1
|
Zhang Y, Tan J, Zhao Y, Guan L, Li S. By activating endothelium histone H4 mediates oleic acid-induced acute respiratory distress syndrome. BMC Pulm Med 2025; 25:3. [PMID: 39757148 DOI: 10.1186/s12890-024-03334-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 10/08/2024] [Indexed: 01/07/2025] Open
Abstract
OBJECTIVE This study investigated pathogenic role and mechanism of extracellular histone H4 during oleic acid (OA)-induced acute respiratory distress syndrome (ARDS). METHODS ARDS was induced by intravenous injection of OA in mice, and evaluated by blood gas, pathological analysis, lung edema, and survival rate. Heparan sulfate (HS) degradation was evaluated using immunofluorescence and flow cytometry. The released von Willebrand factor (vWF) was measured using ELISA. P-selectin translocation and neutrophil infiltration were measured via immunohistochemical analysis. Changes in VE-cadherin were measured by western blot. Blocking antibodies against TLRs were used to investigate the signaling pathway. RESULTS Histone H4 in plasma and BALF increased significantly after OA injection. Histone H4 was closely correlated with the OA dose, which determined the ARDS severity. Pretreatment with histone H4 further aggravated pulmonary edema and death rate, while anti-H4 antibody exerted obvious protective effects. Histone H4 directly activated the endothelia. Endothelial activation was evidently manifested as HS degradation, release of vWF, P-selectin translocation, and VE-Cadherin reduction. The synergistic stimulus of activated endothelia was required for effective neutrophil activation by histone H4. Both TLRs and calcium mediated histone H4-induced endothelial activation. CONCLUSIONS Histone H4 is a pro-inflammatory and pro-thrombotic molecule in OA-induced ARDS in mice.
Collapse
Affiliation(s)
- Yanlin Zhang
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China.
| | - Jingjin Tan
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Yiran Zhao
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Li Guan
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China
| | - Shuqiang Li
- Research Center of Occupational Medicine, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
2
|
Li M, Hao X, Hu Z, Tian J, Shi J, Ma D, Guo M, Li S, Zuo C, Liang Y, Tang M, Mao C, Xu Y, Shi C. Microvascular and cellular dysfunctions in Alzheimer's disease: an integrative analysis perspective. Sci Rep 2024; 14:20944. [PMID: 39251797 PMCID: PMC11385648 DOI: 10.1038/s41598-024-71888-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, characterized by memory loss, cognitive decline, personality changes, and various neurological symptoms. The role of blood-brain barrier (BBB) injury, extracellular matrix (ECM) abnormalities, and oligodendrocytes (ODCs) dysfunction in AD has gained increasing attention, yet the detailed pathogenesis remains elusive. This study integrates single-cell sequencing of AD patients' cerebrovascular system with a genome-wide association analysis. It aims to elucidate the associations and potential mechanisms behind pericytes injury, ECM disorder, and ODCs dysfunction in AD pathogenesis. Finally, we identified that abnormalities in the pericyte PI3K-AKT-FOXO signaling pathway may be involved in the pathogenic process of AD. This comprehensive approach sheds new light on the complex etiology of AD and opens avenues for advanced research into its pathogenesis and therapeutic strategies.
Collapse
Affiliation(s)
- Mengjie Li
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Xiaoyan Hao
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Zhengwei Hu
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Jie Tian
- Zhengzhou Railway Vocational and Technical College, Zhengzhou, 450000, Henan, China
| | - Jingjing Shi
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Dongrui Ma
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Mengnan Guo
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Shuangjie Li
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Chunyan Zuo
- Zhengzhou University, Zhengzhou, 450000, Henan, China
| | | | - Mibo Tang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 Jian-she East Road, Zhengzhou, 450000, Henan, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 Jian-she East Road, Zhengzhou, 450000, Henan, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 Jian-she East Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 1 Jian-she East Road, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
3
|
Breslin JW. Edema and lymphatic clearance: molecular mechanisms and ongoing challenges. Clin Sci (Lond) 2023; 137:1451-1476. [PMID: 37732545 PMCID: PMC11025659 DOI: 10.1042/cs20220314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Resolution of edema remains a significant clinical challenge. Conditions such as traumatic shock, sepsis, or diabetes often involve microvascular hyperpermeability, which leads to tissue and organ dysfunction. Lymphatic insufficiency due to genetic causes, surgical removal of lymph nodes, or infections, leads to varying degrees of tissue swelling that impair mobility and immune defenses. Treatment options are limited to management of edema as there are no specific therapeutics that have demonstrated significant success for ameliorating microvascular leakage or impaired lymphatic function. This review examines current knowledge about the physiological, cellular, and molecular mechanisms that control microvascular permeability and lymphatic clearance, the respective processes for interstitial fluid formation and removal. Clinical conditions featuring edema, along with potential future directions are discussed.
Collapse
Affiliation(s)
- Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, FL, U.S.A
| |
Collapse
|
4
|
Jayant G, Kuperberg S, Somnay K, Wadgaonkar R. The Role of Sphingolipids in Regulating Vascular Permeability in Idiopathic Pulmonary Fibrosis. Biomedicines 2023; 11:1728. [PMID: 37371823 DOI: 10.3390/biomedicines11061728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 05/29/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease that causes scarring and fibrotic transformation of the lung parenchyma, resulting in the progressive loss of respiratory function and, often, death. Current treatments that target profibrotic factors can slow the rate of progression but are unable to ultimately stop it. In the past decade, many studies have shown that increased vascular permeability may be both a predictive and perpetuating factor in fibrogenesis. Consequently, there is a search for therapeutic targets to try and modulate vascular permeability in fibrotic lungs. One such class of targets that show great promise is sphingolipids. Sphingolipids are common in cell membranes and are increasingly recognized as critical to many cell signaling pathways, including those that affect the integrity of the vascular endothelial barrier. In this focused review we look at sphingolipids, particularly the sphingosine-1-phosphate (S1P) axis and its effects on vascular permeability, and how those effects may affect the pathogenesis of IPF. We further examine existing S1P modulators and their potential efficacy as therapeutics for IPF.
Collapse
Affiliation(s)
- Girish Jayant
- SUNY Downstate College of Medicine, Brooklyn, NY 11203, USA
| | | | - Kaumudi Somnay
- NY Presbyterian Hospital Queens, New York, NY 11355, USA
| | - Raj Wadgaonkar
- SUNY Downstate College of Medicine, Brooklyn, NY 11203, USA
| |
Collapse
|
5
|
Teymouri S, Pourbayram Kaleybar S, Hejazian SS, Hejazian SM, Ansarin K, Ardalan M, Zununi Vahed S. The effect of Fingolimod on patients with moderate to severe COVID-19. Pharmacol Res Perspect 2023; 11:e01039. [PMID: 36567519 PMCID: PMC9791159 DOI: 10.1002/prp2.1039] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 12/09/2022] [Indexed: 12/27/2022] Open
Abstract
Hyper-inflammation, cytokine storm, and recruitment of immune cells lead to uncontrollable endothelial cell damage in patients with coronavirus disease 2019 (COVID-19). Sphingosine 1-phosphate (S1P) signaling is needed for endothelial integrity and its decreased serum level is a predictor of clinical severity in COVID-19. In this clinical trial, the effect of Fingolimod, an agonist of S1P, was evaluated on patients with COVID-19. Forty patients with moderate to severe COVID-19 were enrolled and divided into two groups including (1) the control group (n = 21) receiving the national standard regimen for COVID-19 patients and (2) the intervention group (n = 19) that prescribed daily Fingolimod (0.5 mg) for 3 days besides receiving the standard national regimen for COVID-19. The hospitalization period, re-admission rate, intensive care unit (ICU) administration, need for mechanical ventilation, and mortality rate were assessed as primary outcomes in both groups. The results showed that re-admission was significantly decreased in COVID-19 patients who received Fingolimod compared to the controls (p = .04). In addition, the hemoglobin levels of the COVID-19 patients in the intervention group were increased compared to the controls (p = .018). However, no significant differences were found regarding the intubation or mortality rate between the groups (p > .05). Fingolimod could significantly reduce the re-admission rate after hospitalization with COVID-19. Fingolimod may not enhance patients' outcomes with moderate COVID-19. It is necessary to examine these findings in a larger cohort of patients with severe to critical COVID-19.
