1
|
Roudaut M, Caillaud A, Souguir Z, Bray L, Girardeau A, Rimbert A, Croyal M, Lambert G, Patitucci M, Delpouve G, Vandenhaute É, Le May C, Maubon N, Cariou B, Si‐Tayeb K. Human induced pluripotent stem cells-derived liver organoids grown on a Biomimesys® hyaluronic acid-based hydroscaffold as a new model for studying human lipoprotein metabolism. Bioeng Transl Med 2024; 9:e10659. [PMID: 39036087 PMCID: PMC11256179 DOI: 10.1002/btm2.10659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/29/2024] [Accepted: 02/12/2024] [Indexed: 07/23/2024] Open
Abstract
The liver plays a key role in the metabolism of lipoproteins, controlling both production and catabolism. To accelerate the development of new lipid-lowering therapies in humans, it is essential to have a relevant in vitro study model available. The current hepatocyte-like cells (HLCs) models derived from hiPSC can be used to model many genetically driven diseases but require further improvement to better recapitulate the complexity of liver functions. Here, we aimed to improve the maturation of HLCs using a three-dimensional (3D) approach using Biomimesys®, a hyaluronic acid-based hydroscaffold in which hiPSCs may directly form aggregates and differentiate toward a functional liver organoid model. After a 28-day differentiation 3D protocol, we showed that many hepatic genes were upregulated in the 3D model (liver organoids) in comparison with the 2D model (HLCs). Liver organoids, grown on Biomimesys®, exhibited an autonomous cell organization, were composed of different cell types and displayed enhanced cytochromes P450 activities compared to HLCs. Regarding the functional capacities of these organoids, we showed that they were able to accumulate lipids (hepatic steatosis), internalize low-density lipoprotein and secrete apolipoprotein B. Interestingly, we showed for the first time that this model was also able to produce apolipoprotein (a), the apolipoprotein (a) specific of Lp(a). This innovative hiPSC-derived liver organoid model may serve as a relevant model for studying human lipopoprotein metabolism, including Lp(a).
Collapse
Affiliation(s)
- Meryl Roudaut
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
- HCS PharmaLilleFrance
| | - Amandine Caillaud
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | | | - Lise Bray
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | - Aurore Girardeau
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | - Antoine Rimbert
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | - Mikaël Croyal
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
- CRNH‐Ouest Mass Spectrometry Core FacilityNantesFrance
| | - Gilles Lambert
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI)Université de La RéunionSaint‐Denisde La RéunionFrance
| | - Murielle Patitucci
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | | | | | - Cédric Le May
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | | | - Bertrand Cariou
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| | - Karim Si‐Tayeb
- Nantes Université, CHU Nantes, CNRS, Inserm, l'institut du thoraxNantesFrance
| |
Collapse
|
2
|
An C, Sun F, Liu C, Huang S, Xu T, Zhang C, Ge S. IQGAP1 promotes mitochondrial damage and activation of the mtDNA sensor cGAS-STING pathway to induce endothelial cell pyroptosis leading to atherosclerosis. Int Immunopharmacol 2023; 123:110795. [PMID: 37597406 DOI: 10.1016/j.intimp.2023.110795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
Atherosclerosis (AS) is the most common cardiovascular disease and has limited therapeutic options. IQ motif-containing GTPase-activating protein 1 (IQGAP1) is an important scaffolding protein regulating mitochondrial function influencing endothelial cell activity. Evidence suggests that mitochondrial damage can lead to leakage of mtDNA into the cytoplasm to activate the DNA sensor cGAS-STING to mediate pyroptosis. However, whether IQGAP1 induces NLRP3-mediated endothelial cell pyroptosis by regulating mitochondrial function and activating the DNA sensor cGAS-STING, and its underlying mechanisms remain unclear. In vivo, ApoE-/- C57BL/J and Ldlr-/- C57BL/J mice were pre-injected with adeno-associated virus (AAV) by the tail vein to specifically silence IQGAP1 expression and were fed a high-fat diet (HFD) for 12 weeks. IQGAP1 knockdown reduced mtDNA release and decreased the expression of DNA receptors and pyroptosis-related molecules as determined by immunohistochemistry and immunofluorescence. In vitro, palmitic acid (0.3 mmol/L) was incubated with human umbilical vein endothelial cells (HUVECs) for 24 h. Overexpression of IQGAP1 in HUVECs, flow cytometry, and mitochondrial superoxide staining revealed increased levels of ROS. Moreover, the mitochondrial tracker with dsDNA co-localization showed the release of mtDNA into the cytoplasm increased, which activated the DNA receptor cGAS-STING. Protein blotting and TUNEL staining revealed that IQGAP1 promoted NLRP3-mediated pyroptosis. Furthermore, cGAS or STING small-molecule inhibitors RU.521 or C-176 reverse IQGAP1-promoted HUVECs from undergoing NLRP3-mediated pyroptosis. These results suggest that IQGAP1 promotes oxidative stress and mtDNA release, activates the DNA sensor cGAS-STING, and leads to NLRP3-mediated pyroptosis. The present study provides new insights into the mechanisms underlying AS and identifies new pharmacological targets for treatment.
Collapse
Affiliation(s)
- Cheng An
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Fei Sun
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Can Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Shaojun Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, China
| | - Chengxin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China.
| |
Collapse
|
3
|
Batta SPR, Rio M, Lebot C, Baron-menguy C, Le Ruz R, Loirand G, Vion A. ARHGEF18 participates in Endothelial Cell Mechano-sensitivity in Response to Flow.. [DOI: 10.1101/2022.09.10.507283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
AbstractHemodynamic forces play an important role in vascular network development and homeostasis. In physiological condition, shear stress generated by laminar flow promotes endothelial cells (EC) health and induces their alignment in the direction of flow. In contrast, altered hemodynamic forces induce endothelial dysfunction and lead to the development of vascular disorders such as atherosclerosis and aneurysms. Following mechano-sensor activation, Rho protein-mediated cytoskeletal rearrangement is one of the first steps in transforming flow-induced forces into intracellular signals in EC via guanine nucleotide exchange factors (RhoGEFs) that mediate the spatio-temporal activation of these Rho proteins. Here we identified ARHGEF18 as a flow-sensitive RhoGEF specifically activating RhoA. Both ARHGEF18 expression and activity were controlled by shear stress level. ARHGEF18 promotes EC adhesion, focal adhesion formation and migration. ARHGEF18 localized to the tight junction by interacting with ZO-1 and participated to shear stress-induced EC elongation and alignment via its nucleotide exchange activity and the activation of p38 MAPK. Our study therefore characterized ARHGEF18 as the first flow-sensitive RhoA GEF in ECs, whose activity is essential for the maintenance of intercellular junctions and a properly organized endothelial monolayer under physiological flow conditions.
Collapse
|
4
|
Rusu-Zota G, Manole OM, Galeș C, Porumb-Andrese E, Obadă O, Mocanu CV. Kaposi Sarcoma, a Trifecta of Pathogenic Mechanisms. Diagnostics (Basel) 2022; 12:1242. [PMID: 35626397 PMCID: PMC9140574 DOI: 10.3390/diagnostics12051242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/29/2022] [Accepted: 05/13/2022] [Indexed: 01/10/2023] Open
Abstract
Kaposi's sarcoma is a rare disease with four known variants: classic, epidemic, endemic and iatrogenic (transplant-related), all caused by an oncogenic virus named Human Herpes Virus 8. The viral infection in itself, along with the oncogenic properties of HHV8 and with immune system dysfunction, forms the grounds on which Kaposi's Sarcoma may develop. Infection with HHV8 occurs through saliva via close contacts, blood, blood products, solid organ donation and, rarely, vertical transmission. Chronic inflammation and oncogenesis are promoted by a mix of viral genes that directly promote cell survival and transformation or interfere with the regular cell cycle and cell signaling (of particular note: LANA-1, v-IL6, vBCL-2, vIAP, vIRF3, vGPCR, gB, K1, K8.1, K15). The most common development sites for Kaposi's sarcoma are the skin, mucocutaneous zones, lymph nodes and visceral organs, but it can also rarely appear in the musculoskeletal system, urinary system, endocrine organs, heart or eye. Histopathologically, spindle cell proliferation with slit-like vascular spaces, plasma cell and lymphocyte infiltrate are characteristic. The clinical presentation is heterogenic depending on the variant; some patients have indolent disease and others have aggressive disease. The treatment options include highly active antiretroviral therapy, surgery, radiation therapy, chemotherapy, and immunotherapy. A literature search was carried out using the MEDLINE/PubMed, SCOPUS and Google Scholar databases with a combination of keywords with the aim to provide critical, concise, and comprehensive insights into advances in the pathogenic mechanism of Kaposi's sarcoma.
Collapse
Affiliation(s)
- Gabriela Rusu-Zota
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 700115 Iasi, Romania;
| | - Oana Mădălina Manole
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 700115 Iasi, Romania
| | - Cristina Galeș
- Department of Histology, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 700115 Iasi, Romania;
| | - Elena Porumb-Andrese
- Department of Dermatology, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 700115 Iasi, Romania;
| | - Otilia Obadă
- Department of Ophthalmology, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 700115 Iasi, Romania;
| | - Cezar Valentin Mocanu
- Department of Anatomical Pathology, University of Medicine and Pharmacy “Grigore T. Popa” Iasi, 700115 Iasi, Romania;
| |
Collapse
|
5
|
Urade R, Chiu YH, Chiu CC, Wu CY. Small GTPases and Their Regulators: A Leading Road toward Blood Vessel Development in Zebrafish. Int J Mol Sci 2022; 23:4991. [PMID: 35563380 PMCID: PMC9099977 DOI: 10.3390/ijms23094991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 12/26/2022] Open
Abstract
Members of the Ras superfamily have been found to perform several functions leading to the development of eukaryotes. These small GTPases are divided into five major subfamilies, and their regulators can "turn on" and "turn off" signals. Recent studies have shown that this superfamily of proteins has various roles in the process of vascular development, such as vasculogenesis and angiogenesis. Here, we discuss the role of these subfamilies in the development of the vascular system in zebrafish.
Collapse
Affiliation(s)
- Ritesh Urade
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.U.); (Y.-H.C.)
| | - Yan-Hui Chiu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.U.); (Y.-H.C.)
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.U.); (Y.-H.C.)
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan; (R.U.); (Y.-H.C.)
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| |
Collapse
|
6
|
Vav Proteins in Development of the Brain: A Potential Relationship to the Pathogenesis of Congenital Zika Syndrome? Viruses 2022; 14:v14020386. [PMID: 35215978 PMCID: PMC8874935 DOI: 10.3390/v14020386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 12/07/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy can result in a significant impact on the brain and eye of the developing fetus, termed congenital zika syndrome (CZS). At a morphological level, the main serious presentations of CZS are microcephaly and retinal scarring. At a cellular level, many cell types of the brain may be involved, but primarily neuronal progenitor cells (NPC) and developing neurons. Vav proteins have guanine exchange activity in converting GDP to GTP on proteins such as Rac1, Cdc42 and RhoA to stimulate intracellular signaling pathways. These signaling pathways are known to play important roles in maintaining the polarity and self-renewal of NPC pools by coordinating the formation of adherens junctions with cytoskeletal rearrangements. In developing neurons, these same pathways are adopted to control the formation and growth of neurites and mediate axonal guidance and targeting in the brain and retina. This review describes the role of Vavs in these processes and highlights the points of potential ZIKV interaction, such as (i) the binding and entry of ZIKV in cells via TAM receptors, which may activate Vav/Rac/RhoA signaling; (ii) the functional convergence of ZIKV NS2A with Vav in modulating adherens junctions; (iii) ZIKV NS4A/4B protein effects on PI3K/AKT in a regulatory loop via PPI3 to influence Vav/Rac1 signaling in neurite outgrowth; and (iv) the induction of SOCS1 and USP9X following ZIKV infection to regulate Vav protein degradation or activation, respectively, and impact Vav/Rac/RhoA signaling in NPC and neurons. Experiments to define these interactions will further our understanding of the molecular basis of CZS and potentially other developmental disorders stemming from in utero infections. Additionally, Vav/Rac/RhoA signaling pathways may present tractable targets for therapeutic intervention or molecular rationale for disease severity in CZS.
