1
|
Shen W, Hou Y, Yi Y, Li F, He C, Wang J. G-Clamp Heterocycle Modification Containing Interstrand Photo-Cross-Linker to Capture Intracellular MicroRNA Targets. J Am Chem Soc 2024; 146:12778-12789. [PMID: 38679963 DOI: 10.1021/jacs.4c02901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
MicroRNAs (miRNAs) play indispensable roles in post-transcriptional gene regulation. The identification of target mRNAs is essential for dissecting the recognition basis, dynamics, and regulatory mechanism of miRNA-mRNA interactions. However, the lack of an unbiased method for detecting weak miRNA-mRNA interactions remains a long-standing obstacle for miRNA research. Here, we develop and provide proof-of-concept evidence demonstrating a chemical G-clamp-enhanced photo-cross-linking strategy for covalent capture of intracellular miRNA targets in different cell lines. This approach relies on an aryl-diazirine-G-clamp-modified-nucleoside (ARAGON) miRNA probe containing an alkynyl group that improves the thermal stability of miRNA-target mRNA duplex molecules and can rapidly cross-link with the complementary strand upon UV 365 nm activation, enhancing the transient capture of mRNA targets. After validating the accuracy and binding properties of ARAGON-based miRNA probes through the successful enrichment for the known targets of miR-106a, miR-21, and miR-101, we then extend ARAGON's application to screen for previously unknown targets of different miRNAs in various cell lines. Ultimately, results in this study uncover GAB1 as a target of miR-101 in H1299 lung cancer cells and show that miR-101 silencing of GAB1 can promote apoptosis in H1299 cells, suggesting an oncogenic mechanism of GAB1. This study thus provides a powerful and versatile tool for enhanced screening of global miRNA targets in cells to facilitate investigations of miRNA functions in fundamental cellular processes and disease pathogenesis.
Collapse
Affiliation(s)
- Weiguo Shen
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yongkang Hou
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yunpeng Yi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Fei Li
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
- Howard Hughes Medical Institute, The University of Chicago, 929 East 57th Street, Chicago, Illinois 60637, United States
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
2
|
Sanlav G, Baran B, Kum Özşengezer S, Kizmazoğlu D, Altun Z, Aktaş S, Olgun N. S-100 and MATH-1 Protein Expressions Can Be Useful for the Prediction of Clinical Outcome in Neuroblastoma Patients. J Pediatr Hematol Oncol 2024; 46:21-28. [PMID: 37943051 DOI: 10.1097/mph.0000000000002783] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Neuroblastoma (NB) is the most frequent extracranial solid tumor of childhood, remarkable for its broad spectrum of clinical behavior. This diversity in behavior correlates closely with defined clinical and biological features and combinations of prognostic variables are used for risk-group assignment. S-100 proteins have roles in differentiation and were shown to be frequently dysregulated in NB. MATH-1 protein plays role in neuronal cell differentiation through development. However, up to date, there are no studies evaluating the relationship between MATH-1 and NB. Grb2-associated binding (Gab) proteins have roles in the regulation of cell growth and differentiation. Gab1 was reported to be related to poor survival of high-risk NB patients. The aim of this study was to investigate the relationship between differentiation-related S-100, MATH-1, and Gab1 proteins and risk group and/or stages of NB. A significant relation was found between S-100 and early stages of NB. This study also revealed a significant association between MATH-1 and low-risk groups. S-100 and MATH-1 were also shown to provide survival advantages among stages and risk groups. The findings of this study support the assumption that S-100 and MATH-1 can be potential prognostic biomarkers for staging and risk-group assignment of NB patients. These proteins can be useful tools for clinicians to guide through treatment options, especially for the evaluation of tumor differentiation.
Collapse
Affiliation(s)
| | | | | | - Deniz Kizmazoğlu
- Pediatric Oncology, Dokuz Eylul University Institute of Oncology, İzmir, Turkey
| | | | | | - Nur Olgun
- Pediatric Oncology, Dokuz Eylul University Institute of Oncology, İzmir, Turkey
| |
Collapse
|
3
|
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
|
4
|
Antipsychotics impair regulation of glucose metabolism by central glucose. Mol Psychiatry 2022; 27:4741-4753. [PMID: 36241692 DOI: 10.1038/s41380-022-01798-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Hypothalamic detection of elevated circulating glucose triggers suppression of endogenous glucose production (EGP) to maintain glucose homeostasis. Antipsychotics alleviate symptoms associated with schizophrenia but also increase the risk for impaired glucose metabolism. In the current study, we examined whether two acutely administered antipsychotics from different drug classes, haloperidol (first generation antipsychotic) and olanzapine (second generation antipsychotic), affect the ability of intracerebroventricular (ICV) glucose infusion approximating postprandial levels to suppress EGP. The experimental protocol consisted of a pancreatic euglycemic clamp, followed by kinomic and RNA-seq analyses of hypothalamic samples to determine changes in serine/threonine kinase activity and gene expression, respectively. Both antipsychotics inhibited ICV glucose-mediated increases in glucose infusion rate during the clamp, a measure of whole-body glucose metabolism. Similarly, olanzapine and haloperidol blocked central glucose-induced suppression of EGP. ICV glucose stimulated the vascular endothelial growth factor (VEGF) pathway, phosphatidylinositol 3-kinase (PI3K) pathway, and kinases capable of activating KATP channels in the hypothalamus. These effects were inhibited by both antipsychotics. In conclusion, olanzapine and haloperidol impair central glucose sensing. Although results of hypothalamic analyses in our study do not prove causality, they are novel and provide the basis for a multitude of future studies.
Collapse
|
5
|
Zhang Q, Huang X, Xiu Y, Quan Y, Muhetaer H, Liu T. Src Homology 2 Domain Containing Protein Tyrosine Phosphatase-2 (SHP2) Combined with Dental Pulp Stem Cells Promote the Effect of Angiogenesis. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Pulpitis is one of the most important dental diseases. How to improve the blood circulation in the infected and necrotic area of pulp is the current research hotspot in the treatment of pulpitis. Mesenchymal stem cells (MSCs) have similar regeneration and differentiation ability to
the pluripotent stem cells, and can differentiate into various tissues under certain induced conditions. Dental pulp stem cells (DPSCs) extracted from dental pulp, which have stronger proliferation ability and stability, and are more ideal seed cells for the treatment of pulpitis. Research
show that Src homology 2 domain containing SHP2 can promote blood vessel growth. In this subject, we studied the angiogenesis of SHP2 combined with dental pulp stem cells (DPSCs) transplantation. SHP2 and DPSCs were co cultured with human umbilical vein endothelial cells (HUVECs). The proliferation
and migration of endothelial cells were detected by Wound Healing Assays. At the same time, the effect of SHP2+DPSC on endothelial cell angiogenesis was examined by tube formation test. The expression of angiogenesis related cytokines including vascular endothelial growth factor (VEGF),
von willebrand factor (vWF), Angiopoietin-1 (Ang-1) and Cdc42/Rac1 signal pathway were also detected by Western blot. Our results demonstrated that SHP2 combined with DPSCs can advance endothelial cell angiogenesis. Meanwhile, SHP2+ DPSC obviously increased VEGF, Ang-1 and vWF expression.
SHP2+DPSC significantly raise the Cdc42/Rac1 signal pathway in HUVECs. Our data illustrate that SHP2 combined with DPSCs can promote the effect of angiogenesis in pulpitis.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Oncology Rehabilitation, Shenzhen Luohu People’s Hospital, The 3th Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518001, P. R. China
| | - Xing Huang
- Department of Stomatology, Shenzhen Luohu People’s Hospital, The 3th Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518001, P. R. China
| | - Yihong Xiu
- Somatotherapy Department, Shenzhen Kangning Hospital, Shenzhen, Guangdong, 518001, P. R. China
| | - Yaping Quan
- Department of Neurology, Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Guangdong, 518001, P. R. China
| | - Huojia Muhetaer
- Department of Stomatology, Shenzhen Luohu People’s Hospital, The 3th Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518001, P. R. China
| | - Tao Liu
- Department of Oncology Rehabilitation, Shenzhen Luohu People’s Hospital, The 3th Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518001, P. R. China
| |
Collapse
|
6
|
Zhang Y, Wang D, Zhao Z, Liu L, Xia G, Ye T, Chen Y, Xu C, Jin X, Shen C. Nephronectin promotes cardiac repair post myocardial infarction via activating EGFR/JAK2/STAT3 pathway. Int J Med Sci 2022; 19:878-892. [PMID: 35693734 PMCID: PMC9149649 DOI: 10.7150/ijms.71780] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/01/2022] [Indexed: 11/05/2022] Open
Abstract
Background: ECM proteins are instrumental for angiogenesis, which plays momentous roles during development and repair in various organs, including post cardiac insult. After a screening based on an open access RNA-seq database, we identified Nephronectin (NPNT), an extracellular protein, might be involved in cardiac repair post myocardial infarction (MI). However, the specific impact of nephronectin during cardiac repair in MI remains elusive. Methods and Results: In the present study, we established a system overexpressing NPNT locally in mouse heart by utilizing a recombinant adeno-associated virus. One-to-four weeks post MI induction, we observed improved cardiac function, limited infarct size, alleviated cardiac fibrosis, with promoted angiogenesis in infarct border zone in NPNT overexpressed mice. And NPNT treatment enhanced human umbilical vascular endothelial cell (HUVEC) migration and tube formation, putatively through advocating phosphorylation of EGFR/JAK2/STAT3. The migration and capillary-like tube formation events could be readily revoked by EGFR or STAT3 inhibition. Notably, phosphorylation of EGFR, JAK2 and STAT3 were markedly upregulated in AAV2/9-cTnT-NPNT-treated mice with MI. Conclusions: Our study thus identifies the beneficial effects of NPNT on angiogenesis and cardiac repair post MI by enhancing the EGFR/JAK2/STAT3 signaling pathway, implying the potential therapeutic application of NPNT on myocardial dysfunction post MI.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Di Wang
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zhe Zhao
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Liang Liu
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Guofang Xia
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Tianbao Ye
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yu Chen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Congfeng Xu
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Xian Jin
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| |
Collapse
|
7
|
Xu Z, Guo C, Ye Q, Shi Y, Sun Y, Zhang J, Huang J, Huang Y, Zeng C, Zhang X, Ke Y, Cheng H. Endothelial deletion of SHP2 suppresses tumor angiogenesis and promotes vascular normalization. Nat Commun 2021; 12:6310. [PMID: 34728626 PMCID: PMC8564544 DOI: 10.1038/s41467-021-26697-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
SHP2 mediates the activities of multiple receptor tyrosine kinase signaling and its function in endothelial processes has been explored extensively. However, genetic studies on the role of SHP2 in tumor angiogenesis have not been conducted. Here, we show that SHP2 is activated in tumor endothelia. Shp2 deletion and pharmacological inhibition reduce tumor growth and microvascular density in multiple mouse tumor models. Shp2 deletion also leads to tumor vascular normalization, indicated by increased pericyte coverage and vessel perfusion. SHP2 inefficiency impairs endothelial cell proliferation, migration, and tubulogenesis through downregulating the expression of proangiogenic SRY-Box transcription factor 7 (SOX7), whose re-expression restores endothelial function in SHP2-knockdown cells and tumor growth, angiogenesis, and vascular abnormalization in Shp2-deleted mice. SHP2 stabilizes apoptosis signal-regulating kinase 1 (ASK1), which regulates SOX7 expression mediated by c-Jun. Our studies suggest SHP2 in tumor associated endothelial cells is a promising anti-angiogenic target for cancer therapy.
