1
|
Tumurkhuu G, Casanova NG, Kempf CL, Ercan Laguna D, Camp SM, Dagvadorj J, Song JH, Reyes Hernon V, Travelli C, Montano EN, Yu JM, Ishimori M, Wallace DJ, Sammani S, Jefferies C, Garcia JG. eNAMPT/TLR4 inflammatory cascade activation is a key contributor to SLE Lung vasculitis and alveolar hemorrhage. J Transl Autoimmun 2022; 6:100181. [PMID: 36619655 PMCID: PMC9816774 DOI: 10.1016/j.jtauto.2022.100181] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Rationale Effective therapies to reduce the severity and high mortality of pulmonary vasculitis and diffuse alveolar hemorrhage (DAH) in patients with systemic lupus erythematosus (SLE) is a serious unmet need. We explored whether biologic neutralization of eNAMPT (extracellular nicotinamide phosphoribosyl-transferase), a novel DAMP and Toll-like receptor 4 ligand, represents a viable therapeutic strategy in lupus vasculitis. Methods Serum was collected from SLE subjects (n = 37) for eNAMPT protein measurements. In the preclinical pristane-induced murine model of lung vasculitis/hemorrhage, C57BL/6 J mice (n = 5-10/group) were treated with PBS, IgG (1 mg/kg), or the eNAMPT-neutralizing ALT-100 mAb (1 mg/kg, IP or subcutaneously (SQ). Lung injury evaluation (Day 10) included histology/immuno-histochemistry, BAL protein/cellularity, tissue biochemistry, RNA sequencing, and plasma biomarker assessment. Results SLE subjects showed highly significant increases in blood NAMPT mRNA expression and eNAMPT protein levels compared to healthy controls. Preclinical pristane-exposed mice studies showed significantly increased NAMPT lung tissue expression and increased plasma eNAMPT levels accompanied by marked increases in alveolar hemorrhage and lung inflammation (BAL protein, PMNs, activated monocytes). In contrast, ALT-100 mAb-treated mice showed significant attenuation of inflammatory lung injury, alveolar hemorrhage, BAL protein, tissue leukocytes, and plasma inflammatory cytokines (eNAMPT, IL-6, IL-8). Lung RNA sequencing showed pristane-induced activation of inflammatory genes/pathways including NFkB, cytokine/chemokine, IL-1β, and MMP signaling pathways, each rectified in ALT-100 mAb-treated mice. Conclusions These findings highlight the role of eNAMPT/TLR4-mediated inflammatory signaling in the pathobiology of SLE pulmonary vasculitis and alveolar hemorrhage. Biologic neutralization of this novel DAMP appears to serve as a viable strategy to reduce the severity of SLE lung vasculitis.
Collapse
Affiliation(s)
- Gantsetseg Tumurkhuu
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nancy G. Casanova
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Carrie L. Kempf
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Duygu Ercan Laguna
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sara M. Camp
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | | | - Jin H. Song
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Vivian Reyes Hernon
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | | | - Erica N. Montano
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jeong Min Yu
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mariko Ishimori
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Daniel J. Wallace
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Saad Sammani
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Caroline Jefferies
- Department of Medicine, Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joe G.N. Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| |
Collapse
|
2
|
Zemskov EA, Gross CM, Aggarwal S, Zemskova MA, Wu X, Gu C, Wang T, Tang H, Black SM. NF-κB-dependent repression of Sox18 transcription factor requires the epigenetic regulators histone deacetylases 1 and 2 in acute lung injury. Front Physiol 2022; 13:947537. [PMID: 35991176 PMCID: PMC9386230 DOI: 10.3389/fphys.2022.947537] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022] Open
Abstract
In acute lung injury (ALI), the NF-κB-mediated downregulation of Sox18 gene expression leads to the disruption of the pulmonary endothelial barrier. Previous studies have suggested that the action of NF-κB as a transcriptional repressor also requires the action of class I histone deacetylases (HDACs). Thus, the purpose of this study was to investigate and further delineate the mechanism of Sox18 repression during lipopolysaccharide (LPS) induced ALI. Using selective inhibitors and specific siRNA-driven depletion of HDACs 1-3 in human lung microvascular endothelial cells (HLMVEC) we were able to demonstrate a critical role for HDACs 1 and 2 in the LPS-mediated repression of Sox18 gene expression and the loss of endothelial monolayer integrity. Moreover, our data demonstrate that HDAC1 associates with a transcription-repressive complex within the NF-κB-binding site of Sox18 promoter. Further, we were able to show that the selective inhibitor of HDAC1, tacedinaline, significantly reduced the endothelial permeability and injury associated with LPS challenge in the mouse lung. Taken together, our data demonstrate, for the first time, that transcription repressors HDACs 1 and 2 are involved in pathological mechanism of ALI and can be considered as therapeutic targets.
Collapse
Affiliation(s)
- Evgeny A. Zemskov
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Christine M. Gross
- Department of Medicine at Broward Health Medical Center, Fort Lauderdale, FL, United States
| | - Saurabh Aggarwal
- Department of Anesthesiology, The University of Alabama, Birmingham, AL, United States
| | - Marina A. Zemskova
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, AZ, United States
| | - Chenxin Gu
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Haiyang Tang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- College of Veterinary Medicine, Northwest A&F University, Xianyang, China
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port St. Lucie, FL, United States
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| |
Collapse
|
3
|
Quijada H, Bermudez T, Kempf CL, Valera DG, Garcia AN, Camp SM, Song JH, Franco E, Burt JK, Sun B, Mascarenhas JB, Burns K, Gaber A, Oita RC, Reyes Hernon V, Barber C, Moreno-Vinasco L, Sun X, Cress AE, Martin D, Liu Z, Desai AA, Natarajan V, Jacobson JR, Dudek SM, Bime C, Sammani S, Garcia JG. Endothelial eNAMPT amplifies pre-clinical acute lung injury: efficacy of an eNAMPT-neutralising monoclonal antibody. Eur Respir J 2021; 57:2002536. [PMID: 33243842 PMCID: PMC8100338 DOI: 10.1183/13993003.02536-2020] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 11/05/2020] [Indexed: 12/11/2022]
Abstract
RATIONALE The severe acute respiratory syndrome coronavirus 2/coronavirus disease 2019 pandemic has highlighted the serious unmet need for effective therapies that reduce acute respiratory distress syndrome (ARDS) mortality. We explored whether extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a ligand for Toll-like receptor (TLR)4 and a master regulator of innate immunity and inflammation, is a potential ARDS therapeutic target. METHODS Wild-type C57BL/6J or endothelial cell (EC)-cNAMPT -/- knockout mice (targeted EC NAMPT deletion) were exposed to either a lipopolysaccharide (LPS)-induced ("one-hit") or a combined LPS/ventilator ("two-hit")-induced acute inflammatory lung injury model. A NAMPT-specific monoclonal antibody (mAb) imaging probe (99mTc-ProNamptor) was used to detect NAMPT expression in lung tissues. Either an eNAMPT-neutralising goat polyclonal antibody (pAb) or a humanised monoclonal antibody (ALT-100 mAb) were used in vitro and in vivo. RESULTS Immunohistochemical, biochemical and imaging studies validated time-dependent increases in NAMPT lung tissue expression in both pre-clinical ARDS models. Intravenous delivery of either eNAMPT-neutralising pAb or mAb significantly attenuated inflammatory lung injury (haematoxylin and eosin staining, bronchoalveolar lavage (BAL) protein, BAL polymorphonuclear cells, plasma interleukin-6) in both pre-clinical models. In vitro human lung EC studies demonstrated eNAMPT-neutralising antibodies (pAb, mAb) to strongly abrogate eNAMPT-induced TLR4 pathway activation and EC barrier disruption. In vivo studies in wild-type and EC-cNAMPT -/- mice confirmed a highly significant contribution of EC-derived NAMPT to the severity of inflammatory lung injury in both pre-clinical ARDS models. CONCLUSIONS These findings highlight both the role of EC-derived eNAMPT and the potential for biologic targeting of the eNAMPT/TLR4 inflammatory pathway. In combination with predictive eNAMPT biomarker and NAMPT genotyping assays, this offers the opportunity to identify high-risk ARDS subjects for delivery of personalised medicine.
Collapse
Affiliation(s)
- Hector Quijada
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
- Co-first authors
| | - Tadeo Bermudez
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
- Co-first authors
| | - Carrie L. Kempf
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Daniel G. Valera
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Alexander N. Garcia
- Dept of Radiation Oncology, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Sara M. Camp
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Jin H. Song
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Evelyn Franco
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Jessica K. Burt
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Belinda Sun
- Dept of Pathology, University of Arizona Health Sciences, Tucson, AZ, USA
| | | | - Kimberlie Burns
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Amir Gaber
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Radu C. Oita
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | | | - Christy Barber
- Dept of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, USA
| | | | - Xiaoguang Sun
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Anne E. Cress
- Dept of Cellular and Molecular Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Diego Martin
- Houston Methodist Hospital Research Institute, Houston, TX, USA
| | - Zhonglin Liu
- Dept of Medical Imaging, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Ankit A. Desai
- Dept of Medicine, Indiana University, Indianapolis IN, USA
| | | | | | - Steven M. Dudek
- Dept of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Christian Bime
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
| | - Saad Sammani
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
- Co-senior authors
| | - Joe G.N. Garcia
- Dept of Medicine, University of Arizona Health Sciences, Tucson, AZ, USA
- Co-senior authors
| |
Collapse
|
4
|
Zeng Z, Chen W, Moshensky A, Shakir Z, Khan R, Crotty Alexander LE, Ware LB, Aldaz CM, Jacobson JR, Dudek SM, Natarajan V, Machado RF, Singla S. Cigarette Smoke and Nicotine-Containing Electronic-Cigarette Vapor Downregulate Lung WWOX Expression, Which Is Associated with Increased Severity of Murine Acute Respiratory Distress Syndrome. Am J Respir Cell Mol Biol 2021; 64:89-99. [PMID: 33058734 PMCID: PMC7780991 DOI: 10.1165/rcmb.2020-0145oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 09/22/2020] [Indexed: 12/21/2022] Open
Abstract
A history of chronic cigarette smoking is known to increase risk for acute respiratory distress syndrome (ARDS), but the corresponding risks associated with chronic e-cigarette use are largely unknown. The chromosomal fragile site gene, WWOX, is highly susceptible to genotoxic stress from environmental exposures and thus an interesting candidate gene for the study of exposure-related lung disease. Lungs harvested from current versus former/never-smokers exhibited a 47% decrease in WWOX mRNA levels. Exposure to nicotine-containing e-cigarette vapor resulted in an average 57% decrease in WWOX mRNA levels relative to vehicle-treated controls. In separate studies, endothelial (EC)-specific WWOX knockout (KO) versus WWOX flox control mice were examined under ARDS-producing conditions. EC WWOX KO mice exhibited significantly greater levels of vascular leak and histologic lung injury. ECs were isolated from digested lungs of untreated EC WWOX KO mice using sorting by flow cytometry for CD31+ CD45-cells. These were grown in culture, confirmed to be WWOX deficient by RT-PCR and Western blotting, and analyzed by electric cell impedance sensing as well as an FITC dextran transwell assay for their barrier properties during methicillin-resistant Staphylococcus aureus or LPS exposure. WWOX KO ECs demonstrated significantly greater declines in barrier function relative to cells from WWOX flox controls during either methicillin-resistant S. aureus or LPS treatment as measured by both electric cell impedance sensing and the transwell assay. The increased risk for ARDS observed in chronic smokers may be mechanistically linked, at least in part, to lung WWOX downregulation, and this phenomenon may also manifest in the near future in chronic users of e-cigarettes.
