomarker or a drug target for HCC. We assessed the ABL1 expression, genetic alterations and patients' survival from LinkedOmics, GEO, TCGA and Human Protein Atlas. We analyzed PPI, GO and KEGG pathways. GSEA was analyzed for functional comparison. The current drugs targeting ABL1 were statistically analyzed using DRUGSURV and DGIdb database. We found ABL1 is overexpressed in HCC and its higher expression reduces survival probability. Genetic changes of ABL1 are not frequent. We screened out 25 differentially expressed genes correlated with ABL1. The top functions of ABL1 are biological regulation, metabolic process, protein-containing, and protein binding. KEGG pathways showed that ABL1 and correlated with ABL1 significantly genes markedly enriched in the ErbB signaling pathway, and pathways in cancer. We counted the existing drugs targeting ABL1, which indicates that inhibiting ABL1 expression may improve the survival probability of HCC. In conclusion, ABL1 plays a crucial role in the development and progression of this cancerization and is a potential drug target.
Collapse
Affiliation(s)
- Feifei Lan
- Medical Genetics Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xinqia Chen
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zhuolong Xiong
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zitong Cao
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Liangzong Lu
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yueyuan Zhong
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Xuliang Zhan
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yue Yang
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yingqi Shao
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Minhua Li
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zenglei Han
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, China.
| | - Xiao Zhu
- Zhu's Team, Guangdong Medical University, Zhanjiang, China. .,School of Laboratory Medicine and Biomedical Engineering, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
7
|
Luttman JH, Colemon A, Mayro B, Pendergast AM. Role of the ABL tyrosine kinases in the epithelial-mesenchymal transition and the metastatic cascade. Cell Commun Signal 2021; 19:59. [PMID: 34022881 PMCID: PMC8140471 DOI: 10.1186/s12964-021-00739-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
The ABL kinases, ABL1 and ABL2, promote tumor progression and metastasis in various solid tumors. Recent reports have shown that ABL kinases have increased expression and/or activity in solid tumors and that ABL inactivation impairs metastasis. The therapeutic effects of ABL inactivation are due in part to ABL-dependent regulation of diverse cellular processes related to the epithelial to mesenchymal transition and subsequent steps in the metastatic cascade. ABL kinases target multiple signaling pathways required for promoting one or more steps in the metastatic cascade. These findings highlight the potential utility of specific ABL kinase inhibitors as a novel treatment paradigm for patients with advanced metastatic disease. Video abstract.
Collapse
Affiliation(s)
- Jillian Hattaway Luttman
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Ashley Colemon
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Benjamin Mayro
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| |
Collapse
|
8
|
Wang F, Hou W, Chitsike L, Xu Y, Bettler C, Perera A, Bank T, Cotler SJ, Dhanarajan A, Denning MF, Ding X, Breslin P, Qiang W, Li J, Koleske AJ, Qiu W. ABL1, Overexpressed in Hepatocellular Carcinomas, Regulates Expression of NOTCH1 and Promotes Development of Liver Tumors in Mice. Gastroenterology 2020; 159:289-305.e16. [PMID: 32171747 PMCID: PMC7387191 DOI: 10.1053/j.gastro.2020.03.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 01/31/2020] [Accepted: 03/04/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS We investigated whether ABL proto-oncogene 1, non-receptor tyrosine kinase (ABL1) is involved in development of hepatocellular carcinoma (HCC). METHODS We analyzed clinical and gene expression data from The Cancer Genome Atlas. Albumin-Cre (HepWT) mice and mice with hepatocyte-specific disruption of Abl1 (HepAbl-/- mice) were given hydrodynamic injections of plasmids encoding the Sleeping Beauty transposase and transposons with the MET gene and a catenin β1 gene with an N-terminal truncation, which induces development of liver tumors. Some mice were then gavaged with the ABL1 inhibitor nilotinib or vehicle (control) daily for 4 weeks. We knocked down ABL1 with short hairpin RNAs in Hep3B and Huh7 HCC cells and analyzed their proliferation and growth as xenograft tumors in mice. We performed RNA sequencing