Collapse
Affiliation(s)
- Zhiyong Xu
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XThe Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Chunyi Guo
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaoli Ye
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yueli Shi
- grid.13402.340000 0004 1759 700XThe Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Yihui Sun
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Zhang
- grid.13402.340000 0004 1759 700XDepartment of Urology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Huang
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yizhou Huang
- grid.13402.340000 0004 1759 700XDepartment of Gynecology of Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunlai Zeng
- grid.469539.40000 0004 1758 2449Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Xue Zhang
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XDepartment of Respiratory Medicine of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuehai Ke
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XDepartment of Respiratory Medicine of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, China
| | - Hongqiang Cheng
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XDepartment of Cardiology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Ye Q, Jiang H, Lan Y, Wang M, Mao D. Therapeutic Effects and Associated Mechanisms by which "Fuyang Jiedu Huayu" Granules Treat Chronic Liver Failure in the Rat. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:7634673. [PMID: 33959189 PMCID: PMC8075679 DOI: 10.1155/2021/7634673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/23/2020] [Accepted: 04/04/2021] [Indexed: 11/18/2022]
Abstract
AIM Fuyang Jiedu Huayu (FYJDHY) granules are a combination of five traditional Chinese medicines with known therapeutic effects against chronic liver failure (CLF). The aim of the present study was to investigate the efficacy of FYJDHY to ameliorate the effects of carbon tetrachloride- (CCl4-) induced CLF in rats and to explore the possible molecular mechanisms underlying its therapeutic efficacy. METHODS A model of chronic liver failure was established by intraperitoneal injection of 50% carbon tetrachloride into SD rats for 8 weeks. After establishing the model, rats were treated with either low-dose (4.725 kg/d), medium-dose (9.45 kg/d), or high-dose (18.9 g/kg/d) FYJDHY for 2 weeks. After treatment, samples of liver tissue and blood were harvested from rats in each group. Serum ALT, AST, and TBIL levels and prothrombin time were measured using a biochemical analyzer. The expression of Gab1 (Grb2-associated binder 1), TPO (thrombopoietin), and its receptor c-Mpl were measured using quantitative real-time PCR (RT-PCR) and Western blot analysis, and assessment of histological improvement in liver tissue was by H&E-stained tissue sections. RESULTS Compared with the model group, serum ALT, AST, and TBIL levels and PT of rats in the intervention group were significantly reduced (P < 0.05). In addition, FYJDHY alleviated pathological damage to liver tissue and increased the expression of Gab1, TPO, and its receptor c-Mpl in liver tissue, to levels statistically significant compared with the model group (P < 0.05). CONCLUSIONS The therapeutic effect of FYJDHY on CLF may be related to the promotion of angiogenesis and improvement in hemopoietic function in individuals suffering from CLF.
Collapse
Affiliation(s)
- Qianling Ye
- Guangxi University of Traditional Chinese Medicine, NanNing, GuangXi 530222, China
| | - Hainan Jiang
- Guangxi University of Traditional Chinese Medicine, NanNing, GuangXi 530222, China
| | - Yanmei Lan
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, NanNing, GuangXi 530023, China
| | - Minggang Wang
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, NanNing, GuangXi 530023, China
| | - Dewen Mao
- The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, NanNing, GuangXi 530023, China
| |
Collapse
|
9
|
Protein kinase A negatively regulates VEGF-induced AMPK activation by phosphorylating CaMKK2 at serine 495. Biochem J 2021; 477:3453-3469. [PMID: 32869834 DOI: 10.1042/bcj20200555] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/19/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
Activation of AMP-activated protein kinase (AMPK) in endothelial cells by vascular endothelial growth factor (VEGF) via the Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2) represents a pro-angiogenic pathway, whose regulation and function is incompletely understood. This study investigates whether the VEGF/AMPK pathway is regulated by cAMP-mediated signalling. We show that cAMP elevation in endothelial cells by forskolin, an activator of the adenylate cyclase, and/or 3-isobutyl-1-methylxanthine (IBMX), an inhibitor of phosphodiesterases, triggers protein kinase A (PKA)-mediated phosphorylation of CaMKK2 (serine residues S495, S511) and AMPK (S487). Phosphorylation of CaMKK2 by PKA led to an inhibition of its activity as measured in CaMKK2 immunoprecipitates of forskolin/IBMX-treated cells. This inhibition was linked to phosphorylation of S495, since it was not seen in cells expressing a non-phosphorylatable CaMKK2 S495C mutant. Phosphorylation of S511 alone in these cells was not able to inhibit CaMKK2 activity. Moreover, phosphorylation of AMPK at S487 was not sufficient to inhibit VEGF-induced AMPK activation in cells, in which PKA-mediated CaMKK2 inhibition was prevented by expression of the CaMKK2 S495C mutant. cAMP elevation in endothelial cells reduced basal and VEGF-induced acetyl-CoA carboxylase (ACC) phosphorylation at S79 even if AMPK was not inhibited. Together, this study reveals a novel regulatory mechanism of VEGF-induced AMPK activation by cAMP/PKA, which may explain, in part, inhibitory effects of PKA on angiogenic sprouting and play a role in balancing pro- and anti-angiogenic mechanisms in order to ensure functional angiogenesis.
Collapse
|
10
|
Verin AD, Batori R, Kovacs-Kasa A, Cherian-Shaw M, Kumar S, Czikora I, Karoor V, Strassheim D, Stenmark KR, Gerasimovskaya EV. Extracellular adenosine enhances pulmonary artery vasa vasorum endothelial cell barrier function via Gi/ELMO1/Rac1/PKA-dependent signaling mechanisms. Am J Physiol Cell Physiol 2020; 319:C183-C193. [PMID: 32432925 DOI: 10.1152/ajpcell.00505.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The vasa vasorum (VV), the microvascular network around large vessels, has been recognized as an important contributor to the pathological vascular remodeling in cardiovascular diseases. In bovine and rat models of hypoxic pulmonary hypertension (PH), we have previously shown that chronic hypoxia profoundly increased pulmonary artery (PA) VV permeability, associated with infiltration of inflammatory and progenitor cells in the arterial wall, perivascular inflammation, and structural vascular remodeling. Extracellular adenosine was shown to exhibit a barrier-protective effect on VV endothelial cells (VVEC) via cAMP-independent mechanisms, which involved adenosine A1 receptor-mediated activation of Gi-phosphoinositide 3-kinase-Akt pathway and actin cytoskeleton remodeling. Using VVEC isolated from the adventitia of calf PA, in this study we investigated in more detail the mechanisms linking Gi activation to downstream barrier protection pathways. Using a small-interference RNA (siRNA) technique and transendothelial electrical resistance assay, we found that the adaptor protein, engulfment and cell motility 1 (ELMO1), the tyrosine phosphatase Src homology region 2 domain-containing phosphatase-2, and atypical Gi- and Rac1-mediated protein kinase A activation are implicated in VVEC barrier enhancement. In contrast, the actin-interacting GTP-binding protein, girdin, and the p21-activated kinase 1 downstream target, LIM kinase, are not involved in this response. In addition, adenosine-dependent cytoskeletal rearrangement involves activation of cofilin and inactivation of ezrin-radixin-moesin regulatory cytoskeletal proteins, consistent with a barrier-protective mechanism. Collectively, our data indicate that targeting adenosine receptors and downstream barrier-protective pathways in VVEC may have a potential translational significance in developing pharmacological approach for the VV barrier protection in PH.
Collapse
Affiliation(s)
| | - Robert Batori
- Augusta University Vascular Biology Center, Augusta, Georgia
| | | | | | - Sanjiv Kumar
- Augusta University Vascular Biology Center, Augusta, Georgia
| | - Istvan Czikora
- Augusta University Vascular Biology Center, Augusta, Georgia
| | - Vijaya Karoor
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Derek Strassheim
- Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Kurt R Stenmark
- Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | | |
Collapse
|
11
|
Huang J, Cai C, Zheng T, Wu X, Wang D, Zhang K, Xu B, Yan R, Gong H, Zhang J, Shi Y, Xu Z, Zhang X, Zhang X, Shang T, Zhou J, Guo X, Zeng C, Lai EY, Xiao C, Chen J, Wan S, Liu WH, Ke Y, Cheng H. Endothelial Scaffolding Protein ENH (Enigma Homolog Protein) Promotes PHLPP2 (Pleckstrin Homology Domain and Leucine-Rich Repeat Protein Phosphatase 2)-Mediated Dephosphorylation of AKT1 and eNOS (Endothelial NO Synthase) Promoting Vascular Remodeling. Arterioscler Thromb Vasc Biol 2020; 40:1705-1721. [PMID: 32268790 DOI: 10.1161/atvbaha.120.314172] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE A decrease in nitric oxide, leading to vascular smooth muscle cell proliferation, is a common pathological feature of vascular proliferative diseases. Nitric oxide synthesis by eNOS (endothelial nitric oxide synthase) is precisely regulated by protein kinases including AKT1. ENH (enigma homolog protein) is a scaffolding protein for multiple protein kinases, but whether it regulates eNOS activation and vascular remodeling remains unknown. Approach and Results: ENH was upregulated in injured mouse arteries and human atherosclerotic plaques and was associated with coronary artery disease. Neointima formation in carotid arteries, induced by ligation or wire injury, was greatly decreased in endothelium-specific ENH-knockout mice. Vascular ligation reduced AKT and eNOS phosphorylation and nitric oxide production in the endothelium of control but not ENH-knockout mice. ENH was found to interact with AKT1 and its phosphatase PHLPP2 (pleckstrin homology domain and leucine-rich repeat protein phosphatase 2). AKT and eNOS activation were prolonged in VEGF (vascular endothelial growth factor)-induced ENH- or PHLPP2-deficient endothelial cells. Inhibitors of either AKT or eNOS effectively restored ligation-induced neointima formation in ENH-knockout mice. Moreover, endothelium-specific PHLPP2-knockout mice displayed reduced ligation-induced neointima formation. Finally, PHLPP2 was increased in the endothelia of human atherosclerotic plaques and blood cells from patients with coronary artery disease. CONCLUSIONS ENH forms a complex with AKT1 and its phosphatase PHLPP2 to negatively regulate AKT1 activation in the artery endothelium. AKT1 deactivation, a decrease in nitric oxide generation, and subsequent neointima formation induced by vascular injury are mediated by ENH and PHLPP2. ENH and PHLPP2 are thus new proatherosclerotic factors that could be therapeutically targeted.
Collapse
Affiliation(s)
- Jiaqi Huang
- From the Department of Pathology and Pathophysiology and Department of Cardiology, Sir Run Run Shaw Hospital (J.H., K.Z., H.C.), Zhejiang University School of Medicine, Hangzhou, China
| | - Changhong Cai
- Department of Cardiology, Lishui Hospital, Zhejiang University School of Medicine, China. (C.C., C.Z.)
| | - Tianyu Zheng
- Department of Pathology and Pathophysiology (T.Z., X. Wu, R.Y., Y.S., Z.X., X.Z., Y.K.), Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyan Wu
- Department of Pathology and Pathophysiology (T.Z., X. Wu, R.Y., Y.S., Z.X., X.Z., Y.K.), Zhejiang University School of Medicine, Hangzhou, China
| | - Dongfei Wang
- Department of Cardiovascular Science, The First Affiliated Hospital of Zhejiang University (D.W., X.G.), Zhejiang University School of Medicine, Hangzhou, China
| | - Kaijie Zhang
- From the Department of Pathology and Pathophysiology and Department of Cardiology, Sir Run Run Shaw Hospital (J.H., K.Z., H.C.), Zhejiang University School of Medicine, Hangzhou, China
| | - Bocheng Xu
- Institute of Feed Science, College of Animal Sciences, Zhejiang University, Hangzhou, China (B.X.)
| | - Ruochen Yan
- Department of Pathology and Pathophysiology (T.Z., X. Wu, R.Y., Y.S., Z.X., X.Z., Y.K.), Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Gong
- Key Laboratory for Translational Medicine, First Affiliated Hospital, Huzhou University, China (H.G.)
| | - Jie Zhang
- Department of Urology, Sir Run Run Shaw Hospital (J. Zhang), Zhejiang University School of Medicine, Hangzhou, China
| | - Yueli Shi
- Department of Pathology and Pathophysiology (T.Z., X. Wu, R.Y., Y.S., Z.X., X.Z., Y.K.), Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyong Xu
- Department of Pathology and Pathophysiology (T.Z., X. Wu, R.Y., Y.S., Z.X., X.Z., Y.K.), Zhejiang University School of Medicine, Hangzhou, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology (T.Z., X. Wu, R.Y., Y.S., Z.X., X.Z., Y.K.), Zhejiang University School of Medicine, Hangzhou, China
| | - Xuemin Zhang
- Department of Vascular Surgery, Peking University People's Hospital, Peking University Health Science Center, Peking University, Beijing, China (X. Zhang)
| | - Tao Shang
- Department of Vascular Surgery, The First Affiliated Hospital (T.S.)
| | - Jianhong Zhou
- Department of Gynecology, School of Medicine, Zhejiang University, Hangzhou, China (J. Zhou)
| | - Xiaogang Guo
- Department of Cardiovascular Science, The First Affiliated Hospital of Zhejiang University (D.W., X.G.), Zhejiang University School of Medicine, Hangzhou, China
| | - Chunlai Zeng
- Department of Cardiology, Lishui Hospital, Zhejiang University School of Medicine, China. (C.C., C.Z.)
| | - En Yin Lai
- Department of Physiology, School of Basic Medical Sciences (E.Y.L.), Zhejiang University School of Medicine, Hangzhou, China
| | - Changchun Xiao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China (C.X., W.-H.L.).,Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA (C.X.)
| | - Ju Chen
- Department of Medicine and Cardiology, University of California San Diego, La Jolla (J.C.)
| | - Shu Wan
- Brain Center of Zhejiang Hospital, Hangzhou, China (S.W.)
| | - Wen-Hsien Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, China (C.X., W.-H.L.)
| | - Yuehai Ke
- Department of Pathology and Pathophysiology (T.Z., X. Wu, R.Y., Y.S., Z.X., X.Z., Y.K.), Zhejiang University School of Medicine, Hangzhou, China
| | - Hongqiang Cheng
- From the Department of Pathology and Pathophysiology and Department of Cardiology, Sir Run Run Shaw Hospital (J.H., K.Z., H.C.), Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, China (H.C.)
| |
Collapse
|
12
|
Lu NN, Tan C, Sun NH, Shao LX, Liu XX, Gao YP, Tao RR, Jiang Q, Wang CK, Huang JY, Zhao K, Wang GF, Liu ZR, Fukunaga K, Lu YM, Han F. Cholinergic Grb2-Associated-Binding Protein 1 Regulates Cognitive Function. Cereb Cortex 2019; 28:2391-2404. [PMID: 28591834 DOI: 10.1093/cercor/bhx141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 05/21/2017] [Indexed: 12/21/2022] Open
Abstract
Grb2-associated-binding protein 1 (Gab1) is a docking/scaffolding molecule known to play an important role in cell growth and survival. Here, we report that Gab1 is decreased in cholinergic neurons in Alzheimer's disease (AD) patients and in a mouse model of AD. In mice, selective ablation of Gab1 in cholinergic neurons in the medial septum impaired learning and memory and hippocampal long-term potentiation. Gab1 ablation also inhibited SK channels, leading to an increase in firing in septal cholinergic neurons. Gab1 overexpression, on the other hand, improved cognitive function and restored hippocampal CaMKII autorphosphorylation in AD mice. These results suggest that Gab1 plays an important role in the pathophysiology of AD and may represent a novel therapeutic target for diseases involving cholinergic dysfunction.