Collapse
Affiliation(s)
- Soheil Teymouri
- Tuberculosis and Lung Disease Research CenterTabriz University of Medical SciencesTabrizIran
| | - Siamak Pourbayram Kaleybar
- Kidney Research CenterFaculty of MedicineTabriz University of Medical SciencesTabrizIran
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | | | | | - Khalil Ansarin
- Tuberculosis and Lung Disease Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mohammadreza Ardalan
- Kidney Research CenterFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| | - Sepideh Zununi Vahed
- Kidney Research CenterFaculty of MedicineTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
6
|
Sveeggen TM, Abbey CA, Smith RL, Salinas ML, Chapkin RS, Bayless KJ. Annexin A2 modulates phospholipid membrane composition upstream of Arp2 to control angiogenic sprout initiation. FASEB J 2023; 37:e22715. [PMID: 36527391 PMCID: PMC10586062 DOI: 10.1096/fj.202201088r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/10/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
The intersection of protein and lipid biology is of growing importance for understanding how cells address structural challenges during adhesion and migration. While protein complexes engaged with the cytoskeleton play a vital role, support from the phospholipid membrane is crucial for directing localization and assembly of key protein complexes. During angiogenesis, dramatic cellular remodeling is necessary for endothelial cells to shift from a stable monolayer to invasive structures. However, the molecular dynamics between lipids and proteins during endothelial invasion are not defined. Here, we utilized cell culture, immunofluorescence, and lipidomic analyses to identify a novel role for the membrane binding protein Annexin A2 (ANXA2) in modulating the composition of specific membrane lipids necessary for cortical F-actin organization and adherens junction stabilization. In the absence of ANXA2, there is disorganized cortical F-actin, reduced junctional Arp2, excess sprout initiation, and ultimately failed sprout maturation. Furthermore, we observed reduced filipin III labeling of membrane cholesterol in cells with reduced ANXA2, suggesting there is an alteration in phospholipid membrane dynamics. Lipidomic analyses revealed that 42 lipid species were altered with loss of ANXA2, including an accumulation of phosphatidylcholine (16:0_16:0). We found that supplementation of phosphatidylcholine (16:0_16:0) in wild-type endothelial cells mimicked the ANXA2 knock-down phenotype, indicating that ANXA2 regulated the phospholipid membrane upstream of Arp2 recruitment and organization of cortical F-actin. Altogether, these data indicate a novel role for ANXA2 in coordinating events at endothelial junctions needed to initiate sprouting and show that proper lipid modulation is a critical component of these events.
Collapse
Affiliation(s)
- Timothy M. Sveeggen
- Texas A&M Health Science Center, Texas, Bryan, USA
- Interdisciplinary Graduate Program in Genetics, Texas A&M University, College Station, Texas, USA
| | | | | | - Michael L. Salinas
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas, USA
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, Texas, USA
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | | |
Collapse
|
7
|
HDL and Endothelial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:27-47. [DOI: 10.1007/978-981-19-1592-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
8
|
Mascarenhas JB, Gaber AA, Larrinaga TM, Mayfield R, Novak S, Camp SM, Gregorio C, Jacobson JR, Cress AE, Dudek SM, Garcia JGN. EVL is a novel focal adhesion protein involved in the regulation of cytoskeletal dynamics and vascular permeability. Pulm Circ 2021; 11:20458940211049002. [PMID: 34631011 PMCID: PMC8493322 DOI: 10.1177/20458940211049002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022] Open
Abstract
Increases in lung vascular permeability is a cardinal feature of inflammatory disease and represents an imbalance in vascular contractile forces and barrier-restorative forces, with both forces highly dependent upon the actin cytoskeleton. The current study investigates the role of Ena-VASP-like (EVL), a member of the Ena-VASP family known to regulate the actin cytoskeleton, in regulating vascular permeability responses and lung endothelial cell barrier integrity. Utilizing changes in transendothelial electricial resistance (TEER) to measure endothelial cell barrier responses, we demonstrate that EVL expression regulates endothelial cell responses to both sphingosine-1-phospate (S1P), a vascular barrier-enhancing agonist, and to thrombin, a barrier-disrupting stimulus. Total internal reflection fluorescence demonstrates that EVL is present in endothelial cell focal adhesions and impacts focal adhesion size, distribution, and the number of focal adhesions generated in response to S1P and thrombin challenge, with the focal adhesion kinase (FAK) a key contributor in S1P-stimulated EVL-transduced endothelial cell but a limited role in thrombin-induced focal adhesion rearrangements. In summary, these data indicate that EVL is a focal adhesion protein intimately involved in regulation of cytoskeletal responses to endothelial cell barrier-altering stimuli. Keywords: cytoskeleton, vascular barrier, sphingosine-1-phosphate, thrombin, focal adhesion kinase (FAK), Ena-VASP like protein (EVL), cytoskeletal regulatory protein
Collapse
Affiliation(s)
| | - Amir A Gaber
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Tania M Larrinaga
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Rachel Mayfield
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Stefanie Novak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Sara M Camp
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Carol Gregorio
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Jeffrey R Jacobson
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Anne E Cress
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Steven M Dudek
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Joe G N Garcia
- Department of Medicine, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
9
|
Warboys CM, Weinberg PD. S1P in the development of atherosclerosis: roles of hemodynamic wall shear stress and endothelial permeability. Tissue Barriers 2021; 9:1959243. [PMID: 34542010 DOI: 10.1080/21688370.2021.1959243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Atherosclerosis is characterized by focal accumulations of lipid within the arterial wall, thought to arise from effects of hemodynamic wall shear stress (WSS) on endothelial permeability. Identifying pathways that mediate the effects of shear on permeability could therefore provide new therapeutic opportunities. Here, we consider whether the sphingosine-1-phosphate (S1P) pathway could constitute such a route. We review effects of S1P in endothelial barrier function, the influence of WSS on S1P production and signaling, the results of trials investigating S1P in experimental atherosclerosis in mice, and associations between S1P levels and cardiovascular disease in humans. Although it seems clear that S1P reduces endothelial permeability and responds to WSS, the evidence that it influences atherosclerosis is equivocal. The effects of specifically pro- and anti-atherosclerotic WSS profiles on the S1P pathway require investigation, as do influences of S1P on the vesicular pathways likely to dominate low-density lipoprotein transport across endothelium.
Collapse
Affiliation(s)
- Christina M Warboys
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Peter D Weinberg
- Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
10
|
Sharifi-Rad J, Quispe C, Imran M, Rauf A, Nadeem M, Gondal TA, Ahmad B, Atif M, Mubarak MS, Sytar O, Zhilina OM, Garsiya ER, Smeriglio A, Trombetta D, Pons DG, Martorell M, Cardoso SM, Razis AFA, Sunusi U, Kamal RM, Rotariu LS, Butnariu M, Docea AO, Calina D. Genistein: An Integrative Overview of Its Mode of Action, Pharmacological Properties, and Health Benefits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3268136. [PMID: 34336089 PMCID: PMC8315847 DOI: 10.1155/2021/3268136] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/11/2021] [Accepted: 06/28/2021] [Indexed: 12/15/2022]
Abstract
Genistein is an isoflavone first isolated from the brooming plant Dyer's Genista tinctoria L. and is widely distributed in the Fabaceae family. As an isoflavone, mammalian genistein exerts estrogen-like functions. Several biological effects of genistein have been reported in preclinical studies, such as the antioxidant, anti-inflammatory, antibacterial, and antiviral activities, the effects of angiogenesis and estrogen, and the pharmacological activities on diabetes and lipid metabolism. The purpose of this review is to provide up-to-date evidence of preclinical pharmacological activities with mechanisms of action, bioavailability, and clinical evidence of genistein. The literature was researched using the most important keyword "genistein" from the PubMed, Science, and Google Scholar databases, and the taxonomy was validated using The Plant List. Data were also collected from specialized books and other online resources. The main positive effects of genistein refer to the protection against cardiovascular diseases and to the decrease of the incidence of some types of cancer, especially breast cancer. Although the mechanism of protection against cancer involves several aspects of genistein metabolism, the researchers attribute this effect to the similarity between the structure of soy genistein and that of estrogen. This structural similarity allows genistein to displace estrogen from cellular receptors, thus blocking their hormonal activity. The pharmacological activities resulting from the experimental studies of this review support the traditional uses of genistein, but in the future, further investigations are needed on the efficacy, safety, and use of nanotechnologies to increase bioavailability and therapeutic efficacy.