Collapse
|
7
|
Zhou Y, Farooqi AA, Xu B. Comprehensive review on signaling pathways of dietary saponins in cancer cells suppression. Crit Rev Food Sci Nutr 2021:1-26. [PMID: 34751072 DOI: 10.1080/10408398.2021.2000933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Nutrigenomics utilizes high-throughput genomic technologies to reveal changes in gene and protein levels. Excitingly, ever-growing body of scientific findings has provided sufficient evidence about the interplay between diet and genes. Cutting-edge research and advancements in genomics, epigenetics and metabolomics have deepened our understanding on the role of dietary factors in the inhibition of carcinogenesis and metastasis. Dietary saponins, a type of triterpene glycosides, are generally found in Platycodon grandifloras, Dioscorea oppositifolia, asparagus, legumes, and sea cucumber. Wealth of information has started to shed light on pleiotropic mechanistic roles of dietary saponins in cancer prevention and inhibition. In this review, we have attempted to summarize the in vitro research of dietary saponins in the last two decades by searching common databases such as Google Scholar, PubMed, Scopus, and Web of Science. The results showed that dietary saponins exerted anti-cancer activities via regulation of apoptosis, autophagy, arrest cell cycle, anti-proliferation, anti-metastasis, and anti-angiogenesis, by regulation of several critical signaling pathways, including MAPK, PI3K/Akt/mTOR, NF-κB, and VEGF/VEGFR. However, there is no data about the dosage of dietary saponins for practical anti-cancer effects in human bodies. Extensive clinical studies are needed to confirm the effectiveness of dietary saponins for further commercial and medical applications.
Collapse
Affiliation(s)
- Yifan Zhou
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai, Guangdong, China.,Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| | | | - Baojun Xu
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai, Guangdong, China
| |
Collapse
|
8
|
Rodenburg WS, van Buul JD. Rho GTPase signalling networks in cancer cell transendothelial migration. VASCULAR BIOLOGY 2021; 3:R77-R95. [PMID: 34738075 PMCID: PMC8558887 DOI: 10.1530/vb-21-0008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/21/2023]
Abstract
Rho GTPases are small signalling G-proteins that are central regulators of cytoskeleton dynamics, and thereby regulate many cellular processes, including the shape, adhesion and migration of cells. As such, Rho GTPases are also essential for the invasive behaviour of cancer cells, and thus involved in several steps of the metastatic cascade, including the extravasation of cancer cells. Extravasation, the process by which cancer cells leave the circulation by transmigrating through the endothelium that lines capillary walls, is an essential step for metastasis towards distant organs. During extravasation, Rho GTPase signalling networks not only regulate the transmigration of cancer cells but also regulate the interactions between cancer and endothelial cells and are involved in the disruption of the endothelial barrier function, ultimately allowing cancer cells to extravasate into the underlying tissue and potentially form metastases. Thus, targeting Rho GTPase signalling networks in cancer may be an effective approach to inhibit extravasation and metastasis. In this review, the complex process of cancer cell extravasation will be discussed in detail. Additionally, the roles and regulation of Rho GTPase signalling networks during cancer cell extravasation will be discussed, both from a cancer cell and endothelial cell point of view.
Collapse
Affiliation(s)
- Wessel S Rodenburg
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab at Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands.,Leeuwenhoek Centre for Advanced Microscopy, Section Molecular Cytology at Swammerdam Institute for Life Sciences at University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Regulation of Rac1 Activation in Choroidal Endothelial Cells: Insights into Mechanisms in Age-Related Macular Degeneration. Cells 2021; 10:cells10092414. [PMID: 34572063 PMCID: PMC8469925 DOI: 10.3390/cells10092414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
Age-related macular degeneration (AMD) is one of the leading causes of blindness worldwide. Vision loss from the neovascular form is associated with the invasion of choroidal endothelial cells into the neural retina to form vision-threatening macular neovascularization (MNV). Anti-angiogenic agents are the current standard of care but are effective in only ~50% of AMD cases. The molecular mechanisms involved in invasive MNV point to the importance of regulating signaling pathways that lead to pathologic biologic outcomes. In studies testing the effects of AMD-related stresses, activation of the Rho GTPase, Rac1, was found to be important for the choroidal endothelial cell invasion into the neural retina. However, current approaches to prevent Rac1 activation are inefficient and less effective. We summarize active Rac1-mediated mechanisms that regulate choroidal endothelial cell migration. Specifically, we discuss our work regarding the role of a multidomain protein, IQ motif containing GTPase activating protein 1 (IQGAP1), in sustaining pathologic Rac1 activation and a mechanism by which active Rap1, a Ras-like GTPase, may prevent active Rac1-mediated choroidal endothelial cell migration.
Collapse
|
10
|
Zimmer S, Goody PR, Oelze M, Ghanem A, Mueller CF, Laufs U, Daiber A, Jansen F, Nickenig G, Wassmann S. Inhibition of Rac1 GTPase Decreases Vascular Oxidative Stress, Improves Endothelial Function, and Attenuates Atherosclerosis Development in Mice. Front Cardiovasc Med 2021; 8:680775. [PMID: 34422919 PMCID: PMC8377253 DOI: 10.3389/fcvm.2021.680775] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Aims: Oxidative stress and inflammation contribute to atherogenesis. Rac1 GTPase regulates pro-oxidant NADPH oxidase activity, reactive oxygen species (ROS) formation, actin cytoskeleton organization and monocyte adhesion. We investigated the vascular effects of pharmacological inhibition of Rac1 GTPase in mice. Methods and Results: We treated wild-type and apolipoprotein E-deficient (ApoE−/−) mice with Clostridium sordellii lethal toxin (LT), a Rac1 inhibitor, and assessed vascular oxidative stress, expression and activity of involved proteins, endothelial function, macrophage infiltration, and atherosclerosis development. LT-treated wild-type mice displayed decreased vascular NADPH oxidase activity and ROS production. Therapeutic LT doses had no impact on behavior, food intake, body weight, heart rate, blood pressure, vascular and myocardial function, differential blood count, and vascular permeability. ApoE−/− mice were fed a cholesterol-rich diet and were treated with LT or vehicle. LT treatment led to decreased aortic Rac1 GTPase activity, NADPH oxidase activity and ROS production, but had no impact on expression and membrane translocation of NADPH oxidase subunits and RhoA GTPase activity. LT-treated mice showed improved aortic endothelium-dependent vasodilation, attenuated atherosclerotic lesion formation and reduced macrophage infiltration of atherosclerotic plaques. Concomitant treatment of cholesterol-fed ApoE−/− mice with LT, the specific synthetic Rac1 inhibitor NSC 23766 or simvastatin comparably reduced aortic Rac1 activity, NADPH oxidase activity, oxidative stress, endothelial dysfunction, atherosclerosis development, and macrophage infiltration. Conclusions: These findings identify an important role of the small GTPase Rac1 in atherogenesis and provide a potential target for anti-atherosclerotic therapy.
Collapse
Affiliation(s)
- Sebastian Zimmer
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Philip Roger Goody
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Matthias Oelze
- Zentrum für Kardiologie - Kardiologie I, Universitätsmedizin der Johannes Gutenberg-Universität, Mainz, Germany
| | - Alexander Ghanem
- Department of Internal Medicine II - Cardiology and Medical Intensive Care, Asklepius Hospital Nord - Heidberg, Hamburg, Germany
| | - Cornelius F Mueller
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Ulrich Laufs
- Department of Cardiology, University Hospital Leipzig, Leipzig, Germany
| | - Andreas Daiber
- Zentrum für Kardiologie - Kardiologie I, Universitätsmedizin der Johannes Gutenberg-Universität, Mainz, Germany
| | - Felix Jansen
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Georg Nickenig
- Department of Internal Medicine II, Heart Center Bonn, University Hospital Bonn, Bonn, Germany
| | - Sven Wassmann
- Cardiology Pasing, Munich, Germany.,Department of Inernal Medicine III, Saarlang University Medical Center, Homburg, Germany
| |
Collapse
|
11
|
Phng LK, Belting HG. Endothelial cell mechanics and blood flow forces in vascular morphogenesis. Semin Cell Dev Biol 2021; 120:32-43. [PMID: 34154883 DOI: 10.1016/j.semcdb.2021.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 12/21/2022]
Abstract
The vertebrate cardiovascular system is made up by a hierarchically structured network of highly specialised blood vessels. This network emerges during early embryogenesis and evolves in size and complexity concomitant with embryonic growth and organ formation. Underlying this plasticity are actin-driven endothelial cell behaviours, which allow endothelial cells to change their shape and move within the vascular network. In this review, we discuss the cellular and molecular mechanisms involved in vascular network formation and how these intrinsic mechanisms are influenced by haemodynamic forces provided by pressurized blood flow. While most of this review focusses on in vivo evidence from zebrafish embryos, we also mention complementary findings obtained in other experimental systems.
Collapse
Affiliation(s)
- Li-Kun Phng
- Laboratory for Vascular Morphogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan.
| | - Heinz-Georg Belting
- Department of Cell Biology, Biozentrum, University of Basel, Basel 4056, Switzerland.
| |
Collapse
|
12
|
Lauri A, Fasano G, Venditti M, Dallapiccola B, Tartaglia M. In vivo Functional Genomics for Undiagnosed Patients: The Impact of Small GTPases Signaling Dysregulation at Pan-Embryo Developmental Scale. Front Cell Dev Biol 2021; 9:642235. [PMID: 34124035 PMCID: PMC8194860 DOI: 10.3389/fcell.2021.642235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
While individually rare, disorders affecting development collectively represent a substantial clinical, psychological, and socioeconomic burden to patients, families, and society. Insights into the molecular mechanisms underlying these disorders are required to speed up diagnosis, improve counseling, and optimize management toward targeted therapies. Genome sequencing is now unveiling previously unexplored genetic variations in undiagnosed patients, which require functional validation and mechanistic understanding, particularly when dealing with novel nosologic entities. Functional perturbations of key regulators acting on signals' intersections of evolutionarily conserved pathways in these pathological conditions hinder the fine balance between various developmental inputs governing morphogenesis and homeostasis. However, the distinct mechanisms by which these hubs orchestrate pathways to ensure the developmental coordinates are poorly understood. Integrative functional genomics implementing quantitative in vivo models of embryogenesis with subcellular precision in whole organisms contribute to answering these questions. Here, we review the current knowledge on genes and mechanisms critically involved in developmental syndromes and pediatric cancers, revealed by genomic sequencing and in vivo models such as insects, worms and fish. We focus on the monomeric GTPases of the RAS superfamily and their influence on crucial developmental signals and processes. We next discuss the effectiveness of exponentially growing functional assays employing tractable models to identify regulatory crossroads. Unprecedented sophistications are now possible in zebrafish, i.e., genome editing with single-nucleotide precision, nanoimaging, highly resolved recording of multiple small molecules activity, and simultaneous monitoring of brain circuits and complex behavioral response. These assets permit accurate real-time reporting of dynamic small GTPases-controlled processes in entire organisms, owning the potential to tackle rare disease mechanisms.