Collapse
Affiliation(s)
- Nan-Nan Lu
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chao Tan
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ning-He Sun
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ling-Xiao Shao
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiu-Xiu Liu
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China.,School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Yin-Ping Gao
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China.,School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China
| | - Rong-Rong Tao
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Quan Jiang
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cheng-Kun Wang
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ji-Yun Huang
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Kui Zhao
- Department of PET Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guang-Fa Wang
- Department of PET Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhi-Rong Liu
- Department of Neurology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-ku, Sendai, Japan
| | - Ying-Mei Lu
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang, China.,Key Laboratory of Medical Neurobiology of Ministry of Health of China, Department of Neurobiology,Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feng Han
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Bandara N, Gurusinghe S, Kong A, Mitchell G, Wang LX, Lim SY, Strappe P. Generation of a nitric oxide signaling pathway in mesenchymal stem cells promotes endothelial lineage commitment. J Cell Physiol 2019; 234:20392-20407. [PMID: 30997675 DOI: 10.1002/jcp.28640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022]
Abstract
Enhancing differentiation of mesenchymal stem cells (MSCs) to endothelial cells may improve their ability to vascularize tissue and promote wound healing. This study describes a novel role for nitric oxide (NO) in reprogramming MSCs towards an endothelial lineage and highlights the role of Wnt signaling and epigenetic modification by NO. Rat MSCs were transduced with lentiviral vectors expressing endothelial nitric oxide synthase (pLV-eNOS) and a mutated caveolin gene (pLV-CAV-1F92A ) to enhance NO generation resulting in increased in vitro capillary tubule formation and endothelial marker gene expression. An exogenous source of NO could also stimulate CD31 expression in MSCs. NO was associated with an arterial-specific endothelial gene expression profile of Notch1, Dll4, and Hey2 and significantly reduced expression of venous markers. Wnt signaling associated with NO was evident through increased gene expression of Wnt3a and β-catenin protein, and expression of the endothelial marker Pecam-1 could be significantly reduced by treatment with the Wnt signaling inhibitor Dkk-1. The role of NO as an epigenetic modifier was evident with reduced gene expression of the methyltransferase, DNMT1, and bisulfite sequencing of the endothelial Flt1 promoter region in NO-producing MSCs showed significant demethylation compared to control cells. Finally, subcutaneous implantation of NO-producing MSCs seeded in a biomaterial scaffold (NovoSorb®) resulted in survival of transplanted cells and the formation of blood vessels. In summary, this study describes, NO as a potent endothelial programming factor which acts as an epigenetic modifier in MSCs and may provide a novel platform for vascular regenerative therapy.
Collapse
Affiliation(s)
- Nadeeka Bandara
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Saliya Gurusinghe
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia.,School of Agricultural and Wine Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Anne Kong
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Geraldine Mitchell
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia.,Faculty of Health Sciences, Australian Catholic University, Fitzroy, Victoria, Australia
| | - Le-Xin Wang
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | - Padraig Strappe
- School of Health, Medicine and Applied Sciences, Central Queensland University, Rockhampton, Queensland, Australia
| |
Collapse
|
14
|
Prakash R, Izraely S, Thareja NS, Lee RH, Rappaport M, Kawaguchi R, Sagi-Assif O, Ben-Menachem S, Meshel T, Machnicki M, Ohe S, Hoon DS, Coppola G, Witz IP, Carmichael ST. Regeneration Enhances Metastasis: A Novel Role for Neurovascular Signaling in Promoting Melanoma Brain Metastasis. Front Neurosci 2019; 13:297. [PMID: 31024232 PMCID: PMC6465799 DOI: 10.3389/fnins.2019.00297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022] Open
Abstract
Neural repair after stroke involves initiation of a cellular proliferative program in the form of angiogenesis, neurogenesis, and molecular growth signals in the surrounding tissue elements. This cellular environment constitutes a niche in which regeneration of new blood vessels and new neurons leads to partial tissue repair after stroke. Cancer metastasis has similar proliferative cellular events in the brain and other organs. Do cancer and CNS tissue repair share similar cellular processes? In this study, we identify a novel role of the regenerative neurovascular niche induced by stroke in promoting brain melanoma metastasis through enhancing cellular interactions with surrounding niche components. Repair-mediated neurovascular signaling induces metastatic cells to express genes crucial to metastasis. Mimicking stroke-like conditions in vitro displays an enhancement of metastatic migration potential and allows for the determination of cell-specific signals produced by the regenerative neurovascular niche. Comparative analysis of both in vitro and in vivo expression profiles reveals a major contribution of endothelial cells in mediating melanoma metastasis. These results point to a previously undiscovered role of the regenerative neurovascular niche in shaping the tumor microenvironment and brain metastatic landscape.
Collapse
Affiliation(s)
- Roshini Prakash
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sivan Izraely
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nikita S Thareja
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rex H Lee
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maya Rappaport
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Riki Kawaguchi
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Orit Sagi-Assif
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shlomit Ben-Menachem
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tsipi Meshel
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michal Machnicki
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shuichi Ohe
- Department of Translational Molecular Medicine, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Dave S Hoon
- Department of Translational Molecular Medicine, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, CA, United States
| | - Giovanni Coppola
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, United States
| | - Isaac P Witz
- Department of Cell Research and Immunology, School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Dong H, Weng C, Bai R, Sheng J, Gao X, Li L, Xu Z. The regulatory network of miR-141 in the inhibition of angiogenesis. Angiogenesis 2018; 22:251-262. [DOI: 10.1007/s10456-018-9654-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 11/12/2018] [Indexed: 12/15/2022]
|
16
|
Sun J, Huang W, Yang SF, Zhang XP, Yu Q, Zhang ZQ, Yao J, Li KR, Jiang Q, Cao C. Gαi1 and Gαi3mediate VEGF-induced VEGFR2 endocytosis, signaling and angiogenesis. Theranostics 2018; 8:4695-4709. [PMID: 30279732 PMCID: PMC6160771 DOI: 10.7150/thno.26203] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/17/2018] [Indexed: 12/21/2022] Open
Abstract
VEGF binding to VEGFR2 leads to VEGFR2 endocytosis and downstream signaling activation to promote angiogenesis. Methods: Using genetic strategies, we tested the requirement of α subunits of heterotrimeric G proteins (Gαi1/3) in the process. Results: Gαi1/3 are located in the VEGFR2 endocytosis complex (VEGFR2-Ephrin-B2-Dab2-PAR-3), where they are required for VEGFR2 endocytosis and downstream signaling transduction. Gαi1/3 knockdown, knockout or dominant negative mutation inhibited VEGF-induced VEGFR2 endocytosis, and downstream Akt-mTOR and Erk-MAPK activation. Functional studies show that Gαi1/3 shRNA inhibited VEGF-induced proliferation, invasion, migration and vessel-like tube formation of HUVECs. In vivo, Gαi1/3 shRNA lentivirus inhibited alkali burn-induced neovascularization in mouse cornea. Further, oxygen-induced retinopathy (OIR)-induced retinal neovascularization was inhibited by intravitreal injection of Gαi1/3 shRNA lentivirus. Moreover, in vivo angiogenesis by alkali burn and OIR was significantly attenuated in Gαi1/3 double knockout mice. Significantly, Gαi1/3 proteins are upregulated in proliferative retinal tissues of proliferative diabetic retinopathy (PDR) patients. Conclusion: These results provide mechanistic insights into the critical role played by Gαi1/3 proteins in VEGF-induced VEGFR2 endocytosis, signaling and angiogenesis.
Collapse
|
17
|
Zhang XP, Li KR, Yu Q, Yao MD, Ge HM, Li XM, Jiang Q, Yao J, Cao C. Ginsenoside Rh2 inhibits vascular endothelial growth factor-induced corneal neovascularization. FASEB J 2018; 32:3782-3791. [PMID: 29465315 DOI: 10.1096/fj.201701074rr] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
VEGF-induced neovascularization plays a pivotal role in corneal neovascularization (CoNV). The current study investigated the potential effect of ginsenoside Rh2 (GRh2) on neovascularization. In HUVECs, pretreatment with GRh2 largely attenuated VEGF-induced cell proliferation, migration, and vessel-like tube formation in vitro. At the molecular level, GRh2 disrupted VEGF-induced VEGF receptor 2 (VEGFR2)-Grb-2-associated binder 1 (Gab1) association in HUVECs, causing inactivation of downstream AKT and ERK signaling. Gab1 knockdown (by targeted short hairpin RNA) similarly inhibited HUVEC proliferation and migration. Notably, GRh2 was ineffective against VEGF in Gab1-silenced HUVECs. In a mouse cornea alkali burn model, GRh2 eyedrops inhibited alkali-induced neovascularization and inflammatory cell infiltrations in the cornea. Furthermore, alkali-induced corneal expression of mRNAs/long noncoding RNAs in cornea were largely attenuated by GRh2. Overall, GRh2 inhibits VEGF-induced angiogenic effect via inhibiting VEGFR2-Gab1 signaling in vitro. It also alleviates angiogenic and inflammatory responses in alkali burn-treated mouse corneas.-Zhang, X.-P., Li, K.-R., Yu, Q., Yao, M.-D., Ge, H.-M., Li, X.-M., Jiang, Q., Yao, J., Cao, C. Ginsenoside Rh2 inhibits vascular endothelial growth factor-induced corneal neovascularization.
Collapse
Affiliation(s)
- Xiao-Pei Zhang
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Ke-Ran Li
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Qing Yu
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Mu-Di Yao
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Hui-Min Ge
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xiu-Miao Li
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Jin Yao
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Cong Cao
- The Fourth School of Clinical Medicine, The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Neuropsychiatric Diseases Research and Institute of Neuroscience, Soochow University, Suzhou, China; and.,North District, The Municipal Hospital of Suzhou, Suzhou, China
| |
Collapse
|
18
|
Nedvetsky PI, Zhao X, Mathivet T, Aspalter IM, Stanchi F, Metzger RJ, Mostov KE, Gerhardt H. cAMP-dependent protein kinase A (PKA) regulates angiogenesis by modulating tip cell behavior in a Notch-independent manner. Development 2017; 143:3582-3590. [PMID: 27702786 DOI: 10.1242/dev.134767] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 08/08/2016] [Indexed: 01/06/2023]
Abstract
cAMP-dependent protein kinase A (PKA) is a ubiquitously expressed serine/threonine kinase that regulates a variety of cellular functions. Here, we demonstrate that endothelial PKA activity is essential for vascular development, specifically regulating the transition from sprouting to stabilization of nascent vessels. Inhibition of endothelial PKA by endothelial cell-specific expression of dominant-negative PKA in mice led to perturbed vascular development, hemorrhage and embryonic lethality at mid-gestation. During perinatal retinal angiogenesis, inhibition of PKA resulted in hypersprouting as a result of increased numbers of tip cells. In zebrafish, cell autonomous PKA inhibition also increased and sustained endothelial cell motility, driving cells to become tip cells. Although these effects of PKA inhibition were highly reminiscent of Notch inhibition effects, our data demonstrate that PKA and Notch independently regulate tip and stalk cell formation and behavior.