and gene set enrichment analysis of tumors. We knocked down or overexpressed NOTCH1 and MYC in HCC cells and analyzed proliferation. We measured levels of phosphorylated ABL1, MYC, and NOTCH1 by immunohistochemical analysis of an HCC tissue microarray. RESULTS HCC tissues had higher levels of ABL1 than non-tumor liver tissues, which correlated with shorter survival times of patients. HepWT mice with the MET and catenin β1 transposons developed liver tumors and survived a median 64 days; HepAbl-/- mice with these transposons developed tumors that were 50% smaller and survived a median 81 days. Knockdown of ABL1 in human HCC cells reduced proliferation, growth as xenograft tumors in mice, and expression of MYC, which reduced expression of NOTCH1. Knockdown of NOTCH1 or MYC in HCC cells significantly reduced cell growth. NOTCH1 or MYC overexpression in human HCC cells promoted proliferation and rescued the phenotype caused by ABL1 knockdown. The level of phosphorylated (activated) ABL1 correlated with levels of MYC and NOTCH1 in human HCC specimens. Nilotinib decreased expression of MYC and NOTCH1 in HCC cell lines, reduced the growth of xenograft tumors in mice, and slowed growth of liver tumors in mice with MET and catenin β1 transposons, reducing tumor levels of MYC and NOTCH1. CONCLUSIONS HCC samples have increased levels of ABL1 compared with nontumor liver tissues, and increased levels of ABL1 correlate with shorter survival times of patients. Loss or inhibition of ABL1 reduces proliferation of HCC cells and slows growth of liver tumors in mice. Inhibitors of ABL1 might be used for treatment of HCC.
Collapse
Affiliation(s)
- Fang Wang
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Wei Hou
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Lennox Chitsike
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Yingchen Xu
- Department of General Surgery, Beijing Tongren Hospital, Capital Medical University
| | - Carlee Bettler
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Aldeb Perera
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Thomas Bank
- Departments of Surgery and Cancer Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Scott J. Cotler
- Department of Medicine, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Asha Dhanarajan
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Mitchell F. Denning
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Xianzhong Ding
- Department of Pathology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Peter Breslin
- Departments of Molecular/Cellular Physiology and Oncology Institute, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA,Department of Biology, Loyola University Chicago Stritch School of Medicine, 2160 South 1st Avenue., Maywood, IL 60153, USA
| | - Wenan Qiang
- Department of Obstetrics and Gynecology and Pathology, Northwestern University
| | - Jun Li
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame
| | | | - Wei Qiu
- Department of Surgery, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois; Department of Cancer Biology, Loyola University Chicago Stritch School of Medicine, Chicago, Illinois.
| |
Collapse
|
9
|
Hu Y, Lyu W, Lowery LA, Koleske AJ. Regulation of MT dynamics via direct binding of an Abl family kinase. J Cell Biol 2019; 218:3986-3997. [PMID: 31699690 PMCID: PMC6891085 DOI: 10.1083/jcb.201812144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/02/2019] [Accepted: 09/30/2019] [Indexed: 12/20/2022] Open
Abstract
Genetic studies revealed that Abl family kinases interact functionally with microtubules, but the mechanism by which Abl kinases regulate microtubules remains unclear. Hu et al. provide the first evidence that the Abl family kinase Abl2 directly binds microtubules to regulate microtubule dynamics. Abl family kinases are essential regulators of cell shape and movement. Genetic studies revealed functional interactions between Abl kinases and microtubules (MTs), but the mechanism by which Abl family kinases regulate MTs remains unclear. Here, we report that Abl2 directly binds to MTs and regulates MT behaviors. Abl2 uses its C-terminal half to bind MTs, an interaction mediated in part through electrostatic binding to tubulin C-terminal tails. Using purified proteins, we found that Abl2 binds growing MTs and promotes MT polymerization and stability. In cells, knockout of Abl2 significantly impairs MT growth, and this defect can be rescued via reexpression of Abl2. Stable reexpression of an Abl2 fragment containing the MT-binding domain alone was sufficient to restore MT growth at the cell edge. These results show Abl2 uses its C-terminal half to bind MTs and directly regulate MT dynamics.