Collapse
Affiliation(s)
- Antonella Lauri
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | | | | | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
13
|
Vion AC, Perovic T, Petit C, Hollfinger I, Bartels-Klein E, Frampton E, Gordon E, Claesson-Welsh L, Gerhardt H. Endothelial Cell Orientation and Polarity Are Controlled by Shear Stress and VEGF Through Distinct Signaling Pathways. Front Physiol 2021; 11:623769. [PMID: 33737879 PMCID: PMC7960671 DOI: 10.3389/fphys.2020.623769] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Vascular networks form, remodel and mature under the influence of multiple signals of mechanical or chemical nature. How endothelial cells read and interpret these signals, and how they integrate information when they are exposed to both simultaneously is poorly understood. Here, we show using flow-induced shear stress and VEGF-A treatment on endothelial cells in vitro, that the response to the magnitude of a mechanical stimulus is influenced by the concentration of a chemical stimulus, and vice versa. By combining different flow levels and different VEGF-A concentrations, front-rear polarity of endothelial cells against the flow direction was established in a flow and VEGF-A dose-response while their alignment with the flow displayed a biphasic response depending on the VEGF-A dose (perpendicular at physiological dose, aligned at no or pathological dose of VEGF-A). The effect of pharmaceutical inhibitors demonstrated that while VEGFR2 is essential for both polarity and orientation establishment in response to flow with and without VEGF-A, different downstream effectors were engaged depending on the presence of VEGF-A. Thus, Src family inhibition (c-Src, Yes, Fyn together) impaired alignment and polarity without VEGF-A while FAK inhibition modified polarity and alignment only when endothelial cells were exposed to VEGF-A. Studying endothelial cells in the aortas of VEGFR2Y949F mutant mice and SRC iEC-KO mice confirmed the role of VEGFR2 and specified the role of c-SRC in vivo. Endothelial cells of VEGFR2Y949F mutant mice lost their polarity and alignment while endothelial cells from SRC iEC-KO mice only showed reduced polarity. We propose here that VEGFR2 is a sensor able to integrate chemical and mechanical information simultaneously and that the underlying pathways and mechanisms activated will depend on the co-stimulation. Flow alone shifts VEGFR2 signaling toward a Src family pathway activation and a junctional effect (both in vitro and in vivo) while flow and VEGF-A together shift VEGFR2 signaling toward focal adhesion activation (in vitro) both modifying cell responses that govern orientation and polarity.
Collapse
Affiliation(s)
- Anne-Clémence Vion
- Integrative Vascular Biology Laboratory, Max Delbruck Center for Molecular Medicine, Berlin, Germany.,Université de Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France
| | - Tijana Perovic
- Integrative Vascular Biology Laboratory, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Charlie Petit
- Integrative Vascular Biology Laboratory, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Irene Hollfinger
- Integrative Vascular Biology Laboratory, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Eireen Bartels-Klein
- Integrative Vascular Biology Laboratory, Max Delbruck Center for Molecular Medicine, Berlin, Germany
| | - Emmanuelle Frampton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Emma Gordon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.,Beijer and Science for Life Laboratories, Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Lena Claesson-Welsh
- Beijer and Science for Life Laboratories, Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max Delbruck Center for Molecular Medicine, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
14
|
Connor K, Murray DW, Jarzabek MA, Tran NL, White K, Dicker P, Sweeney KJ, O’Halloran PJ, MacCarthy B, Shiels LP, Lodi F, Lambrechts D, Sarkaria JN, Schiffelers RM, Symons M, Byrne AT. Targeting the RhoGEF βPIX/COOL-1 in Glioblastoma: Proof of Concept Studies. Cancers (Basel) 2020; 12:cancers12123531. [PMID: 33256106 PMCID: PMC7761123 DOI: 10.3390/cancers12123531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/15/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) is an incurable disease with a 14-month average life-expectancy following diagnosis, and clinical management has not improved in four decades. GBM mortality is due to rapid tumour growth and invasion into surrounding normal brain. Invasive cells make complete surgical removal of the tumour impossible, and result in disease relapse. Thus, it is imperative that any new treatment strategy takes these invading cells into consideration. Bevacizumab (Bev), which prevents the formation of new blood vessels, is an FDA approved therapy, but it has failed to increase overall survival in GBM and has even been shown to increase tumour invasion in some cases. Complementary anti-invasive therapies are therefore urgently required to enhance bevacizumab efficacy. We have identified βPIX/COOL-1, a RhoGEF protein which plays an important role in GBM cell invasion and angiogenesis and could be a useful target in this setting. Abstract Glioblastoma (GBM), a highly invasive and vascular malignancy is shown to rapidly develop resistance and evolve to a more invasive phenotype following bevacizumab (Bev) therapy. Rho Guanine Nucleotide Exchange Factor proteins (RhoGEFs) are mediators of key components in Bev resistance pathways, GBM and Bev-induced invasion. To identify GEFs with enhanced mRNA expression in the leading edge of GBM tumours, a cohort of GEFs was assessed using a clinical dataset. The GEF βPix/COOL-1 was identified, and the functional effect of gene depletion assessed using 3D-boyden chamber, proliferation, and colony formation assays in GBM cells. Anti-angiogenic effects were assessed in endothelial cells using tube formation and wound healing assays. In vivo effects of βPix/COOL-1-siRNA delivered via RGD-Nanoparticle in combination with Bev was studied in an invasive model of GBM. We found that siRNA-mediated knockdown of βPix/COOL-1 in vitro decreased cell invasion, proliferation and increased apoptosis in GBM cell lines. Moreover βPix/COOL-1 mediated endothelial cell migration in vitro. Mice treated with βPix/COOL-1 siRNA-loaded RGD-Nanoparticle and Bev demonstrated a trend towards improved median survival compared with Bev monotherapy. Our hypothesis generating study suggests that the RhoGEF βPix/COOL-1 may represent a target of vulnerability in GBM, in particular to improve Bev efficacy.
Collapse
Affiliation(s)
- Kate Connor
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - David W. Murray
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Monika A. Jarzabek
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Nhan L. Tran
- Department of Cancer Biology and Neurological Surgery, Mayo Clinic Arizona, Scottsdale, AZ 85054, USA;
| | - Kieron White
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Patrick Dicker
- Epidemiology & Public Health, Royal College of Surgeons in Ireland, Dublin 2, Ireland;
| | - Kieron J. Sweeney
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
- National Neurosurgical Department, Beaumont Hospital, Dublin 9, Ireland
| | - Philip J. O’Halloran
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
- National Neurosurgical Department, Beaumont Hospital, Dublin 9, Ireland
| | - Brian MacCarthy
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Liam P. Shiels
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
| | - Francesca Lodi
- Center for Cancer Biology, Laboratory for Translational Genetics, Vlaams Instituut voor Biotechnologie (VIB), B-3000 Leuven, Belgium; (F.L.); (D.L.)
| | - Diether Lambrechts
- Center for Cancer Biology, Laboratory for Translational Genetics, Vlaams Instituut voor Biotechnologie (VIB), B-3000 Leuven, Belgium; (F.L.); (D.L.)
| | - Jann N. Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA;
| | - Raymond M. Schiffelers
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, 100 3584 Utrecht, The Netherlands;
| | - Marc Symons
- Department of Oncology & Cell Biology, Feinstein Institute for Medical Research at North Shore-LIJ, Manhasset, NY 11030, USA;
| | - Annette T. Byrne
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland; (K.C.); (D.W.M.); (M.A.J.); (K.W.); (K.J.S.); (P.J.O.); (B.M.); (L.P.S.)
- Correspondence: ; Tel.: +353-1-402-8673
| |
Collapse
|
15
|
Cross-Talk between NADPH Oxidase and Mitochondria: Role in ROS Signaling and Angiogenesis. Cells 2020; 9:cells9081849. [PMID: 32781794 PMCID: PMC7466096 DOI: 10.3390/cells9081849] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis, a new vessel formation from the pre-existing ones, is essential for embryonic development, wound repair and treatment of ischemic heart and limb diseases. However, dysregulated angiogenesis contributes to various pathologies such as diabetic retinopathy, atherosclerosis and cancer. Reactive oxygen species (ROS) derived from NADPH oxidase (NOX) as well as mitochondria play an important role in promoting the angiogenic switch from quiescent endothelial cells (ECs). However, how highly diffusible ROS produced from different sources and location can communicate with each other to regulate angiogenesis remains unclear. To detect a localized ROS signal in distinct subcellular compartments in real time in situ, compartment-specific genetically encoded redox-sensitive fluorescence biosensors have been developed. Recently, the intercellular communication, “cross-talk”, between ROS derived from NOX and mitochondria, termed “ROS-induced ROS release”, has been proposed as a mechanism for ROS amplification at distinct subcellular compartments, which are essential for activation of redox signaling. This “ROS-induced ROS release” may represent a feed-forward mechanism of localized ROS production to maintain sustained signaling, which can be targeted under pathological conditions with oxidative stress or enhanced to promote therapeutic angiogenesis. In this review, we summarize the recent knowledge regarding the role of the cross-talk between NOX and mitochondria organizing the sustained ROS signaling involved in VEGF signaling, neovascularization and tissue repair.
Collapse
|
16
|
Vav2 pharmaco-mimetic mice reveal the therapeutic value and caveats of the catalytic inactivation of a Rho exchange factor. Oncogene 2020; 39:5098-5111. [PMID: 32528129 PMCID: PMC7610363 DOI: 10.1038/s41388-020-1353-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/29/2020] [Accepted: 06/03/2020] [Indexed: 11/20/2022]
Abstract
The current paradigm holds that the inhibition of Rho guanosine nucleotide exchange factors (GEFs), the enzymes that stimulate Rho GTPases, can be a valuable therapeutic strategy to treat Rho-dependent tumors. However, formal validation of this idea using in vivo models is still missing. In this context, it is worth remembering that many Rho GEFs can mediate both catalysis-dependent and independent responses, thus raising the possibility that the inhibition of their catalytic activities might not be sufficient per se to block tumorigenic processes. On the other hand, the inhibition of these enzymes can trigger collateral side effects that could preclude the practical implementation of anti-GEF therapies. To address those issues, we have generated mouse models to mimic the effect of the systemic application of an inhibitor for the catalytic activity of the Rho GEF Vav2 at the organismal level. Our results indicate that lowering the catalytic activity of Vav2 below specific thresholds is sufficient to block skin tumor initiation, promotion, and progression. They also reveal that the negative side effects typically induced by the loss of Vav2 can be bypassed depending on the overall level of Vav2 inhibition achieved in vivo. These data underscore the pros and cons of anti-Rho GEF therapies for cancer treatment. They also support the idea that Vav2 could represent a viable drug target.
Collapse
|
17
|
Abstract
The Rho GTPase Cdc42 is a central regulator of cell polarity in diverse cell types. The activity of Cdc42 is dynamically controlled in time and space to enable distinct polarization events, which generally occur along a single axis in response to spatial cues. Our understanding of the mechanisms underlying Cdc42 polarization has benefited largely from studies of the budding yeast Saccharomyces cerevisiae, a genetically tractable model organism. In budding yeast, Cdc42 activation occurs in two temporal steps in the G1 phase of the cell cycle to establish a proper growth site. Here, we review findings in budding yeast that reveal an intricate crosstalk among polarity proteins for biphasic Cdc42 regulation. The first step of Cdc42 activation may determine the axis of cell polarity, while the second step ensures robust Cdc42 polarization for growth. Biphasic Cdc42 polarization is likely to ensure the proper timing of events including the assembly and recognition of spatial landmarks and stepwise assembly of a new ring of septins, cytoskeletal GTP-binding proteins, at the incipient bud site. Biphasic activation of GTPases has also been observed in mammalian cells, suggesting that biphasic activation could be a general mechanism for signal-responsive cell polarization. Cdc42 activity is necessary for polarity establishment during normal cell division and development, but its activity has also been implicated in the promotion of aging. We also discuss negative polarity signaling and emerging concepts of Cdc42 signaling in cellular aging.
Collapse
Affiliation(s)
- Kristi E Miller
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210.,Present address: Department of Biochemistry and Cell Biology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755
| | - Pil Jung Kang
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210
| | - Hay-Oak Park
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
18
|
Harde E, Nicholson L, Furones Cuadrado B, Bissen D, Wigge S, Urban S, Segarra M, Ruiz de Almodóvar C, Acker-Palmer A. EphrinB2 regulates VEGFR2 during dendritogenesis and hippocampal circuitry development. eLife 2019; 8:49819. [PMID: 31868584 PMCID: PMC6927743 DOI: 10.7554/elife.49819] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/06/2019] [Indexed: 12/12/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is an angiogenic factor that play important roles in the nervous system, although it is still unclear which receptors transduce those signals in neurons. Here, we show that in the developing hippocampus VEGFR2 (also known as KDR or FLK1) is expressed specifically in the CA3 region and it is required for dendritic arborization and spine morphogenesis in hippocampal neurons. Mice lacking VEGFR2 in neurons (Nes-cre Kdrlox/-) show decreased dendritic arbors and spines as well as a reduction in long-term potentiation (LTP) at the associational-commissural – CA3 synapses. Mechanistically, VEGFR2 internalization is required for VEGF-induced spine maturation. In analogy to endothelial cells, ephrinB2 controls VEGFR2 internalization in neurons. VEGFR2-ephrinB2 compound mice (Nes-cre Kdrlox/+ Efnb2lox/+) show reduced dendritic branching, reduced spine head size and impaired LTP. Our results demonstrate the functional crosstalk of VEGFR2 and ephrinB2 in vivo to control dendritic arborization, spine morphogenesis and hippocampal circuitry development.