Collapse
Affiliation(s)
- Pavel I Nedvetsky
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Xiaocheng Zhao
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Thomas Mathivet
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Irene M Aspalter
- Vascular Biology Laboratory, London Research Institute - Cancer Research UK, Lincoln's Inn Fields Laboratories, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Fabio Stanchi
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ross J Metzger
- Department of Anatomy, University of California San Francisco, Genentech Hall, 600 16th Street, San Francisco, CA 94143-2140, USA
| | - Keith E Mostov
- Department of Anatomy, University of California San Francisco, Genentech Hall, 600 16th Street, San Francisco, CA 94143-2140, USA
| | - Holger Gerhardt
- Vascular Patterning Laboratory, Vesalius Research Center, VIB, Leuven, Belgium Vascular Patterning Laboratory, Vesalius Research Center, Department of Oncology, KU Leuven, Leuven, Belgium Vascular Biology Laboratory, London Research Institute - Cancer Research UK, Lincoln's Inn Fields Laboratories, 44 Lincoln's Inn Fields, London WC2A 3LY, UK Integrative Vascular Biology Lab, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, Berlin 13125, Germany DZHK (German Center for Cardiovascular Research), partner site Berlin Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
19
|
Hu L, Liu R. Expression of Gab1 Is Associated with Poor Prognosis of Patients with Epithelial Ovarian Cancer. TOHOKU J EXP MED 2017; 239:177-84. [PMID: 27302321 DOI: 10.1620/tjem.239.177] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Growth factor receptor-bound protein-2 (Grb2) can act as the scaffold protein recruiting other molecules to the stimulated receptors. Grb2-associated binding protein 1 (Gab1) is involved in cell proliferation, and its expression may enhance the carcinogenesis and cancer progression. However, the function of Gab1 remains to be investigated. Epithelial ovarian cancer (EOC) is the most lethal malignancy in the female reproductive system with increasing incidence and unsatisfied overall survival (OS). We investigated the expression of Gab1 in EOC tissues and the correlations between Gab1 expression and the clinicopathological characteristics of patients with EOC using Spearman rank test. The staining results were evaluated based on both the percentage of Gab1-positive tumor cells and the staining intensity for Gab1 expression. Kaplan-Meier survival analysis and Cox proportional hazards analysis were used to compare the postoperative OS between EOC patients with high Gab1 expression and those with low Gab1 expression. The high expression of Gab1 was positively correlated with advanced FIGO stage and lymph node metastasis of EOC. Univariate analysis showed that advanced FIGO stage, pathological grade, lymph node metastasis or Gab1 expression were associated with poor OS. Moreover, multivariate analysis revealed that Gab1 expression could be an independent prognostic factor for the poor OS of EOC patients (P = 0.042). We propose that Gab1 expression is correlated with poor prognosis of EOC patients and may act as an independent prognostic indicator.
Collapse
Affiliation(s)
- Lingling Hu
- Department of Gynecology, Linyi People's Hospital
| | | |
Collapse
|
20
|
Zhang Y, Xu Y, Liu S, Guo X, Cen D, Xu J, Li H, Li K, Zeng C, Lu L, Zhou Y, Shen H, Cheng H, Zhang X, Ke Y. Scaffolding protein Gab1 regulates myeloid dendritic cell migration in allergic asthma. Cell Res 2016; 26:1226-1241. [PMID: 27811945 DOI: 10.1038/cr.2016.124] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 08/13/2016] [Accepted: 08/18/2016] [Indexed: 12/14/2022] Open
Abstract
Asthma is a common allergic disorder involving a complex interplay among multiple genetic and environmental factors. Recent studies identified genetic variants of human GAB1 as a novel asthma susceptibility factor. However, the functions of Gab1 in lung remain largely unexplored. In this study, we first observed an elevation of Gab1 level in peripheral blood mononuclear cells from asthmatic patients during acute exacerbation compared with convalescence. Mice with a selectively disrupted Gab1 in myeloid dendritic cells (mDCs) considerably attenuated allergic inflammation in experimental models of asthma. Further investigations revealed a prominent reduction in CCL19-mediated migration of Gab1-deficient mDCs to draining lymph nodes and subsequent impairment of Th2-driven adaptive activation. Mechanistically, Gab1 is an essential component of the CCL19/CCR7 chemokine axis that regulates mDC migration during asthmatic responses. Together, these findings provide the first evidence for the roles of Gab1 in lung, giving us deeper understanding of asthmatic pathogenesis.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Yun Xu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Shuwan Liu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Xiaohong Guo
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Dong Cen
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Jiaqi Xu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Heyuan Li
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China
| | - Kaijun Li
- Lishui Central Hospital, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, China
| | - Chunlai Zeng
- Lishui Central Hospital, Affiliated Lishui Hospital of Zhejiang University, Lishui, Zhejiang 323000, China
| | - Linrong Lu
- The Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Yiting Zhou
- Department of Biochemistry and Molecular Biology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, China
| | - Xue Zhang
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, China
| | - Yuehai Ke
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
21
|
CdGAP/ARHGAP31, a Cdc42/Rac1 GTPase regulator, is critical for vascular development and VEGF-mediated angiogenesis. Sci Rep 2016; 6:27485. [PMID: 27270835 PMCID: PMC4895392 DOI: 10.1038/srep27485] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023] Open
Abstract
Mutations in the CdGAP/ARHGAP31 gene, which encodes a GTPase-activating protein for Rac1 and Cdc42, have been reported causative in the Adams-Oliver developmental syndrome often associated with vascular defects. However, despite its abundant expression in endothelial cells, CdGAP function in the vasculature remains unknown. Here, we show that vascular development is impaired in CdGAP-deficient mouse embryos at E15.5. This is associated with superficial vessel defects and subcutaneous edema, resulting in 44% embryonic/perinatal lethality. VEGF-driven angiogenesis is defective in CdGAP(-/-) mice, showing reduced capillary sprouting from aortic ring explants. Similarly, VEGF-dependent endothelial cell migration and capillary formation are inhibited upon CdGAP knockdown. Mechanistically, CdGAP associates with VEGF receptor-2 and controls VEGF-dependent signaling. Consequently, CdGAP depletion results in impaired VEGF-mediated Rac1 activation and reduced phosphorylation of critical intracellular mediators including Gab1, Akt, PLCγ and SHP2. These findings are the first to demonstrate the importance of CdGAP in embryonic vascular development and VEGF-induced signaling, and highlight CdGAP as a potential therapeutic target to treat pathological angiogenesis and vascular dysfunction.
Collapse
|
22
|
Yamauchi H, Miura S, Owada T, Saitoh SI, Machii H, Yamada S, Ishigami A, Takeishi Y. Senescence marker protein-30 deficiency impairs angiogenesis under ischemia. Free Radic Biol Med 2016; 94:66-73. [PMID: 26912033 DOI: 10.1016/j.freeradbiomed.2016.02.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 02/02/2016] [Accepted: 02/17/2016] [Indexed: 01/09/2023]
Abstract
Aging decreases collateral-dependent flow recovery following acute arterial obstruction. However, the mechanisms are partially understood, therefore critical management has been lacked in clinical setting. Senescence marker protein-30 (SMP30) is a novel aging marker, which is assumed to act as an anti-aging factor in various organs. Therefore, we studied the effect of SMP30 on ischemia-induced collateral growth in SMP30 knockout (KO) mice, young and old C57BL/6 mice. The SMP30 expression in gastrocnemius tissue was decreased in old mice compared to that of young mice. The recovery of cutaneous blood flow in hind limb after femoral artery ligation and tissue capillary density recoveries were suppressed in SMP30 KO and old mice compared to those in young mice. Nitric oxide generation induced by l-arginine and GSH/GSSG in aorta of SMP30 KO and old mice were lower than those in young mice. The levels of NADPH oxidase activity and superoxide production in the ischemic tissue were higher in SMP30 KO and old mice than in young mice. The phosphorylated eNOS and Akt levels and VEGF levels in ischemic muscle were lower in SMP30 KO and old mice than in young mice. Deficiency of SMP30 exacerbates oxidative stress related to NADPH oxidase activity enhancement and impairs eNOS activity, which leads to rarefaction of angiogenesis induced by ischemia. These results suggest that SMP30 plays a key role in disrupting collateral growth under ischemia in aging.
Collapse
Affiliation(s)
- Hiroyuki Yamauchi
- Department of Cardiology and Hematology, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| | - Shunsuke Miura
- Department of Cardiology and Hematology, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| | - Takashi Owada
- Department of Cardiology and Hematology, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| | - Shu-Ichi Saitoh
- Department of Cardiology and Hematology, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan.
| | - Hirofumi Machii
- Department of Cardiology and Hematology, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| | - Shinya Yamada
- Department of Cardiology and Hematology, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yasuchika Takeishi
- Department of Cardiology and Hematology, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| |
Collapse
|
23
|
Bandara N, Gurusinghe S, Chen H, Chen S, Wang LX, Lim SY, Strappe P. Minicircle DNA-mediated endothelial nitric oxide synthase gene transfer enhances angiogenic responses of bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther 2016; 7:48. [PMID: 27036881 PMCID: PMC4818467 DOI: 10.1186/s13287-016-0307-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/24/2016] [Accepted: 03/11/2016] [Indexed: 11/10/2022] Open
Abstract
Background Non-viral-based gene modification of adult stem cells with endothelial nitric oxide synthase (eNOS) may enhance production of nitric oxide and promote angiogenesis. Nitric oxide (NO) derived from endothelial cells is a pleiotropic diffusible gas with positive effects on maintaining vascular tone and promoting wound healing and angiogenesis. Adult stem cells may enhance angiogenesis through expression of bioactive molecules, and their genetic modification to express eNOS may promote NO production and subsequent cellular responses. Methods Rat bone marrow-derived mesenchymal stem cells (rBMSCs) were transfected with a minicircle DNA vector expressing either green fluorescent protein (GFP) or eNOS. Transfected cells were analysed for eNOS expression and NO production and for their ability to form in vitro capillary tubules and cell migration. Transcriptional activity of angiogenesis-associated genes, CD31, VEGF-A, PDGFRα, FGF2, and FGFR2, were analysed by quantitative polymerase chain reaction. Results Minicircle vectors expressing GFP (MC-GFP) were used to transfect HEK293T cells and rBMSCs, and were compared to a larger parental vector (P-GFP). MC-GFP showed significantly higher transfection in HEK293T cells (55.51 ± 3.3 %) and in rBMSC (18.65 ± 1.05 %) compared to P-GFP in HEK293T cells (43.4 ± 4.9 %) and rBMSC (15.21 ± 0.22 %). MC-eNOS vectors showed higher transfection efficiency (21 ± 3 %) compared to P-eNOS (9 ± 1 %) and also generated higher NO levels. In vitro capillary tubule formation assays showed both MC-eNOS and P-eNOS gene-modified rBMSCs formed longer (14.66 ± 0.55 mm and 13.58 ± 0.68 mm, respectively) and a greater number of tubules (56.33 ± 3.51 and 51 ± 4, respectively) compared to controls, which was reduced with the NOS inhibitor L-NAME. In an in vitro wound healing assay, MC-eNOS transfected cells showed greater migration which was also reversed by L-NAME treatment. Finally, gene expression analysis in MC-eNOS transfected cells showed significant upregulation of the endothelial-specific marker CD31 and enhanced expression of VEGFA and FGF-2 and their corresponding receptors PDGFRα and FGFR2, respectively. Conclusions A novel eNOS-expressing minicircle vector can efficiently transfect rBMSCs and produce sufficient NO to enhance in vitro models of capillary formation and cell migration with an accompanying upregulation of CD31, angiogenic growth factor, and receptor gene expression.