Collapse
Affiliation(s)
- Zhenguo Zeng
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Weiguo Chen
- Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois, Chicago, Illinois
| | | | - Zaid Shakir
- Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois, Chicago, Illinois
| | - Raheel Khan
- Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois, Chicago, Illinois
| | | | | | - C. M. Aldaz
- MD Anderson Cancer Center, University of Texas, Houston, Texas; and
| | - Jeffrey R. Jacobson
- Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois, Chicago, Illinois
| | - Steven M. Dudek
- Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois, Chicago, Illinois
| | - Viswanathan Natarajan
- Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois, Chicago, Illinois
| | - Roberto F. Machado
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sunit Singla
- Division of Pulmonary, Critical Care, Sleep and Allergy Medicine, University of Illinois, Chicago, Illinois
| |
Collapse
|
5
|
FAK and Pyk2 activity promote TNF-α and IL-1β-mediated pro-inflammatory gene expression and vascular inflammation. Sci Rep 2019; 9:7617. [PMID: 31110200 PMCID: PMC6527705 DOI: 10.1038/s41598-019-44098-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 05/09/2019] [Indexed: 01/07/2023] Open
Abstract
Protein tyrosine kinase (PTK) activity has been implicated in pro-inflammatory gene expression following tumor necrosis factor-α (TNF-α) or interkeukin-1β (IL-1β) stimulation. However, the identity of responsible PTK(s) in cytokine signaling have not been elucidated. To evaluate which PTK is critical to promote the cytokine-induced inflammatory cell adhesion molecule (CAM) expression including VCAM-1, ICAM-1, and E-selectin in human aortic endothelial cells (HAoECs), we have tested pharmacological inhibitors of major PTKs: Src and the focal adhesion kinase (FAK) family kinases - FAK and proline-rich tyrosine kinase (Pyk2). We found that a dual inhibitor of FAK/Pyk2 (PF-271) most effectively reduced all three CAMs upon TNF-α or IL-1β stimulation compared to FAK or Src specific inhibitors (PF-228 or Dasatinib), which inhibited only VCAM-1 expression. In vitro inflammation assays showed PF-271 reduced monocyte attachment and transmigration on HAoECs. Furthermore, FAK/Pyk2 activity was not limited to CAM expression but was also required for expression of various pro-inflammatory molecules including MCP-1 and IP-10. Both TNF-α and IL-1β signaling requires FAK/Pyk2 activity to activate ERK and JNK MAPKs leading to inflammatory gene expression. Knockdown of either FAK or Pyk2 reduced TNF-α-stimulated ERK and JNK activation and CAM expression, suggesting that activation of ERK or JNK is specific through FAK and Pyk2. Finally, FAK/Pyk2 activity is required for VCAM-1 expression and macrophage recruitment to the vessel wall in a carotid ligation model in ApoE-/- mice. Our findings define critical roles of FAK/Pyk2 in mediating inflammatory cytokine signaling and implicate FAK/Pyk2 inhibitors as potential therapeutic agents to treat vascular inflammatory disease such as atherosclerosis.
Collapse
|
6
|
Singla S, Sysol JR, Dille B, Jones N, Chen J, Machado RF. Hemin Causes Lung Microvascular Endothelial Barrier Dysfunction by Necroptotic Cell Death. Am J Respir Cell Mol Biol 2017; 57:307-314. [PMID: 28421813 DOI: 10.1165/rcmb.2016-0287oc] [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: 12/30/2022] Open
Abstract
Hemin, the oxidized prosthetic moiety of hemoglobin, has been implicated in the pathogenesis of acute chest syndrome in patients with sickle cell disease by virtue of its endothelial-activating properties. In this study, we examined whether hemin can cause lung microvascular endothelial barrier dysfunction. By assessing transendothelial resistance using electrical cell impedance sensing, and by directly measuring trans-monolayer fluorescein isothiocyanate-dextran flux, we found that hemin does cause endothelial barrier dysfunction in a concentration-dependent manner. Pretreatment with either a Toll-like receptor 4 inhibitor, TAK-242, or an antioxidant, N-acetylcysteine, abrogated this effect. Increased monolayer permeability was found to be associated with programmed cell death by necroptosis, as evidenced by Trypan blue staining, terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, Western blotting for activated forms of key effectors of cell death pathways, and studies utilizing specific inhibitors of necroptosis and apoptosis. Further studies examining the role of endothelial cell necroptosis in promoting noncardiogenic pulmonary edema during acute chest syndrome are warranted and may open a new avenue of potential treatments for this devastating disease.
Collapse
Affiliation(s)
- Sunit Singla
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Justin R Sysol
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Benjamin Dille
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Nicole Jones
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Jiwang Chen
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, Department of Medicine, University of Illinois, Chicago, Illinois
| |
Collapse
|
7
|
NLRP3 protects alveolar barrier integrity by an inflammasome-independent increase of epithelial cell adherence. Sci Rep 2016; 6:30943. [PMID: 27476670 PMCID: PMC4967923 DOI: 10.1038/srep30943] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/11/2016] [Indexed: 12/17/2022] Open
Abstract
Bacterial pneumonia is a major cause of acute lung injury and acute respiratory distress syndrome, characterized by alveolar barrier disruption. NLRP3 is best known for its ability to form inflammasomes and to regulate IL-1β and IL-18 production in myeloid cells. Here we show that NLRP3 protects the integrity of the alveolar barrier in a mouse model of Streptococcus pneumoniae-induced pneumonia, and ex vivo upon treatment of isolated perfused and ventilated lungs with the purified bacterial toxin, pneumolysin. We reveal that the preserving effect of NLRP3 on the lung barrier is independent of inflammasomes, IL-1β and IL-18. NLRP3 improves the integrity of alveolar epithelial cell monolayers by enhancing cellular adherence. Collectively, our study uncovers a novel function of NLRP3 by demonstrating that it protects epithelial barrier function independently of inflammasomes.
Collapse
|
8
|
Haines RJ, Beard RS, Chen L, Eitnier RA, Wu MH. Interleukin-1β Mediates β-Catenin-Driven Downregulation of Claudin-3 and Barrier Dysfunction in Caco2 Cells. Dig Dis Sci 2016; 61:2252-2261. [PMID: 27074920 PMCID: PMC5517031 DOI: 10.1007/s10620-016-4145-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/22/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND IL-1β is a cytokine involved in mediating epithelial barrier dysfunction in the gut. It is known that IL-1β mediates activation of non-muscle myosin light chain kinase in epithelial cells, but the precise mechanism by which epithelial barrier dysfunction is induced by IL-1β is not understood. METHODS AND RESULTS Using a Caco2 cell model, we show that the expression of the tight junction protein, claudin-3, is transcriptionally downregulated by IL-1β treatment. In addition, after assessing protein and mRNA expression, and protein localization, we show that inhibition of nmMLCK rescues IL-1β-mediated decrease in claudin-3 expression as well as junction protein redistribution. Using chromatin immunoprecipitation assays, we also show that β-catenin targeting of the claudin-3 promoter occurs as a consequence of IL-1β-mediated epithelial barrier dysfunction, and inhibition of nmMLCK interferes with this interaction. CONCLUSIONS Taken together, these data represent the first line of evidence demonstrating nmMLCK regulation of claudin-3 expression in response to IL-1β-treated epithelial cells.
Collapse
Affiliation(s)
- R J Haines
- Department of Surgery, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., MDC 8, Office MDC 2012, Tampa, FL, 33612, USA
- James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - R S Beard
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
| | - L Chen
- Department of Surgery, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., MDC 8, Office MDC 2012, Tampa, FL, 33612, USA
| | - R A Eitnier
- Department of Surgery, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., MDC 8, Office MDC 2012, Tampa, FL, 33612, USA
| | - M H Wu
- Department of Surgery, University of South Florida, Morsani College of Medicine, 12901 Bruce B. Downs Blvd., MDC 8, Office MDC 2012, Tampa, FL, 33612, USA.
- James A. Haley Veterans' Hospital, Tampa, FL, USA.
| |
Collapse
|
9
|
Good RB, Gilbane AJ, Trinder SL, Denton CP, Coghlan G, Abraham DJ, Holmes AM. Endothelial to Mesenchymal Transition Contributes to Endothelial Dysfunction in Pulmonary Arterial Hypertension. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1850-8. [PMID: 25956031 DOI: 10.1016/j.ajpath.2015.03.019] [Citation(s) in RCA: 250] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 02/03/2015] [Accepted: 03/03/2015] [Indexed: 12/16/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by lung endothelial cell dysfunction and vascular remodeling. Normally, the endothelium forms an integral cellular barrier to regulate vascular homeostasis. During embryogenesis endothelial cells exhibit substantial plasticity that contribute to cardiac development by undergoing endothelial-to-mesenchymal transition (EndoMT). We determined the presence of EndoMT in the pulmonary vasculature in vivo and the functional effects on pulmonary artery endothelial cells (PAECs) undergoing EndoMT in vitro. Histologic assessment of patients with systemic sclerosis-associated PAH and the hypoxia/SU5416 mouse model identified the presence von Willebrand factor/α-smooth muscle actin-positive endothelial cells in up to 5% of pulmonary vessels. Induced EndoMT in PAECs by inflammatory cytokines IL-1β, tumor necrosis factor α, and transforming growth factor β led to actin cytoskeleton reorganization and the development of a mesenchymal morphology. Induced EndoMT cells exhibited up-regulation of mesenchymal markers, including collagen type I and α-smooth muscle actin, and a reduction in endothelial cell and junctional proteins, including von Willebrand factor, CD31, occludin, and vascular endothelial-cadherin. Induced EndoMT monolayers failed to form viable biological barriers and induced enhanced leak in co-culture with PAECs. Induced EndoMT cells secreted significantly elevated proinflammatory cytokines, including IL-6, IL-8, and tumor necrosis factor α, and supported higher immune transendothelial migration compared with PAECs. These findings suggest that EndoMT may contribute to the development of PAH.
Collapse
Affiliation(s)
- Robert B Good
- Division of Medicine, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Adrian J Gilbane
- Division of Medicine, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Sarah L Trinder
- Division of Medicine, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Christopher P Denton
- Division of Medicine, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Gerry Coghlan
- National Pulmonary Hypertension Service, Royal Free Hospital National Health Service Foundation Trust, London, United Kingdom
| | - David J Abraham
- Division of Medicine, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Alan M Holmes
- Division of Medicine, University College London Medical School, Royal Free Campus, London, United Kingdom.
| |
Collapse
|
10
|
Kása A, Csortos C, Verin AD. Cytoskeletal mechanisms regulating vascular endothelial barrier function in response to acute lung injury. Tissue Barriers 2015; 3:e974448. [PMID: 25838980 DOI: 10.4161/21688370.2014.974448] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/04/2014] [Indexed: 01/11/2023] Open
Abstract
Endothelial cells (EC) form a semi-permeable barrier between the interior space of blood vessels and the underlying tissues. In acute lung injury (ALI) the EC barrier is weakened leading to increased vascular permeability. It is widely accepted that EC barrier integrity is critically dependent upon intact cytoskeletal structure and cell junctions. Edemagenic agonists, like thrombin or endotoxin lipopolysaccharide (LPS), induced cytoskeletal rearrangement, and EC contractile responses leading to disruption of intercellular contacts and EC permeability increase. The highly clinically-relevant cytoskeletal mechanisms of EC barrier dysfunction are currently under intense investigation and will be described and discussed in the current review.