Collapse
Affiliation(s)
- Eva Harde
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Max Planck Institute for Brain Research, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - LaShae Nicholson
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Max Planck Institute for Brain Research, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Beatriz Furones Cuadrado
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Diane Bissen
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Max Planck Institute for Brain Research, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Sylvia Wigge
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Severino Urban
- Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Marta Segarra
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany
| | - Carmen Ruiz de Almodóvar
- Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany.,European Center for Angioscience, Medicine Faculty Mannheim, Heidelberg University, Heidelberg, Germany.,Institute for Transfusion Medicine and Immunology, Medicine Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience, University of Frankfurt, Frankfurt, Germany.,Max Planck Institute for Brain Research, Frankfurt, Germany.,Buchmann Institute for Molecular Life Sciences (BMLS), University of Frankfurt, Frankfurt, Germany.,Cardio-Pulmonary Institute (CPI), Frankfurt, Germany
| |
Collapse
|
19
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
20
|
Koohini Z, Koohini Z, Teimourian S. Slit/Robo Signaling Pathway in Cancer; a New Stand Point for Cancer Treatment. Pathol Oncol Res 2019; 25:1285-1293. [PMID: 30610466 DOI: 10.1007/s12253-018-00568-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/17/2018] [Indexed: 10/27/2022]
Abstract
Angiogenesis and metastasis are two critical steps for cancer cells survival and migration. The microenvironment of tumor sphere induces new blood vessels formation for enhancing tumor mass. Preexisting capillaries and postcapillary venules in tumors bring about new blood vessels. ROBO1-ROBO4 are transmembrane receptors family which act as guidance molecules of the nervous system. The SLITs family is secreted glycoproteins that bind to these receptors. SLIT-ROBO signaling pathway plays an important role in neurogenesis and immune response. Linkage between ROBOs and their ligands (SLITs) induce chemorepllent signal for regulation of axon guidance and leukocyte cell migration, recent finding shows that it is also involved in endothelial cell migration and angiogenesis in various type of cancers. In this article we review recent finding of SLIT-ROBO pathway in angiogenesis and metastasis.
Collapse
Affiliation(s)
- Zahra Koohini
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zohreh Koohini
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Mesenchyme-specific deletion of Tgf-β1 in the embryonic lung disrupts branching morphogenesis and induces lung hypoplasia. J Transl Med 2019; 99:1363-1375. [PMID: 31028279 PMCID: PMC7422700 DOI: 10.1038/s41374-019-0256-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/06/2019] [Accepted: 03/13/2019] [Indexed: 01/08/2023] Open
Abstract
Proper lung development depends on the precise temporal and spatial expression of several morphogenic factors, including Fgf10, Fgf9, Shh, Bmp4, and Tgf-β. Over- or under-expression of these molecules often leads to aberrant embryonic or postnatal lung development. Herein, we deleted the Tgf-β1 gene specifically within the lung embryonic mesenchymal compartment at specific gestational stages to determine the contribution of this cytokine to lung development. Mutant embryos developed severe lung hypoplasia and died at birth due to the inability to breathe. Despite the markedly reduced lung size, proliferation and differentiation of the lung epithelium was not affected by the lack of mesenchymal expression of the Tgf-β1 gene, while apoptosis was significantly increased in the mutant lung parenchyma. Lack of mesenchymal expression of the Tgf-β1 gene was also associated with reduced lung branching morphogenesis, with accompanying inhibition of the local FGF10 signaling pathway as well as abnormal development of the vascular system. To shed light on the mechanism of lung hypoplasia, we quantified the phosphorylation of 226 proteins in the mutant E12.5 lung compared with control. We identified five proteins, Hrs, Vav2, c-Kit, the regulatory subunit of Pi3k (P85), and Fgfr1, that were over- or under-phosphorylated in the mutant lung, suggesting that they could be indispensable effectors of the TGF-β signaling program during embryonic lung development. In conclusion, we have uncovered novel roles of the mesenchyme-specific Tgf-β1 ligand in embryonic mouse lung development and generated a mouse model that may prove helpful to identify some of the key pathogenic mechanisms underlying lung hypoplasia in humans.
Collapse
|
22
|
Li B, Li Y, Wang Q, Li F, Li F. Corosolic acid impairs human lung adenocarcinoma A549 cells proliferation by inhibiting cell migration. Oncol Lett 2019; 17:5747-5753. [PMID: 31186800 DOI: 10.3892/ol.2019.10262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 01/15/2019] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to investigate the anticancer effects of corosolic acid (CA) in the human lung adenocarcinoma A549 cell line. A549 cells were treated with increasing concentrations of CA, prior to assessing cell viability, migration rate, vascular endothelial growth factor receptor 2 (VEGFR2) kinase activity and cytoskeleton structure. In addition, in vivo imaging system was used to analyze the anticancer effects of CA in vivo. Results demonstrated that CA exhibited a low cytotoxicity with a half maximal inhibitory concentration of 65 µM. In addition, 4 µM CA efficiently inhibited A549 cell migration. Furthermore, CA inhibited VEGFR2 kinase activity and disrupted tubulin structure. Data also revealed that CA inhibited A549 cell proliferation in a xenograft mouse model. In conclusion, results from the present study suggested that CA may be used as a novel potential therapy for lung cancer.
Collapse
Affiliation(s)
- Biao Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Yongjie Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Qiongyu Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Fan Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| | - Fu Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
23
|
Janardhan HP, Trivedi CM. Establishment and maintenance of blood-lymph separation. Cell Mol Life Sci 2019; 76:1865-1876. [PMID: 30758642 PMCID: PMC6482084 DOI: 10.1007/s00018-019-03042-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/15/2019] [Accepted: 02/05/2019] [Indexed: 02/07/2023]
Abstract
Hippocratic Corpus, a collection of Greek medical literature, described the functional anatomy of the lymphatic system in the fifth century B.C. Subsequent studies in cadavers and surgical patients firmly established that lymphatic vessels drain extravasated interstitial fluid, also known as lymph, into the venous system at the bilateral lymphovenous junctions. Recent advances revealed that lymphovenous valves and platelet-mediated hemostasis at the lymphovenous junctions maintain life-long separation of the blood and lymphatic vascular systems. Here, we review murine models that exhibit failure of blood-lymph separation to highlight the novel mechanisms and molecular targets for the modulation of lymphatic disorders. Specifically, we focus on the transcription factors, cofactors, and signaling pathways that regulate lymphovenous valve development and platelet-mediated lymphovenous hemostasis, which cooperate to maintain blood-lymph separation.
Collapse
Affiliation(s)
- Harish P Janardhan
- Division of Cardiovascular Medicine, University of Massachusetts Medical School, The Albert Sherman Center, AS7-1047, 368 Plantation St, Worcester, MA, 01605, USA
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Chinmay M Trivedi
- Division of Cardiovascular Medicine, University of Massachusetts Medical School, The Albert Sherman Center, AS7-1047, 368 Plantation St, Worcester, MA, 01605, USA.
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
- The Li-Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
24
|
Tang Y, Zhou X. Antagonistic effects of exogenous Slit2 on VEGF-induced choroidal endothelial cell migration and tube formation. Exp Ther Med 2019; 17:2443-2450. [PMID: 30906431 PMCID: PMC6425150 DOI: 10.3892/etm.2019.7235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) is involved in the pathogenesis of choroidal neovascularization. The aim of the present study was to assess the effects of exogenous slit guidance ligand 2 (Slit2) on VEGF-induced choroidal endothelial cell (CEC) migration and tube formation. The protein and mRNA expression levels of Slit2, roundabout guidance receptor (Robo) 1 and Robo4 in CECs were evaluated by immunocytochemistry and reverse transcription-polymerase chain reaction analyses, respectively. Western blot analysis was used to assess Robo4 protein levels in CECs exposed to increasing concentrations (0, 50, 75, 100, 125 and 150 ng/ml) of exogenous Slit2. The effects of exogenous Slit2 (125 ng/ml) on VEGF-induced CEC migration and tube formation were also examined. CECs expressed Slit2 and Robo4, but lacked Robo1 expression, at the mRNA and protein levels. Robo4 protein expression increased significantly following treatment with 50–150 ng/ml exogenous Slit2. No significant difference in Robo4 protein expression was observed in CECs treated with 125 and 150 ng/ml Slit2. VEGF-induced CEC migration and tube formation were significantly reduced following treatment with 125 ng/ml exogenous Slit2. In conclusion, these results indicate that Robo4 is expressed in CECs. In addition, exogenous Slit2 may regulate Robo4 expression and partially inhibit VEGF-induced CEC migration and tube formation.
Collapse
Affiliation(s)
- Yanling Tang
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xiyuan Zhou
- Department of Ophthalmology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
25
|
Flentje A, Kalsi R, Monahan TS. Small GTPases and Their Role in Vascular Disease. Int J Mol Sci 2019; 20:ijms20040917. [PMID: 30791562 PMCID: PMC6413073 DOI: 10.3390/ijms20040917] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 01/31/2019] [Accepted: 02/07/2019] [Indexed: 12/18/2022] Open
Abstract
Over eighty million people in the United States have cardiovascular disease that can affect the heart causing myocardial infarction; the carotid arteries causing stroke; and the lower extremities leading to amputation. The treatment for end-stage cardiovascular disease is surgical—either endovascular therapy with balloons and stents—or open reconstruction to reestablish blood flow. All interventions damage or destroy the protective inner lining of the blood vessel—the endothelium. An intact endothelium is essential to provide a protective; antithrombotic lining of a blood vessel. Currently; there are no agents used in the clinical setting that promote reendothelialization. This process requires migration of endothelial cells to the denuded vessel; proliferation of endothelial cells on the denuded vessel surface; and the reconstitution of the tight adherence junctions responsible for the formation of an impermeable surface. These processes are all regulated in part and are dependent on small GTPases. As important as the small GTPases are for reendothelialization, dysregulation of these molecules can result in various vascular pathologies including aneurysm formation, atherosclerosis, diabetes, angiogenesis, and hypertension. A better understanding of the role of small GTPases in endothelial cell migration is essential to the development for novel agents to treat vascular disease.
Collapse
Affiliation(s)
- Alison Flentje
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| | - Richa Kalsi
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| | - Thomas S Monahan
- Division of Vascular Surgery, Department of Surgery, University of Maryland School of Medicine, 22 South Greene Street, Suite S10B00, Baltimore, MD 21201, USA.
| |
Collapse
|
26
|
Guan T, Huang K, Liu Y, Hou S, Hu C, Li Y, Zhang J, Zhao J, Zhang J, Wang R, Huang Y. Aristolochic acid inhibits Slit2-induced migration and tube formation via inactivation of Robo1/Robo2-NCK1/NCK2 signaling pathway in human umbilical vein endothelial cells. Toxicol Lett 2018; 300:51-58. [PMID: 30381256 DOI: 10.1016/j.toxlet.2018.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 10/15/2018] [Accepted: 10/22/2018] [Indexed: 11/28/2022]
Abstract
Robo1/Robo2-NCK1/NCK2 signaling pathway controls endothelial cell sprouting and migration induced by Slit2 or VEGF, but whether it is involved in peritubular capillary (PTC) rarefaction of Aristolochic acid nephropathy (AAN) is unclear. In the present study, we evaluated whether AA exerts antiangiogenic effects by targeting this signaling pathways in HUVECs. HUVECs or lentivirus-mediated NCK1-overexpressing HUVECs were stimulated with AA (1, 2 or 3 μg/ml) in the absence or presence of 6 nM Slit2. Our results showed that AAІ (1-3 μg/ml) dose-dependently inhibited the migration and tube formation of HUVECs. This inhibition was in parallel with down-regulated mRNA and protein expression of Slit2/Robo1/Robo2-NCK1/NCK2 signaling pathway. Importantly, overexpression of NCK1 rescued AAІ-impaired angiogenesis, as evidenced by the increase of cell migration and tube formation of HUVECs in response to Slit2. The down-regulation of NCK2 and decreased activation of Rac1 was also restored by overexpression of NCK1. Taken together, our findings show that AA inhibits Slit2-induced migration and tube formation via inactivation of Robo1/Robo2-NCK1/NCK2 signaling pathway in HUVECs, and NCK1 might be a potential agent for vascular remodeling in AAN and diseases associated with impaired angiogenesis.