Collapse
Affiliation(s)
- Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique 1110939, Chile
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar-, 23561 Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Nadeem
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Vehari-, Pakistan
| | | | - Bashir Ahmad
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar-, 25120 KPK, Pakistan
| | - Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72341, Saudi Arabia
| | | | - Oksana Sytar
- Department of Plant Biology Department, Institute of Biology, Taras Shevchenko National University of Kyiv, Volodymyrska Str., 64, Kyiv 01033, Ukraine
- Department of Plant Physiology, Slovak University of Agriculture, A. Hlinku 2, 94976 Nitra, Slovakia
| | - Oxana Mihailovna Zhilina
- Department of Organic Chemistry, Pyatigorsk Medical-Pharmaceutical Institute (PMPI), Branch of Volgograd State Medical University, Ministry of Health of Russia, Pyatigorsk 357532, Russia
| | - Ekaterina Robertovna Garsiya
- Department of Pharmacognosy, Botany and Technology of Phytopreparations, Pyatigorsk Medical-Pharmaceutical Institute (PMPI), Branch of Volgograd State Medical University, Ministry of Health of Russia, Pyatigorsk 357532, Russia
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Domenico Trombetta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Daniel Gabriel Pons
- Grupo Multidisciplinar de Oncología Traslacional (GMOT), Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears (UIB), Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma 07122, Spain
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
- Unidad de Desarrollo Tecnológico, Universidad de Concepción UDT, Concepción 4070386, Chile
| | - Susana M Cardoso
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Usman Sunusi
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Bayero University Kano, PMB 3011 Kano, Nigeria
| | - Ramla Muhammad Kamal
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Pharmacology, Federal University Dutse, PMB 7156 Dutse Jigawa State, Nigeria
| | - Lia Sanda Rotariu
- Banat's University of Agricultural Sciences and Veterinary Medicine "King Michael I of Romania" from Timisoara, Romania
| | - Monica Butnariu
- Banat's University of Agricultural Sciences and Veterinary Medicine "King Michael I of Romania" from Timisoara, Romania
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
11
|
Uehara K, Uehara A. Immunohistochemical study of dissociation and association of adherens junctions in splenic sinus endothelial cells. Cell Tissue Res 2021; 384:25-33. [PMID: 33660049 DOI: 10.1007/s00441-021-03426-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 01/25/2021] [Indexed: 11/26/2022]
Abstract
It is not yet clear whether cellular junctions between splenic sinus endothelial cells are open or closed. In order to clarify this, immunolocalization of thrombomodulin (TM), endothelial protein C receptor (EPCR), protease-activated receptor 1 (PAR1), sphingosine 1-phosphate receptor 1 (S1P1), β-catenin phosphorylated at Try142 (β-catenin Y142) and β-catenin phosphorylated at Try654 (β-catenin Y654), which are related proteins that regulate dissociation and association of the adherens junctions of endothelial cells, are examined in rats using laser microscopy and electron microscopy. TM, EPCR, PAR1 and S1P1 were colocalized in the entire circumference of the endothelial cells, as well as in the caveolar membranes and junctional membranes of adjacent endothelial cells. These molecules may protect the adherens junctions of the endothelial cells. On the other hand, β-catenin Y142 and β-catenin Y 654 colocalized with α-catenin and β-catenin, respectively and in addition, β-catenin Y142 and β-catenin Y 654 were localized in the vicinity of the adherens junctions of the endothelial cells from immunogold electron microcopy. The adherens junctions are considered to be partially dissociated at the site where β-catenin Y142 and β-catenin Y 654 are localized. Thus, the system that protects the adherens junctions and the system that dissociates them may concurrently coexist in the endothelial cells and dissociation and association of the adherens junctions may be constantly repeated at the cell boundary of the endothelial cells.
Collapse
Affiliation(s)
- Kiyoko Uehara
- Department of Cell Biology, Fukuoka University School of Medicine, Jonan-ku, Fukuoka, 814-0180, Japan.
| | - Akira Uehara
- Department of Physiology, Fukuoka University School of Medicine, Jonan-ku, Fukuoka, 814-0180, Japan
| |
Collapse
|
12
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
13
|
Klomp JE, Shaaya M, Matsche J, Rebiai R, Aaron JS, Collins KB, Huyot V, Gonzalez AM, Muller WA, Chew TL, Malik AB, Karginov AV. Time-Variant SRC Kinase Activation Determines Endothelial Permeability Response. Cell Chem Biol 2019; 26:1081-1094.e6. [PMID: 31130521 DOI: 10.1016/j.chembiol.2019.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/25/2019] [Accepted: 04/05/2019] [Indexed: 12/31/2022]
Abstract
In the current model of endothelial barrier regulation, the tyrosine kinase SRC is purported to induce disassembly of endothelial adherens junctions (AJs) via phosphorylation of VE cadherin, and thereby increase junctional permeability. Here, using a chemical biology approach to temporally control SRC activation, we show that SRC exerts distinct time-variant effects on the endothelial barrier. We discovered that the immediate effect of SRC activation was to transiently enhance endothelial barrier function as the result of accumulation of VE cadherin at AJs and formation of morphologically distinct reticular AJs. Endothelial barrier enhancement via SRC required phosphorylation of VE cadherin at Y731. In contrast, prolonged SRC activation induced VE cadherin phosphorylation at Y685, resulting in increased endothelial permeability. Thus, time-variant SRC activation differentially phosphorylates VE cadherin and shapes AJs to fine-tune endothelial barrier function. Our work demonstrates important advantages of synthetic biology tools in dissecting complex signaling systems.
Collapse
Affiliation(s)
- Jennifer E Klomp
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Mark Shaaya
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Jacob Matsche
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Rima Rebiai
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Jesse S Aaron
- Advanced Imaging Center at Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Kerrie B Collins
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Vincent Huyot
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Annette M Gonzalez
- Department of Pathology, The Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - William A Muller
- Department of Pathology, The Feinberg School of Medicine at Northwestern University, Chicago, IL 60611, USA
| | - Teng-Leong Chew
- Advanced Imaging Center at Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Asrar B Malik
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | - Andrei V Karginov
- Department of Pharmacology, The University of Illinois College of Medicine, 835 S. Wolcott Avenue, Chicago, IL 60612, USA.
| |
Collapse
|
14
|
Wang L, Chung J, Gill SE, Mehta S. Quantification of adherens junction disruption and contiguous paracellular protein leak in human lung endothelial cells under septic conditions. Microcirculation 2019; 26:e12528. [PMID: 30636088 DOI: 10.1111/micc.12528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 12/12/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Sepsis is associated with dysfunction of MVEC resulting in organ edema and inflammation. VE-cadherin, a component of MVEC adherens junctions, may be disrupted in sepsis. However, the direct connection between individual MVEC VE-cadherin disruption and increased paracellular permeability is uncertain. METHODS Human pulmonary MVEC were cultured on a biotin matrix and treated with cytomix, as a model of sepsis, vs PBS. MVEC permeability was assessed by trans-MVEC monolayer leak of Oregon green 488-conjugated avidin, which bound subcellular biotin to localize sites of paracellular leak. Leak was correlated with individual cell-specific MVEC surface VE-cadherin continuity by fluorescence microscopy. RESULTS Cytomix treatment reduced total MVEC VE-cadherin density, disrupted surface VE-cadherin continuity, was associated with intercellular gap formation, and enhanced paracellular avidin leak. Cytomix-induced MVEC paracellular avidin leak was strongly correlated temporally and was highly contiguous with focal MVEC surface VE-cadherin disruption. Total cellular VE-cadherin density was less strongly correlated with MVEC paracellular avidin leak and individual cell-specific focal surface VE-cadherin discontinuity. CONCLUSIONS These data support a mechanistic link between septic human lung MVEC VE-cadherin disruption and contiguous paracellular protein leak, and will permit more detailed assessment of individual cell-specific mechanisms of septic MVEC barrier dysfunction.