Collapse
Key Words
- AJ, adherens junction
- ALI, Acute Lung Injury
- ARDS, Acute Respiratory Distress Syndrome
- CPI-17, PKC potentiated inhibitory protein of 17 kDa
- CaD, caldesmon
- EC, endothelial cells
- GJ, gap junction
- HSP-27, small heat shock actin-capping protein of 27 kDa
- IL, interleukin
- LPS, lipopolysaccharide
- MLC, myosin light chain
- MLCK, Ca2+/calmodulin (CaM) dependent MLC kinase
- MLCP, myosin light chain phosphatase
- MT, microtubules
- MYPT1, myosin phosphatase targeting subunit 1
- PKA, protein kinase A
- PKC, protein kinase C
- SM, smooth muscle
- TJ, tight junction
- TLR4, toll-like receptor 4
- TNFα, tumor necrosis factor α
- acute lung injury
- barrier function
- cytoskeleton
- endothelial junctions
- pulmonary endothelium
- thrombin
Collapse
Affiliation(s)
- Anita Kása
- Vascular Biology Center; Georgia Regents University ; Augusta, GA USA
| | - Csilla Csortos
- Department of Medical Chemistry; Faculty of Medicine; University of Debrecen ; Debrecen, Hungary
| | - Alexander D Verin
- Vascular Biology Center; Georgia Regents University ; Augusta, GA USA ; Division of Pulmonary; Medicine Medical College of Georgia; Georgia Regents University; Augusta, GA USA
| |
Collapse
|
11
|
Arnold KM, Goeckeler ZM, Wysolmerski RB. Loss of Focal Adhesion Kinase Enhances Endothelial Barrier Function and Increases Focal Adhesions. Microcirculation 2013; 20:637-49. [DOI: 10.1111/micc.12063] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/16/2013] [Indexed: 12/30/2022]
|
12
|
Usatyuk PV, Kotha SR, Parinandi NL, Natarajan V. Phospholipase D signaling mediates reactive oxygen species-induced lung endothelial barrier dysfunction. Pulm Circ 2013; 3:108-15. [PMID: 23662182 PMCID: PMC3641713 DOI: 10.4103/2045-8932.109925] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Reactive oxygen species (ROS) have emerged as critical players in the pathophysiology of pulmonary disorders and diseases. Earlier, we have demonstrated that ROS stimulate lung endothelial cell (EC) phospholipase D (PLD) that generates phosphatidic acid (PA), a second messenger involved in signal transduction. In the current study, we investigated the role of PLD signaling in the ROS-induced lung vascular EC barrier dysfunction. Our results demonstrated that hydrogen peroxide (H2O2), a typical physiological ROS, induced PLD activation and altered the barrier function in bovine pulmonary artery ECs (BPAECs). 1-Butanol, the quencher of PLD, generated PA leading to the formation of physiologically inactive phosphatidyl butanol but not its biologically inactive analog, 2-butanol, blocked the H2O2-mediated barrier dysfunction. Furthermore, cell permeable C2 ceramide, an inhibitor of PLD but not the C2 dihydroceramide, attenuated the H2O2-induced PLD activation and enhancement of paracellular permeability of Evans blue conjugated albumin across the BPAEC monolayers. In addition, transfection of BPAECs with adenoviral constructs of hPLD1 and mPLD2 mutants attenuated the H2O2-induced barrier dysfunction, cytoskeletal reorganization and distribution of focal adhesion proteins. For the first time, this study demonstrated that the PLD-generated intracellular bioactive lipid signal mediator, PA, played a critical role in the ROS-induced barrier dysfunction in lung vascular ECs. This study also underscores the importance of PLD signaling in vascular leak and associated tissue injury in the etiology of lung diseases among critically ill patients encountering oxygen toxicity and excess ROS production during ventilator-assisted breathing.
Collapse
Affiliation(s)
- Peter V Usatyuk
- Department of Pharmacology and Institute for Personalized Respiratory Medicine, University of Illinois, Chicago, IL, USA
| | | | | | | |
Collapse
|
13
|
Adyshev DM, Moldobaeva NK, Elangovan VR, Garcia JGN, Dudek SM. Differential involvement of ezrin/radixin/moesin proteins in sphingosine 1-phosphate-induced human pulmonary endothelial cell barrier enhancement. Cell Signal 2011; 23:2086-96. [PMID: 21864676 DOI: 10.1016/j.cellsig.2011.08.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 07/30/2011] [Accepted: 08/01/2011] [Indexed: 01/07/2023]
Abstract
Endothelial cell (EC) barrier dysfunction induced by inflammatory agonists is a frequent pathophysiologic event in multiple diseases. The platelet-derived phospholipid sphingosine-1 phosphate (S1P) reverses this dysfunction by potently enhancing the EC barrier through a process involving Rac GTPase-dependent cortical actin rearrangement as an integral step. In this study we explored the role of the ezrin, radixin, and moesin (ERM) family of actin-binding linker protein in modulating S1P-induced human pulmonary EC barrier enhancement. S1P induces ERM translocation to the EC periphery and promotes ERM phosphorylation on a critical threonine residue (Ezrin-567, Radixin-564, Moesin-558). This phosphorylation is dependent on activation of PKC isoforms and Rac1. The majority of ERM phosphorylation on these critical threonine residues after S1P occurs in moesin and ezrin. Baseline radixin phosphorylation is higher than in the other two ERM proteins but does not increase after S1P. S1P-induced moesin and ezrin threonine phosphorylation is not mediated by the barrier enhancing receptor S1PR1 because siRNA downregulation of S1PR1 fails to inhibit these phosphorylation events, while stimulation of EC with the S1PR1-specific agonist SEW2871 fails to induce these phosphorylation events. Silencing of either all ERM proteins or radixin alone (but not moesin alone) reduced S1P-induced Rac1 activation and phosphorylation of the downstream Rac1 effector PAK1. Radixin siRNA alone, or combined siRNA for all three ERM proteins, dramatically attenuates S1P-induced EC barrier enhancement (measured by transendothelial electrical resistance (TER), peripheral accumulation of di-phospho-MLC, and cortical cytoskeletal rearrangement. In contrast, moesin depletion has the opposite effects on these parameters. Ezrin silencing partially attenuates S1P-induced EC barrier enhancement and cytoskeletal changes. Thus, despite structural similarities and reported functional redundancy, the ERM proteins differentially modulate S1P-induced alterations in lung EC cytoskeleton and permeability. These results suggest that ERM activation is an important regulatory event in EC barrier responses to S1P.
Collapse
Affiliation(s)
- Djanybek M Adyshev
- Institute for Personalized Respiratory Medicine, Department of Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
14
|
Sharma S, Singh M, Sharma PL. Mechanism of attenuation of diabetes mellitus and hypercholesterolemia induced vascular endothelial dysfunction by protein tyrosine phosphatase inhibition. Vascul Pharmacol 2011; 54:80-7. [PMID: 21237289 DOI: 10.1016/j.vph.2010.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Revised: 11/16/2010] [Accepted: 12/31/2010] [Indexed: 11/28/2022]
Abstract
The study has been designed to investigate downstream mechanisms in the PTPase inhibition mediated attenuation of diabetes mellitus and hypercholesterolemia-induced vascular endothelial dysfunction. Diabetes mellitus was induced in rats using streptozotocin (55 mg/kg, i.v. once), while hypercholesterolemia was produced by feeding high cholesterol diet. After 4 weeks of streptozotocin and Cholesterol rich diet administration, vascular endothelium dysfunction was assessed, in terms of attenuation of acetylcholine-induced, endothelium-dependent relaxation (Isolated Aortic Ring Preparation), a decrease in serum nitrate/nitrite level, as well as mRNA expression of eNOS (rtPCR) and disruption of integrity of vascular endothelium (Electron microscopy). After 14 days of daily administration, sodium orthovanadate (8 mg/kg, p.o., 16 mg/kg, p.o and 24 mg/kg, p.o) and atorvastatin (30 mg/kg, p.o) (positive control) significantly improved acetylcholine-induced endothelium-dependent relaxation, serum nitrate/nitrite level, mRNA expression of eNOS and maintained integrity of vascular endothelium. However, this ameliorative effect of SOV was significantly blocked by UCN-01, (PDK inhibitor) and L-NAME (Inhibitor of eNOS). Therefore, it may be concluded that sodium orthovanadate, a specific inhibitor of PTPase, may stimulate PDK and eNOS and consequently improve vascular endothelium dysfunction. Thus, inhibition of PTPase might be a useful approach in the therapeutics of vascular endothelium dysfunction.
Collapse
Affiliation(s)
- Saurabh Sharma
- Department of Pharmacology, ISF College of Pharmacy, Ferozpur Road, Moga-142001, Punjab, India.
| | | | | |
Collapse
|
15
|
Acute Lung Injury: The Injured Lung Endothelium, Therapeutic Strategies for Barrier Protection, and Vascular Biomarkers. TEXTBOOK OF PULMONARY VASCULAR DISEASE 2010. [PMCID: PMC7120335 DOI: 10.1007/978-0-387-87429-6_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
16
|
Wang L, Chiang ET, Simmons JT, Garcia JGN, Dudek SM. FTY720-induced human pulmonary endothelial barrier enhancement is mediated by c-Abl. Eur Respir J 2010; 38:78-88. [PMID: 21071472 DOI: 10.1183/09031936.00047810] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Strategies to improve pulmonary endothelial barrier function are needed to reverse the devastating effects of vascular leak in acute respiratory distress syndrome. FTY720 is a pharmaceutical analogue of the potent barrier-enhancing phospholipid sphingosine 1-phosphate (S1P). FTY720 decreases vascular permeability by an incompletely characterised mechanism that differs from S1P. Here, we describe its barrier-promoting effects on intracellular signalling and junctional assembly formation in human pulmonary endothelium. Permeability of cultured human pulmonary endothelial cells was assessed using transendothelial electrical resistance and dextran transwell assays. Junctional complex formation was assessed using membrane fractionation and immunofluorescence. Pharmacological inhibitors and small interfering (si)RNA were utilised to determine the effects of individual components on permeability. Unlike S1P, FTY720 failed to induce membrane translocation of adherens junction or tight junction proteins. β-catenin, occludin, claudin-5 or zona occludens protein (ZO)-1/ZO-2 siRNAs did not alter FTY720-induced barrier enhancement. FTY720 induced focal adhesion kinase (FAK) phosphorylation and focal adhesion formation, with FAK siRNA partially attenuating the prolonged phase of barrier enhancement. Inhibition of Src, protein kinase (PK)A, PKG, PKC or protein phosphatase 2A failed to alter FTY720-induced barrier enhancement. FTY720 increased c-Abl tyrosine kinase activity and c-Abl siRNA attenuated peak barrier enhancement after FTY720. FTY720 enhances endothelial barrier function by a novel pathway involving c-Abl signalling.