Collapse
Affiliation(s)
- Tao Guan
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Ke Huang
- Department of Dermatology, Rheumatic immunology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing, 400037, PR China
| | - Yuanyuan Liu
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Shihui Hou
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Chengfang Hu
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Yi Li
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Jingbo Zhang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Jun Zhang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Rupeng Wang
- Department of Dermatology, Rheumatic immunology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing, 400037, PR China
| | - Yunjian Huang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China.
| |
Collapse
|
27
|
Hilfenhaus G, Nguyen DP, Freshman J, Prajapati D, Ma F, Song D, Ziyad S, Cuadrado M, Pellegrini M, Bustelo XR, Iruela-Arispe ML. Vav3-induced cytoskeletal dynamics contribute to heterotypic properties of endothelial barriers. J Cell Biol 2018; 217:2813-2830. [PMID: 29858212 PMCID: PMC6080943 DOI: 10.1083/jcb.201706041] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 04/12/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022] Open
Abstract
Through multiple cell-cell and cell-matrix interactions, epithelial and endothelial sheets form tight barriers. Modulators of the cytoskeleton contribute to barrier stability and act as rheostats of vascular permeability. In this study, we sought to identify cytoskeletal regulators that underlie barrier diversity across vessels. To achieve this, we correlated functional and structural barrier features to gene expression of endothelial cells (ECs) derived from different vascular beds. Within a subset of identified candidates, we found that the guanosine nucleotide exchange factor Vav3 was exclusively expressed by microvascular ECs and was closely associated with a high-resistance barrier phenotype. Ectopic expression of Vav3 in large artery and brain ECs significantly enhanced barrier resistance and cortical rearrangement of the actin cytoskeleton. Mechanistically, we found that the barrier effect of Vav3 is dependent on its Dbl homology domain and downstream activation of Rap1. Importantly, inactivation of Vav3 in vivo resulted in increased vascular leakage, highlighting its function as a key regulator of barrier stability.
Collapse
Affiliation(s)
- Georg Hilfenhaus
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Dai Phuong Nguyen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Jonathan Freshman
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Divya Prajapati
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Dana Song
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Safiyyah Ziyad
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Myriam Cuadrado
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, and Centro de Investigación Biomédica en Red de Cáncer, Consejo Superior de Investigaciones Científicas, and University of Salamanca, Campus Unamuno, Salamanca, Spain
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA.,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| | - Xosé R Bustelo
- Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, and Centro de Investigación Biomédica en Red de Cáncer, Consejo Superior de Investigaciones Científicas, and University of Salamanca, Campus Unamuno, Salamanca, Spain
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA .,Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
28
|
Sepúlveda-Ramírez SP, Toledo-Jacobo L, Henson JH, Shuster CB. Cdc42 controls primary mesenchyme cell morphogenesis in the sea urchin embryo. Dev Biol 2018; 437:140-151. [PMID: 29555242 PMCID: PMC5973877 DOI: 10.1016/j.ydbio.2018.03.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/05/2018] [Accepted: 03/15/2018] [Indexed: 12/15/2022]
Abstract
In the sea urchin embryo, gastrulation is characterized by the ingression and directed cell migration of primary mesenchyme cells (PMCs), as well as the primary invagination and convergent extension of the endomesoderm. Like all cell shape changes, individual and collective cell motility is orchestrated by Rho family GTPases and their modulation of the actomyosin cytoskeleton. And while endomesoderm specification has been intensively studied in echinoids, much less is known about the proximate regulators driving cell motility. Toward these ends, we employed anti-sense morpholinos, mutant alleles and pharmacological inhibitors to assess the role of Cdc42 during sea urchin gastrulation. While inhibition of Cdc42 expression or activity had only mild effects on PMC ingression, PMC migration, alignment and skeletogenesis were disrupted in the absence of Cdc42, as well as elongation of the archenteron. PMC migration and patterning of the larval skeleton relies on the extension of filopodia, and Cdc42 was required for filopodia in vivo as well as in cultured PMCs. Lastly, filopodial extension required both Arp2/3 and formin actin-nucleating factors, supporting models of filopodial nucleation observed in other systems. Together, these results suggest that Cdc42 plays essential roles during PMC cell motility and organogenesis.
Collapse
Affiliation(s)
- Silvia P Sepúlveda-Ramírez
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, United States; University of Washington Friday Harbor Laboratories, Friday Harbor, WA 98250, United States
| | - Leslie Toledo-Jacobo
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, United States; University of Washington Friday Harbor Laboratories, Friday Harbor, WA 98250, United States
| | - John H Henson
- University of Washington Friday Harbor Laboratories, Friday Harbor, WA 98250, United States; Department of Biology, Dickinson College, Carlisle, PA 17013, United States
| | - Charles B Shuster
- Department of Biology, New Mexico State University, Las Cruces, NM 88003, United States; University of Washington Friday Harbor Laboratories, Friday Harbor, WA 98250, United States.
| |
Collapse
|
29
|
Acevedo A, González-Billault C. Crosstalk between Rac1-mediated actin regulation and ROS production. Free Radic Biol Med 2018; 116:101-113. [PMID: 29330095 DOI: 10.1016/j.freeradbiomed.2018.01.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/03/2018] [Accepted: 01/05/2018] [Indexed: 02/08/2023]
Abstract
The small RhoGTPase Rac1 is implicated in a variety of events related to actin cytoskeleton rearrangement. Remarkably, another event that is completely different from those related to actin regulation has the same relevance; the Rac1-mediated production of reactive oxygen species (ROS) through NADPH oxidases (NOX). Each outcome involves different Rac1 downstream effectors; on one hand, events related to the actin cytoskeleton require Rac1 to bind to WAVEs proteins and PAKs that ultimately promote actin branching and turnover, on the other, NOX-derived ROS production demands active Rac1 to be bound to a cytosolic activator of NOX. How Rac1-mediated signaling ends up promoting actin-related events, NOX-derived ROS, or both is poorly understood. Rac1 regulators, including scaffold proteins, are known to exert tight control over its functions. Hence, evidence of Rac1 regulatory events leading to both actin remodeling and NOX-mediated ROS generation are discussed. Moreover, cellular functions linked to physiological and pathological conditions that exhibit crosstalk between Rac1 outcomes are analyzed, while plausible roles in neuronal functions (and dysfunctions) are highlighted. Together, discussed evidence shed light on cellular mechanisms which requires Rac1 to direct either actin- and/or ROS-related events, helping to understand crucial roles of Rac1 dual functionality.
Collapse
Affiliation(s)
- Alejandro Acevedo
- FONDAP Geroscience Center for Brain Health and Metabolism, Santiago, Chile.
| | - Christian González-Billault
- FONDAP Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024, Chile; The Buck Institute for Research on Aging, Novato, USA.
| |
Collapse
|
30
|
Group-I PAKs-mediated phosphorylation of HACE1 at serine 385 regulates its oligomerization state and Rac1 ubiquitination. Sci Rep 2018; 8:1410. [PMID: 29362425 PMCID: PMC5780496 DOI: 10.1038/s41598-018-19471-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/28/2017] [Indexed: 12/21/2022] Open
Abstract
The regulation of Rac1 by HACE1-mediated ubiquitination and proteasomal degradation is emerging as an essential element in the maintenance of cell homeostasis. However, how the E3 ubiquitin ligase activity of HACE1 is regulated remains undetermined. Using a proteomic approach, we identified serine 385 as a target of group-I PAK kinases downstream Rac1 activation by CNF1 toxin from pathogenic E. coli. Moreover, cell treatment with VEGF also promotes Ser-385 phosphorylation of HACE1. We have established in vitro that HACE1 is a direct target of PAK1 kinase activity. Mechanistically, we found that the phospho-mimetic mutant HACE1(S385E), as opposed to HACE1(S385A), displays a lower capacity to ubiquitinate Rac1 in cells. Concomitantly, phosphorylation of Ser-385 plays a pivotal role in controlling the oligomerization state of HACE1. Finally, Ser-385 phosphorylated form of HACE1 localizes in the cytosol away from its target Rac1. Together, our data point to a feedback inhibition of HACE1 ubiquitination activity on Rac1 by group-I PAK kinases.
Collapse
|
31
|
Li Z, Yang A, Yin X, Dong S, Luo F, Dou C, Lan X, Xie Z, Hou T, Xu J, Xing J. Mesenchymal stem cells promote endothelial progenitor cell migration, vascularization, and bone repair in tissue‐engineered constructs
via
activating CXCR2‐Src‐PKL/Vav2‐Rac1. FASEB J 2018; 32:2197-2211. [DOI: 10.1096/fj.201700895r] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Zhilin Li
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
- Department of SpineLanzhou General Hospital, Lanzhou Command of the Chinese People's Liberation Army (CPLA)LanzhouChina
| | - Aijun Yang
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Xiaolong Yin
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Shiwu Dong
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Department of Biomedical Materials ScienceCollege of Biomedical Engineering, Third Military Medical UniversityChongqingChina
| | - Fei Luo
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Ce Dou
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Xu Lan
- Department of SpineLanzhou General Hospital, Lanzhou Command of the Chinese People's Liberation Army (CPLA)LanzhouChina
| | - Zhao Xie
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Tianyong Hou
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Jianzhong Xu
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Junchao Xing
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| |
Collapse
|
32
|
Cao J, Ehling M, März S, Seebach J, Tarbashevich K, Sixta T, Pitulescu ME, Werner AC, Flach B, Montanez E, Raz E, Adams RH, Schnittler H. Polarized actin and VE-cadherin dynamics regulate junctional remodelling and cell migration during sprouting angiogenesis. Nat Commun 2017; 8:2210. [PMID: 29263363 PMCID: PMC5738342 DOI: 10.1038/s41467-017-02373-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 11/24/2017] [Indexed: 02/07/2023] Open
Abstract
VEGFR-2/Notch signalling regulates angiogenesis in part by driving the remodelling of endothelial cell junctions and by inducing cell migration. Here, we show that VEGF-induced polarized cell elongation increases cell perimeter and decreases the relative VE-cadherin concentration at junctions, triggering polarized formation of actin-driven junction-associated intermittent lamellipodia (JAIL) under control of the WASP/WAVE/ARP2/3 complex. JAIL allow formation of new VE-cadherin adhesion sites that are critical for cell migration and monolayer integrity. Whereas at the leading edge of the cell, large JAIL drive cell migration with supportive contraction, lateral junctions show small JAIL that allow relative cell movement. VEGFR-2 activation initiates cell elongation through dephosphorylation of junctional myosin light chain II, which leads to a local loss of tension to induce JAIL-mediated junctional remodelling. These events require both microtubules and polarized Rac activity. Together, we propose a model where polarized JAIL formation drives directed cell migration and junctional remodelling during sprouting angiogenesis. The formation of new blood vessels requires both polarized cell migration and coordinated control of endothelial cell contacts. Here, Cao and colleagues describe at the sub-cellular level the cytoskeletal and cell junction dynamics regulating these processes upon VEGF-induced cell elongation.