Collapse
Affiliation(s)
- Yuhan Hu
- Department of Cell Biology, Yale University, New Haven, CT
| | - Wanqing Lyu
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT
| | | | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT .,Department of Neuroscience, Yale University, New Haven, CT
| |
Collapse
|
10
|
Lu Y, Lv F, Kong M, Chen X, Duan Y, Chen X, Sun D, Fang M, Xu Y. A cAbl-MRTF-A Feedback Loop Contributes to Hepatic Stellate Cell Activation. Front Cell Dev Biol 2019; 7:243. [PMID: 31681772 PMCID: PMC6805704 DOI: 10.3389/fcell.2019.00243] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/03/2019] [Indexed: 12/13/2022] Open
Abstract
Trans-differentiation of quiescent hepatic stellate cells (HSC) to myofibroblasts is a hallmark event in liver fibrosis. Previous studies have led to the discovery that myocardin-related transcription factor A (MRTF-A) is a key regulator of HSC trans-differentiation or, activation. In the present study we investigated the interplay between MRTF-A and c-Abl (encoded by Abl1), a tyrosine kinase, in this process. We report that hepatic expression levels of c-Abl were down-regulated in MRTF-A knockout (KO) mice compared to wild type (WT) littermates in several different models of liver fibrosis. MRTF-A deficiency also resulted in c-Abl down-regulation in freshly isolated HSCs from the fibrotic livers of mice. MRTF-A knockdown or inhibition repressed c-Abl in cultured HSCs in vitro. Further analyses revealed that MRTF-A directly bound to the Abl1 promoter to activate transcription by interacting with Sp1. Reciprocally, pharmaceutical inhibition of c-Abl suppressed MRTF-A activity. Mechanistically, c-Abl activated extracellular signal-regulated kinase (ERK), which in turn phosphorylated MRTF-A and promoted MRTF-A nuclear trans-localization. In conclusion, our data suggest that a c-Abl-MRTF-A positive feedback loop contributes to HSC activation and liver fibrosis.
Collapse
Affiliation(s)
- Yunjie Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Fangqiao Lv
- Department of Cell Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ming Kong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xuyang Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yunfei Duan
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xuemin Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Donglin Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Mingming Fang
- Institute of Biomedical Research, Liaocheng University, Liaocheng, China.,Department of Clinical Medicine and Laboratory Center for Experimental Medicine, Jiangsu Vocational College of Medicine, Nanjing, China
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Institute of Biomedical Research, Liaocheng University, Liaocheng, China
| |
Collapse
|
11
|
Tan FH, Putoczki TL, Stylli SS, Luwor RB. Ponatinib: a novel multi-tyrosine kinase inhibitor against human malignancies. Onco Targets Ther 2019; 12:635-645. [PMID: 30705592 PMCID: PMC6343508 DOI: 10.2147/ott.s189391] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Human malignancies are often the result of overexpressed and constitutively active receptor and non-receptor tyrosine kinases, which ultimately lead to the mediation of key tumor-driven pathways. Several tyrosine kinases (ie, EGFR, FGFR, PDGFR, VEGFR), are aberrantly activated in most common tumors, including leukemia, glioblastoma, gastrointestinal stromal tumors, non-small-cell lung cancer, and head and neck cancers. Iclusig™ (ponatinib, previously known as AP24534) is an orally active multi-tyrosine kinase inhibitor and is currently approved by the US Food and Drug Administration for patients with chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia, specifically targeting the BCR-ABL gene mutation, T315I. Due to ponatinib's unique multi-targeted characteristics, further studies have demonstrated its ability to target other important tyrosine kinases (FGFR, PDGFR, SRC, RET, KIT, and FLT1) in other human malignancies. This review focuses on the available data of ponatinib and its molecular targets for treatment in various cancers, with a discussion on the broader potential of this agent in other cancer indications.