Collapse
Affiliation(s)
- L Wang
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | |
Collapse
|
17
|
Grinnell KL, Casserly B, Harrington EO. Role of protein tyrosine phosphatase SHP2 in barrier function of pulmonary endothelium. Am J Physiol Lung Cell Mol Physiol 2009; 298:L361-70. [PMID: 20023173 DOI: 10.1152/ajplung.00374.2009] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Pulmonary edema is mediated in part by disruption of interendothelial cell contacts. Protein tyrosine phosphatases (PTP) have been shown to affect both cell-extracellular matrix and cell-cell junctions. The SH2 domain-containing nonreceptor PTP, SHP2, is involved in intercellular signaling through direct interaction with adherens junction proteins. In this study, we examined the role of SHP2 in pulmonary endothelial barrier function. Inhibition of SHP2 promoted edema formation in rat lungs and increased monolayer permeability in cultured lung endothelial cells. In addition, pulmonary endothelial cells demonstrated a decreased level of p190RhoGAP activity following inhibition of SHP2, events that were accompanied by a concomitant increase in RhoA activity. Furthermore, immunofluorescence microscopy confirmed enhanced actin stress fiber formation and diminished interendothelial staining of adherens junction complex-associated proteins upon SHP2 inhibition. Finally, immunoprecipitation and immunoblot analyses demonstrated increased tyrosine phosphorylation of VE-cadherin, beta-catenin, and p190RhoGAP proteins, as well as decreased association between p120-catenin and VE-cadherin proteins. Our findings suggest that SHP2 supports basal pulmonary endothelial barrier function by coordinating the tyrosine phosphorylation profile of VE-cadherin, beta-catenin, and p190RhoGAP and the activity of RhoA, signaling molecules important in adherens junction complex integrity.
Collapse
Affiliation(s)
- K L Grinnell
- Vascular Research Laboratory, Providence VA Medical Center, 830 Chalkstone Ave., Providence, RI 02908, USA
| | | | | |
Collapse
|
18
|
Monaghan-Benson E, Burridge K. The regulation of vascular endothelial growth factor-induced microvascular permeability requires Rac and reactive oxygen species. J Biol Chem 2009; 284:25602-11. [PMID: 19633358 DOI: 10.1074/jbc.m109.009894] [Citation(s) in RCA: 168] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vascular permeability is a complex process involving the coordinated regulation of multiple signaling pathways in the endothelial cell. It has long been documented that vascular endothelial growth factor (VEGF) greatly enhances microvascular permeability; however, the molecular mechanisms controlling VEGF-induced permeability remain unknown. Treatment of microvascular endothelial cells with VEGF led to an increase in reactive oxygen species (ROS) production. ROS are required for VEGF-induced permeability as treatment with the free radical scavenger, N-acetylcysteine, inhibited this effect. Additionally, treatment with VEGF caused ROS-dependent tyrosine phosphorylation of both vascular-endothelial (VE)-cadherin and beta-catenin. Rac1 was required for the VEGF-induced increase in permeability and adherens junction protein phosphorylation. Knockdown of Rac1 inhibited VEGF-induced ROS production consistent with Rac lying upstream of ROS in this pathway. Collectively, these data suggest that VEGF leads to a Rac-mediated generation of ROS, which, in turn, elevates the tyrosine phosphorylation of VE-cadherin and beta-catenin, ultimately regulating adherens junction integrity.
Collapse
Affiliation(s)
- Elizabeth Monaghan-Benson
- Department of Cell and Developmental Biology and Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina 27599-7295, USA.
| | | |
Collapse
|
19
|
Pendyala S, Usatyuk PV, Gorshkova IA, Garcia JG, Natarajan V. Regulation of NADPH oxidase in vascular endothelium: the role of phospholipases, protein kinases, and cytoskeletal proteins. Antioxid Redox Signal 2009; 11:841-60. [PMID: 18828698 PMCID: PMC2850292 DOI: 10.1089/ars.2008.2231] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The generation of reactive oxygen species (ROS) in the vasculature plays a major role in the genesis of endothelial cell (EC) activation and barrier function. Of the several potential sources of ROS in the vasculature, the endothelial NADPH oxidase family of proteins is a major contributor of ROS associated with lung inflammation, ischemia/reperfusion injury, sepsis, hyperoxia, and ventilator-associated lung injury. The NADPH oxidase in lung ECs has most of the components found in phagocytic oxidase, and recent studies show the expression of several homologues of Nox proteins in vascular cells. Activation of NADPH oxidase of nonphagocytic vascular cells is complex and involves assembly of the cytosolic (p47(phox), p67(phox), and Rac1) and membrane-associated components (Noxes and p22(phox)). Signaling pathways leading to NADPH oxidase activation are not completely defined; however, they do appear to involve the cytoskeleton and posttranslation modification of the components regulated by protein kinases, protein phosphatases, and phospholipases. Furthermore, several key components regulating NADPH oxidase recruitment, assembly, and activation are enriched in lipid microdomains to form a functional signaling platform. Future studies on temporal and spatial localization of Nox isoforms will provide new insights into the role of NADPH oxidase-derived ROS in the pathobiology of lung diseases.
Collapse
|
20
|
Ramis-Conde I, Chaplain MAJ, Anderson ARA, Drasdo D. Multi-scale modelling of cancer cell intravasation: the role of cadherins in metastasis. Phys Biol 2009; 6:016008. [PMID: 19321920 DOI: 10.1088/1478-3975/6/1/016008] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Transendothelial migration is a crucial process of the metastatic cascade in which a malignant cell attaches itself to the endothelial layer forming the inner wall of a blood or lymph vessel and creates a gap through which it enters into the bloodstream (or lymphatic system) and then is transported to distant parts of the body. In this process both biological pathways involving cell adhesion molecules such as VE-cadherin and N-cadherin, and the biophysical properties of the cells play an important role. In this paper, we present one of the first mathematical models considering the problem of cancer cell intravasation. We use an individual force-based multi-scale approach which accounts for intra- and inter-cellular protein pathways and for the physical properties of the cells, and a modelling framework which accounts for the biological shape of the vessel. Using our model, we study the influence of different protein pathways in the achievement of transendothelial migration and give quantitative simulation results comparable with real experiments.
Collapse
Affiliation(s)
- Ignacio Ramis-Conde
- French National Institute for Research in Computer Science and Control, Le Chesnay, France.
| | | | | | | |
Collapse
|
21
|
Wittchen ES. Endothelial signaling in paracellular and transcellular leukocyte transmigration. Front Biosci (Landmark Ed) 2009; 14:2522-45. [PMID: 19273217 DOI: 10.2741/3395] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
As the primary physical barrier between blood and tissue compartments within the body, blood vessel endothelial cells and integrity of the cell junctions connecting them must be carefully regulated to support leukocyte transendothelial migration only when necessary. Leukocytes utilize two independent routes across the endothelium: the paracellular route involves migration in-between adjacent endothelial cells and requires the transient disassembly of endothelial cell junctions, while the transcellular route occurs directly through an individual endothelial cell, likely requiring the formation of a channel or pore. In this review, I will first summarize the signaling events that are transduced by leukocyte engagement of endothelial cell-surface receptors like ICAM-1 and VCAM-1. Some of these signals include activation of GTPases, production of reactive oxygen species, and phosphorylation of target proteins. These signaling pathways converge to cause junctional disruption, cytoskeletal remodeling, and/or the membrane fusion events that are associated with leukocyte transendothelial migration. The review will conclude with a detailed discussion of the newly characterized transmigratory cup structure, and the recent advances made towards understanding the mechanisms of transcellular transendothelial migration.
Collapse
Affiliation(s)
- Erika S Wittchen
- Department of Cell and Developmental Biology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7295, USA.
| |
Collapse
|
22
|
|
23
|
Aghajanian A, Wittchen ES, Allingham MJ, Garrett TA, Burridge K. Endothelial cell junctions and the regulation of vascular permeability and leukocyte transmigration. J Thromb Haemost 2008; 6:1453-60. [PMID: 18647230 PMCID: PMC2868262 DOI: 10.1111/j.1538-7836.2008.03087.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The endothelial lining of the vasculature forms the physical barrier between the blood and underlying tissues. Junctions between adjacent endothelial cells are dynamically modulated to sustain vascular homeostasis and to support the transendothelial migration of leukocytes during inflammation. A variety of factors initiate intracellular signaling pathways that regulate the opening and resealing of junctional complexes. This review focuses on three primary signaling pathways initiated within endothelial cells by the binding of vasoactive factors and leukocyte adhesion: Rho GTPases, reactive oxygen species, and tyrosine phosphorylation of junctional proteins. These pathways converge to regulate junctional permeability, either by affecting the stability of junctional proteins or by modulating their interactions. Although much progress has been made in understanding the relationships of these pathways, many questions remain to be answered. A full understanding of the signaling cascades that affect endothelial junctions should identify novel therapeutic targets for diseases that involve excessive permeability or inappropriate leukocyte infiltration into tissues.
Collapse
Affiliation(s)
- A Aghajanian
- Department of Cell and Developmental Biology and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
24
|
|
25
|
Troyanovsky B, Alvarez DF, King JA, Schaphorst KL. Thrombin enhances the barrier function of rat microvascular endothelium in a PAR-1-dependent manner. Am J Physiol Lung Cell Mol Physiol 2007; 294:L266-75. [PMID: 18083763 DOI: 10.1152/ajplung.00107.2007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Thrombin is a multifunctional coagulation protease with pro- and anti-inflammatory vascular effects. We questioned whether thrombin may have segmentally differentiated effects on pulmonary endothelium. In cultured rat endothelial cells, rat thrombin (10 U/ml) recapitulated the previously reported decrease in transmonolayer electrical resistance (TER), F-actin stress fiber formation, paracellular gap formation, and increased permeability. In contrast, in rat pulmonary microvascular endothelial cells (PMVEC), isolated on the basis of Griffonia simplicifolia lectin recognition, thrombin increased TER, induced fewer stress fibers, and decreased permeability. To assess for differential proteinase-activated receptor (PAR) expression as a basis for the different responses, PAR family expression was analyzed. Both pulmonary artery endothelial cells and PMVEC expressed PAR-1 and PAR-2; however, only PMVEC expressed PAR-3, as shown by both RT-PCR and Western analysis. PAR-1 activating peptides (PAR-APs: SFLLRN-NH(2) and TFLLRN-NH(2)) were used to confirm a role for the PAR-1 receptor. PAR-APs (25-250 muM) also increased TER, formed fewer stress fibers, and did not induce paracellular gaps in PMVEC in contrast to that shown in pulmonary artery endothelial cells. These results were confirmed in isolated perfused rat lung preparations. PAR-APs (100 mug/ml) induced a 60% increase in the filtration coefficient over baseline. However, by transmission electron microscopy, perivascular fluid cuffs were seen only along conduit veins and arteries without evidence of intra-alveolar edema. We conclude that thrombin exerts a segmentally differentiated effect on endothelial barrier function in vitro, which corresponds to a pattern of predominant perivascular fluid cuff formation in situ. This may indicate a distinct role for thrombin in the microcirculation.
Collapse
Affiliation(s)
- B Troyanovsky
- Center for Lung Biology, University of South Alabama College of Medicine, 307 North University Drive, Mobile, AL 36688-0002, USA
| | | | | | | |
Collapse
|
26
|
Solodushko V, Parker JC, Fouty B. Pulmonary microvascular endothelial cells form a tighter monolayer when grown in chronic hypoxia. Am J Respir Cell Mol Biol 2007; 38:491-7. [PMID: 18048805 DOI: 10.1165/rcmb.2007-0127oc] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Unique among the vascular beds, loss of endothelial integrity in the pulmonary microcirculation due to injury can lead to rapidly fatal hypoxemia. The ability to regain confluence and re-establish barrier function is central to restoring proper gas exchange. The adult respiratory distress syndrome (ARDS) is a heterogeneous disease, however, meaning that endothelial cells within different regions of the lung do not likely see the same oxygen tension as they attempt to proliferate and re-establish an intact endothelial monolayer; the effect of hypoxia on the integrity of this newly formed endothelial monolayer is not clear. Immortalized human pulmonary microvascular endothelial cells (PMVEC) (ST1.6R cells) were sparsely plated and grown to confluence over 4 days in either normoxia (21% oxygen) or hypoxia (5% oxygen). Confluence attained in a hypoxic environment resulted in a tighter, less permeable endothelial monolayer (as determined by an increase in transendothelial electrical resistance, decreased permeability to fluorescently labeled macromolecules, and decreased hydraulic conductance). PMVEC grown to confluence under hypoxia had decreased RhoA activity; consistent with this finding, inhibition of Rho kinase, a well-described downstream target of RhoA, markedly increased electrical resistance in normoxic, but not hypoxic, PMVEC. These results were confirmed in primary human and rat PMVEC. These data suggest that PMVEC grown to confluence under hypoxia form a tighter monolayer than similar cells grown under normoxia. This tighter barrier appears to be due, in part, to the inhibition of RhoA activity in hypoxic cells.