Collapse
Affiliation(s)
- Lefeng Wang
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Division of Respirology, Western University, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada
| | - Justin Chung
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Division of Respirology, Western University, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada.,Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Sanjay Mehta
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Division of Respirology, Western University, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada
| |
Collapse
|
15
|
Dennhardt S, Finke KR, Huwiler A, Coldewey SM. Sphingosine-1-phosphate promotes barrier-stabilizing effects in human microvascular endothelial cells via AMPK-dependent mechanisms. Biochim Biophys Acta Mol Basis Dis 2019; 1865:774-781. [PMID: 30660683 DOI: 10.1016/j.bbadis.2018.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 01/01/2023]
Abstract
Breakdown of the endothelial barrier is a critical step in the development of organ failure in severe inflammatory conditions such as sepsis. Endothelial cells from different tissues show phenotypic variations which are often neglected in endothelial research. Sphingosine-1-phosphate (S1P) and AMP-dependent kinase (AMPK) have been shown to protect the endothelium and phosphorylation of AMPK by S1P was shown in several cell types. However, the role of the S1P-AMPK interrelationship for endothelial barrier stabilization has not been investigated. To assess the role of the S1P-AMPK signalling axis in this context, we established an in vitro model allowing real-time monitoring of endothelial barrier function in human microvascular endothelial cells (HMEC-1) and murine glomerular endothelial cells (GENCs) with the electric cell-substrate impedance sensing (ECIS™) system. Following the disruption of the cell barrier by co-administration of LPS, TNF-α, IL-1ß, IFN-γ, and IL-6, we demonstrated self-recovery of the disrupted barrier in HMEC-1, while the barrier remained compromised in GENCs. Under physiological conditions we observed a rapid phosphorylation of AMPK in HMEC-1 stimulated with S1P, but not in GENCs. Consistently, S1P enhanced the basal endothelial barrier in HMEC-1 exclusively. siRNA-mediated knockdown of AMPK in HMEC-1 led to a less pronounced barrier enhancement. Thus we present evidence for a functional role of AMPK in S1P-mediated barrier stabilization in HMEC-1 and we provide insight into cell-type specific differences of the S1P-AMPK-interrelationship, which might influence the development of interventional strategies targeting endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Sophie Dennhardt
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Septomics Research Centre, Jena University Hospital, Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Karl R Finke
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, Bern, Switzerland
| | - Sina M Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Septomics Research Centre, Jena University Hospital, Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany.
| |
Collapse
|
16
|
Fong LY, Ng CT, Yong YK, Hakim MN, Ahmad Z. Asiatic acid stabilizes cytoskeletal proteins and prevents TNF-α-induced disorganization of cell-cell junctions in human aortic endothelial cells. Vascul Pharmacol 2018; 117:15-26. [PMID: 30114509 DOI: 10.1016/j.vph.2018.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/12/2018] [Accepted: 08/11/2018] [Indexed: 12/22/2022]
Abstract
Endothelial hyperpermeability represents an initiating step in early atherosclerosis and it often occurs as a result of endothelial barrier dysfunction. Asiatic acid, a major triterpene isolated from Centella asiatica (L.) Urban, has previously been demonstrated to protect against tumor necrosis factor (TNF)-α-induced endothelial barrier dysfunction. The present study aimed to investigate the mechanisms underlying the barrier protective effect of asiatic acid in human aortic endothelial cells (HAECs). The localization of F-actin, diphosphorylated myosin light chain (diphospho-MLC), adherens junctions (AJs) and tight junctions (TJs) was studied using immunocytochemistry techniques and confocal microscopy. Their total protein expressions were examined using western blot analysis. The endothelial permeability was assessed using In Vitro Vascular Permeability Assay kits. In addition, intracellular redistribution of the junctional proteins was evaluated using subcellular fractionation kits. We show that asiatic acid stabilized F-actin and diphospho-MLC at the cell periphery and prevented their rearrangement stimulated by TNF-α. However, asiatic acid failed to attenuate cytochalasin D-induced increased permeability. Besides, asiatic acid abrogated TNF-α-induced structural reorganization of vascular endothelial (VE)-cadherin and β-catenin by preserving their reticulum structures at cell-cell contact areas. In addition, asiatic acid also inhibited TNF-α-induced redistribution of occludin and zona occludens (ZO)-1 in different subcellular fractions. In conclusion, the barrier-stabilizing effect of asiatic acid might be associated with preservation of AJs and prevention of TJ redistribution caused by TNF-α. This study provides evidence to support the potential use of asiatic acid in the prevention of early atherosclerosis, which is initiated by endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Lai Yen Fong
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Department of Pre-clinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor, Malaysia.
| | - Chin Theng Ng
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia; Physiology Unit, Faculty of Medicine, AIMST University, 08100 Bedong, Kedah, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Muhammad Nazrul Hakim
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Zuraini Ahmad
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
17
|
Chang F, Flavahan S, Flavahan NA. Superoxide inhibition restores endothelium-dependent dilatation in aging arteries by enhancing impaired adherens junctions. Am J Physiol Heart Circ Physiol 2018; 314:H805-H811. [PMID: 29351453 DOI: 10.1152/ajpheart.00681.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Endothelium-dependent, nitric oxide-mediated dilatation is impaired in aging arteries. The dysfunction reflects increased production of reactive oxygen species (ROS), is reversed by inhibiting superoxide with superoxide dismutase (SOD) mimics, and is assumed to reflect superoxide-mediated inactivation of nitric oxide. However, the dysfunction also reflects Src-dependent degradation and loss of vascular-endothelial (VE)-cadherin from adherens junctions, resulting in a selective impairment in the ability of the junctions to amplify endothelial dilatation. Experiments therefore tested the hypothesis that SOD mimics might restore endothelial dilation in aging arteries by inhibiting Src and protecting endothelial adherens junctions. Tail arteries from young and aging Fisher 344 rats were processed for functional (pressure myograph), biochemical (immunoblot), and morphological (immunofluorescence) analyses. Cell-permeable SOD mimics [manganese(III) tetrakis(1-methyl-4-pyridyl)porphyrin (MnTMPyP) or tempol] did not affect acetylcholine-induced dilatation in young arteries but increased responses and restored normal dilator function in aging arteries. In aging arteries, MnTMPyP decreased Src activity (immunoblots of Tyr416 phosphorylated compared with total Src), increased the intensity and width of VE-cadherin staining at endothelial junctions, and increased VE-cadherin levels in Triton X-100-insoluble lysates, which represents the junctional protein. Because of aging-induced junctional disruption, inhibiting VE-cadherin clustering at adherens junctions with a function-blocking antibody does not affect acetylcholine-induced dilatation in aging arteries. However, the antibody prevented SOD mimics from restoring acetylcholine-induced dilatation in aging arteries. Therefore, SOD mimics improve impaired adherens junctions in aging endothelium, which is essential for SOD mimics to restore endothelium-dependent dilatation in aging arteries. The results suggest an important new pathological role for ROS in aging endothelium, namely, disruption of adherens junctions. NEW & NOTEWORTHY Aging-induced endothelial dysfunction is reversed by SOD mimics. This study demonstrates that they improve impaired adherens junctions in aging endothelium and that their restoration of endothelial dilatation is dependent on increased junctional activity. The results suggest a novel role for oxygen radicals in vascular aging, namely, disruption of adherens junctions.