Collapse
Affiliation(s)
- Fiona H Tan
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia, .,Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Tracy L Putoczki
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia, .,Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, VIC 3050, Australia
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia, .,Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Rodney B Luwor
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3050, Australia,
| |
Collapse
|
12
|
Ratcliffe CDH, Siddiqui N, Coelho PP, Laterreur N, Cookey TN, Sonenberg N, Park M. HGF-induced migration depends on the PI(3,4,5)P 3-binding microexon-spliced variant of the Arf6 exchange factor cytohesin-1. J Cell Biol 2018; 218:285-298. [PMID: 30404949 PMCID: PMC6314551 DOI: 10.1083/jcb.201804106] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 09/19/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022] Open
Abstract
Splice variants of the Arf6 guanine exchange factor cytohesin-1 display differential affinity for PI(4,5)P2 and PI(3,4,5)P3. Ratcliffe et al. show that the specific lipid binding of the diglycine variant of cytohesin-1 is needed for HGF-dependent cell migration and establishment of the leading edge, thereby regulating cancer cell migration following activation of the proto-oncogenic receptor tyrosine kinase Met. Differential inclusion or skipping of microexons is an increasingly recognized class of alternative splicing events. However, the functional significance of microexons and their contribution to signaling diversity is poorly understood. The Met receptor tyrosine kinase (RTK) modulates invasive growth and migration in development and cancer. Here, we show that microexon switching in the Arf6 guanine nucleotide exchange factor cytohesin-1 controls Met-dependent cell migration. Cytohesin-1 isoforms, differing by the inclusion of an evolutionarily conserved three-nucleotide microexon in the pleckstrin homology domain, display differential affinity for PI(4,5)P2 (triglycine) and PI(3,4,5)P3 (diglycine). We show that selective phosphoinositide recognition by cytohesin-1 isoforms promotes distinct subcellular localizations, whereby the triglycine isoform localizes to the plasma membrane and the diglycine to the leading edge. These data highlight microexon skipping as a mechanism to spatially restrict signaling and provide a mechanistic link between RTK-initiated phosphoinositide microdomains and Arf6 during signal transduction and cancer cell migration.
Collapse
Affiliation(s)
- Colin D H Ratcliffe
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Nadeem Siddiqui
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Paula P Coelho
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Nancy Laterreur
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Tumini N Cookey
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Nahum Sonenberg
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada .,Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
13
|
Tripathi R, Liu Z, Plattner R. EnABLing Tumor Growth and Progression: Recent progress in unraveling the functions of ABL kinases in solid tumor cells. CURRENT PHARMACOLOGY REPORTS 2018; 4:367-379. [PMID: 30746323 PMCID: PMC6368175 DOI: 10.1007/s40495-018-0149-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to summarize our current knowledge regarding how ABL family kinases are activated in solid tumors and impact on solid tumor development/progression, with a focus on recent advances in the field. RECENT FINDINGS Although ABL kinases are known drivers of human leukemia, emerging data also implicates the kinases in a large number of solid tumor types where they promote diverse processes such as proliferation, survival, cytoskeletal reorganization, cellular polarity, EMT (epithelial-mesenchymal-transition), metabolic reprogramming, migration, invasion and metastasis via unique signaling pathways. ABL1 and ABL2 appear to have overlapping but also unique roles in driving these processes. In some tumor types, the kinases may act to integrate pro- and anti-proliferative and -invasive signals, and also may serve as a switch during EMT/MET (mesenchymal-epithelial) transitions. CONCLUSIONS Most data indicate that targeting ABL kinases may be effective for reducing tumor growth and preventing metastasis; however, ABL kinases also may have a tumor suppressive role in some tumor types and in some cellular contexts. Understanding the functions of ABL kinases in solid tumors is critical for developing successful clinical trials aimed at targeting ABL kinases for the treatment of solid tumors.