Collapse
Affiliation(s)
- Jiahui Cao
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Manuel Ehling
- Max Planck Institute for Molecular Biomedicine and Westfälische Wilhelms University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Sigrid März
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Jochen Seebach
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Katsiaryna Tarbashevich
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, D-48149, Münster, Germany
| | - Tomas Sixta
- Department of Cybernetics, Czech Technical University, 16627, Prague 6, Czech Republic
| | - Mara E Pitulescu
- Max Planck Institute for Molecular Biomedicine and Westfälische Wilhelms University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Ann-Cathrin Werner
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, D-81377 Munich, Germany
| | - Boris Flach
- Department of Cybernetics, Czech Technical University, 16627, Prague 6, Czech Republic
| | - Eloi Montanez
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, D-81377 Munich, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, D-48149, Münster, Germany
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine and Westfälische Wilhelms University of Münster, Faculty of Medicine, D-48149, Münster, Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms University of Münster, Faculty of Medicine, D-48149, Münster, Germany.
| |
Collapse
|
33
|
Combedazou A, Gayral S, Colombié N, Fougerat A, Laffargue M, Ramel D. Small GTPases orchestrate cell-cell communication during collective cell movement. Small GTPases 2017; 11:103-112. [PMID: 28980871 DOI: 10.1080/21541248.2017.1366965] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Collective cell migration is a critical mechanism involved in cell movement during various physiological and pathological processes such as angiogenesis and metastasis formation. During collective movement, cells remain functionally connected and can coordinate individual cell behaviors to ensure efficient migration. A cell-cell communication process ensures this complex coordination. Although the mechanisms regulating cell-cell communication remain unclear, recent findings indicate that it is based on acto-myosin cytoskeleton tension transmission from cell to cell through adherens junctions. As for single cell migration, small GTPases of the Rho and Rab families have been shown to be critical regulators of collective motion. Here, we discuss our current understanding on how these small GTPases are themselves regulated and how they control cell-cell communication during collective migration. Moreover, we also shed light on the key role of cell-cell communication and RhoGTPases in the physiological context of endothelial cell migration during angiogenesis.
Collapse
Affiliation(s)
- Anne Combedazou
- LBCMCP, Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, France
| | - Stéphanie Gayral
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, France
| | - Nathalie Colombié
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, France
| | - Anne Fougerat
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, France
| | - Muriel Laffargue
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, France
| | - Damien Ramel
- INSERM, U1048, I2MC and Université Toulouse III, Toulouse, France
| |
Collapse
|
34
|
Duran CL, Howell DW, Dave JM, Smith RL, Torrie ME, Essner JJ, Bayless KJ. Molecular Regulation of Sprouting Angiogenesis. Compr Physiol 2017; 8:153-235. [PMID: 29357127 DOI: 10.1002/cphy.c160048] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term angiogenesis arose in the 18th century. Several studies over the next 100 years laid the groundwork for initial studies performed by the Folkman laboratory, which were at first met with some opposition. Once overcome, the angiogenesis field has flourished due to studies on tumor angiogenesis and various developmental models that can be genetically manipulated, including mice and zebrafish. In addition, new discoveries have been aided by the ability to isolate primary endothelial cells, which has allowed dissection of various steps within angiogenesis. This review will summarize the molecular events that control angiogenesis downstream of biochemical factors such as growth factors, cytokines, chemokines, hypoxia-inducible factors (HIFs), and lipids. These and other stimuli have been linked to regulation of junctional molecules and cell surface receptors. In addition, the contribution of cytoskeletal elements and regulatory proteins has revealed an intricate role for mobilization of actin, microtubules, and intermediate filaments in response to cues that activate the endothelium. Activating stimuli also affect various focal adhesion proteins, scaffold proteins, intracellular kinases, and second messengers. Finally, metalloproteinases, which facilitate matrix degradation and the formation of new blood vessels, are discussed, along with our knowledge of crosstalk between the various subclasses of these molecules throughout the text. Compr Physiol 8:153-235, 2018.
Collapse
Affiliation(s)
- Camille L Duran
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - David W Howell
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Jui M Dave
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Rebecca L Smith
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Melanie E Torrie
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Jeffrey J Essner
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, Iowa, USA
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| |
Collapse
|
35
|
Komarova YA, Kruse K, Mehta D, Malik AB. Protein Interactions at Endothelial Junctions and Signaling Mechanisms Regulating Endothelial Permeability. Circ Res 2017; 120:179-206. [PMID: 28057793 DOI: 10.1161/circresaha.116.306534] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/31/2022]
Abstract
The monolayer of endothelial cells lining the vessel wall forms a semipermeable barrier (in all tissue except the relatively impermeable blood-brain and inner retinal barriers) that regulates tissue-fluid homeostasis, transport of nutrients, and migration of blood cells across the barrier. Permeability of the endothelial barrier is primarily regulated by a protein complex called adherens junctions. Adherens junctions are not static structures; they are continuously remodeled in response to mechanical and chemical cues in both physiological and pathological settings. Here, we discuss recent insights into the post-translational modifications of junctional proteins and signaling pathways regulating plasticity of adherens junctions and endothelial permeability. We also discuss in the context of what is already known and newly defined signaling pathways that mediate endothelial barrier leakiness (hyperpermeability) that are important in the pathogenesis of cardiovascular and lung diseases and vascular inflammation.
Collapse
Affiliation(s)
- Yulia A Komarova
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Kevin Kruse
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Dolly Mehta
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago
| | - Asrar B Malik
- From the Department of Pharmacology and the Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago.
| |
Collapse
|
36
|
Korayem AH, Mujica PE, Aramoto H, Durán RG, Nepali PR, Kim DD, Harris AL, Sánchez FA, Durán WN. Endothelial cAMP deactivates ischemia-reperfusion-induced microvascular hyperpermeability via Rap1-mediated mechanisms. Am J Physiol Heart Circ Physiol 2017; 313:H179-H189. [PMID: 28476918 DOI: 10.1152/ajpheart.00002.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/18/2017] [Accepted: 04/18/2017] [Indexed: 12/11/2022]
Abstract
Approaches to reduce excessive edema due to the microvascular hyperpermeability that occurs during ischemia-reperfusion (I/R) are needed to prevent muscle compartment syndrome. We tested the hypothesis that cAMP-activated mechanisms actively restore barrier integrity in postischemic striated muscle. We found, using I/R in intact muscles and hypoxia-reoxygenation (H/R, an I/R mimic) in human microvascular endothelial cells (HMVECs), that hyperpermeability can be deactivated by increasing cAMP levels through application of forskolin. This effect was seen whether or not the hyperpermeability was accompanied by increased mRNA expression of VEGF, which occurred only after 4 h of ischemia. We found that cAMP increases in HMVECs after H/R, suggesting that cAMP-mediated restoration of barrier function is a physiological mechanism. We explored the mechanisms underlying this effect of cAMP. We found that exchange protein activated by cAMP 1 (Epac1), a downstream effector of cAMP that stimulates Rap1 to enhance cell adhesion, was activated only at or after reoxygenation. Thus, when Rap1 was depleted by small interfering RNA, H/R-induced hyperpermeability persisted even when forskolin was applied. We demonstrate that 1) VEGF mRNA expression is not involved in hyperpermeability after brief ischemia, 2) elevation of cAMP concentration at reperfusion deactivates hyperpermeability, and 3) cAMP activates the Epac1-Rap1 pathway to restore normal microvascular permeability. Our data support the novel concepts that 1) different hyperpermeability mechanisms operate after brief and prolonged ischemia and 2) cAMP concentration elevation during reperfusion contributes to deactivation of I/R-induced hyperpermeability through the Epac-Rap1 pathway. Endothelial cAMP management at reperfusion may be therapeutic in I/R injury.NEW & NOTEWORTHY Here, we demonstrate that 1) stimulation of cAMP production deactivates ischemia-reperfusion-induced hyperpermeability in muscle and endothelial cells; 2) VEGF mRNA expression is not enhanced by brief ischemia, suggesting that VEGF mechanisms do not activate immediate postischemic hyperpermeability; and 3) deactivation mechanisms operate via cAMP-exchange protein activated by cAMP 1-Rap1 to restore integrity of the endothelial barrier.
Collapse
Affiliation(s)
- Adam H Korayem
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Newark, New Jersey.,Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey; and
| | - Patricio E Mujica
- Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey; and
| | - Haruo Aramoto
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Newark, New Jersey
| | - Ricardo G Durán
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Newark, New Jersey
| | - Prerna R Nepali
- Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey; and
| | - David D Kim
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Newark, New Jersey
| | - Andrew L Harris
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Newark, New Jersey.,Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey; and
| | - Fabiola A Sánchez
- Instituto de Inmunología, Escuela de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Walter N Durán
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Newark, New Jersey; .,Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, Newark, New Jersey; and
| |
Collapse
|
37
|
Kim YM, Kim SJ, Tatsunami R, Yamamura H, Fukai T, Ushio-Fukai M. ROS-induced ROS release orchestrated by Nox4, Nox2, and mitochondria in VEGF signaling and angiogenesis. Am J Physiol Cell Physiol 2017; 312:C749-C764. [PMID: 28424170 DOI: 10.1152/ajpcell.00346.2016] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/10/2017] [Accepted: 04/10/2017] [Indexed: 01/07/2023]
Abstract
Reactive oxygen species (ROS) derived from NADPH oxidase (NOX) and mitochondria play a critical role in growth factor-induced switch from a quiescent to an angiogenic phenotype in endothelial cells (ECs). However, how highly diffusible ROS produced from different sources can coordinate to stimulate VEGF signaling and drive the angiogenic process remains unknown. Using the cytosol- and mitochondria-targeted redox-sensitive RoGFP biosensors with real-time imaging, here we show that VEGF stimulation in human ECs rapidly increases cytosolic RoGFP oxidation within 1 min, followed by mitochondrial RoGFP oxidation within 5 min, which continues at least for 60 min. Silencing of Nox4 or Nox2 or overexpression of mitochondria-targeted catalase significantly inhibits VEGF-induced tyrosine phosphorylation of VEGF receptor type 2 (VEGFR2-pY), EC migration and proliferation at the similar extent. Exogenous hydrogen peroxide (H2O2) or overexpression of Nox4, which produces H2O2, increases mitochondrial ROS (mtROS), which is prevented by Nox2 siRNA, suggesting that Nox2 senses Nox4-derived H2O2 to promote mtROS production. Mechanistically, H2O2 increases S36 phosphorylation of p66Shc, a key mtROS regulator, which is inhibited by siNox2, but not by siNox4. Moreover, Nox2 or Nox4 knockdown or overexpression of S36 phosphorylation-defective mutant p66Shc(S36A) inhibits VEGF-induced mtROS, VEGFR2-pY, EC migration, and proliferation. In summary, Nox4-derived H2O2 in part activates Nox2 to increase mtROS via pSer36-p66Shc, thereby enhancing VEGFR2 signaling and angiogenesis in ECs. This may represent a novel feed-forward mechanism of ROS-induced ROS release orchestrated by the Nox4/Nox2/pSer36-p66Shc/mtROS axis, which drives sustained activation of angiogenesis signaling program.
Collapse
Affiliation(s)
- Young-Mee Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Departments of Medicine (Cardiology) and Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Seok-Jo Kim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.,Department of Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Ryosuke Tatsunami
- School of Pharmacy, Hokkaido Pharmaceutical University, Hokkaido, Japan; and.,Department of Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Departments of Medicine (Cardiology) and Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia; .,Department of Pharmacology, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
38
|
Tang DD, Gerlach BD. The roles and regulation of the actin cytoskeleton, intermediate filaments and microtubules in smooth muscle cell migration. Respir Res 2017; 18:54. [PMID: 28390425 PMCID: PMC5385055 DOI: 10.1186/s12931-017-0544-7] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 04/05/2017] [Indexed: 02/06/2023] Open
Abstract
Smooth muscle cell migration has been implicated in the development of respiratory and cardiovascular systems; and airway/vascular remodeling. Cell migration is a polarized cellular process involving a protrusive cell front and a retracting trailing rear. There are three cytoskeletal systems in mammalian cells: the actin cytoskeleton, the intermediate filament network, and microtubules; all of which regulate all or part of the migrated process. The dynamic actin cytoskeleton spatially and temporally regulates protrusion, adhesions, contraction, and retraction from the cell front to the rear. c-Abl tyrosine kinase plays a critical role in regulating actin dynamics and migration of airway smooth muscle cells and nonmuscle cells. Recent studies suggest that intermediate filaments undergo reorganization during migration, which coordinates focal adhesion dynamics, cell contraction, and nucleus rigidity. In particular, vimentin intermediate filaments undergo phosphorylation and reorientation in smooth muscle cells, which may regulate cell contraction and focal adhesion assembly/disassembly. Motile cells are characterized by a front-rear polarization of the microtubule framework, which regulates all essential processes leading to cell migration through its role in cell mechanics, intracellular trafficking, and signaling. This review recapitulates our current knowledge how the three cytoskeletal systems spatially and temporally modulate the migratory properties of cells. We also summarize the potential role of migration-associated biomolecules in lung and vascular diseases.