Collapse
Affiliation(s)
- Fumin Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Sheila Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| | - Nicholas A Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University , Baltimore, Maryland
| |
Collapse
|
18
|
Shepherd J, Fisher M, Welford A, McDonald DM, Kanthou C, Tozer GM. The protective role of sphingosine-1-phosphate against the action of the vascular disrupting agent combretastatin A-4 3- O-phosphate. Oncotarget 2017; 8:95648-95661. [PMID: 29221156 PMCID: PMC5707050 DOI: 10.18632/oncotarget.21172] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
Solid tumours vary in sensitivity to the vascular disrupting agent combretastatin A-4 3-O-phosphate (CA4P), but underlying factors are poorly understood. The signaling sphingolipid, sphingosine-1-phosphate (S1P), promotes vascular barrier integrity by promoting assembly of VE-cadherin/β-catenin complexes. We tested the hypothesis that tumour pre-treatment with S1P would render tumours less susceptible to CA4P. S1P (1μM) pretreatment attenuated an increase in endothelial cell (HUVEC) monolayer permeability induced by 10μM CA4P. Intravenously administered S1P (8mg/kg/hr for 20 minutes then 2mg/kg/hr for 40 minutes), reduced CA4P-induced (30mg/kg) blood flow shut-down in fibrosarcoma tumours in SCID mice (n≥7 per group), as measured by tumour retention of an intravenously administered fluorescent lectin. A trend towards in vivo protection was also found using laser Doppler flowmetry. Immunohistochemical staining of tumours ex vivo revealed disrupted patterns of VE-cadherin in vasculature of mice treated with CA4P, which were decreased by pretreatment with S1P. S1P treatment also stabilized N-cadherin junctions between endothelial cells and smooth muscle cells in culture, and stabilized tubulin filaments in HUVEC monolayers. We conclude that the rapid shutdown of tumour microvasculature by CA4P is due in part to disruption of adherens junctions and that S1P has a protective effect on both adherens junctions and the endothelial cell cytoskeleton.
Collapse
Affiliation(s)
- Joanna Shepherd
- Current/Present address: School of Clinical Dentistry, The University of Sheffield, Claremont Crescent, Sheffield, UK
| | - Matthew Fisher
- Tumour Microcirculation Group, The University of Sheffield, Department of Oncology and Metabolism, School of Medicine, Sheffield, UK
| | - Abigail Welford
- Tumour Microcirculation Group, The University of Sheffield, Department of Oncology and Metabolism, School of Medicine, Sheffield, UK
| | - Donald M McDonald
- UCSF Comprehensive Cancer Center, Cardiovascular Research Institute, and Department of Anatomy, University of California, San Francisco, CA, USA
| | - Chryso Kanthou
- Tumour Microcirculation Group, The University of Sheffield, Department of Oncology and Metabolism, School of Medicine, Sheffield, UK
| | - Gillian M Tozer
- Tumour Microcirculation Group, The University of Sheffield, Department of Oncology and Metabolism, School of Medicine, Sheffield, UK
| |
Collapse
|
19
|
Chang F, Flavahan S, Flavahan NA. Impaired activity of adherens junctions contributes to endothelial dilator dysfunction in ageing rat arteries. J Physiol 2017; 595:5143-5158. [PMID: 28561330 DOI: 10.1113/jp274189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022] Open
Abstract
KEY POINTS Ageing-induced endothelial dysfunction contributes to organ dysfunction and progression of cardiovascular disease. VE-cadherin clustering at adherens junctions promotes protective endothelial functions, including endothelium-dependent dilatation. Ageing increased internalization and degradation of VE-cadherin, resulting in impaired activity of adherens junctions. Inhibition of VE-cadherin clustering at adherens junctions (function-blocking antibody; FBA) reduced endothelial dilatation in young arteries but did not affect the already impaired dilatation in old arteries. After junctional disruption with the FBA, dilatation was similar in young and old arteries. Src tyrosine kinase activity and tyrosine phosphorylation of VE-cadherin were increased in old arteries. Src inhibition increased VE-cadherin at adherens junctions and increased endothelial dilatation in old, but not young, arteries. Src inhibition did not increase dilatation in old arteries treated with the VE-cadherin FBA. Ageing impairs the activity of adherens junctions, which contributes to endothelial dilator dysfunction. Restoring the activity of adherens junctions could be of therapeutic benefit in vascular ageing. ABSTRACT Endothelial dilator dysfunction contributes to pathological vascular ageing. Experiments assessed whether altered activity of endothelial adherens junctions (AJs) might contribute to this dysfunction. Aortas and tail arteries were isolated from young (3-4 months) and old (22-24 months) F344 rats. VE-cadherin immunofluorescent staining at endothelial AJs and AJ width were reduced in old compared to young arteries. A 140 kDa VE-cadherin species was present on the cell surface and in TTX-insoluble fractions, consistent with junctional localization. Levels of the 140 kDa VE-cadherin were decreased, whereas levels of a TTX-soluble 115 kDa VE-cadherin species were increased in old compared to young arteries. Acetylcholine caused endothelium-dependent dilatation that was decreased in old compared to young arteries. Disruption of VE-cadherin clustering at AJs (function-blocking antibody, FBA) inhibited dilatation to acetylcholine in young, but not old, arteries. After the FBA, there was no longer any difference in dilatation between old and young arteries. Src activity and tyrosine phosphorylation of VE-cadherin were increased in old compared to young arteries. In old arteries, Src inhibition (saracatinib) increased: (i) 140 kDa VE-cadherin in the TTX-insoluble fraction, (ii) VE-cadherin intensity at AJs, (iii) AJ width, and (iv) acetylcholine dilatation. In old arteries treated with the FBA, saracatinib no longer increased acetylcholine dilatation. Saracatinib did not affect dilatation in young arteries. Therefore, ageing impairs AJ activity, which appears to reflect Src-induced phosphorylation, internalization and degradation of VE-cadherin. Moreover, impaired AJ activity can account for the endothelial dilator dysfunction in old arteries. Restoring endothelial AJ activity may be a novel therapeutic approach to vascular ageing.
Collapse
Affiliation(s)
- Fumin Chang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sheila Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas A Flavahan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
20
|
Zhou H, Hu S, Jin Q, Shi C, Zhang Y, Zhu P, Ma Q, Tian F, Chen Y. Mff-Dependent Mitochondrial Fission Contributes to the Pathogenesis of Cardiac Microvasculature Ischemia/Reperfusion Injury via Induction of mROS-Mediated Cardiolipin Oxidation and HK2/VDAC1 Disassociation-Involved mPTP Opening. J Am Heart Assoc 2017; 6:JAHA.116.005328. [PMID: 28288978 PMCID: PMC5524036 DOI: 10.1161/jaha.116.005328] [Citation(s) in RCA: 242] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Background The cardiac microvascular system ischemia/reperfusion injury following percutaneous coronary intervention is a clinical thorny problem. This study explores the mechanisms by which ischemia/reperfusion injury induces cardiac microcirculation collapse. Methods and Results In wild‐type mice, mitochondrial fission factor (Mff) expression increased in response to acute microvascular ischemia/reperfusion injury. Compared with wild‐type mice, homozygous Mff‐deficient (Mffgt) mice exhibited a smaller infarcted area, restored cardiac function, improved blood flow, and reduced microcirculation perfusion defects. Histopathology analysis demonstrated that cardiac microcirculation endothelial cells (CMECs) in Mffgt mice had an intact endothelial barrier, recovered phospho‐endothelial nitric oxide synthase production, opened lumen, undivided mitochondrial structures, and less CMEC death. In vitro, Mff‐deficient CMECs (derived from Mffgt mice or Mff small interfering RNA–treated) demonstrated less mitochondrial fission and mitochondrial‐dependent apoptosis compared with cells derived from wild‐type mice. The loss of Mff inhibited mitochondrial permeability transition pore opening via blocking the oligomerization of voltage‐dependent anion channel 1 and subsequent hexokinase 2 separation from mitochondria. Moreover, Mff deficiency reduced the cyt‐c leakage into the cytoplasm by alleviating cardiolipin oxidation resulting from damage to the electron transport chain complexes and mitochondrial reactive oxygen species overproduction. Conclusions This evidence clearly illustrates that microcirculatory ischemia/reperfusion injury can be attributed to Mff‐dependent mitochondrial fission via voltage‐dependent anion channel 1/hexokinase 2–mediated mitochondrial permeability transition pore opening and mitochondrial reactive oxygen species/cardiolipin involved cyt‐c release.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qinhua Jin
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Chen Shi
- Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Qiang Ma
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
21
|
Abstract
Sphingosine-1-phosphate (S1P), a simple, bioactive sphingolipid metabolite, plays a key role, both intracellularly and extracellularly, in various cellular processes such as proliferation, survival, migration, inflammation, angiogenesis, and endothelial barrier integrity. The cellular S1P level is low and is tightly regulated by its synthesis and degradation. Sphingosine Kinases (SphKs) 1 and 2, catalyze the ATP-dependent phosphorylation of sphingosine to S1P, while the degradation is mediated by the reversible dephosphorylation catalyzed by the S1P phosphatases and lipid phosphate phosphatases and the irreversible degradation to hexadecenal and ethanolamine phosphate by sphingosine-1-phosphate lyase (S1PL). As a ligand for specific G-protein-coupled receptors, S1P1-5, which are differentially expressed in different cell types, S1P generates downstream signals that play crucial role in developmental and disease related pathologies. In addition to acting extracellularly on receptors located on the plasma membrane, S1P can also act intracellularly, independently of S1P1-5, affecting calcium homeostasis and cell proliferation. The SphKs /S1P /S1PL metabolic pathway is implicated in numerous human pathologies including respiratory disorders, thereby raising the possibility that manipulating intracellular S1P levels could offer therapeutic potential in ameliorating lung diseases. This review focuses on the prospects of targeting S1P signaling and S1P metabolizing enzymes using small molecule inhibitors, receptor agonists, and antagonists in the treatment of lung diseases.