Collapse
Affiliation(s)
- Rakshamani Tripathi
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, Kentucky 40536
| | - Zulong Liu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, Kentucky 40536
| | - Rina Plattner
- Department of Pharmacology and Nutritional Sciences, University of Kentucky School of Medicine, Lexington, Kentucky 40536
| |
Collapse
|
14
|
Golden BO, Griess B, Mir S, Fitzgerald M, Kuperwasser C, Domann F, Teoh-Fitzgerald M. Extracellular superoxide dismutase inhibits hepatocyte growth factor-mediated breast cancer-fibroblast interactions. Oncotarget 2017; 8:107390-107408. [PMID: 29296173 PMCID: PMC5746075 DOI: 10.18632/oncotarget.22379] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/25/2017] [Indexed: 11/25/2022] Open
Abstract
We have previously shown tumor suppressive effects of extracellular superoxide dismutase, EcSOD in breast cancer cells. In this study, an RTK signaling array revealed an inhibitory effect of EcSOD on c-Met phosphorylation and its downstream kinase c-Abl in MDA-MB231 cells. Moreover, an extracellular protein array showed that thrombospondin 1 (TSP-1), a scavenger of the c-Met ligand, hepatocyte growth factor (HGF) is significantly up-regulated in EcSOD overexpressing cells (Ec.20). We further determined the effects of EcSOD on HGF/c-Met-mediated cancer-fibroblast interactions by co-culturing normal fibroblasts (RMF) or RMF which overexpresses HGF (RMF-HGF) with MDA-MB231 cells. We observed that while RMF-HGF significantly promoted Matrigel growth of MDA-MB231, overexpression of EcSOD inhibited the HGF-stimulated growth. Similarly, a SOD mimetic, MnTE-2-PyP, inhibited HGF-induced growth and invasion of MDA-MB231. In addition, a long-term heterotypic co-culture study not only showed that Ec.20 cells are resistant to RMF-HGF-induced invasive stimulation but RMF-HGF that were co-cultured with Ec.20 cells showed an attenuated phenotype, suggesting an oxidative-mediated reciprocal interaction between the two cell types. In addition, we demonstrated that RMF-HGF showed an up-regulation of an ROS-generating enzyme, NADPH oxidase 4 (Nox4). Targeting this pro-oxidant significantly suppressed the activated phenotype of RMF-HGF in a collagen contraction assay, suggesting that RMF-HGF contributes to the oxidative tumor microenvironment. We have further shown that scavenging ROS with EcSOD significantly inhibited RMF-HGF-stimulated orthotopic tumor growth of MDA-MB231. This study suggests the loss of EcSOD in breast cancer plays a pivotal role in promoting the HGF/c-Met-mediated cancer-fibroblast interactions.
Collapse
Affiliation(s)
- Briana Ormsbee Golden
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Brandon Griess
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shakeel Mir
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Matthew Fitzgerald
- Department of Surgery-General Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Charlotte Kuperwasser
- Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Frederick Domann
- Free Radical and Radiation Biology Program, Radiation Oncology, University of Iowa, Iowa City, IA 52241, USA
| | - Melissa Teoh-Fitzgerald
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
15
|
Fredriksson-Lidman K, Van Itallie CM, Tietgens AJ, Anderson JM. Sorbin and SH3 domain-containing protein 2 (SORBS2) is a component of the acto-myosin ring at the apical junctional complex in epithelial cells. PLoS One 2017; 12:e0185448. [PMID: 28961272 PMCID: PMC5621683 DOI: 10.1371/journal.pone.0185448] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/12/2017] [Indexed: 12/30/2022] Open
Abstract
SORBS2 is a scaffolding protein associated with Abl/Arg non-receptor tyrosine kinase pathways and is known to interact with actin and several other cytoskeletal proteins in various cell types. Previous BioID proximity labeling of tight and adherens junction proteins suggested that SORBS2 is a component of the apical junction complex of epithelial cells. We asked whether SORBS2 plays a previously unappreciated role in controlling perijunctional actin and tight junction barrier function. Using super resolution imaging we confirmed that SORBS2 is localized at the apical junction complex but farther from the membrane than ZO-1 and located partially overlapping both the tight- and adherens junctions with a periodic concentration that alternates with myosin IIB in polarized epithelial cells. Overexpression of GFP-SORBS2 recruited alpha-actinin, vinculin and N-WASP, and possibly CIP4 to cellular junctions. However, CRISPR-Cas9 knock-out of SORBS2 did not alter the localization- or immunofluorescent staining intensity of these or several other junctional- and cytoskeletal proteins. SORBS2 knock-out also did not affect the barrier function as measured by TER and dextran flux; nor did it change actin-dependent junction re-assembly as measured by Ca2+-switch and Latrunculin-B wash-out assays. The kinetics of HGF-induced cell scattering and wound healing, and dextran flux increase induced by PDGF also were unaffected by SORBS2 knock-out. SORBS2 concentrates with apical junctional actin that accumulates in response to knock-down of ZO-1 and ZO-2. In spite of our finding that SORBS2 is clearly a component of the apical junction complex, it does not appear to be required for either normal tight- or adherens junction assembly, structure or function or for growth factor-mediated changes in tight junction dynamics.