Collapse
Affiliation(s)
- Nadeeka Bandara
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia.,O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia
| | - Saliya Gurusinghe
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia
| | - Haiying Chen
- Central laboratory and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, 252000, PR China
| | - Shuangfeng Chen
- Central laboratory and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, 252000, PR China
| | - Le-Xin Wang
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia.,Central laboratory and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, 252000, PR China
| | - Shiang Y Lim
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, Fitzroy, VIC, 3065, Australia.,Department of Surgery, St. Vincent's Hospital, University of Melbourne, Melbourne, VIC, 3002, Australia
| | - Padraig Strappe
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia.
| |
Collapse
|
24
|
Xu S, Ha CH, Wang W, Xu X, Yin M, Jin FQ, Mastrangelo M, Koroleva M, Fujiwara K, Jin ZG. PECAM1 regulates flow-mediated Gab1 tyrosine phosphorylation and signaling. Cell Signal 2015; 28:117-124. [PMID: 26706435 DOI: 10.1016/j.cellsig.2015.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/09/2015] [Accepted: 12/14/2015] [Indexed: 01/25/2023]
Abstract
Endothelial dysfunction, characterized by impaired activation of endothelial nitric oxide (NO) synthase (eNOS) and ensued decrease of NO production, is a common mechanism of various cardiovascular pathologies, including hypertension and atherosclerosis. Laminar blood flow-mediated specific signaling cascades modulate vascular endothelial cells (ECs) structure and functions. We have previously shown that flow-stimulated Gab1 (Grb2-associated binder-1) tyrosine phosphorylation mediates eNOS activation in ECs, which in part confers laminar flow atheroprotective action. However, the molecular mechanisms whereby flow regulates Gab1 tyrosine phosphorylation and its downstream signaling events remain unclear. Here we show that platelet endothelial cell adhesion molecule-1 (PECAM1), a key molecule in an endothelial mechanosensing complex, specifically mediates Gab1 tyrosine phosphorylation and its downstream Akt and eNOS activation in ECs upon flow rather than hepatocyte growth factor (HGF) stimulation. Small interfering RNA (siRNA) targeting PECAM1 abolished flow- but not HGF-induced Gab1 tyrosine phosphorylation and Akt, eNOS activation as well as Gab1 membrane translocation. Protein-tyrosine phosphatase SHP2, which has been shown to interact with Gab1, was involved in flow signaling and HGF signaling, as SHP2 siRNA diminished the flow- and HGF-induced Gab1 tyrosine phosphorylation, membrane localization and downstream signaling. Pharmacological inhibition of PI3K decreased flow-, but not HGF-mediated Gab1 phosphorylation and membrane localization as well as eNOS activation. Finally, we observed that flow-mediated Gab1 and eNOS phosphorylation in vivo induced by voluntary wheel running was reduced in PECAM1 knockout mice. These results demonstrate a specific role of PECAM1 in flow-mediated Gab1 tyrosine phosphorylation and eNOS signaling in ECs.
Collapse
Affiliation(s)
- Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Chang Hoon Ha
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Weiye Wang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Xiangbin Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Meimei Yin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Felix Q Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Michael Mastrangelo
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Marina Koroleva
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Keigi Fujiwara
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| |
Collapse
|
25
|
Hwang S, Lee HJ, Kim G, Won KJ, Park YS, Jo I. CCN1 acutely increases nitric oxide production via integrin αvβ3-Akt-S6K-phosphorylation of endothelial nitric oxide synthase at the serine 1177 signaling axis. Free Radic Biol Med 2015; 89:229-40. [PMID: 26393424 DOI: 10.1016/j.freeradbiomed.2015.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 07/14/2015] [Accepted: 08/05/2015] [Indexed: 11/30/2022]
Abstract
Although CCN1 (also known as cysteine-rich, angiogenic inducer 61, CYR61) has been reported to promote angiogenesis and neovascularization in endothelial cells (ECs), its effects on endothelial nitric oxide (NO) production have never been studied. Using human umbilical vein ECs, we investigated whether and how CCN1 regulates NO production. CCN1 acutely increased NO production in a time- and dose-dependent manner, which was accompanied by increased phosphorylation of endothelial NO synthase (eNOS) at serine 1177 (eNOS-Ser(1177)), but not that of eNOS-Thr(495) or eNOS-Ser(114). The level of total eNOS expression was unaltered. Treatment with either LY294002, a selective inhibitor of phosphoinositide 3-kinase known as an upstream kinase of Akt, or H-89, an inhibitor of protein kinase A, mitogen- and stress-activated protein kinase 1, Rho-associated protein kinase 2, and ribosomal protein S6 kinase (S6K), inhibited CCN1-stimulated eNOS-Ser(1177) phosphorylation and subsequent NO production. Ectopic expression of small interfering RNA against Akt and S6K significantly inhibited the effects of CCN1. Consistently, CCN1 increased the phosphorylation of Akt-Ser(473) and S6K-Thr(389). However, CCN1 did not alter the expression or secretion of VEGF, a known downstream factor of CCN1 and a potential upstream factor of Akt-mediated eNOS-Ser(1177) phosphorylation. Furthermore, neutralization of integrin αvβ3 with corresponding antibody completely reversed all of the observed effects of CCN1. Moreover, CCN1 increased acetylcholine-induced relaxation in the rat aortas. Finally, we also found that CCN1-stimulated eNOS-Ser(1177) phosphorylation and NO production are true for other types of EC tested. In conclusion, CCN1 acutely increases NO production via activation of a signaling axis in integrin αvβ3-Akt-S6K-eNOS-Ser(1177) phosphorylation, suggesting an important role for CCN1 in vasodilation.
Collapse
Affiliation(s)
- Soojin Hwang
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Hyeon-Ju Lee
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Gyungah Kim
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Kyung-Jong Won
- Department of Medical Science, School of Medicine, Konkuk University, Chungju 380-701, Republic of Korea
| | - Yoon Shin Park
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea
| | - Inho Jo
- Department of Molecular Medicine, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea.
| |
Collapse
|
26
|
Zhao J, Yin M, Deng H, Jin FQ, Xu S, Lu Y, Mastrangelo MA, Luo H, Jin ZG. Cardiac Gab1 deletion leads to dilated cardiomyopathy associated with mitochondrial damage and cardiomyocyte apoptosis. Cell Death Differ 2015; 23:695-706. [PMID: 26517531 DOI: 10.1038/cdd.2015.143] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 09/01/2015] [Accepted: 09/18/2015] [Indexed: 01/28/2023] Open
Abstract
A vital step in the development of heart failure is the transition from compensatory cardiac hypertrophy to decompensated dilated cardiomyopathy (DCM) during cardiac remodeling under mechanical or pathological stress. However, the molecular mechanisms underlying the development of DCM and heart failure remain incompletely understood. In the present study, we investigate whether Gab1, a scaffolding adaptor protein, protects against hemodynamic stress-induced DCM and heat failure. We first observed that the protein levels of Gab1 were markedly reduced in hearts from human patients with DCM and from mice with experimental viral myocarditis in which DCM developed. Next, we generated cardiac-specific Gab1 knockout mice (Gab1-cKO) and found that Gab-cKO mice developed DCM in hemodynamic stress-dependent and age-dependent manners. Under transverse aorta constriction (TAC), Gab1-cKO mice rapidly developed decompensated DCM and heart failure, whereas Gab1 wild-type littermates exhibited adaptive left ventricular hypertrophy without changes in cardiac function. Mechanistically, we showed that Gab1-cKO mouse hearts displayed severe mitochondrial damages and increased cardiomyocyte apoptosis. Loss of cardiac Gab1 in mice impaired Gab1 downstream MAPK signaling pathways in the heart under TAC. Gene profiles further revealed that ablation of Gab1 in heart disrupts the balance of anti- and pro-apoptotic genes in cardiomyocytes. These results demonstrate that cardiomyocyte Gab1 is a critical regulator of the compensatory cardiac response to aging and hemodynamic stress. These findings may provide new mechanistic insights and potential therapeutic target for DCM and heart failure.
Collapse
Affiliation(s)
- J Zhao
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - M Yin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - H Deng
- Center for Heart Lung Innovation/Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - F Q Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - S Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Y Lu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - M A Mastrangelo
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - H Luo
- Center for Heart Lung Innovation/Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Z G Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|
27
|
Burgoyne JR, Rudyk O, Cho HJ, Prysyazhna O, Hathaway N, Weeks A, Evans R, Ng T, Schröder K, Brandes RP, Shah AM, Eaton P. Deficient angiogenesis in redox-dead Cys17Ser PKARIα knock-in mice. Nat Commun 2015; 6:7920. [PMID: 26258640 DOI: 10.1038/ncomms8920] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 06/24/2015] [Indexed: 02/05/2023] Open
Abstract
Angiogenesis is essential for tissue development, wound healing and tissue perfusion, with its dysregulation linked to tumorigenesis, rheumatoid arthritis and heart disease. Here we show that pro-angiogenic stimuli couple to NADPH oxidase-dependent generation of oxidants that catalyse an activating intermolecular-disulphide between regulatory-RIα subunits of protein kinase A (PKA), which stimulates PKA-dependent ERK signalling. This is crucial to blood vessel growth as 'redox-dead' Cys17Ser RIα knock-in mice fully resistant to PKA disulphide-activation have deficient angiogenesis in models of hind limb ischaemia and tumour-implant growth. Disulphide-activation of PKA represents a new therapeutic target in diseases with aberrant angiogenesis.
Collapse
Affiliation(s)
- Joseph R Burgoyne
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Olena Rudyk
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Hyun-Ju Cho
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Oleksandra Prysyazhna
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Natasha Hathaway
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Amanda Weeks
- King's College London, Division of Imaging Sciences, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| | - Rachel Evans
- King's College London, Division of Cancer Studies, 2nd Floor, New Hunt's House, Guy's Medical School Campus, London SE1 1UL, UK
| | - Tony Ng
- King's College London, Division of Cancer Studies, 2nd Floor, New Hunt's House, Guy's Medical School Campus, London SE1 1UL, UK
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Theodor-Stern Kai 7, 60590 Frankfurt, Germany
| | - Ajay M Shah
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The James Black Centre, Denmark Hill Campus, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Philip Eaton
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, Saint Thomas' Hospital, London SE1 7EH, UK
| |
Collapse
|
28
|
Shp2 promotes metastasis of prostate cancer by attenuating the PAR3/PAR6/aPKC polarity protein complex and enhancing epithelial-to-mesenchymal transition. Oncogene 2015; 35:1271-82. [DOI: 10.1038/onc.2015.184] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 03/19/2015] [Accepted: 04/24/2015] [Indexed: 12/13/2022]
|
29
|
Yan L, Ge Q, Xi C, Zhang X, Guo Y. Genetic variations of VEGF gene were associated with tetralogy of fallot risk in a Chinese Han population. Genet Test Mol Biomarkers 2015; 19:264-71. [PMID: 25894981 DOI: 10.1089/gtmb.2014.0303] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVES Tetralogy of Fallot (TOF) is one of the most common forms of congenital heart disease. In this study, we aimed at investigating the associations between genetic variations of vascular endothelial growth factor (VEGF) gene and the risk of TOF in a Chinese Han population. Our findings may contribute to a deeper understanding of TOF pathogenesis and better diagnostic and therapeutic suggestions. METHODS A total of 165 TOF patients and 240 controls from a Chinese Han population in Shenyang and Harbin were recruited in the current study. Nine single-nucleotide polymorphisms (SNPs) (-2578C/A, -460T/C, -1154G/A, -634G/C, 534C/T, +398G/A, +963C/T, 752C/T, 913G/A) were genotyped by the MALDI-TOF MassARRAY system. Individual SNPs as well as their haplotypes were analyzed for their associations with TOF risk, using odds ratios and the 95% confidence interval under codominant and dominant models. RESULTS In the single SNP analyses, the mutant homozygous genotypes of -2578C/A (rs699947) and +963C/T (rs3025039) were related with an increased risk of TOF. In addition, carriers with the mutant A allele of -1154G/A (rs1570360) were supposed to have a significantly elevated TOF risk. Similarly, compared with the wild homozygote GG carriers, the GC carrier of -634G/C (rs2010963) revealed a significant relationship with susceptibility of TOF, but not for the mutant homozygote CC carriers. However, no significant association was found for the other five SNPs. Meanwhile, haplotype analysis revealed that CCA and ATA in block 1 (-2578C/A, -460T/C, and -1154G/A) and TTG and TCA in block 3 (+963C/T, 752C/T, and 913G/A) were significantly related with an increased TOF risk compared with the most common haplotypes. CONCLUSION In summary, our results suggested that VEGF variants (-2578C/A, -1154G/A, -634G/C, +963G/A) were involved in the susceptibility of TOF. However, validation of our study needs further study in various ethnics to reveal the functional relationship between VEGF polymorphisms and TOF risk, which may contribute to diagnosis and therapy of TOF.