Collapse
Affiliation(s)
- David L Ebenezer
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, IL, USA
| | - Panfeng Fu
- Department of Pharmacology, University of Illinois at Chicago, IL, USA
| | - Viswanathan Natarajan
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, IL, USA; Department of Pharmacology, University of Illinois at Chicago, IL, USA; Department of Medicine, University of Illinois at Chicago, IL, USA; Department of Bioengineering, University of Illinois at Chicago, IL, USA.
| |
Collapse
|
22
|
Lung remodeling associated with recovery from acute lung injury. Cell Tissue Res 2016; 367:495-509. [DOI: 10.1007/s00441-016-2521-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022]
|
23
|
Tsuneki M, Madri JA. CD44 Influences Fibroblast Behaviors Via Modulation of Cell-Cell and Cell-Matrix Interactions, Affecting Survivin and Hippo Pathways. J Cell Physiol 2016; 231:731-43. [PMID: 26248063 DOI: 10.1002/jcp.25123] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/04/2015] [Indexed: 12/17/2022]
Abstract
CD44 has been studied in a wide variety of cell types, in a diverse array of cell behaviors and in a diverse range of signaling pathways. We now document a role for CD44 in mediating fibroblast behaviors via regulation of N-cadherin, extracellular matrix expression, Survivin and the Hippo pathway. Here, we report our findings on the roles of CD44 in modulating proliferation, apoptosis, migration and invasion of murine wild-type (WT-FB) and CD44 knockout dermal fibroblasts (CD44KO-FB). As we have documented in microvascular endothelial cells lacking CD44, we found persistent increased proliferation, reduced activation of cleaved caspase 3, increased initial attachment, but decreased strength of cell attachment in high cell density, post confluent CD44KO-FB cultures. Additionally, we found that siRNA knock-down of CD44 mimicked the behaviors of CD44KO-FB, restoring the decreases in N-cadherin, collagen type I, fibronectin, Survivin, nuclear fractions of YAP and phospho-YAP and decreased levels of cleaved caspase 3 to the levels observed in CD44KO-FB. Interestingly, plating CD44KO-FB on collagen type I or fibronectin resulted in significant decreases in secondary proliferation rates compared to plating cells on non-coated dishes, consistent with increased cell adhesion compared to their effects on WT-FB. Lastly, siRNA knockdown of CD44 in WT-FB resulted in increased fibroblast migration compared to WT-FB, albeit at reduced rates compared to CD44KO-FB. These results are consistent with CD44's pivotal role in modulating several diverse behaviors important for adhesion, proliferation, apoptosis, migration and invasion during development, growth, repair, maintenance and regression of a wide variety of mesenchymal tissues.
Collapse
Affiliation(s)
- Masayuki Tsuneki
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut.,Division of Cancer Biology, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Joseph A Madri
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
24
|
Birukova AA, Shah AS, Tian Y, Moldobaeva N, Birukov KG. Dual role of vinculin in barrier-disruptive and barrier-enhancing endothelial cell responses. Cell Signal 2016; 28:541-51. [PMID: 26923917 DOI: 10.1016/j.cellsig.2016.02.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 02/15/2016] [Accepted: 02/21/2016] [Indexed: 01/31/2023]
Abstract
Endothelial cell (EC) barrier disruption induced by edemagenic agonists such as thrombin is a result of increased actomyosin contraction and enforcement of focal adhesions (FA) anchoring contracting stress fibers, which leads to cell retraction and force-induced disruption of cell junctions. In turn, EC barrier enhancement by oxidized phospholipids (OxPAPC) and other agonists is a result of increased tethering forces due to enforcement of the peripheral actin rim and enhancement of cell-cell adherens junction (AJ) complexes promoting EC barrier integrity. This study tested participation of the mechanosensitive adaptor, vinculin, which couples FA and AJ to actin cytoskeleton, in control of the EC permeability response to barrier disruptive (thrombin) and barrier enhancing (OxPAPC) stimulation. OxPAPC and thrombin induced different patterns of FA remodeling. Knockdown of vinculin attenuated both, OxPAPC-induced decrease and thrombin-induced increase in EC permeability. Thrombin stimulated the vinculin association with FA protein talin and suppressed the interaction with AJ protein, VE-cadherin. In contrast, OxPAPC stimulated the vinculin association with VE-cadherin. Thrombin and OxPAPC induced different levels of myosin light chain (MLC) phosphorylation and caused different patterns of intracellular phospho-MLC distribution. Thrombin-induced talin-vinculin and OxPAPC-induced VE-cadherin-vinculin association were abolished by myosin inhibitor blebbistatin. Expression of the vinculin mutant unable to interact with actin attenuated EC permeability changes and MLC phosphorylation caused by both, thrombin and OxPAPC. These data suggest that the specific vinculin interaction with FA or AJ in different contexts of agonist stimulation is defined by development of regional actyomyosin-based tension and participates in both, the barrier-disruptive and barrier-enhancing endothelial responses.
Collapse
Affiliation(s)
- Anna A Birukova
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Alok S Shah
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Yufeng Tian
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Nurgul Moldobaeva
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
25
|
van Buul JD, Timmerman I. Small Rho GTPase-mediated actin dynamics at endothelial adherens junctions. Small GTPases 2016; 7:21-31. [PMID: 26825121 DOI: 10.1080/21541248.2015.1131802] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
VE-cadherin-based cell-cell junctions form the major restrictive barrier of the endothelium to plasma proteins and blood cells. The function of VE-cadherin and the actin cytoskeleton are intimately linked. Vascular permeability factors and adherent leukocytes signal through small Rho GTPases to tightly regulate actin cytoskeletal rearrangements in order to open and re-assemble endothelial cell-cell junctions in a rapid and controlled manner. The Rho GTPases are activated by guanine nucleotide exchange factors (GEFs), conferring specificity and context-dependent control of cell-cell junctions. Although the molecular mechanisms that couple cadherins to actin filaments are beginning to be elucidated, specific stimulus-dependent regulation of the actin cytoskeleton at VE-cadherin-based junctions remains unexplained. Accumulating evidence has suggested that depending on the vascular permeability factor and on the subcellular localization of GEFs, cell-cell junction dynamics and organization are differentially regulated by one specific Rho GTPase. In this Commentary, we focus on new insights how the junctional actin cytoskeleton is specifically and locally regulated by Rho GTPases and GEFs in the endothelium.