Collapse
Affiliation(s)
- Karin Fredriksson-Lidman
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Christina M. Van Itallie
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Amber J. Tietgens
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - James M. Anderson
- Laboratory of Tight Junction Structure and Function, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
16
|
HGF/Met Signaling in Cancer Invasion: The Impact on Cytoskeleton Remodeling. Cancers (Basel) 2017; 9:cancers9050044. [PMID: 28475121 PMCID: PMC5447954 DOI: 10.3390/cancers9050044] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/25/2017] [Accepted: 05/02/2017] [Indexed: 12/21/2022] Open
Abstract
The invasion of cancer cells into surrounding tissue and the vasculature is essential for tumor metastasis. Increasing evidence indicates that hepatocyte growth factor (HGF) induces cancer cell migration and invasion. A broad spectrum of mechanisms underlies cancer cell migration and invasion. Cytoskeletal reorganization is of central importance in the development of the phenotype of cancer cells with invasive behavior. Through their roles in cell mechanics, intracellular trafficking, and signaling, cytoskeleton proteins participate in all essential events leading to cell migration. HGF has been involved in cytoskeleton assembly and reorganization, and its role in regulating cytoskeleton dynamics is still expanding. This review summarizes our current understanding of the role of HGF in regulating cytoskeleton remodeling, distribution, and interactions.
Collapse
|
17
|
Abstract
The Abelson tyrosine kinases were initially identified as drivers of leukemia in mice and humans. The Abl family kinases Abl1 and Abl2 regulate diverse cellular processes during development and normal homeostasis, and their functions are subverted during inflammation, cancer and other pathologies. Abl kinases can be activated by multiple stimuli leading to cytoskeletal reorganization required for cell morphogenesis, motility, adhesion and polarity. Depending on the cellular context, Abl kinases regulate cell survival and proliferation. Emerging data support important roles for Abl kinases in pathologies linked to inflammation. Among these are neurodegenerative diseases and inflammatory pathologies. Unexpectedly, Abl kinases have also been identified as important players in mammalian host cells during microbial pathogenesis. Thus, the use of Abl kinase inhibitors might prove to be effective in the treatment of pathologies beyond leukemia and solid tumors. In this Cell Science at a Glance article and in the accompanying poster, we highlight the emerging roles of Abl kinases in the regulation of cellular processes in normal cells and diverse pathologies ranging from cancer to microbial pathogenesis.
Collapse
Affiliation(s)
- Aaditya Khatri
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jun Wang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ann Marie Pendergast
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
18
|
Odenthal J, Takes R, Friedl P. Plasticity of tumor cell invasion: governance by growth factors and cytokines. Carcinogenesis 2016; 37:1117-1128. [PMID: 27664164 DOI: 10.1093/carcin/bgw098] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/15/2016] [Accepted: 09/22/2016] [Indexed: 01/01/2023] Open
Abstract
Tumor cell migration, the basis for metastatic dissemination, is an adaptive process which depends upon coordinated cell interaction with the environment, influencing cell-matrix and cell-cell adhesion, cytoskeletal dynamics and extracellular matrix remodeling. Growth factors and cytokines, released within the reactive tumor microenvironment and their intracellular effector signals strongly impact mechanocoupling functions in tumor cells and thereby control the mode and extent of tumor invasion, including collective and single-cell migration and their interconversions. Besides their role in controlling tumor cell growth and survival, cytokines and growth factors thus provide complex orchestration of the metastatic cascade and tumor cell adaptation to environmental challenge. We here review the mechanisms by which growth factors and cytokines control the reciprocal interactions between tumor cells and their microenvironment, and the consequences for the efficacy and plasticity of invasion programs and metastasis.