Collapse
Affiliation(s)
- Liru Yan
- 1 Department of Developmental Pediatrics, Shengjing Hospital, China Medical University , Shenyang, Liaoning Province, China
| | | | | | | | | |
Collapse
|
30
|
Jiang B, Deng Q, Huo Y, Li W, Shibuya M, Luo J. Endothelial Gab1 deficiency aggravates splenomegaly in portal hypertension independent of angiogenesis. Am J Physiol Gastrointest Liver Physiol 2015; 308:G416-26. [PMID: 25501549 DOI: 10.1152/ajpgi.00292.2014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Certain pathological changes, including angiogenesis, actively contribute to the pathogenesis of splenomegaly in portal hypertension (PH), although the detailed molecular and cellular mechanisms remain elusive. In this study, we demonstrated that endothelial Grb-2-associated binder 1 (Gab1) plays a negative role in PH-associated splenomegaly independent of angiogenesis. PH, which was induced by partial portal vein ligation, significantly enhanced Gab1 expression in endothelial cells in a time-dependent manner. Compared with controls, endothelium-specific Gab1 knockout (EGKO) mice exhibited a significant increase in spleen size while their PH levels remained similar. Pathological analysis indicated that EGKO mice developed more severe hyperactive white pulp and fibrosis in the enlarged spleen but less angiogenesis in both the spleen and mesenteric tissues. Mechanistic studies showed that the phosphorylation of endothelial nitric oxide synthase (eNOS) in EGKO mice was significantly lower than in controls. In addition, the dysregulation of fibrosis and inflammation-related transcription factors [e.g., Krüppel-like factor (KLF) 2 and KLF5] and the upregulation of cytokine genes (e.g., TNF-α and IL-6) were observed in EGKO mice. We thus propose that endothelial Gab1 mediates multiple pathways in inhibition of the pathogenesis of splenomegaly in PH via prevention of endothelial dysfunction and overproduction of proinflammatory/profibrotic cytokines.
Collapse
Affiliation(s)
- Beibei Jiang
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Qiuping Deng
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Yingqing Huo
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China
| | - Wei Li
- People's Hospital, Peking University, Beijing, China; and
| | - Masabumi Shibuya
- Institute of Physiology and Medicine, Jobu University, Takasaki, Japan
| | - Jincai Luo
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China;
| |
Collapse
|
31
|
Wang W, Xu S, Yin M, Jin ZG. Essential roles of Gab1 tyrosine phosphorylation in growth factor-mediated signaling and angiogenesis. Int J Cardiol 2014; 181:180-4. [PMID: 25528308 DOI: 10.1016/j.ijcard.2014.10.148] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 09/08/2014] [Accepted: 10/18/2014] [Indexed: 12/16/2022]
Abstract
Growth factors and their downstream receptor tyrosine kinases (RTKs) mediate a number of biological processes controlling cell function. Adaptor (docking) proteins, which consist exclusively of domains and motifs that mediate molecular interactions, link receptor activation to downstream effectors. Recent studies have revealed that Grb2-associated-binders (Gab) family members (including Gab1, Gab2, and Gab3), when phosphorylated on tyrosine residues, provide binding sites for multiple effector proteins, such as Src homology-2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2) and phosphatidylinositol 3-kinase (PI3K) regulatory subunit p85, thereby playing important roles in transducing RTKs-mediated signals into pathways with diversified biological functions. Here, we provide an up-to-date overview on the domain structure and biological functions of Gab1, the most intensively studied Gab family protein, in growth factor signaling and biological functions, with a special focus on angiogenesis.
Collapse
Affiliation(s)
- Weiye Wang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Meimei Yin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Zheng Gen Jin
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14642, USA.
| |
Collapse
|
32
|
Kumarswamy R, Volkmann I, Beermann J, Napp LC, Jabs O, Bhayadia R, Melk A, Ucar A, Chowdhury K, Lorenzen JM, Gupta SK, Batkai S, Thum T. Vascular importance of the miR-212/132 cluster. Eur Heart J 2014; 35:3224-31. [PMID: 25217442 DOI: 10.1093/eurheartj/ehu344] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
RATIONALE Many processes in endothelial cells including angiogenic responses are regulated by microRNAs. However, there is limited information available about their complex cross-talk in regulating certain endothelial functions. AIM The objective of this study is to identify endothelial functions of the pro-hypertrophic miR-212/132 cluster and its cross-talk with other microRNAs during development and disease. METHODS AND RESULTS We here show that anti-angiogenic stimulation by transforming growth factor-beta activates the microRNA-212/132 cluster by derepression of their transcriptional co-activator cAMP response element-binding protein (CREB)-binding protein (CBP) which is a novel target of a previously identified pro-angiogenic miRNA miR-30a-3p in endothelial cells. Surprisingly, despite having the same seed-sequence, miR-212 and miR-132 exerted differential effects on endothelial transcriptome regulation and cellular functions with stronger endothelial inhibitory effects caused by miR-212. These differences could be attributed to additional auxiliary binding of miR-212 to its targets. In vivo, deletion of the miR-212/132 cluster increased endothelial vasodilatory function, improved angiogenic responses during postnatal development and in adult mice. CONCLUSION Our results identify (i) a novel miRNA-cross-talk involving miR-30a-3p and miR-212, which led to suppression of important endothelial genes such as GAB1 and SIRT1 finally culminating in impaired endothelial function; and (ii) microRNAs may have different biological roles despite having the same seed sequence.
Collapse
Affiliation(s)
- Regalla Kumarswamy
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Ingo Volkmann
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Julia Beermann
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Lars Christian Napp
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Olga Jabs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Raj Bhayadia
- Department of Kidney, Liver and Metabolic Diseases, Children's Hospital, Hannover Medical School, Hannover, Germany
| | - Anette Melk
- Department of Kidney, Liver and Metabolic Diseases, Children's Hospital, Hannover Medical School, Hannover, Germany
| | - Ahmet Ucar
- Department of Molecular Cell Biology, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany Division of Developmental Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kamal Chowdhury
- Department of Molecular Cell Biology, Max Planck Institute of Biophysical Chemistry, Göttingen, Germany
| | - Johan M Lorenzen
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany Integrated Research and Treatment Center Transplantation, Hannover Medical School, Hannover, Germany
| | - Shashi Kumar Gupta
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Sandor Batkai
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany Integrated Research and Treatment Center Transplantation, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany Integrated Research and Treatment Center Transplantation, Hannover Medical School, Hannover, Germany National Heart and Lung Institute, Imperial College London, London, UK REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany
| |
Collapse
|
33
|
Wang H, Yang Z, Gu J. Therapeutic Targeting of Angiogenesis with a Recombinant CTT Peptide–Endostatin Mimic–Kringle 5 Protein. Mol Cancer Ther 2014; 13:2674-87. [PMID: 25127900 DOI: 10.1158/1535-7163.mct-14-0266] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
MESH Headings
- Angiogenesis Inhibitors/pharmacology
- Animals
- Biomimetic Materials/chemistry
- Biomimetic Materials/pharmacology
- Carcinoma, Lewis Lung/blood supply
- Carcinoma, Lewis Lung/drug therapy
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation/drug effects
- Endostatins/chemistry
- Endostatins/pharmacology
- HEK293 Cells
- HeLa Cells
- Humans
- Kringles
- Liver Neoplasms, Experimental/blood supply
- Liver Neoplasms, Experimental/drug therapy
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Neovascularization, Pathologic/drug therapy
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/pharmacology
- Random Allocation
- Recombinant Fusion Proteins/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Houbin Wang
- National Key Laboratory of Protein and Plant Gene Research, LSC, Peking University, Beijing, China
| | - Zhigang Yang
- Department of Hematology, Affiliated Hospital of Guangdong Medical College, Guangzhou, China
| | - Jun Gu
- National Key Laboratory of Protein and Plant Gene Research, LSC, Peking University, Beijing, China. Department of Hematology, Affiliated Hospital of Guangdong Medical College, Guangzhou, China.
| |
Collapse
|
34
|
Deng Q, Huo Y, Luo J. Endothelial mechanosensors: the gatekeepers of vascular homeostasis and adaptation under mechanical stress. SCIENCE CHINA-LIFE SCIENCES 2014; 57:755-62. [PMID: 25104447 DOI: 10.1007/s11427-014-4705-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 07/05/2014] [Indexed: 01/27/2023]
Abstract
Endothelial cells (ECs) not only serve as a barrier between blood and extravascular space to modulate the exchange of fluid, macromolecules and cells, but also play a critical role in regulation of vascular homeostasis and adaptation under mechanical stimulus via intrinsic mechanotransduction. Recently, with the dissection of microdomains responsible for cellular responsiveness to mechanical stimulus, a lot of mechanosensing molecules (mechanosensors) and pathways have been identified in ECs. In addition, there is growing evidence that endothelial mechanosensors not only serve as key vascular gatekeepers, but also contribute to the pathogenesis of various vascular disorders. This review focuses on recent findings in endothelial mechanosensors in subcellular microdomains and their roles in regulation of physiological and pathological functions under mechanical stress.
Collapse
Affiliation(s)
- QiuPing Deng
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | | | | |
Collapse
|
35
|
Li S, Hsu DDF, Li B, Luo X, Alderson N, Qiao L, Ma L, Zhu HH, He Z, Suino-Powell K, Ji K, Li J, Shao J, Xu HE, Li T, Feng GS. Cytoplasmic tyrosine phosphatase Shp2 coordinates hepatic regulation of bile acid and FGF15/19 signaling to repress bile acid synthesis. Cell Metab 2014; 20:320-32. [PMID: 24981838 PMCID: PMC4365973 DOI: 10.1016/j.cmet.2014.05.020] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/18/2014] [Accepted: 05/16/2014] [Indexed: 02/06/2023]
Abstract
Bile acid (BA) biosynthesis is tightly controlled by intrahepatic negative feedback signaling elicited by BA binding to farnesoid X receptor (FXR) and also by enterohepatic communication involving ileal BA reabsorption and FGF15/19 secretion. However, how these pathways are coordinated is poorly understood. We show here that nonreceptor tyrosine phosphatase Shp2 is a critical player that couples and regulates the intrahepatic and enterohepatic signals for repression of BA synthesis. Ablating Shp2 in hepatocytes suppressed signal relay from FGFR4, receptor for FGF15/19, and attenuated BA activation of FXR signaling, resulting in elevation of systemic BA levels and chronic hepatobiliary disorders in mice. Acting immediately downstream of FGFR4, Shp2 associates with FRS2α and promotes the receptor activation and signal relay to several pathways. These results elucidate a molecular mechanism for the control of BA homeostasis by Shp2 through the orchestration of multiple signals in hepatocytes.
Collapse
Affiliation(s)
- Shuangwei Li
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA
| | - Diane D F Hsu
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA
| | - Bing Li
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xiaolin Luo
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA
| | - Nazilla Alderson
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA
| | - Liping Qiao
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0983, USA
| | - Lina Ma
- Molecular Neuroscience Laboratory, The Salk Institute for Biological Sciences, La Jolla, CA 92186, USA
| | - Helen H Zhu
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA
| | - Zhao He
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA
| | - Kelly Suino-Powell
- Laboratory of Structural Sciences, Van Andel Research Institute, 333 Bostwick Avenue Northeast, Grand Rapids, MI 49503, USA
| | - Kaihong Ji
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA
| | - Jiefu Li
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA
| | - Jianhua Shao
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093-0983, USA
| | - H Eric Xu
- Laboratory of Structural Sciences, Van Andel Research Institute, 333 Bostwick Avenue Northeast, Grand Rapids, MI 49503, USA; VARI-SIMM Center, Center for Structure and Function of Drug Targets, CAS-Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Tiangang Li
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Gen-Sheng Feng
- Department of Pathology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093-0864, USA.
| |
Collapse
|
36
|
Combined detection of Gab1 and Gab2 expression predicts clinical outcome of patients with glioma. Med Oncol 2014; 31:77. [PMID: 24998422 DOI: 10.1007/s12032-014-0077-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 06/12/2014] [Indexed: 10/25/2022]
Abstract
Grb2-associated binder 1 (Gab1) and Gab2 play important roles in cancer cell signaling. In particular, it has been demonstrated that the upregulation of Gab2 may be correlated with the World Health Organization (WHO) grade of gliomas and that patients with high Gab2 expression levels exhibited shorter survival time. However, the prognostic value of combined expression of Gab1 and Gab2 has not been explored. Gab1 and Gab2 expression in human gliomas and non-neoplastic brain tissues was measured by immunohistochemistry. Both the expression levels of Gab1 and Gab2 proteins in glioma tissues were significantly higher than those in non-neoplastic brain tissues (both P < 0.001). In addition, the overexpression of Gab1 and Gab2 proteins were both significantly associated with advanced WHO grades (both P < 0.001) and low KPS (both P = 0.01). Moreover, the overall survival of patients with high Gab1 protein expression or high Gab2 protein expression was obviously lower than those with low expressions (both P < 0.001). Notably, glioma patients with combined overexpression of Gab1 and Gab2 proteins (Gab1-high/Gab2-high) had shortest overall survival (P < 0.001). Furthermore, multivariate analysis showed that Gab1 expression (P = 0.01), Gab2 expression (P = 0.02), and combined expression of Gab1 and Gab2 (Gab1/Gab2, P = 0.006) were all independent prognostic factors for overall survival in glioma patients. Gab1 and Gab2 proteins are differentially expressed in glioma patients and closely correlated with the biological behavior of this malignancy. Combination of Gab1 and Gab2 expression may represent a promising biomarker for prognostication of human gliomas.