Collapse
Affiliation(s)
- Jaap D van Buul
- a Department of Molecular Cell Biology , Sanquin Research and Landsteiner Laboratory, Academic Medical Center Amsterdam, University of Amsterdam , Amsterdam , the Netherlands
| | - Ilse Timmerman
- b Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory , Academic Medical Center Amsterdam, University of Amsterdam , Amsterdam , the Netherlands
| |
Collapse
|
26
|
Visualization of adherent cell monolayers by cryo-electron microscopy: A snapshot of endothelial adherens junctions. J Struct Biol 2015; 192:470-477. [DOI: 10.1016/j.jsb.2015.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/08/2015] [Accepted: 10/09/2015] [Indexed: 01/05/2023]
|
27
|
Krishnan R, Park JA, Seow CY, Lee PVS, Stewart AG. Cellular Biomechanics in Drug Screening and Evaluation: Mechanopharmacology. Trends Pharmacol Sci 2015; 37:87-100. [PMID: 26651416 DOI: 10.1016/j.tips.2015.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/12/2015] [Accepted: 10/23/2015] [Indexed: 12/14/2022]
Abstract
The study of mechanobiology is now widespread. The impact of cell and tissue mechanics on cellular responses is well appreciated. However, knowledge of the impact of cell and tissue mechanics on pharmacological responsiveness, and its application to drug screening and mechanistic investigations, have been very limited in scope. We emphasize the need for a heightened awareness of the important bidirectional influence of drugs and biomechanics in all living systems. We propose that the term 'mechanopharmacology' be applied to approaches that employ in vitro systems, biomechanically appropriate to the relevant (patho)physiology, to identify new drugs and drug targets. This article describes the models and techniques that are being developed to transform drug screening and evaluation, ranging from a 2D environment to the dynamic 3D environment of the target expressed in the disease of interest.
Collapse
Affiliation(s)
- Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jin-Ah Park
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chun Y Seow
- Center for Heart Lung Innovation, St Pauls Hospital, University of British Columbia, Vancouver, Canada
| | - Peter V-S Lee
- Department of Mechanical Engineering, University of Melbourne, Melbourne, Australia
| | - Alastair G Stewart
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
28
|
Regulation of Endothelial Adherens Junctions by Tyrosine Phosphorylation. Mediators Inflamm 2015; 2015:272858. [PMID: 26556953 PMCID: PMC4628659 DOI: 10.1155/2015/272858] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/16/2015] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells form a semipermeable, regulated barrier that limits the passage of fluid, small molecules, and leukocytes between the bloodstream and the surrounding tissues. The adherens junction, a major mechanism of intercellular adhesion, is comprised of transmembrane cadherins forming homotypic interactions between adjacent cells and associated cytoplasmic catenins linking the cadherins to the cytoskeleton. Inflammatory conditions promote the disassembly of the adherens junction and a loss of intercellular adhesion, creating openings or gaps in the endothelium through which small molecules diffuse and leukocytes transmigrate. Tyrosine kinase signaling has emerged as a central regulator of the inflammatory response, partly through direct phosphorylation and dephosphorylation of the adherens junction components. This review discusses the findings that support and those that argue against a direct effect of cadherin and catenin phosphorylation in the disassembly of the adherens junction. Recent findings indicate a complex interaction between kinases, phosphatases, and the adherens junction components that allow a fine regulation of the endothelial permeability to small molecules, leukocyte migration, and barrier resealing.
Collapse
|
29
|
Chichger H, Braza J, Duong H, Harrington EO. SH2 domain-containing protein tyrosine phosphatase 2 and focal adhesion kinase protein interactions regulate pulmonary endothelium barrier function. Am J Respir Cell Mol Biol 2015; 52:695-707. [PMID: 25317600 DOI: 10.1165/rcmb.2013-0489oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Enhanced protein tyrosine phosphorylation is associated with changes in vascular permeability through formation and dissolution of adherens junctions and regulation of stress fiber formation. Inhibition of the protein tyrosine phosphorylase SH2 domain-containing protein tyrosine phosphatase 2 (SHP2) increases tyrosine phosphorylation of vascular endothelial cadherin and β-catenin, resulting in disruption of the endothelial monolayer and edema formation in the pulmonary endothelium. Vascular permeability is a hallmark of acute lung injury (ALI); thus, enhanced SHP2 activity offers potential therapeutic value for the pulmonary vasculature in diseases such as ALI, but this has not been characterized. To assess whether SHP2 activity mediates protection against edema in the endothelium, we assessed the effect of molecular activation of SHP2 on lung endothelial barrier function in response to the edemagenic agents LPS and thrombin. Both LPS and thrombin reduced SHP2 activity, correlated with decreased focal adhesion kinase (FAK) phosphorylation (Y(397) and Y(925)) and diminished SHP2 protein-protein associations with FAK. Overexpression of constitutively active SHP2 (SHP2(D61A)) enhanced baseline endothelial monolayer resistance and completely blocked LPS- and thrombin-induced permeability in vitro and significantly blunted pulmonary edema formation induced by either endotoxin (LPS) or Pseudomonas aeruginosa exposure in vivo. Chemical inhibition of FAK decreased SHP2 protein-protein interactions with FAK concomitant with increased permeability; however, overexpression of SHP2(D61A) rescued the endothelium and maintained FAK activity and FAK-SHP2 protein interactions. Our data suggest that SHP2 activation offers the pulmonary endothelium protection against barrier permeability mediators downstream of the FAK signaling pathway. We postulate that further studies into the promotion of SHP2 activation in the pulmonary endothelium may offer a therapeutic approach for patients suffering from ALI.
Collapse
Affiliation(s)
- Havovi Chichger
- 1 Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island; and
| | | | | | | |
Collapse
|
30
|
Mooren OL, Kim J, Li J, Cooper JA. Role of N-WASP in Endothelial Monolayer Formation and Integrity. J Biol Chem 2015; 290:18796-805. [PMID: 26070569 DOI: 10.1074/jbc.m115.668285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 11/06/2022] Open
Abstract
Endothelial cells (ECs) form a monolayer that serves as a barrier between the blood and the underlying tissue. ECs tightly regulate their cell-cell junctions, controlling the passage of soluble materials and immune cells across the monolayer barrier. We studied the role of N-WASP, a key regulator of Arp2/3 complex and actin assembly, in EC monolayers. We report that N-WASP regulates endothelial monolayer integrity by affecting the organization of cell junctions. Depletion of N-WASP resulted in an increase in transendothelial electrical resistance, a measure of monolayer integrity. N-WASP depletion increased the width of cell-cell junctions and altered the organization of F-actin and VE-cadherin at junctions. N-WASP was not present at cell-cell junctions in monolayers under resting conditions, but it was recruited following treatment with sphingosine-1-phosphate. Taken together, our results reveal a novel role for N-WASP in remodeling EC junctions, which is critical for monolayer integrity and function.
Collapse
Affiliation(s)
- Olivia L Mooren
- From the Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| | - Joanna Kim
- From the Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| | - Jinmei Li
- From the Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| | - John A Cooper
- From the Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri 63110
| |
Collapse
|
31
|
Fu P, Usatyuk PV, Lele A, Harijith A, Gregorio CC, Garcia JGN, Salgia R, Natarajan V. c-Abl mediated tyrosine phosphorylation of paxillin regulates LPS-induced endothelial dysfunction and lung injury. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1025-38. [PMID: 25795725 PMCID: PMC4437005 DOI: 10.1152/ajplung.00306.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/19/2015] [Indexed: 01/11/2023] Open
Abstract
Paxillin is phosphorylated at multiple residues; however, the role of tyrosine phosphorylation of paxillin in endothelial barrier dysfunction and acute lung injury (ALI) remains unclear. We used siRNA and site-specific nonphosphorylable mutants of paxillin to abrogate the function of paxillin to determine its role in lung endothelial permeability and ALI. In vitro, lipopolysaccharide (LPS) challenge of human lung microvascular endothelial cells (HLMVECs) resulted in enhanced tyrosine phosphorylation of paxillin at Y31 and Y118 with no significant change in Y181 and significant barrier dysfunction. Knockdown of paxillin with siRNA attenuated LPS-induced endothelial barrier dysfunction and destabilization of VE-cadherin. LPS-induced paxillin phosphorylation at Y31 and Y118 was mediated by c-Abl tyrosine kinase, but not by Src and focal adhesion kinase. c-Abl siRNA significantly reduced LPS-induced endothelial barrier dysfunction. Transfection of HLMVECs with paxillin Y31F, Y118F, and Y31/118F double mutants mitigated LPS-induced barrier dysfunction and VE-cadherin destabilization. In vivo, the c-Abl inhibitor AG957 attenuated LPS-induced pulmonary permeability in mice. Together, these results suggest that c-Abl mediated tyrosine phosphorylation of paxillin at Y31 and Y118 regulates LPS-mediated pulmonary vascular permeability and injury.