Collapse
Affiliation(s)
- Julia Odenthal
- Department of Otorhinolaryngology and Head and Neck Surgery, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands.,Department of Cell Biology, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Robert Takes
- Department of Otorhinolaryngology and Head and Neck Surgery, Radboud University Medical Center, 6525 EX Nijmegen, The Netherlands
| | - Peter Friedl
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands, .,Department of Genitourinary Medical Oncology - Research, Houston, TX 77030, USA and.,Cancer Genomics Center, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
19
|
Koubek EJ, Santy LC. ARF1 and ARF6 regulate recycling of GRASP/Tamalin and the Rac1-GEF Dock180 during HGF-induced Rac1 activation. Small GTPases 2016; 9:242-259. [PMID: 27562622 DOI: 10.1080/21541248.2016.1219186] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Hepatocyte growth factor (HGF) is a potent signaling factor that acts on epithelial cells, causing them to dissociate and scatter. This migration is coordinated by a number of small GTPases, such as ARF6 and Rac1. Active ARF6 is required for HGF-stimulated migration and intracellular levels of ARF6-GTP and Rac1-GTP increase following HGF treatment. During migration, cross talk between ARF6 and Rac1 occurs through formation of a multi-protein complex containing the ARF-GEF cytohesin-2, the scaffolding protein GRASP/Tamalin, and the Rac1-GEF Dock180. Previously, the role of ARF6 in this process was unclear. We have now found that ARF6 and ARF1 regulate trafficking of GRASP and Dock180 to the plasma membrane following HGF treatment. Trafficking of GRASP and Dock180 is impaired by blocking ARF6-mediated recycling pathways and is required for HGF-stimulated Rac1 activation. Finally, HGF treatment stimulates association of GRASP and Dock180. Inhibition of ARF6 trafficking pathways traps GRASP and Dock180 as a complex in the cell.
Collapse
Affiliation(s)
- Emily J Koubek
- a Department of Biochemistry and Molecular Biology , The Pennsylvania State University , University Park , PA , USA
| | - Lorraine C Santy
- a Department of Biochemistry and Molecular Biology , The Pennsylvania State University , University Park , PA , USA
| |
Collapse
|
20
|
Zhang P, Guo ZF, Xu YM, Li YS, Song JG. N-Butylphthalide (NBP) ameliorated cerebral ischemia reperfusion-induced brain injury via HGF-regulated TLR4/NF-κB signaling pathway. Biomed Pharmacother 2016; 83:658-666. [PMID: 27468961 DOI: 10.1016/j.biopha.2016.07.040] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 07/06/2016] [Accepted: 07/08/2016] [Indexed: 12/28/2022] Open
Abstract
N-Butylphthalide (NBP) has been known to have potential neuroprotective effects in Alzheimer's disease and stroke animal models. Hepatocyte-growth factor (HGF), with strong angiogenic properties, exerted protective role in brain injury. The present study was aimed to investigate the possible anti-inflammatory effects of NBP on the brain injury of rats with cerebral ischemia reperfusion (IR) and astrocytes activation induced by lipopolysaccharide (LPS) treatment. Our results showed that cerebral IR induced brain damage with down-regulation of HGF and astrocytes activation. NBP treatment significantly increased HGF expression and activated cMet/PI3K/AKT signaling pathway, stimulating mTOR activity and suppressing apoptosis in brain tissues. Also NBP inhibited pro-inflammatory cytokines expression, including IL-6, IL-1β, and TNFα, via TLR4/NF-κB suppression. Anti-HGF treatment enhanced TLR4 expression while HGF could suppress TLR4 activation and its down-streaming signals, attenuating inflammation finally. Notably, NBP up-regulated HGF and down-regulated TLR4 expression significantly in the astrocytes combined with the treatment of TLR4 inhibitor than the cells only treated with TLR4 inhibitor, suggesting that NBP could further suppress TLR4 activation, suggesting that NBP might impede TLR4 through up-regulating HGF expression. These results suggested that NBP treatment significantly ameliorated cerebral IR-induced brain injury by inhibiting TLR4/NF-κB-associated inflammation regulated by HGF.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan Province, 450052 PR China
| | - Zhen-Fang Guo
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan Province, 450052 PR China
| | - Yu-Ming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan Province, 450052 PR China.
| | - Yu-Sheng Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan Province, 450052 PR China
| | - Jing-Gui Song
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe Road, Zhengzhou City, Henan Province, 450052 PR China
| |
Collapse
|
21
|
Abstract
The Abelson (ABL) tyrosine kinases were identified as drivers of leukemia in mice and humans. Emerging data has shown a role for the ABL family kinases, ABL1 and ABL2, in the progression of several solid tumors. This review will focus on recent reports of the involvement of the ABL kinases in tumor progression using mouse models as well as recent data generated from genomic and proteomic studies linking enhanced expression and hyper-activation of the ABL kinases to some human cancers. Preclinical studies on small molecule inhibitors of the ABL kinases suggest that their use may have beneficial effects for the treatment of selected solid tumors.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710 USA
| | - Ann Marie Pendergast
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710 USA
| |
Collapse
|