Collapse
Affiliation(s)
- Victor Solodushko
- Center for Lung Biology; Department of Pharmacology; University of South Alabama School of Medicine, Mobile, Alabama 36688, USA
| | | | | |
Collapse
|
27
|
Allingham MJ, van Buul JD, Burridge K. ICAM-1-Mediated, Src- and Pyk2-Dependent Vascular Endothelial Cadherin Tyrosine Phosphorylation Is Required for Leukocyte Transendothelial Migration. THE JOURNAL OF IMMUNOLOGY 2007; 179:4053-64. [PMID: 17785844 DOI: 10.4049/jimmunol.179.6.4053] [Citation(s) in RCA: 263] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Leukocyte transendothelial migration (TEM) has been modeled as a multistep process beginning with rolling adhesion, followed by firm adhesion, and ending with either transcellular or paracellular passage of the leukocyte across the endothelial monolayer. In the case of paracellular TEM, endothelial cell (EC) junctions are transiently disassembled to allow passage of leukocytes. Numerous lines of evidence demonstrate that tyrosine phosphorylation of adherens junction proteins, such as vascular endothelial cadherin (VE-cadherin) and beta-catenin, correlates with the disassembly of junctions. However, the role of tyrosine phosphorylation in the regulation of junctions during leukocyte TEM is not completely understood. Using human leukocytes and EC, we show that ICAM-1 engagement leads to activation of two tyrosine kinases, Src and Pyk2. Using phospho-specific Abs, we show that engagement of ICAM-1 induces phosphorylation of VE-cadherin on tyrosines 658 and 731, which correspond to the p120-catenin and beta-catenin binding sites, respectively. These phosphorylation events require the activity of both Src and Pyk2. We find that inhibition of endothelial Src with PP2 or SU6656 blocks neutrophil transmigration (71.1 +/- 3.8% and 48.6 +/- 3.8% reduction, respectively), whereas inhibition of endothelial Pyk2 also results in decreased neutrophil transmigration (25.5 +/- 6.0% reduction). Moreover, overexpression of the nonphosphorylatable Y658F or Y731F mutants of VE-cadherin impairs transmigration of neutrophils compared with overexpression of wild-type VE-cadherin (32.7 +/- 7.1% and 38.8 +/- 6.5% reduction, respectively). Our results demonstrate that engagement of ICAM-1 by leukocytes results in tyrosine phosphorylation of VE-cadherin, which is required for efficient neutrophil TEM.
Collapse
Affiliation(s)
- Michael J Allingham
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | | | | |
Collapse
|
28
|
Birukova AA, Alekseeva E, Mikaelyan A, Birukov KG. HGF attenuates thrombin-induced endothelial permeability by Tiam1-mediated activation of the Rac pathway and by Tiam1/Rac-dependent inhibition of the Rho pathway. FASEB J 2007; 21:2776-86. [PMID: 17428964 DOI: 10.1096/fj.06-7660com] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Reorganization of the endothelial cell (EC) cytoskeleton and cell adhesive complexes provides a structural basis for increased vascular permeability implicated in the pathogenesis of many diseases, including asthma, sepsis, and acute respiratory distress syndrome (ARDS). We have recently described the barrier-protective effects of hepatocyte growth factor (HGF) on the human pulmonary EC. In the present study, we explored the involvement of Rac-GTPase and Rac-specific nucleotide exchange factor Tiam1 in the mechanisms of EC barrier protection by HGF. HGF protected EC monolayers from thrombin-induced hyperpermeability, disruption of intercellular junctions, and formation of stress fibers and paracellular gaps by inhibiting thrombin-induced activation of Rho GTPase, Rho association with nucleotide exchange factor p115-RhoGEF, and myosin light chain phosphorylation, which was opposed by stimulation of Rac-dependent signaling. The pharmacological Rac inhibitor or silencing RNA (siRNA) based depletion of either Rac or Tiam1 significantly attenuated HGF-induced peripheral translocation of Rac effector cortactin, cortical actin ring formation, and EC barrier enhancement. Moreover, Tiam1 knockdown using the siRNA approach, attenuated the protective effect of HGF against thrombin-induced activation of Rho signaling, monolayer disruption, and EC hyperpermeability. This study demonstrates the Tiam1/Rac-dependent mechanism of HGF-induced EC barrier protection and provides novel mechanistic insights into regulation of EC permeability via dynamic interactions between Rho- and Tiam1/Rac-mediated pathways.
Collapse
Affiliation(s)
- Anna A Birukova
- Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, 929 East 57th St., CIS Bldg., W410, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
29
|
Chiang ET, Persaud-Sawin DA, Kulkarni S, Garcia JGN, Imani F. Bluetongue virus and double-stranded RNA increase human vascular permeability: role of p38 MAPK. J Clin Immunol 2006; 26:406-16. [PMID: 16786433 DOI: 10.1007/s10875-006-9024-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Endothelial cell (EC) involvement in viral hemorrhagic fevers has been clearly established. However, virally activated mechanisms leading to endothelial activation and dysfunction are not well understood. Several different potential mechanisms such as direct viral infection, alterations in procoagulant/anticoagulant balance, and increased cytokine production have been suggested. We utilized a model of EC barrier dysfunction and vascular endothelial leakage to explore the effect of bluetongue virus (BTV), a hemorrhagic fever virus of ruminants, on human lung endothelial cell barrier properties. Infection of human lung EC with BTV induced a significant and dose-dependent decrease in trans-endothelial electrical resistance (TER). Furthermore, decreases in TER occurred in conjunction with cytoskeletal rearrangement, suggesting a direct mechanism for viral infection-mediated endothelial barrier disruption. Interestingly, double-stranded RNA (dsRNA) mimicked the effects of BTV on endothelial barrier properties. Both BTV- and dsRNA-induced endothelial barrier dysfunction was blocked by treatment with a pharmacological inhibitor of p38 MAPK. The induction of vascular permeability by dsRNA treatment or BTV infection was concomitent with induction of inflammatory cytokines. Taken together, our data suggest that the presence of dsRNA during viral infections and subsequent activation of p38 MAPK is a potential molecular pathway for viral induction of hemorrhagic fevers. Collectively, our data suggest that inhibition of p38 MAPK may be a possible therapeutic approach to alter viral-induced acute hemorrhagic diseases.
Collapse
Affiliation(s)
- Eddie T Chiang
- University of Chicago, Pritzker School of Medicine, Chicago, Illinois 60637, USA
| | | | | | | | | |
Collapse
|
30
|
Abstract
The microvascular endothelial cell monolayer localized at the critical interface between the blood and vessel wall has the vital functions of regulating tissue fluid balance and supplying the essential nutrients needed for the survival of the organism. The endothelial cell is an exquisite “sensor” that responds to diverse signals generated in the blood, subendothelium, and interacting cells. The endothelial cell is able to dynamically regulate its paracellular and transcellular pathways for transport of plasma proteins, solutes, and liquid. The semipermeable characteristic of the endothelium (which distinguishes it from the epithelium) is crucial for establishing the transendothelial protein gradient (the colloid osmotic gradient) required for tissue fluid homeostasis. Interendothelial junctions comprise a complex array of proteins in series with the extracellular matrix constituents and serve to limit the transport of albumin and other plasma proteins by the paracellular pathway. This pathway is highly regulated by the activation of specific extrinsic and intrinsic signaling pathways. Recent evidence has also highlighted the importance of the heretofore enigmatic transcellular pathway in mediating albumin transport via transcytosis. Caveolae, the vesicular carriers filled with receptor-bound and unbound free solutes, have been shown to shuttle between the vascular and extravascular spaces depositing their contents outside the cell. This review summarizes and analyzes the recent data from genetic, physiological, cellular, and morphological studies that have addressed the signaling mechanisms involved in the regulation of both the paracellular and transcellular transport pathways.
Collapse
Affiliation(s)
- Dolly Mehta
- Center of Lung and Vascular Biology, Dept. of Pharmacology (M/C 868), University of Illinois, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | | |
Collapse
|
31
|
Usatyuk PV, Natarajan V. Regulation of reactive oxygen species-induced endothelial cell-cell and cell-matrix contacts by focal adhesion kinase and adherens junction proteins. Am J Physiol Lung Cell Mol Physiol 2005; 289:L999-1010. [PMID: 16040628 DOI: 10.1152/ajplung.00211.2005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Oxidants, generated by activated neutrophils, have been implicated in the pathophysiology of vascular disorders and lung injury; however, mechanisms of oxidant-mediated endothelial barrier dysfunction are unclear. Here, we have investigated the role of focal adhesion kinase (FAK) in regulating hydrogen peroxide (H2O2)-mediated tyrosine phosphorylation of intercellular adhesion proteins and barrier function in endothelium. Treatment of bovine pulmonary artery endothelial cells (BPAECs) with H2O2increased tyrosine phosphorylation of FAK, paxillin, β-catenin, and vascular endothelial (VE)-cadherin and decreased transendothelial electrical resistance (TER), an index of cell-cell adhesion and/or cell-matrix adhesion. To study the role of FAK in H2O2-induced TER changes, BPAECs were transfected with vector or FAK wild-type or FAK-related non-kinase (FRNK) plasmids. Overexpression of FRNK reduced FAK expression and attenuated H2O2-mediated tyrosine phosphorylation of FAK, paxillin, β-catenin, and VE-cadherin and cell-cell adhesion. Additionally, FRNK prevented H2O2-induced distribution of FAK, paxillin, β-catenin, or VE-cadherin toward focal adhesions and cell-cell adhesions but not actin stress fiber formation. These results suggest that activation of FAK by H2O2is an important event in oxidant-mediated VE barrier function regulated by cell-cell and cell-matrix contacts.
Collapse
Affiliation(s)
- Peter V Usatyuk
- Dept. of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, C/S Bldg., Rm. 408, 929 E. 57th Street, Chicago, IL 60637, USA
| | | |
Collapse
|
32
|
Ye SQ, Zhang LQ, Adyshev D, Usatyuk PV, Garcia AN, Lavoie TL, Verin AD, Natarajan V, Garcia JGN. Pre-B-cell-colony-enhancing factor is critically involved in thrombin-induced lung endothelial cell barrier dysregulation. Microvasc Res 2005; 70:142-51. [PMID: 16188281 DOI: 10.1016/j.mvr.2005.08.003] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2005] [Revised: 08/15/2005] [Accepted: 08/16/2005] [Indexed: 11/26/2022]
Abstract
Prior genomic and genetic studies identified pre-B-cell colony-enhancing factor (PBEF) as a novel candidate gene and biomarker in acute lung injury (ALI). As increased vascular permeability is a cardinal feature of ALI, we assessed the role of PBEF in in vitro vascular barrier regulation using confluent human pulmonary artery endothelial cell (HPAEC) monolayers. Reductions in PBEF protein expression (>70%) by siRNA significantly attenuated EC barrier dysfunction induced by the potent edemagenic agent, thrombin, reflected by reductions in transendothelial electric resistance (TER, approximately 60% reduction). Furthermore, PBEF siRNA blunted thrombin-mediated increases in Ca(2+) entry, polymerized actin formation, and myosin light chain phosphorylation, events critical to the thrombin-mediated permeability response. Finally, PBEF siRNA also significantly inhibited thrombin-stimulated increase of IL-8 secretion in HPAEC, a chemokine known to induce actin fiber formation and intercellular gap formation of endothelial cells. Taken together, these studies demonstrate that PBEF may be required for complete expression of the thrombin-induced inflammatory response and reveal potentially novel role for PBEF in the regulation of EC Ca(2+)-dependent cytoskeletal rearrangement and endothelial barrier dysfunction. Ongoing studies will continue to address the molecular mechanisms by which PBEF contributes to ALI susceptibility.