Collapse
|
37
|
Liu S, Lai L, Zuo Q, Dai F, Wu L, Wang Y, Zhou Q, Liu J, Liu J, Li L, Lin Q, Creighton CJ, Costello MG, Huang S, Jia C, Liao L, Luo H, Fu J, Liu M, Yi Z, Xiao J, Li X. PKA turnover by the REGγ-proteasome modulates FoxO1 cellular activity and VEGF-induced angiogenesis. J Mol Cell Cardiol 2014; 72:28-38. [PMID: 24560667 PMCID: PMC4237316 DOI: 10.1016/j.yjmcc.2014.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 02/04/2014] [Accepted: 02/11/2014] [Indexed: 01/08/2023]
Abstract
The REGγ-proteasome serves as a short-cut for the destruction of certain intact mammalian proteins in the absence of ubiquitin- and ATP. The biological roles of the proteasome activator REGγ are not completely understood. Here we demonstrate that REGγ controls degradation of protein kinase A catalytic subunit-α (PKAca) both in primary human umbilical vein endothelial cells (HUVECs) and mouse embryonic fibroblast cells (MEFs). Accumulation of PKAca in REGγ-deficient HUVECs or MEFs results in phosphorylation and nuclear exclusion of the transcription factor FoxO1, indicating that REGγ is involved in preserving FoxO1 transcriptional activity. Consequently, VEGF-induced expression of the FoxO1 responsive genes, VCAM-1 and E-Selectin, was tightly controlled by REGγ in a PKA dependent manner. Functionally, REGγ is crucial for the migration of HUVECs. REGγ(-/-) mice display compromised VEGF-instigated neovascularization in cornea and aortic ring models. Implanted matrigel plugs containing VEGF in REGγ(-/-) mice induced fewer capillaries than in REGγ(+/+) littermates. Taken together, our study identifies REGγ as a novel angiogenic factor that plays an important role in VEGF-induced expression of VCAM-1 and E-Selectin by antagonizing PKA signaling. Identification of the REGγ-PKA-FoxO1 pathway in endothelial cells (ECs) provides another potential target for therapeutic intervention in vascular diseases.
Collapse
Affiliation(s)
- Shuang Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Department of Hematology, Guangdong No. 2 Provincial People's Hospital, No.1 Shiliugang Rd, Guangzhou, Guangdong 510317, China
| | - Li Lai
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qiuhong Zuo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Fujun Dai
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Lin Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Yan Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qingxia Zhou
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Jian Liu
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jiang Liu
- Institute of Aging Research, School of Medicine, Hangzhou Normal University, Hangzhou, Zhejiang 310036, China
| | - Lei Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Qingxiang Lin
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Chad J Creighton
- Department of Medicine, Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, USA
| | - Myra Grace Costello
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Caifeng Jia
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Lujian Liao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Honglin Luo
- The James Hogg Research Centre for Cardiovascular and Pulmonary Research, University of British Columbia-St. Paul's Hospital, 1081 Burrard St., Vancouver, British Columbia V6Z 1Y6, Canada
| | - Junjiang Fu
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Luzhou Medical College, Luzhou 646000, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China; Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education), Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China.
| | - Jianru Xiao
- Department of Orthopaedic Oncology, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China.
| | - Xiaotao Li
- Department of Molecular and Cellular Biology, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Orthopaedic Oncology, Changzheng Hospital, The Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China; Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China.
| |
Collapse
|
38
|
Sun L, Chen C, Jiang B, Li Y, Deng Q, Sun M, An X, Yang X, Yang Y, Zhang R, Lu Y, Zhu DS, Huo Y, Feng GS, Zhang Y, Luo J. Grb2-associated binder 1 is essential for cardioprotection against ischemia/reperfusion injury. Basic Res Cardiol 2014; 109:420. [PMID: 24951957 DOI: 10.1007/s00395-014-0420-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 12/20/2022]
Abstract
We have shown recently that endothelial Grb-2-associated binder 1 (Gab1), an intracellular scaffolding adaptor, has a protective effect against limb ischemia via mediating angiogenic signaling pathways. However, the role of Gab1 in cardiac ischemia/reperfusion (I/R) injury remains unknown. In this study, we show that Gab1 is required for cardioprotection against I/R injury. I/R injury led to remarkable phosphorylation of Gab1 in cardiomyocytes. Compared with controls, the mice with cardiomyocyte-specific deletion of Gab1 gene (CGKO mice) exhibited an increase in infarct size and a decrease in cardiac function after I/R injury. Consistently, in hearts of CGKO mice subjected to I/R, the activation of caspase 3 and myocardial apoptosis was markedly enhanced whereas the activation of protein kinase B (Akt) and mitogen-activated protein kinase (MAPK), which are critical for cardiomyocyte survival, was attenuated. Oxidative stress is regarded as a major contributor to myocardial I/R injury. To examine the role of Gab1 in oxidative stress directly, isolated adult cardiomyocytes were subject to oxidant hydrogen peroxide and the cardioprotective effects of Gab1 were confirmed. Furthermore, we found that the phosphorylation of Gab1 and Gab1-mediated activation of Akt and MAPK by oxidative stress was suppressed by ErbB receptor and Src kinase inhibitors, accompanied by an increase in apoptotic cell death. In conclusion, our results suggest that Gab1 is essential for cardioprotection against I/R oxidative injury via mediating survival signaling.
Collapse
Affiliation(s)
- Lulu Sun
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, 100871, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Deng J, Liu X, Rong L, Ni C, Li X, Yang W, Lu Y, Yan X, Qin C, Zhang L, Qin Z. IFNγ-responsiveness of endothelial cells leads to efficient angiostasis in tumours involving down-regulation of Dll4. J Pathol 2014; 233:170-82. [PMID: 24615277 DOI: 10.1002/path.4340] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 02/12/2014] [Accepted: 02/13/2014] [Indexed: 12/16/2023]
Abstract
Although IFNγ is regarded as a key cytokine in angiostatic response, our poor understanding of its effective cellular target drastically limits its clinical trials against angiogenesis-related disorders. Here, we investigated the effect of IFNγ on endothelial cells (ECs) and possible molecular mechanisms in angiostasis. By employing Tie2(IFNγR) mice, in which IFNγR expression was reconstituted under the control of Tie2 promoter in IFNγR-deficient mice, we found that the response of ECs to IFNγ was highly effective in inhibiting blood supply and retarding tumour growth. Interestingly, the expression of IFNγR on Tie2(-) cells did not inhibit, but promoted tumour growth in control wild-type mice. Mechanism studies showed that IFNγ reacting on ECs down-regulated the delta-like ligand 4 (Dll4)/Notch signalling pathway. Accordingly, overexpression of Dll4 in human ECs diminished the effect of IFNγ on ECs. This study demonstrates that the action of IFNγ on ECs, but not other cells, is highly effective for tumour angiostasis, which involves down-regulating Dll4. It provides insights for EC-targeted angiostatic therapy in treating angiogenesis-associated disorders in the clinic.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Calcium-Binding Proteins
- Cell Line, Tumor
- Down-Regulation
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation, Neoplastic
- Human Umbilical Vein Endothelial Cells/metabolism
- Human Umbilical Vein Endothelial Cells/pathology
- Humans
- Intercellular Signaling Peptides and Proteins/genetics
- Intercellular Signaling Peptides and Proteins/metabolism
- Interferon-gamma/metabolism
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, SCID
- Mice, Transgenic
- Neoplasms/blood supply
- Neoplasms/genetics
- Neoplasms/metabolism
- Neoplasms/pathology
- Neovascularization, Pathologic
- Pericytes/metabolism
- Pericytes/pathology
- Promoter Regions, Genetic
- Receptor, TIE-2/genetics
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- Signal Transduction
- Time Factors
- Transfection
- Tumor Burden
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Jingjing Deng
- Key Laboratory of Protein and Peptide Pharmaceuticals; Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of the Chinese Academy of Sciences, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang Q, Shen B, Chen L, Zheng P, Feng H, Hao Q, Liu X, Liu L, Xu S, Chen J, Teng J. Extracellular calumenin suppresses ERK1/2 signaling and cell migration by protecting fibulin-1 from MMP-13-mediated proteolysis. Oncogene 2014; 34:1006-18. [DOI: 10.1038/onc.2014.52] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/06/2014] [Accepted: 01/20/2014] [Indexed: 01/04/2023]
|
41
|
Hu Z, Xiong Y, Han X, Geng C, Jiang B, Huo Y, Luo J. Acute mechanical stretch promotes eNOS activation in venous endothelial cells mainly via PKA and Akt pathways. PLoS One 2013; 8:e71359. [PMID: 23977025 PMCID: PMC3743752 DOI: 10.1371/journal.pone.0071359] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 06/28/2013] [Indexed: 11/19/2022] Open
Abstract
In the vasculature, physiological levels of nitric oxide (NO) protect against various stressors, including mechanical stretch. While endothelial NO production in response to various stimuli has been studied extensively, the precise mechanism underlying stretch-induced NO production in venous endothelial cells remains incompletely understood. Using a model of continuous cellular stretch, we found that stretch promoted phosphorylation of endothelial NO synthase (eNOS) at Ser1177, Ser633 and Ser615 and NO production in human umbilical vein endothelial cells. Although stretch activated the kinases AMPKα, PKA, Akt, and ERK1/2, stretch-induced eNOS activation was only inhibited by kinase-specific inhibitors of PKA and PI3K/Akt, but not of AMPKα and Erk1/2. Similar results were obtained with knockdown by shRNAs targeting the PKA and Akt genes. Furthermore, inhibition of PKA preferentially attenuated eNOS activation in the early phase, while inhibition of the PI3K/Akt pathway reduced eNOS activation in the late phase, suggesting that the PKA and PI3K/Akt pathways play distinct roles in a time-dependent manner. Finally, we investigated the role of these pathways in stretch-induced endothelial exocytosis and leukocyte adhesion. Interestingly, we found that inhibition of the PI3K/Akt pathway increased stretch-induced Weibel-Palade body exocytosis and leukocyte adhesion, while inhibition of the PKA pathway had the opposite effects, suggesting that the exocytosis-promoting effect of PKA overwhelms the inhibitory effect of PKA-mediated NO production. Taken together, the results suggest that PKA and Akt are important regulators of eNOS activation in venous endothelial cells under mechanical stretch, while playing different roles in the regulation of stretch-induced endothelial exocytosis and leukocyte adhesion.
Collapse
Affiliation(s)
- Zhenqian Hu
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Yan Xiong
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Xiaofan Han
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Chenyang Geng
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Beibei Jiang
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Yingqing Huo
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
| | - Jincai Luo
- Laboratory of Vascular Biology, Institute of Molecular Medicine, Peking University, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Beijing, China
- * E-mail:
| |
Collapse
|
42
|
AGGF1 is a novel anti-inflammatory factor associated with TNF-α-induced endothelial activation. Cell Signal 2013; 25:1645-53. [DOI: 10.1016/j.cellsig.2013.04.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/14/2013] [Accepted: 04/14/2013] [Indexed: 12/21/2022]
|
43
|
Hypertensive stretch regulates endothelial exocytosis of Weibel-Palade bodies through VEGF receptor 2 signaling pathways. Cell Res 2013; 23:820-34. [PMID: 23609797 DOI: 10.1038/cr.2013.56] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Regulated endothelial exocytosis of Weibel-Palade bodies (WPBs), the first stage in leukocyte trafficking, plays a pivotal role in inflammation and injury. Acute mechanical stretch has been closely associated with vascular inflammation, although the precise mechanism is unknown. Here, we show that hypertensive stretch regulates the exocytosis of WPBs of endothelial cells (ECs) through VEGF receptor 2 (VEGFR2) signaling pathways. Stretch triggers a rapid release (within minutes) of von Willebrand factor and interleukin-8 from WPBs in cultured human ECs, promoting the interaction between leukocytes and ECs through the translocation of P-selectin to the cell membrane. We further show that hypertensive stretch significantly induces P-selectin translocation of intact ECs and enhances leukocyte adhesion both ex vivo and in vivo. Stretch-induced endothelial exocytosis is mediated via a VEGFR2/PLCγ1/calcium pathway. Interestingly, stretch also induces a negative feedback via a VEGFR2/Akt/nitric oxide pathway. Such dual effects are confirmed using pharmacological and genetic approaches in carotid artery segments, as well as in acute hypertensive mouse models. These studies reveal mechanical stretch as a potent agonist for endothelial exocytosis, which is modulated by VEGFR2 signaling. Thus, VEGFR2 signaling pathways may represent novel therapeutic targets in limiting hypertensive stretch-related inflammation.