Collapse
Affiliation(s)
- Dale D Tang
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA.
| | - Brennan D Gerlach
- Department of Molecular and Cellular Physiology, Albany Medical College, 47 New Scotland Avenue, MC-8, Albany, NY, 12208, USA
| |
Collapse
|
39
|
Li JJ, Sun ZJ, Yuan YM, Yin FF, Bian YG, Long LY, Zhang XL, Xie D. EphB3 Stimulates Cell Migration and Metastasis in a Kinase-dependent Manner through Vav2-Rho GTPase Axis in Papillary Thyroid Cancer. J Biol Chem 2016; 292:1112-1121. [PMID: 27986811 DOI: 10.1074/jbc.m116.750349] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/28/2016] [Indexed: 12/18/2022] Open
Abstract
Eph receptors, the largest subfamily of transmembrane tyrosine kinase receptors, have been increasingly implicated in various physiologic and pathologic processes, and the roles of the Eph family members during tumorigenesis have recently attracted growing attentions. In the present study, we explored the function of EphB3, one member of Eph family, in papillary thyroid cancer (PTC). We found that the expression of EphB3 was significantly elevated in PTC. Either overexpression of EphB3 or activation of EphB3 by EfnB1-Fc/EfnB2-Fc stimulated in vitro migration of PTC cells. In contrast, siRNA-mediated knockdown of EphB3 or EphB3-Fc treatment, which only blocked EphB3-mediated forward signaling, inhibited migration and metastasis of PTC cells. A mechanism study revealed that EphB3 knockdown led to suppressed activity of Rac1 and enhanced activity of RhoA. Moreover, we found that Vav2, an important regulator of Rho family GTPases, was activated by EphB3 in a kinase-dependent manner. Altogether, our work suggested that EphB3 acted as a tumor promoter in PTC by increasing the in vitro migration as well as the in vivo metastasis of PTC cells through regulating the activities of Vav2 and Rho GTPases in a kinase-dependent manner.
Collapse
Affiliation(s)
- Jing-Jing Li
- From the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China and
| | - Zhi-Jian Sun
- From the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China and
| | - Yan-Mei Yuan
- From the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China and
| | - Fen-Fen Yin
- From the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China and
| | - Yao-Gang Bian
- the Department of General Surgery, Fengxian Hospital Affiliated to Southern Medical University, 6600 Nanfeng Road, Shanghai 201499, China
| | - Ling-Yun Long
- From the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China and
| | - Xue-Li Zhang
- the Department of General Surgery, Fengxian Hospital Affiliated to Southern Medical University, 6600 Nanfeng Road, Shanghai 201499, China
| | - Dong Xie
- From the Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China and
| |
Collapse
|
40
|
Yassin S, Hu J, Xu H, Li C, Setrerrahmane S. In vitro and in vivo activities of an antitumor peptide HM-3: A special dose-efficacy relationship on an HCT‑116 xenograft model in nude mice. Oncol Rep 2016; 36:2951-2959. [PMID: 27633584 DOI: 10.3892/or.2016.5077] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/29/2016] [Indexed: 11/06/2022] Open
Abstract
Anti-angiogenesis is an important therapy for cancer treatment. Peptide HM-3 is an integrin antagonist with anti-angiogenic and antitumor activity. Previous research found that HM-3 at an effective dose inhibited tumor growth whereas at higher doses, the inhibitory effect gradually decreased. In the present study, three human tumor cell lines, human colorectal cancer cell (HCT-116) and human hepatic cancer cell (Hep G-2 and SMMC-7721), were selected and their interactions with HM-3 were compared with western blot and flow cytometric assays. The effect of HM-3 on the migration of two tumor cell lines (HCT-116 and Hep G-2) was also evaluated and a bell-shaped dose-efficacy curve was found for both cell lines. Furthermore, in vivo imaging in BALB/c nude mice confirmed that HM-3 had a short half-life and targeted the tumor tissue. Moreover, on an HCT-116 xenograft model in BALB/c nude mice, HM-3 at 3 mg/kg inhibited tumor growth with an inhibition rate of 71.5% (by tumor mass) whereas at 12 and 48 mg/kg, the inhibition rates were 59.2 and 36.0%, respectively. Immunohistochemistry analyses found that both sunitinib (60 mg/kg) and HM-3 (3 and 48 mg/kg) decreased microvascular density and increased percent of HIF-1α and VEGF expressing cells. The present study investigated the effect of tumor microenvironments on the antitumor effect of HM-3 and concluded that HM-3 inhibited angiogenesis and thereafter tumor growth by directly inhibiting HUVEC migration. The special dose-efficacy curves for antitumor effect and for cell migration inhibition were correlated. The present study also confirmed that the effective dose has to be strictly defined for better clinical applications of anti‑angiogenic drugs such as HM-3.
Collapse
Affiliation(s)
- Sitelbanat Yassin
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Jialiang Hu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Hanmei Xu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Ce Li
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| | - Sarra Setrerrahmane
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
41
|
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: 2.5] [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
|
42
|
Guillonneau M, Paris F, Dutoit S, Estephan H, Bénéteau E, Huot J, Corre I. Oxidative stress disassembles the p38/NPM/PP2A complex, which leads to modulation of nucleophosmin-mediated signaling to DNA damage response. FASEB J 2016; 30:2899-914. [PMID: 27142525 DOI: 10.1096/fj.201500194r] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/26/2016] [Indexed: 11/11/2022]
Abstract
Oxidative stress is a leading cause of endothelial dysfunction. The p38 MAPK pathway plays a determinant role in allowing cells to cope with oxidative stress and is tightly regulated by a balanced interaction between p38 protein and its interacting partners. By using a proteomic approach, we identified nucleophosmin (NPM) as a new partner of p38 in HUVECs. Coimmunoprecipitation and microscopic analyses confirmed the existence of a cytosolic nucleophosmin (NPM)/p38 interaction in basal condition. Oxidative stress, which was generated by exposure to 500 µM H2O2, induces a rapid dephosphorylation of NPM at T199 that depends on phosphatase PP2A, another partner of the NPM/p38 complex. Blocking PP2A activity leads to accumulation of NPM-pT199 and to an increased association of NPM with p38. Concomitantly to its dephosphorylation, oxidative stress promotes translocation of NPM to the nucleus to affect the DNA damage response. Dephosphorylated NPM impairs the signaling of oxidative stress-induced DNA damage via inhibition of the phosphorylation of ataxia-telangiectasia mutated and DNA-dependent protein kinase catalytic subunit. Overall, these results suggest that the p38/NPM/PP2A complex acts as a dynamic sensor, allowing endothelial cells to react rapidly to acute oxidative stress.-Guillonneau, M., Paris, F., Dutoit, S., Estephan, H., Bénéteau, E., Huot, J., Corre, I. Oxidative stress disassembles the p38/NPM/PP2A complex, which leads to modulation of nucleophosmin-mediated signaling to DNA damage response.
Collapse
Affiliation(s)
- Maëva Guillonneau
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 6299, Nantes, France; INSERM, Unité Mixte de Recherche 892, Nantes, France; Université de Nantes, Nantes, France; and Le Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval et le Centre de Recherche sur le Cancer de l'Université Laval, Québec City, Québec, Canada
| | - François Paris
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 6299, Nantes, France; INSERM, Unité Mixte de Recherche 892, Nantes, France; Université de Nantes, Nantes, France; and
| | - Soizic Dutoit
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 6299, Nantes, France; INSERM, Unité Mixte de Recherche 892, Nantes, France; Université de Nantes, Nantes, France; and
| | - Hala Estephan
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 6299, Nantes, France; INSERM, Unité Mixte de Recherche 892, Nantes, France; Université de Nantes, Nantes, France; and
| | - Elise Bénéteau
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 6299, Nantes, France; INSERM, Unité Mixte de Recherche 892, Nantes, France; Université de Nantes, Nantes, France; and
| | - Jacques Huot
- Le Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval et le Centre de Recherche sur le Cancer de l'Université Laval, Québec City, Québec, Canada
| | - Isabelle Corre
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche 6299, Nantes, France; INSERM, Unité Mixte de Recherche 892, Nantes, France; Université de Nantes, Nantes, France; and Le Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval et le Centre de Recherche sur le Cancer de l'Université Laval, Québec City, Québec, Canada
| |
Collapse
|
43
|
Wang Y, Yan F, Ye Q, Wu X, Jiang F. PTP1B inhibitor promotes endothelial cell motility by activating the DOCK180/Rac1 pathway. Sci Rep 2016; 6:24111. [PMID: 27052191 PMCID: PMC4823726 DOI: 10.1038/srep24111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/21/2016] [Indexed: 12/14/2022] Open
Abstract
Promoting endothelial cell (EC) migration is important not only for therapeutic angiogenesis, but also for accelerating re-endothelialization after vessel injury. Several recent studies have shown that inhibition of protein tyrosine phosphatase 1B (PTP1B) may promote EC migration and angiogenesis by enhancing the vascular endothelial growth factor receptor-2 (VEGFR2) signalling. In the present study, we demonstrated that PTP1B inhibitor could promote EC adhesion, spreading and migration, which were abolished by the inhibitor of Rac1 but not RhoA GTPase. PTP1B inhibitor significantly increased phosphorylation of p130Cas, and the interactions among p130Cas, Crk and DOCK180; whereas the phosphorylation levels of focal adhesion kinase, Src, paxillin, or Vav2 were unchanged. Gene silencing of DOCK180, but not Vav2, abrogated the effects of PTP1B inhibitor on EC motility. The effects of PTP1B inhibitor on EC motility and p130Cas/DOCK180 activation persisted in the presence of the VEGFR2 antagonist. In conclusion, we suggest that stimulation of the DOCK180 pathway represents an alternative mechanism of PTP1B inhibitor-stimulated EC motility, which does not require concomitant VEGFR2 activation as a prerequisite. Therefore, PTP1B inhibitor may be a useful therapeutic strategy for promoting EC migration in cardiovascular patients in which the VEGF/VEGFR functions are compromised.
Collapse
Affiliation(s)
- Yuan Wang
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Feng Yan
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Qing Ye
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xiao Wu
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | - Fan Jiang
- Key Laboratory of Cardiovascular Remodelling and Function Research (Chinese Ministry of Education and Chinese Ministry of Health) and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
44
|
Wang H, Hartnett ME. Regulation of signaling events involved in the pathophysiology of neovascular AMD. Mol Vis 2016; 22:189-202. [PMID: 27013848 PMCID: PMC4789180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/25/2016] [Indexed: 11/04/2022] Open
Abstract
Neovascular age-related macular degeneration (AMD) is a complex disease in which an individual's genetic predisposition is affected by aging and environmental stresses, which trigger signaling pathways involving inflammation, oxidation, and/or angiogenesis in the RPE cells and choroidal endothelial cells (CECs), to lead to vision loss from choroidal neovascularization. Antiangiogenic therapies have greatly improved clinical outcomes in the last decade; however, vision improves in less than half of patients treated for neovascular AMD, and treatments remain inadequate for atrophic AMD. Many studies focus on genetic predisposition or the association of outcomes in trials of human neovascular AMD but are unable to evaluate the effects between different cell types involved in AMD and the signaling events that take place to cause pathologic biologic events. This manuscript complements other reviews in that it describes what is known generally in human AMD studies and clinical trials testing methods to inhibit vascular endothelial growth factor (VEGF inhibitors) and presents pathologic signaling events that develop in two important cell types, the RPE cells and the CECs, when stimulated by stresses or placed into conditions similar to what is currently understood to occur in neovascular AMD. This manuscript complements other reviews by discussing signaling events that are activated by cell-cell or cell-matrix interactions. These considerations are particularly important when considering growth factors, such as VEGF, which are important in physiologic and pathologic processes, or GTPases that are present but active only if GTP bound. In either case, it is essential to understand the role of signaling activation to distinguish what is pathologic from what is physiologic. Particularly important is the essential role of activated Rac1 in CEC transmigration of the RPE monolayer, an important step in blindness associated with neovascular AMD. Other concepts discussed include the importance of feed-forward loops that overwhelm mechanisms that seek to restore homeostasis in cells and the importance of regulating, instead of abolishing, signaling events in a chronic, complex disease, such as neovascular AMD. These concepts are important as we move to the next stages in developing treatments for neovascular AMD. A novel therapeutic strategy that will be discussed is activating an isoform of the GTPase, Rap1, which can regulate downstream signaling and a pathologic feed-forward loop leading to Rac1 activation and migration of CECs.