Collapse
Affiliation(s)
- Shui Q Ye
- Department of Medicine, Section of Pulmonary/Critical Care, University of Chicago Pritzker School of Medicine, 5841 S. Maryland Avenue, MC 6076, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
Focal adhesions composed of integrins provide an important structural basis for anchoring the endothelial lining to its surrounding matrices in the vascular wall. Complex molecular reactions occur at the endothelial cell-matrix contact sites in response to physical and chemical stress present in the circulatory system. Recent experimental evidence points to the importance of focal adhesions in the regulation of microvascular barrier function. On one hand, the adhesive interaction between integrins and their extracellular ligands is essential to the maintenance of endothelial barrier properties, and interruption of integrin-matrix binding leads to leaky microvessels. On the other hand, focal adhesion assembly and activation serve as important signalling events in modulating endothelial permeability under stimulatory conditions in the presence of angiogenic factors, inflammatory mediators, or physical forces. The focal responses show distinctive patterns with different temporal characteristics, whereas focal adhesion kinase (FAK) plays a central role in initiating and integrating various signalling pathways that ultimately affect the barrier function. The molecular basis of focal adhesion-dependent microvascular permeability is currently under investigation, and advances in the technologies of computerized quantitative microscopy and intact microvessel imaging should aid the establishment of a functional significance for focal adhesions in the physiological regulation of microvascular permeability.
Collapse
Affiliation(s)
- Mack H Wu
- Department of Surgery, University of California at Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
34
|
Fiala M, Eshleman AJ, Cashman J, Lin J, Lossinsky AS, Suarez V, Yang W, Zhang J, Popik W, Singer E, Chiappelli F, Carro E, Weinand M, Witte M, Arthos J. Cocaine increases human immunodeficiency virus type 1 neuroinvasion through remodeling brain microvascular endothelial cells. J Neurovirol 2005; 11:281-91. [PMID: 16036808 DOI: 10.1080/13550280590952835] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Cocaine is a suspected cofactor in human immunodeficiency virus (HIV)-associated dementia but cocaine's effects are not clear. Herein the authors describe investigations of the mechanisms by which cocaine increases HIV-1 invasion through brain microvascular endothelial cells (BMVECs). Cocaine binds to a site on BMVECs, which is not a biogenic amine transporter, a binding site for estrogen, or a muscarinic receptor and for which benztropine and tamoxifen have the highest affinity. Cocaine treatment of BMVECs disrupts intercellular junctions and induces cell ruffling, which could account for their increased permeability and decreased electrical resistance. HIV-1 enters BMVECs by macropinocytosis and is transported to lysosomes and inactivated. In cocaine-treated BMVECs, the virus enters and persists in large cytoplasmic "lakes." Cocaine exposure of BMVECs up-regulates transcription of genes important in cytoskeleton organization, signal transduction, cell swelling, vesicular trafficking, and cell adhesion. The toxicity of cocaine for the blood-brain barrier may lead to increased virus neuroinvasion and neurovascular complications of cocaine abuse.
Collapse
Affiliation(s)
- Milan Fiala
- Department of Medicine, West Los Angeles VA Medical Center and UCLA School of Medicine, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Guo M, Wu MH, Granger HJ, Yuan SY. Focal adhesion kinase in neutrophil-induced microvascular hyperpermeability. Microcirculation 2005; 12:223-32. [PMID: 15824042 DOI: 10.1080/10739680590905251] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Recent experimental evidence indicates an essential role of focal adhesion kinase (FAK) in mediating endothelial adhesion, contraction, and migration under physical stress and chemical stimulation. However, the functional impact of FAK on microvascular barrier property during inflammation has not been revealed. The aim of this study was to explore the potential contribution of FAK to neutrophil-dependent microvascular hyperpermeability. METHODS The apparent permeability coefficient of albumin was measured in intact, isolated porcine coronary venules during stimulation by C5a-activated neutrophils. In parallel, the transendothelial flux of albumin was quantified in cultured venular endothelial cell monolayers exposed to C5a-activated neutrophils. Western blotting and immunocytochemistry were performed to assess FAK tyrosine phosphorylation and distribution in endothelial cells, respectively. To specify the signaling effect of FAK on neutrophil-elicited endothelial hyperpermeability, FAK-related nonkinase (FRNK) was expressed, purified, and directly transfected into the endothelium of venules, and the permeability response to neutrophils was measured during inhibition of FAK. RESULTS C5a-activated neutrophils induced a time- and concentration-dependent increase in venular permeability. Transfection of venules with FRNK did not alter the basal barrier function but greatly attenuated neutrophil-induced hyperpermeability in a dose-related manner. A similar permeability response to neutrophils was observed in venular endothelial cell monolayers, which was diminished after FRNK transfection. In addition, Western blot analysis showed that activated neutrophils caused a concentration-dependent increase in FAK tyrosine phosphorylation with a time course correlating with that of venular hyperpermeability. Transfection of FRNK blocked neutrophil-evoked FAK tyrosine phosphorylation. Furthermore, immunofluorescence microscopy revealed a significant morphological change of FAK from a punctuated, dot-like pattern under normal conditions to an elongated, dash-like staining that aligned with the longitudinal axis of cells upon neutrophil stimulation. CONCLUSION The results suggest that focal adhesion kinase significantly contributes to the endothelial hyperpermeability response to neutrophil activation. Phosphorylation of FAK may play an important signaling role in the regulation of microvascular barrier function during inflammation.
Collapse
Affiliation(s)
- Mingzhang Guo
- Department of Surgery, Texas A & M University Health Science Center, Temple, Texas, USA.
| | | | | | | |
Collapse
|
36
|
Verin AD. Tyrosine phosphorylation and endothelial cell barrier regulation. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:955-7. [PMID: 15793276 PMCID: PMC1602377 DOI: 10.1016/s0002-9440(10)62316-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Alexander D Verin
- The Johns Hopkins Asthma and Allergy Center, 5501 Hopkins Bayview Circle, 5A.50B, Baltimore, MD 21224-6801, USA.
| |
Collapse
|
37
|
Linz-McGillem LA, Moitra J, Garcia JGN. Cytoskeletal rearrangement and caspase activation in sphingosine 1-phosphate-induced lung capillary tube formation. Stem Cells Dev 2005; 13:496-508. [PMID: 15588507 DOI: 10.1089/scd.2004.13.496] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Angiogenesis is a multistep process involving the endothelial cell (EC) cytoskeleton in migration, proliferation, and barrier stabilization. Although precise intracellular pathways by which angiogenic tube formation occurs remain poorly understood, we speculated that interactions between the cytoskeleton and apoptosis are involved and explored cytoskeletal rearrangement and caspase activation in human lung microvascular EC capillary-like tube formation induced by sphingosine 1-phosphate (Sph 1-P) and vascular endothelial growth factor (VEGF). Sph 1-P and VEGF enhance tube formation quantified by a Tube Immaturity Index (TII) generated from the ratio of cell number to tube length, with concomitant morphologic and actomyosin network changes. Angiogenesis was temporally grouped into three stages with early changes characterized by cortical actin localization, whereas midstage tube development demonstrated elongated EC with peripheral actin labeling with transcellular stress fibers. Late tube formation was characterized by broad actin distribution and presence of caspase-positive EC. Phosphorylated MLC immunoreactivity was present at all stages, suggesting that coordinate Rho kinase and MLCK involvement is important to Sph 1-P-induced cell motility; however, chemical inhibition of either MLCK or Rho kinase failed to alter early tube formation. To address whether gaps created by apoptosis expand the lumen, Sph 1-P-induced tubes were differentiated in the presence of caspase inhibitor z-Val-Ala-Asp-fluoromethylketone (zVAD-FMK). Capillary-like tube maturation, but not length, was decreased by zVAD-FMK treatment. These studies suggest that Sph 1-P may induce EC tube formation by regulating early cytoskeletal rearrangement, whereas EC apoptosis within capillary-like tubes is necessary for late stage Sph 1-P-induced tube maturation and lumen formation.
Collapse
Affiliation(s)
- L A Linz-McGillem
- Division of Pulmonary and Critical Care Medicine, Center for Translational Respiratory Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | | | | |
Collapse
|
38
|
Lim YP, Wong CY, Ooi LL, Druker BJ, Epstein RJ. Selective Tyrosine Hyperphosphorylation of Cytoskeletal and Stress Proteins in Primary Human Breast Cancers. Clin Cancer Res 2004; 10:3980-7. [PMID: 15217928 DOI: 10.1158/1078-0432.ccr-03-0663] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Small-molecule growth factor receptor inhibitors block cell growth in vitro and downstream signaling in vivo, but controlled trials in patients with advanced solid tumors have yielded disappointing response rates. To clarify this discrepancy, we compared the patterns of tyrosine phosphoprotein expression in human cancer cells and primary tumors. EXPERIMENTAL DESIGN Immunoaffinity chromatography, two-dimensional electrophoresis, and antiphosphotyrosine immunoblotting were combined with mass spectrometry to determine the phosphoproteomic signatures of 40 matched normal and malignant tissues from patients with breast or liver cancer. The identities and abundance of the detected tyrosine phosphoproteins were compared with those of ligand-responsive A431 cells. RESULTS Patterns of tyrosine-phosphorylated proteins are similar among normal tissues of the same origin but vary markedly between different tissues. Primary breast tumors exhibit a strikingly homogeneous tyrosine phosphorylation profile, whereas liver cancers display greater phosphoproteomic diversity. The main breast-tumor-specific tyrosine phosphoproteins are cytoskeletal molecules (actin, tubulin, and vimentin) and molecular chaperones (Hsp70, Hsc71, and Grp75). In contrast, control studies in ligand-stimulated A431 human cancer cells revealed an additional phosphorylated subset of promitogenic phosphoproteins (Grb2, Shc, Jnk2, phospholipase C-gamma, and phosphatidylinositol 3'-kinase). CONCLUSIONS Identification of cytoskeletal and stress proteins as the most abundant tyrosine phosphoproteins in breast tumors implicates these molecules, rather than promitogenic effectors, as the prime stoichiometric substrates for kinase-inhibitory anticancer drugs in vivo. Because phosphorylated cytoskeletal proteins and chaperones mediate cell motility and apoptotic resistance, respectively, these data raise the intriguing possibility that small-molecule tyrosine kinase inhibitors may be of greatest value either as adjuvant antimetastatic/-invasive drugs or as chemo-/radiosensitizers.