Collapse
|
44
|
Chade AR, Stewart N. Angiogenic cytokines in renovascular disease: do they have potential for therapeutic use? JOURNAL OF THE AMERICAN SOCIETY OF HYPERTENSION : JASH 2013; 7:180-90. [PMID: 23428409 PMCID: PMC3605220 DOI: 10.1016/j.jash.2013.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 01/11/2013] [Indexed: 10/27/2022]
Abstract
Experimental and clinical studies suggest that the damage of the renal microvascular function and architecture may participate in the early steps of renal injury in chronic renal disease, irrespective of the cause. This supporting evidence has provided the impetus to targeting the renal microvasculature as an attempt to interfere with the progressive nature of the disease process. Chronic renovascular disease is often associated with renal microvascular dysfunction, damage, loss, and defective renal angiogenesis associated with progressive renal dysfunction and damage. It is possible that damage of the renal microvasculature in renovascular disease constitutes an initiating event for renal injury and contributes towards progressive and later on irreversible renal injury. Recent studies have suggested that protection of the renal microcirculation can slow or halt the progression of renal injury in this disease. This brief review will focus on the therapeutic potential and feasibility of using angiogenic cytokines to protect the kidney microvasculature in chronic renovascular disease. There is limited but provocative evidence showing that stimulation of vascular proliferation and repair using vascular endothelial growth factor or hepatocyte growth factor can slow the progression of renal damage, stabilize renal function, and protect the renal parenchyma. Such interventions may potentially constitute a sole strategy to preserve renal function and/or a co-adjuvant tool to improve the success of current therapeutic approaches in renovascular disease.
Collapse
Affiliation(s)
- Alejandro R Chade
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216-4505, USA.
| | | |
Collapse
|
45
|
Abstract
In this review we summarize the current understanding of signal transduction downstream of vascular endothelial growth factor A (VEGFA) and its receptor VEGFR2, and the relationship between these signal transduction pathways and the hallmark responses of VEGFA, angiogenesis and vascular permeability. These physiological responses involve a number of effectors, including extracellular signal-regulated kinases (ERKs), Src, phosphoinositide 3 kinase (PI3K)/Akt, focal adhesion kinase (FAK), Rho family GTPases, endothelial NO and p38 mitogen-activated protein kinase (MAPK). Several of these factors are involved in the regulation of both angiogenesis and vascular permeability. Tumour angiogenesis primarily relies on VEGFA-driven responses, which to a large extent result in a dysfunctional vasculature. The reason for this remains unclear, although it appears that certain aspects of the VEGFA-stimulated angiogenic milieu (high level of microvascular density and permeability) promote tumour expansion. The high degree of redundancy and complexity of VEGFA-driven tumour angiogenesis may explain why tumours commonly develop resistance to anti-angiogenic therapy targeting VEGFA signal transduction.
Collapse
Affiliation(s)
- L Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
46
|
Gab docking proteins in cardiovascular disease, cancer, and inflammation. Int J Inflam 2013; 2013:141068. [PMID: 23431498 PMCID: PMC3566608 DOI: 10.1155/2013/141068] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 12/11/2012] [Indexed: 12/23/2022] Open
Abstract
The docking proteins of the Grb2-associated binder (Gab) family have emerged as crucial signaling compartments in metazoans. In mammals, the Gab proteins, consisting of Gab1, Gab2, and Gab3, are involved in the amplification and integration of signal transduction evoked by a variety of extracellular stimuli, including growth factors, cytokines, antigens, and other molecules. Gab proteins lack the enzymatic activity themselves; however, when phosphorylated on tyrosine residues, they provide binding sites for multiple Src homology-2 (SH2) domain-containing proteins, such as SH2-containing protein tyrosine phosphatase 2 (SHP2), phosphatidylinositol 3-kinase regulatory subunit p85, phospholipase Cγ, Crk, and GC-GAP. Through these interactions, the Gab proteins transduce signals from activated receptors into pathways with distinct biological functions, thereby contributing to signal diversification. They are known to play crucial roles in numerous physiological processes through their associations with SHP2 and p85. In addition, abnormal Gab protein signaling has been linked to human diseases including cancer, cardiovascular disease, and inflammatory disorders. In this paper, we provide an overview of the structure, effector functions, and regulation of the Gab docking proteins, with a special focus on their associations with cardiovascular disease, cancer, and inflammation.
Collapse
|
47
|
Stewart N, Chade AR. Renoprotective effects of hepatocyte growth factor in the stenotic kidney. Am J Physiol Renal Physiol 2012; 304:F625-33. [PMID: 23269649 DOI: 10.1152/ajprenal.00504.2012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal microvascular (MV) damage and loss contribute to the progression of renal injury in renal artery stenosis (RAS). Hepatocyte growth factor (HGF) is a powerful angiogenic and antifibrotic cytokine that we showed to be decreased in the stenotic kidney. We hypothesized that renal HGF therapy will improve renal function mainly by protecting the renal microcirculation. Unilateral RAS was induced in 15 pigs. Six weeks later, single-kidney RBF and GFR were quantified in vivo using multidetector computed tomography (CT). Then, intrarenal rh-HGF or vehicle was randomly administered into the stenotic kidney (RAS, n = 8; RAS+HGF, n = 7). Pigs were observed for 4 additional weeks before CT studies were repeated. Renal MV density was quantified by 3D micro-CT ex vivo and histology, and expression of angiogenic and inflammatory factors, apoptosis, and fibrosis was determined. HGF therapy improved RBF and GFR compared with vehicle-treated pigs. This was accompanied by improved renal expression of angiogenic cytokines (VEGF, p-Akt) and tissue-healing promoters (SDF-1, CXCR4, MMP-9), reduced MV remodeling, apoptosis, and fibrosis, and attenuated renal inflammation. However, HGF therapy did not improve renal MV density, which was similarly reduced in RAS and RAS+HGF compared with controls. Using a clinically relevant animal model of RAS, we showed novel therapeutic effects of a targeted renal intervention. Our results show distinct actions on the existing renal microcirculation and promising renoprotective effects of HGF therapy in RAS. Furthermore, these effects imply plasticity of the stenotic kidney to recuperate its function and underscore the importance of MV integrity in the progression of renal injury in RAS.
Collapse
Affiliation(s)
- Nicholas Stewart
- Department of Physiology and Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | | |
Collapse
|
48
|
Hota PK, Buck M. Plexin structures are coming: opportunities for multilevel investigations of semaphorin guidance receptors, their cell signaling mechanisms, and functions. Cell Mol Life Sci 2012; 69:3765-805. [PMID: 22744749 PMCID: PMC11115013 DOI: 10.1007/s00018-012-1019-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 04/09/2012] [Accepted: 04/11/2012] [Indexed: 01/13/2023]
Abstract
Plexin transmembrane receptors and their semaphorin ligands, as well as their co-receptors (Neuropilin, Integrin, VEGFR2, ErbB2, and Met kinase) are emerging as key regulatory proteins in a wide variety of developmental, regenerative, but also pathological processes. The diverse arenas of plexin function are surveyed, including roles in the nervous, cardiovascular, bone and skeletal, and immune systems. Such different settings require considerable specificity among the plexin and semaphorin family members which in turn are accompanied by a variety of cell signaling networks. Underlying the latter are the mechanistic details of the interactions and catalytic events at the molecular level. Very recently, dramatic progress has been made in solving the structures of plexins and of their complexes with associated proteins. This molecular level information is now suggesting detailed mechanisms for the function of both the extracellular as well as the intracellular plexin regions. Specifically, several groups have solved structures for extracellular domains for plexin-A2, -B1, and -C1, many in complex with semaphorin ligands. On the intracellular side, the role of small Rho GTPases has been of particular interest. These directly associate with plexin and stimulate a GTPase activating (GAP) function in the plexin catalytic domain to downregulate Ras GTPases. Structures for the Rho GTPase binding domains have been presented for several plexins, some with Rnd1 bound. The entire intracellular domain structure of plexin-A1, -A3, and -B1 have also been solved alone and in complex with Rac1. However, key aspects of the interplay between GTPases and plexins remain far from clear. The structural information is helping the plexin field to focus on key questions at the protein structural, cellular, as well as organism level that collaboratoria of investigations are likely to answer.
Collapse
Affiliation(s)
- Prasanta K. Hota
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
| | - Matthias Buck
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Neuroscience, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Department of Pharmacology, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Comprehensive Cancer Center, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106 USA
| |
Collapse
|
49
|
Yang Y, Wu J, Demir A, Castillo-Martin M, Melamed RD, Zhang G, Fukunaga-Kanabis M, Perez-Lorenzo R, Zheng B, Silvers DN, Brunner G, Wang S, Rabadan R, Cordon-Cardo C, Celebi JT. GAB2 induces tumor angiogenesis in NRAS-driven melanoma. Oncogene 2012; 32:3627-37. [PMID: 22926523 DOI: 10.1038/onc.2012.367] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 07/02/2012] [Accepted: 07/04/2012] [Indexed: 12/17/2022]
Abstract
GAB2 is a scaffold protein with diverse upstream and downstream effectors. MAPK and PI3K signaling pathways are known effectors of GAB2. It is amplified and overexpressed in a variety of human tumors including melanoma. Here we show a previously undescribed role for GAB2 in NRAS-driven melanoma. Specifically, we found that GAB2 is co-expressed with mutant NRAS in melanoma cell lines and tumor samples and its expression correlated with metastatic potential. Co-expression of GAB2(WT) and NRAS(G12D) in melanocytes and in melanoma cells increased anchorage-independent growth by providing GAB2-expressing cells a survival advantage through upregulation of BCL-2 family of anti-apoptotic factors. Of note, collaboration of GAB2 with mutant NRAS enhanced tumorigenesis in vivo and led to an increased vessel density with strong CD34 and VEGFR2 activity. We found that GAB2 facilitiated an angiogenic switch by upregulating HIF-1α and VEGF levels. This angiogenic response was significantly suppressed with the MEK inhibitor PD325901. These data suggest that GAB2-mediated signaling cascades collaborate with NRAS-driven downstream activation for conferring an aggressive phenotype in melanoma. Second, we show that GAB2/NRAS signaling axis is non-linear and non-redundant in melanocytes and melanoma, and thus are acting independent of each other. Finally, we establish a link between GAB2 and angiogenesis in melanoma for the first time. In conclusion, our findings provide evidence that GAB2 is a novel regulator of tumor angiogenesis in NRAS-driven melanoma through regulation of HIF-1α and VEGF expressions mediated by RAS-RAF-MEK-ERK signaling.
Collapse
Affiliation(s)
- Y Yang
- Department of Dermatology, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Sun Z, Li X, Massena S, Kutschera S, Padhan N, Gualandi L, Sundvold-Gjerstad V, Gustafsson K, Choy WW, Zang G, Quach M, Jansson L, Phillipson M, Abid MR, Spurkland A, Claesson-Welsh L. VEGFR2 induces c-Src signaling and vascular permeability in vivo via the adaptor protein TSAd. ACTA ACUST UNITED AC 2012; 209:1363-77. [PMID: 22689825 PMCID: PMC3405501 DOI: 10.1084/jem.20111343] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
VEGFR2 activates c-Src and induces vascular permeability by binding to the adaptor protein TSAd Regulation of vascular endothelial (VE) growth factor (VEGF)–induced permeability is critical in physiological and pathological processes. We show that tyrosine phosphorylation of VEGF receptor 2 (VEGFR2) at Y951 facilitates binding of VEGFR2 to the Rous sarcoma (Src) homology 2-domain of T cell–specific adaptor (TSAd), which in turn regulates VEGF-induced activation of the c-Src tyrosine kinase and vascular permeability. c-Src was activated in vivo and in vitro in a VEGF/TSAd-dependent manner, and was regulated via increased phosphorylation at pY418 and reduced phosphorylation at pY527. Tsad silencing blocked VEGF-induced c-Src activation, but did not affect pathways involving phospholipase Cγ, extracellular regulated kinase, and endothelial nitric oxide. VEGF-induced rearrangement of VE–cadherin–positive junctions in endothelial cells isolated from mouse lungs, or in mouse cremaster vessels, was dependent on TSAd expression, and TSAd formed a complex with VE-cadherin, VEGFR2, and c-Src at endothelial junctions. Vessels in tsad−/− mice showed undisturbed flow and pressure, but impaired VEGF-induced permeability, as measured by extravasation of Evans blue, dextran, and microspheres in the skin and the trachea. Histamine-induced extravasation was not affected by TSAd deficiency. We conclude that TSAd is required for VEGF-induced, c-Src-mediated regulation of endothelial cell junctions and for vascular permeability.
Collapse
Affiliation(s)
- Zuyue Sun
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|