Collapse
|
45
|
Park-Windhol C, D'Amore PA. Disorders of Vascular Permeability. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:251-81. [PMID: 26907525 DOI: 10.1146/annurev-pathol-012615-044506] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The endothelial barrier maintains vascular and tissue homeostasis and modulates many physiological processes, such as angiogenesis. Vascular barrier integrity can be disrupted by a variety of soluble permeability factors, and changes in barrier function can exacerbate tissue damage during disease progression. Understanding endothelial barrier function is critical for vascular homeostasis. Many of the signaling pathways promoting vascular permeability can also be triggered during disease, resulting in prolonged or uncontrolled vascular leak. It is believed that recovery of the normal vasculature requires diminishing this hyperpermeable state. Although the molecular mechanisms governing vascular leak have been studied over the last few decades, recent advances have identified new therapeutic targets that have begun to show preclinical and clinical promise. These approaches have been successfully applied to an increasing number of disease conditions. New perspectives regarding how vascular leak impacts the progression of various diseases are highlighted in this review.
Collapse
Affiliation(s)
- Cindy Park-Windhol
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114; , .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Patricia A D'Amore
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114; , .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115.,Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
46
|
van Buul JD, Timmerman I. Small Rho GTPase-mediated actin dynamics at endothelial adherens junctions. Small GTPases 2016; 7:21-31. [PMID: 26825121 DOI: 10.1080/21541248.2015.1131802] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
VE-cadherin-based cell-cell junctions form the major restrictive barrier of the endothelium to plasma proteins and blood cells. The function of VE-cadherin and the actin cytoskeleton are intimately linked. Vascular permeability factors and adherent leukocytes signal through small Rho GTPases to tightly regulate actin cytoskeletal rearrangements in order to open and re-assemble endothelial cell-cell junctions in a rapid and controlled manner. The Rho GTPases are activated by guanine nucleotide exchange factors (GEFs), conferring specificity and context-dependent control of cell-cell junctions. Although the molecular mechanisms that couple cadherins to actin filaments are beginning to be elucidated, specific stimulus-dependent regulation of the actin cytoskeleton at VE-cadherin-based junctions remains unexplained. Accumulating evidence has suggested that depending on the vascular permeability factor and on the subcellular localization of GEFs, cell-cell junction dynamics and organization are differentially regulated by one specific Rho GTPase. In this Commentary, we focus on new insights how the junctional actin cytoskeleton is specifically and locally regulated by Rho GTPases and GEFs in the endothelium.
Collapse
Affiliation(s)
- Jaap D van Buul
- a Department of Molecular Cell Biology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam , Amsterdam , the Netherlands
| | - Ilse Timmerman
- b Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory , Academic Medical Center Amsterdam, University of Amsterdam , Amsterdam , the Netherlands
| |
Collapse
|
47
|
Veluthakal R, Tunduguru R, Arora DK, Sidarala V, Syeda K, Vlaar CP, Thurmond DC, Kowluru A. VAV2, a guanine nucleotide exchange factor for Rac1, regulates glucose-stimulated insulin secretion in pancreatic beta cells. Diabetologia 2015. [PMID: 26224100 PMCID: PMC4591202 DOI: 10.1007/s00125-015-3707-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AIMS/HYPOTHESIS Rho GTPases (Ras-related C3 botulinum toxin substrate 1 [Rac1] and cell division cycle 42 [Cdc42]) have been shown to regulate glucose-stimulated insulin secretion (GSIS) via cytoskeletal remodelling, trafficking and fusion of insulin-secretory granules with the plasma membrane. GTP loading of these G proteins, which is facilitated by GDP/GTP exchange factors, is a requisite step in the regulation of downstream effector proteins. Guanine nucleotide exchange factor VAV2 (VAV2), a member of the Dbl family of proteins, has been identified as one of the GDP/GTP exchange factors for Rac1. Despite recent evidence on the regulatory roles of VAV2 in different cell types, roles of this guanine nucleotide exchange factor in the signalling events leading to GSIS remain undefined. Using immunological, short interfering RNA (siRNA), pharmacological and microscopic approaches we investigated the role of VAV2 in GSIS from islet beta cells. METHODS Co-localisation of Rac1 and VAV2 was determined by Triton X-114 phase partition and confocal microscopy. Glucose-induced actin remodelling was quantified by live cell imaging using the LifeAct-GFP fluorescent biosensor. Rac1 activation was determined by G protein linked immunosorbent assay (G-LISA). RESULTS Western blotting indicated that VAV2 is expressed in INS-1 832/13 beta cells, normal rat islets and human islets. Vav2 siRNA markedly attenuated GSIS in INS-1 832/13 cells. Ehop-016, a newly discovered small molecule inhibitor of the VAV2-Rac1 interaction, or siRNA-mediated knockdown of VAV2 markedly attenuated glucose-induced Rac1 activation and GSIS in INS-1 832/13 cells. Pharmacological findings were recapitulated in primary rat islets. A high glucose concentration promoted co-localisation of Rac1 and VAV2. Real-time imaging in live cells indicated a significant inhibition of glucose-induced cortical actin remodelling by Ehop-016. CONCLUSIONS/INTERPRETATION Our data provide the first evidence to implicate VAV2 in glucose-induced Rac1 activation, actin remodelling and GSIS in pancreatic beta cells.
Collapse
Affiliation(s)
- Rajakrishnan Veluthakal
- Beta Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, 4646 John R, Detroit, MI, 48201, USA
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ragadeepthi Tunduguru
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daleep Kumar Arora
- Immunobiology and Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Vaibhav Sidarala
- Beta Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, 4646 John R, Detroit, MI, 48201, USA
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy, Wayne State University, Detroit, MI, USA
| | - Khadija Syeda
- Beta Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, 4646 John R, Detroit, MI, 48201, USA
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy, Wayne State University, Detroit, MI, USA
| | - Cornelis P Vlaar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, Puerto Rico
| | - Debbie C Thurmond
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anjaneyulu Kowluru
- Beta Cell Biochemistry Laboratory, John D. Dingell VA Medical Center, 4646 John R, Detroit, MI, 48201, USA.
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy, Wayne State University, Detroit, MI, USA.
- B-4237 Research Service, John D. Dingell VA Medical Center, 4646 John R, Detroit, MI, 48201, USA.
| |
Collapse
|
48
|
Hernández-García R, Iruela-Arispe ML, Reyes-Cruz G, Vázquez-Prado J. Endothelial RhoGEFs: A systematic analysis of their expression profiles in VEGF-stimulated and tumor endothelial cells. Vascul Pharmacol 2015; 74:60-72. [PMID: 26471833 DOI: 10.1016/j.vph.2015.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 12/18/2022]
Abstract
Rho guanine nucleotide exchange factors (RhoGEFs) integrate cell signaling inputs into morphological and functional responses. However, little is known about the endothelial repertoire of RhoGEFs and their regulation. Thus, we assessed the expression of 81 RhoGEFs (70 homologous to Dbl and 11 of the DOCK family) in endothelial cells. Further, in the case of DH-RhoGEFs, we also determined their responses to VEGF exposure in vitro and in the context of tumors. A phylogenetic analysis revealed the existence of four groups of DH-RhoGEFs and two of the DOCK family. Among them, we found that the most abundant endothelial RhoGEFs were: Tuba, FGD5, Farp1, ARHGEF17, TRIO, P-Rex1, ARHGEF15, ARHGEF11, ABR, Farp2, ARHGEF40, ALS, DOCK1, DOCK7 and DOCK6. Expression of RASGRF2 and PREX2 increased significantly in response to VEGF, but most other RhoGEFs were unaffected. Interestingly murine endothelial cells isolated from tumors showed that all four phylogenetic subgroups of DH-RhoGEFs were altered when compared to non-tumor endothelial cells. In summary, our results provide a detailed assessment of RhoGEFs expression profiles in the endothelium and set the basis to systematically address their regulation in vascular signaling.
Collapse
Affiliation(s)
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell, and Developmental Biology and Molecular Biology Institute,University of California,Los Angeles, CA,USA
| | | | | |
Collapse
|
49
|
Hasan SS, Siekmann AF. The same but different: signaling pathways in control of endothelial cell migration. Curr Opin Cell Biol 2015; 36:86-92. [DOI: 10.1016/j.ceb.2015.07.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/16/2015] [Accepted: 07/19/2015] [Indexed: 11/24/2022]
|
50
|
Lewis-Tuffin LJ, Feathers R, Hari P, Durand N, Li Z, Rodriguez FJ, Bakken K, Carlson B, Schroeder M, Sarkaria JN, Anastasiadis PZ. Src family kinases differentially influence glioma growth and motility. Mol Oncol 2015; 9:1783-98. [PMID: 26105207 DOI: 10.1016/j.molonc.2015.06.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 12/27/2022] Open
Abstract
Src-family kinase (SFK) signaling impacts multiple tumor-related properties, particularly in the context of the brain tumor glioblastoma. Consequently, the pan-SFK inhibitor dasatinib has emerged as a therapeutic strategy, despite physiologic limitations to its effectiveness in the brain. We investigated the importance of individual SFKs (Src, Fyn, Yes, and Lyn) to glioma tumor biology by knocking down individual SFK expression both in culture (LN229, SF767, GBM8) and orthotopic xenograft (GBM8) contexts. We evaluated the effects of these knockdowns on tumor cell proliferation, migration, and motility-related signaling in culture, as well as overall survival in the orthotopic xenograft model. The four SFKs differed significantly in their importance to these properties. In culture, Src, Fyn, and Yes knockdown generally reduced growth and migration and altered motility-related phosphorylation patterns while Lyn knockdown did so to a lesser extent. However the details of these effects varied significantly depending on the cell line: in no case were conclusions about the role of a particular SFK applicable to all of the measures or all of the cell types examined. In the orthotopic xenograft model, mice implanted with non-target or Src or Fyn knockdown cells showed no differences in survival. In contrast, mice implanted with Yes knockdown cells had longer survival, associated with reduced tumor cell proliferation. Those implanted with Lyn knockdown cells had shorter survival, associated with higher overall tumor burden. Together, our results suggest that Yes signaling directly affects tumor cell biology in a pro-tumorigenic manner, while Lyn signaling affects interactions between tumor cells and the microenvironment in an anti-tumor manner. In the context of therapeutic targeting of SFKs, these results suggest that pan-SFK inhibitors may not produce the intended therapeutic benefit when Lyn is present.
Collapse
Affiliation(s)
- Laura J Lewis-Tuffin
- Department of Cancer Cell Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Ryan Feathers
- Department of Cancer Cell Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Priya Hari
- Department of Cancer Cell Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Nisha Durand
- Department of Cancer Cell Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Zhimin Li
- Department of Cancer Cell Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA
| | - Fausto J Rodriguez
- Department of Pathology, Johns Hopkins Hospital, 1800 Orleans Street, Baltimore, MD 21231, USA
| | - Katie Bakken
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Brett Carlson
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Mark Schroeder
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Panos Z Anastasiadis
- Department of Cancer Cell Biology, Mayo Clinic, 4500 San Pablo Road South, Jacksonville, FL 32224, USA.
| |
Collapse
|