Collapse
Affiliation(s)
- Yoon-Pin Lim
- Division of Medical Sciences and Department of Surgical Oncology, National Cancer Centre, Singapore
| | | | | | | | | |
Collapse
|
39
|
Dudek SM, Jacobson JR, Chiang ET, Birukov KG, Wang P, Zhan X, Garcia JGN. Pulmonary Endothelial Cell Barrier Enhancement by Sphingosine 1-Phosphate. J Biol Chem 2004; 279:24692-700. [PMID: 15056655 DOI: 10.1074/jbc.m313969200] [Citation(s) in RCA: 234] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We recently reported the critical importance of Rac GTPase-dependent cortical actin rearrangement in the augmentation of pulmonary endothelial cell (EC) barrier function by sphingosine 1-phosphate (S1P). We now describe functional roles for the actin-binding proteins cortactin and EC myosin light chain kinase (MLCK) in mediating this response. Antisense down-regulation of cortactin protein expression significantly inhibits S1P-induced barrier enhancement in cultured human pulmonary artery EC as measured by transendothelial electrical resistance (TER). Immunofluorescence studies reveal rapid, Rac-dependent translocation of cortactin to the expanded cortical actin band following S1P challenge, where colocalization with EC MLCK occurs within 5 min. Adenoviral overexpression of a Rac dominant negative mutant attenuates TER elevation by S1P. S1P also induces a rapid increase in cortactin tyrosine phosphorylation (within 30 s) critical to subsequent barrier enhancement, since EC transfected with a tyrosine-deficient mutant cortactin exhibit a blunted TER response. Direct binding of EC MLCK to the cortactin Src homology 3 domain appears essential to S1P barrier regulation, since cortactin blocking peptide inhibits both S1P-induced MLC phosphorylation and peak S1P-induced TER values. These data support novel roles for the cytoskeletal proteins cortactin and EC MLCK in mediating lung vascular barrier augmentation evoked by S1P.
Collapse
Affiliation(s)
- Steven M Dudek
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Sharma M, Sharma R, McCarthy ET, Savin VJ. The focal segmental glomerulosclerosis permeability factor: biochemical characteristics and biological effects. Exp Biol Med (Maywood) 2004; 229:85-98. [PMID: 14709781 DOI: 10.1177/153537020422900111] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is characterized by steroid resistant nephrotic syndrome and progression to end-stage renal disease. Proteinuria in certain patients with FSGS may be caused by a circulating factor (FSGS permeability factor [FSPF]). The current report documents the biochemical characteristics and the biological and molecular effects of 70% ammonium sulfate supernatant of plasma from patients with recurrence of FSGS after transplantation (FSGS 70% supernatant). FS permeability activity, defined as the capacity of plasma from patients with FSGS to increase albumin permeability (P(alb)) of isolated glomeruli, was assessed in vitro. Permeability activity was not affected by lyophilization. FSPF bound strongly to matrices containing Mono-Q anion exchanger or protein A. It eluted from matrix-bound Cibacron blue F3GA over a wide range of salt concentrations, indicating a potential binding with other proteins, such as albumin. FSPF caused a maximal increase in P(alb) within 2 mins of incubation in vitro. Cellular proteins isolated from glomeruli with increased P(alb) showed decreased tyrosine phosphorylation of focal adhesion kinase, paxillin, and other proteins. Tyrosine phosphatase ]inhibition prevented the increase in P(alb). Intravenous administration of as little as 3 mg protein in FSGS 70% supernatant increased P(alb), while 9 mg or more were required to produce proteinuria. We conclude that FSPF is a low-molecular-weight protein, carries an anionic charge, and binds to protein A. Effects of FSPF on the glomerular permeability barrier are rapid and dose dependent and involve signaling through altered phosphorylation of cellular proteins. Identification of these biochemical and biological characteristics may be used to design strategies for removing FSPF from circulation and for purification and identification of this factor.
Collapse
Affiliation(s)
- Mukut Sharma
- Nephrology Division, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | | | | | |
Collapse
|
41
|
Alvarez DF, Gjerde EAB, Townsley MI. Role of EETs in regulation of endothelial permeability in rat lung. Am J Physiol Lung Cell Mol Physiol 2004; 286:L445-51. [PMID: 14578116 DOI: 10.1152/ajplung.00150.2003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study tested the hypothesis that epoxyeicosatrienoic acids (EETs) derived from arachidonic acid via P-450 epoxygenases are soluble factors linking depletion of endoplasmic reticulum Ca2+ stores and store-dependent regulation of endothelial cell (EC) permeability in rat lung. EC permeability was measured via the capillary filtration coefficient ( Kf,c) in isolated, perfused rat lungs. 14,15-EET and 5,6-EET increased EC permeability, a response that was significantly different from that of 8,9-EET, 11,12-EET, and vehicle control. The permeability response to 14,15-EET was not significantly attenuated by the nonspecific Ca2+ channel blocker Gd3+ ( P = 0.068). In lungs perfused with low [Ca2+], 14,15-EET tended to increase EC permeability, although a significant increase in Kf,c was observed only following Ca2+ add-back. As positive control, we showed that the 3.7-fold increase in Kf,c evoked by thapsigargin (TG), a known activator of store depletion-induced Ca2+ entry, was blocked by both Gd3+ and low [Ca2+] buffer. Nonetheless, the permeability response to TG could not be blocked by the phospholipase A2 inhibitors mepacrine or methyl arachidonyl fluorophosphonate or the P-450 epoxygenase inhibitors 17-octadecynoic acid or propargyloxyphenyl hexanoic acid. Similarly, combined pretreatment with ibuprofen and dicyclohexylurea to block EET metabolism had no effect on the permeability response to TG. We conclude that EETs have a heterogeneous impact on EC permeability. Despite a requirement for Ca2+ entry with both TG and 14,15-EET, our data suggest that distinct signaling pathways or heterogeneity in EC responsiveness is responsible for the observed EC injury evoked by EETs and store depletion in the isolated rat lung.
Collapse
Affiliation(s)
- Diego F Alvarez
- Department of Physiology, University of Southern Alabama, Mobile, AL 36688, USA
| | | | | |
Collapse
|
42
|
Birukova AA, Smurova K, Birukov KG, Kaibuchi K, Garcia JGN, Verin AD. Role of Rho GTPases in thrombin-induced lung vascular endothelial cells barrier dysfunction. Microvasc Res 2004; 67:64-77. [PMID: 14709404 DOI: 10.1016/j.mvr.2003.09.007] [Citation(s) in RCA: 224] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thrombin-induced barrier dysfunction of pulmonary endothelial monolayer is associated with dramatic cytoskeletal reorganization, activation of actomyosin contraction, and gap formation. Phosphorylation of regulatory myosin light chains (MLC) is a key mechanism of endothelial cell (EC) contraction and barrier dysfunction, which is triggered by Ca(2+)/calmodulin-dependent MLC kinase (MLCK) and Rho-associated kinase (Rho-kinase). The role of MLCK in EC barrier regulation has been previously described; however, Rho-mediated pathway in thrombin-induced pulmonary EC dysfunction is not yet precisely characterized. Here, we demonstrate that thrombin-induced decreases in transendothelial electrical resistance (TER) indicating EC barrier dysfunction are universal for human and bovine pulmonary endothelium, and involve membrane translocation and direct activation of small GTPase Rho and its downstream target Rho-kinase. Transient Rho membrane translocation coincided with translocation of upstream Rho activator, guanosine nucleotide exchange factor p115-RhoGEF. Rho mediated activation of downstream target, Rho-kinase induced phosphorylation of the EC MLC phosphatase (MYPT1) at Thr(686) and Thr(850), resulting in MYPT1 inactivation, accumulation of diphospho-MLC, actin remodeling, and cell contraction. The specific Rho-kinase inhibitor, Y27632, abolished MYPT1 phosphorylation, MLC phosphorylation, significantly attenuated stress fiber formation and thrombin-induced TER decrease. Furthermore, expression of dominant-negative Rho and Rho-kinase abolished thrombin-induced stress fiber formation and MLC phosphorylation. Our data, which provide comprehensive analysis of Rho-mediated signal transduction in pulmonary EC, demonstrate involvement of guanosine nucleotide exchange factor, p115-RhoGEF, in thrombin-mediated Rho regulation, and suggest Rho, Rho-kinase, and MYPT1 as potential pharmacological and gene therapy targets critical for prevention of thrombin-induced EC barrier disruption and pulmonary edema associated with acute lung injury.
Collapse
Affiliation(s)
- Anna A Birukova
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
43
|
Bogatcheva NV, Dudek SM, Garcia JGN, Verin AD. Mitogen-Activated Protein Kinases in Endothelial Pathophysiology. J Investig Med 2003. [DOI: 10.1177/108155890305100630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Endothelial cells continuously respond to extracellular stimuli such as chemical signals produced by circulating blood elements or mechanical forces such as shear stress. Proinflammatory cytokines, mitogens, reactive oxygen species, and shear stress trigger signal molecules to initiate multiple intracellular pathways, which often converge at mitogen-activated protein (MAP) kinase activation. The MAP kinase superfamily represents a burgeoning area of clinical investigation for treatment of various inflammatory and oncologic diseases and plays an essential role in mediating response to infection, ischemia/reperfusion injury, and vessel healing and remodeling through regulation of such diverse phenomena as endothelial cell proliferation, migration, apoptosis, and endothelial barrier function. The downstream effects of MAP kinase activation include modulation of gene expression via up-regulation of various transcription factors. In addition to these sustained effects, MAP kinases coordinate more immediate responses that affect dynamic cytoskeletal rearrangements necessary for cell migration and regulation of barrier function. This review discusses the important regulatory roles of MAP kinases in the vital physiologic functions of endothelium, focusing mainly on the role of MAP kinases in the maintenance of endothelial barrier.
Collapse
Affiliation(s)
| | - Steven M. Dudek
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joe G. N. Garcia
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alexander D. Verin
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
44
|
Wu MH, Guo M, Yuan SY, Granger HJ. Focal adhesion kinase mediates porcine venular hyperpermeability elicited by vascular endothelial growth factor. J Physiol 2003; 552:691-9. [PMID: 12949227 PMCID: PMC2343443 DOI: 10.1113/jphysiol.2003.048405] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Focal adhesion kinase (FAK) is known to mediate endothelial cell adhesion and migration in response to vascular endothelial growth factor (VEGF). The aim of this study was to explore a potential role for FAK in VEGF regulation of microvascular endothelial barrier function. The apparent permeability coefficient of albumin (Pa) was measured in intact isolated porcine coronary venules. Treating the vessels with VEGF induced a time- and concentration-dependent increase in Pa. Inhibition of FAK through direct delivery of FAK-related non-kinase (FRNK) into venular endothelium did not alter basal barrier function but significantly attenuated VEGF-elicited hyperpermeability. Furthermore, cultured human umbilical vein endothelial monolayers displayed a similar hyperpermeability response to VEGF which was greatly attenuated by FRNK. Western blot analysis showed that VEGF promoted FAK phosphorylation in a time course correlating with that of venular hyperpermeability. The phosphorylation response was blocked by FRNK treatment. In addition, VEGF stimulation caused a significant morphological change of FAK from a punctate pattern to an elongated, dash-like staining that aligned with the longitudinal axis of the cells. Taken together, the results suggest that FAK contributes to VEGF-elicited vascular hyperpermeability. Phosphorylation of FAK may play an important role in the signal transduction of vascular barrier response to VEGF.
Collapse
Affiliation(s)
- Mack H Wu
- Cardiovascular Research Institute and Department of Medical Physiology, College of Medicine, Texas A&M University System Health Science Center, 702 Southwest HK Dodgen Loop, Temple, TX 76504, USA.
| | | | | | | |
Collapse
|
45
|
Usatyuk PV, Fomin VP, Shi S, Garcia JGN, Schaphorst K, Natarajan V. Role of Ca2+ in diperoxovanadate-induced cytoskeletal remodeling and endothelial cell barrier function. Am J Physiol Lung Cell Mol Physiol 2003; 285:L1006-17. [PMID: 12882766 DOI: 10.1152/ajplung.00408.2002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diperoxovanadate (DPV), a potent inhibitor of protein tyrosine phosphatases and activator of tyrosine kinases, alters endothelial barrier function via signaling pathways that are incompletely understood. One potential pathway is Src kinase-mediated tyrosine phosphorylation of proteins such as cortactin that regulate endothelial cell (EC) cytoskeleton assembly. As DPV modulates endothelial cell signaling via protein tyrosine phosphorylation, we determined the role of DPV-induced intracellular free calcium concentration ([Ca2+]i) in activation of Src kinase, cytoskeletal remodeling, and barrier function in bovine pulmonary artery endothelial cells (BPAECs). DPV in a dose- and time-dependent fashion increased [Ca2+]i, which was partially blocked by the calcium channel blockers nifedipine and Gd3+. Treatment of cells with thapsigargin released Ca2+ from the endoplasmic reticulum, and subsequent addition of DPV caused no further change in [Ca2+]i. These data suggest that DPV-induced [Ca2+]i includes Ca release from the endoplasmic reticulum and Ca influx through store-operated calcium entry. Furthermore, DPV induced an increase in protein tyrosine phosphorylation, phosphorylation of Src and cortactin, actin remodeling, and altered transendothelial electrical resistance in BPAECs. These DPV-mediated effects were significantly attenuated by BAPTA (25 microM), a chelator of [Ca2+]i. Immunofluorescence studies reveal that the DPV-mediated colocalization of cortactin with peripheral actin was also prevented by BAPTA. Chelation of extracellular Ca2+ by EGTA had marginal effects on DPV-induced phosphorylation of Src and cortactin; actin stress fibers formation, however, affected EC barrier function. These data suggest that DPV-induced changes in [Ca2+]i regulate endothelial barrier function using signaling pathways that involve Src and cytoskeleton remodeling.
Collapse
Affiliation(s)
- Peter V Usatyuk
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | | | | | | | | | | |
Collapse
|
46
|
Borbiev T, Verin AD, Birukova A, Liu F, Crow MT, Garcia JGN. Role of CaM kinase II and ERK activation in thrombin-induced endothelial cell barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 2003; 285:L43-54. [PMID: 12788788 DOI: 10.1152/ajplung.00460.2001] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
We have previously shown that thrombin-induced endothelial cell barrier dysfunction involves cytoskeletal rearrangement and contraction, and we have elucidated the important role of endothelial cell myosin light chain kinase and the actin- and myosin-binding protein caldesmon. We evaluated the contribution of calmodulin (CaM) kinase II and extracellular signal-regulated kinase (ERK) activation in thrombin-mediated bovine pulmonary artery endothelial cell contraction and barrier dysfunction. Similar to thrombin, infection with a constitutively active adenoviral alpha-CaM kinase II construct induced significant ERK activation, indicating that CaM kinase II activation lies upstream of ERK. Thrombin-induced ERK-dependent caldesmon phosphorylation (Ser789) was inhibited by either KN-93, a specific CaM kinase II inhibitor, or U0126, an inhibitor of MEK activation. Immunofluorescence microscopy studies revealed phosphocaldesmon colocalization within thrombin-induced actin stress fibers. Pretreatment with either U0126 or KN-93 attenuated thrombin-mediated cytoskeletal rearrangement and evoked declines in transendothelial electrical resistance while reversing thrombin-induced dissociation of myosin from nondenaturing caldesmon immunoprecipitates. These results strongly suggest the involvement of CaM kinase II and ERK activities in thrombin-mediated caldesmon phosphorylation and both contractile and barrier regulation.
Collapse
Affiliation(s)
- Talaibek Borbiev
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, Maryland 21224, USA
| | | | | | | | | | | |
Collapse
|
47
|
Young BA, Sui X, Kiser TD, Hyun SW, Wang P, Sakarya S, Angelini DJ, Schaphorst KL, Hasday JD, Cross AS, Romer LH, Passaniti A, Goldblum SE. Protein tyrosine phosphatase activity regulates endothelial cell-cell interactions, the paracellular pathway, and capillary tube stability. Am J Physiol Lung Cell Mol Physiol 2003; 285:L63-75. [PMID: 12626337 DOI: 10.1152/ajplung.00423.2002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Protein tyrosine phosphorylation is tightly regulated through the actions of both protein tyrosine kinases and protein tyrosine phosphatases. In this study, we demonstrate that protein tyrosine phosphatase inhibition promotes tyrosine phosphorylation of endothelial cell-cell adherens junction proteins, opens an endothelial paracellular pathway, and increases both transendothelial albumin flux and neutrophil migration. Tyrosine phosphatase inhibition with sodium orthovanadate or phenylarsine oxide induced dose- and time-dependent increases in [14C]bovine serum albumin flux across postconfluent bovine pulmonary artery endothelial cell monolayers. These increases in albumin flux were coincident with actin reorganization and intercellular gap formation in both postconfluent monolayers and preformed endothelial cell capillary tubes. Vanadate (25 microM) increased tyrosine phosphorylation of endothelial cell proteins 12-fold within 1 h. Tyrosine phosphorylated proteins were immunolocalized to the intercellular boundaries, and several were identified as the endothelial cell-cell adherens junction proteins, vascular-endothelial cadherin, and beta-, gamma-, and p120-catenin as well as platelet endothelial cell adhesion molecule-1. Of note, these tyrosine phosphorylation events were not associated with disassembly of the adherens junction complex or its uncoupling from the actin cytoskeleton. The dose and time requirements for vanadate-induced increases in phosphorylation were comparable with those defined for increments in transendothelial [14C]albumin flux and neutrophil migration, and pretreatment with the tyrosine kinase inhibitor herbimycin A protected against these effects. These data suggest that protein tyrosine phosphatases and their substrates, which localize to the endothelial cell-cell boundaries, regulate adherens junctional integrity, the movement of macromolecules and cells through the endothelial paracellular pathway, and capillary tube stability.
Collapse
Affiliation(s)
- Bradford A Young
- Division of Infectious Diseases, Department of Veterans Affairs Medical Center, Baltimore 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Schaphorst KL, Chiang E, Jacobs KN, Zaiman A, Natarajan V, Wigley F, Garcia JGN. Role of sphingosine-1 phosphate in the enhancement of endothelial barrier integrity by platelet-released products. Am J Physiol Lung Cell Mol Physiol 2003; 285:L258-67. [PMID: 12626332 DOI: 10.1152/ajplung.00311.2002] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In vitro and in vivo evidence indicates that circulating platelets affect both vascular integrity and hemostasis. How platelets enhance the permeability barrier of the vascular endothelium is not well understood. We measured the effect of isolated human platelets on human pulmonary artery endothelial cell (EC) barrier integrity by monitoring transmonolayer electrical resistance. EC barrier function was significantly increased by the addition of platelets ( approximately 40% maximum, 2.5 x 106 platelets/ml). Platelet supernatants, derived from 2.5 x 106 platelets/ml, reproduced the barrier enhancement and reversed the barrier dysfunction produced by the edemagenic agonist thrombin, which implicates a soluble barrier-promoting factor. The barrier-enhancing effect of platelet supernatants was heat stable but was attenuated by either charcoal delipidation (suggesting a vasoactive lipid mediator) or pertussis toxin, implying involvement of a Gialpha-coupled receptor signal transduction pathway. Sphingosine-1-phosphate (S1P), a sphingolipid that is released from activated platelets, is known to ligate G protein-coupled EC differentiation gene (EDG) receptors, increase EC electrical resistance, and reorganize the actin cytoskeleton (Garcia JG, Liu F, Verin AD, Birukova A, Dechert MA, Gerthoffer WT, Bamberg JR, and English D. J Clin Invest 108: 689-701, 2001). Infection of EC with an adenoviral vector expressing an antisense oligonucleotide directed against EDG-1 but not infection with control vector attenuated the barrier-enhancing effect of both platelet supernatants and S1P. These results indicate that a major physiologically relevant vascular barrier-protective mediator produced by human platelets is S1P.
Collapse
Affiliation(s)
- Kane L Schaphorst
- Division of Pulmonary and Critical Care Medicine and Center for Translational Respiratory Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21224-6801, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
The permeability of exchange microvessels is regulated through complex interactions between signaling molecules and structural proteins in the endothelium. Endothelial barrier integrity is maintained by adhesive interactions occurring at the cell-cell and cell-matrix contacts via junctional proteins and focal adhesion complexes that are anchored to the cytoskeleton. Cyclic AMP (cAMP) and cAMP-dependent kinase counteract with the nitric oxide (NO)-cyclic GMP (cGMP) pathway to protect the basal barrier function. Upon stimulation by physical stress, growth factors, or inflammatory agents, endothelial cells undergo a series of intracellular signaling reactions involving activation of protein kinase C (PKC), protein kinase G (PKG), mitogen-activated protein kinases (MAPK), and/or protein tyrosine kinases. The phosphorylation cascades trigger biochemical and conformational changes in the barrier structure and ultimately lead to an opening of the paracellular pathway. In particular, myosin light chain kinase (MLCK) activation and subsequent myosin light chain (MLC) phosphorylation in endothelial cells directly result in cell contraction and shape changes. The phosphorylation of beta-catenin may cause disorganization of adherens junctions or dissociation of vascular endothelial (VE)-cadherin-catenin complex from its cytoskeletal anchor, leading to loose or opened intercellular junctions. Additionally, focal adhesion kinase (FAK) phosphorylation-coupled focal adhesion assembly and redistribution provide an anchorage support for the conformational changes occurring in the cells and at the cell junctions. The Src family tyrosine kinases may serve as common signals that coordinate these molecular events to facilitate the paracellular transport of macromolecules. The critical roles of protein kinases in endothelial hyperpermeability implicate the therapeutic significance of protein kinase inhibitors in the prevention and treatment of diseases and injuries that are associated with microvascular barrier dysfunction.
Collapse
Affiliation(s)
- Sarah Y Yuan
- Department of Surgery, Cardiovascular Research Institute, Texas A & M University Health Science Center College of Medicine, 702 Southwest HK Dodgen Loop, Temple, TX 76504, USA.
| |
Collapse
|
50
|
Bogatcheva NV, Garcia JGN, Verin AD. Role of tyrosine kinase signaling in endothelial cell barrier regulation. Vascul Pharmacol 2002; 39:201-12. [PMID: 12747960 DOI: 10.1016/s1537-1891(03)00009-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Phosphorylation of proteins on tyrosine acts as a reversible and specific trigger mechanism, forming or disrupting regulatory connections between proteins. Tyrosine kinases and phosphatases participate in multiple cellular processes, and considerable evidence now supports a role for tyrosine phosphorylation in vascular permeability. A semipermeable barrier between the vascular compartment and the interstitium is maintained by the integrity of endothelial monolayer, controlling movement of fluids, macromolecules and leucocytes. Barrier function is regulated by the adjustment of paracellular gaps between endothelial cells (ECs) by two antagonistic forces, centripetal cytoskeletal tension and opposing cell-cell and cell-matrix adhesion forces. Both cytoskeletal filaments and adhesion sites are intimately linked in complex machinery which is regulated by multiple signaling events including protein phosphorylation and/or protein translocation to specific intracellular positions. Tyrosine kinases occupy key positions in the mechanism controlling cell responses mediated through various cell surface receptors, which use tyrosine phosphorylation to transduce extracellular signal.
Collapse
Affiliation(s)
- Natalia V Bogatcheva
- Johns Hopkins Asthma and Allergy Center, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, 5501 Hopkins, Bayview Circle, Baltimore, MD 21224-6801, USA
| | | | | |
Collapse
|