Collapse
Affiliation(s)
- Panfeng Fu
- Department of Pharmacology, University of Illinois, Chicago, Illinois;
| | - Peter V Usatyuk
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - Abhishek Lele
- Department of Pharmacology, University of Illinois, Chicago, Illinois
| | - Anantha Harijith
- Department of Pediatrics, University of Illinois, Chicago, Illinois
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona College of Medicine, Tucson, Arizona; and
| | - Ravi Salgia
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois, Chicago, Illinois; Department of Medicine, College of Medicine, University of Illinois, Chicago, Illinois
| |
Collapse
|
32
|
Sukriti S, Tauseef M, Yazbeck P, Mehta D. Mechanisms regulating endothelial permeability. Pulm Circ 2015; 4:535-51. [PMID: 25610592 DOI: 10.1086/677356] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/03/2014] [Indexed: 12/26/2022] Open
Abstract
The endothelial monolayer partitioning underlying tissue from blood components in the vessel wall maintains tissue fluid balance and host defense through dynamically opening intercellular junctions. Edemagenic agonists disrupt endothelial barrier function by signaling the opening of the intercellular junctions leading to the formation of protein-rich edema in the interstitial tissue, a hallmark of tissue inflammation that, if left untreated, causes fatal diseases, such as acute respiratory distress syndrome. In this review, we discuss how intercellular junctions are maintained under normal conditions and after stimulation of endothelium with edemagenic agonists. We have focused on reviewing the new concepts dealing with the alteration of adherens junctions after inflammatory stimulus.
Collapse
Affiliation(s)
- Sukriti Sukriti
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Mohammad Tauseef
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Pascal Yazbeck
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Dolly Mehta
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
33
|
FTY720, a sphingosine-1-phosphate (S1P) receptor modulator, protects sinusoid endothelial cells from radiation injury in vitro. Hepatol Int 2014; 9:149-54. [PMID: 25788389 DOI: 10.1007/s12072-014-9590-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/20/2014] [Indexed: 01/28/2023]
Abstract
PURPOSE To determine whether FTY720, a sphingosine-1-phosphate (S1P) receptor modulator, would protect sinusoid endothelial cells (SECs) from radiation injury in vitro. MATERIALS AND METHODS The effect of FTY720 on the viability of irradiated human liver SECs were examined by MTT assay or FACS analysis. The effect of FTY720 on the survival of hepatocellular carcinoma cell line, HepG2 and McA-RH7777, were determined by clonogenic assays. The activation of Akt pathway was tested by western bolt. RESULTS FTY720 increases the survival of irradiated SECs; in contrast, it does not appear to be radioprotective of tumor cells. Furthermore, the activation of Akt pathway was confirmed in the protective effect of FTY720 on SECs. CONCLUSION These results suggest that FTY720 will be a potential therapeutic protector for the SEC apoptosis during RILD.
Collapse
|
34
|
Petridou NI, Skourides PA. FAK transduces extracellular forces that orient the mitotic spindle and control tissue morphogenesis. Nat Commun 2014; 5:5240. [PMID: 25341507 DOI: 10.1038/ncomms6240] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 09/11/2014] [Indexed: 01/08/2023] Open
Abstract
Spindle orientation is critical for proper morphogenesis of organs and tissues as well as for the maintenance of tissue morphology. Although significant progress has been made in understanding the mechanisms linking the cell cortex to the spindle and the well-documented role that extracellular forces play in spindle orientation, how such forces are transduced to the cortex remains poorly understood. Here we report that focal adhesion kinase (FAK) is necessary for correct spindle orientation and as a result, indispensable for proper epithelial morphogenesis in the vertebrate embryo. We show that FAK's role in spindle orientation is dependent on its ability to localize at focal adhesions and its interaction with paxillin, but is kinase activity independent. Finally, we present evidence that FAK is required for external force-induced spindle reorientation, suggesting that FAK's involvement in this process stems from a role in the transduction of external forces to the cell cortex.
Collapse
Affiliation(s)
- Nicoletta I Petridou
- Laboratory of Developmental Biology and BioImaging, Department of Biological Sciences, University of Cyprus, University Ave 1, Nicosia 2109, Cyprus
| | - Paris A Skourides
- Laboratory of Developmental Biology and BioImaging, Department of Biological Sciences, University of Cyprus, University Ave 1, Nicosia 2109, Cyprus
| |
Collapse
|
35
|
Wilkerson BA, Argraves KM. The role of sphingosine-1-phosphate in endothelial barrier function. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1841:1403-1412. [PMID: 25009123 PMCID: PMC4169319 DOI: 10.1016/j.bbalip.2014.06.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/19/2014] [Accepted: 06/26/2014] [Indexed: 02/08/2023]
Abstract
Loss of endothelial barrier function is implicated in the etiology of metastasis, atherosclerosis, sepsis and many other diseases. Studies suggest that sphingosine-1-phosphate (S1P), particularly HDL-bound S1P (HDL-S1P) is essential for endothelial barrier homeostasis and that HDL-S1P may be protective against the loss of endothelial barrier function in disease. This review summarizes evidence providing mechanistic insights into how S1P maintains endothelial barrier function, highlighting the recent findings that implicate the major S1P carrier, HDL, in the maintenance of the persistent S1P-signaling needed to maintain endothelial barrier function. We review the mechanisms proposed for HDL maintenance of persistent S1P-signaling, the evidence supporting these mechanisms and the remaining fundamental questions.
Collapse
Affiliation(s)
- Brent A Wilkerson
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave., BSB650, Charleston, SC 29425, USA
| | - Kelley M Argraves
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave., BSB650, Charleston, SC 29425, USA.
| |
Collapse
|
36
|
Gulino-Debrac D. Mechanotransduction at the basis of endothelial barrier function. Tissue Barriers 2014; 1:e24180. [PMID: 24665386 PMCID: PMC3879236 DOI: 10.4161/tisb.24180] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/01/2013] [Accepted: 03/02/2013] [Indexed: 01/03/2023] Open
Abstract
Destabilization of cell-cell contacts involved in the maintenance of endothelial barrier function can lead to increased endothelial permeability. This increase in endothelial permeability results in an anarchical movement of fluid, solutes and cells outside the vasculature and into the surrounding tissues, thereby contributing to various diseases such as stroke or pulmonary edema. Thus, a better understanding of the molecular mechanisms regulating endothelial cell junction integrity is required for developing new therapies for these diseases. In this review, we describe the mechanotransduction mechanism at the basis of adherens junction strengthening at endothelial cell-cell contacts. More particularly, we report on the emerging role of α-catenin and EPLIN that act as a mechanotransmitter of myosin-IIgenerated traction forces. The interplay between α-catenin, EPLIN and the myosin-II machinery initiates the junctional recruitment of vinculin and α-actinin leading to a drastic remodeling of the actin cytoskeleton and to cortical actin ring reshaping. The pathways initiated by tyrosine phosphorylation of VE-cadherin at the basis of endothelial cell-cell junction remodeling is also reported, as it may be interrelated to α-catenin/ EPLIN-mediated mechanotransduction mechanisms. We also describe the junctional mechanosensory complex composed of PECAM-1, VE-cadherin and VEGFR2 that is able to transmit signaling pathway under the onset of shear stress. This mechanosensing mechanism, involved in the earliest events promoting atherogenesis, is required for endothelial cell alignment along flow direction.
Collapse
Affiliation(s)
- Danielle Gulino-Debrac
- Biology of Cancer and Infection Laboratory; U INSERM 1036, iRTSV; Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA); Université Joseph Fourier; Grenoble, France
| |
Collapse
|
37
|
Abstract
Increased endothelial permeability and reduction of alveolar liquid clearance capacity are two leading pathogenic mechanisms of pulmonary edema, which is a major complication of acute lung injury, severe pneumonia, and acute respiratory distress syndrome, the pathologies characterized by unacceptably high rates of morbidity and mortality. Besides the success in protective ventilation strategies, no efficient pharmacological approaches exist to treat this devastating condition. Understanding of fundamental mechanisms involved in regulation of endothelial permeability is essential for development of barrier protective therapeutic strategies. Ongoing studies characterized specific barrier protective mechanisms and identified intracellular targets directly involved in regulation of endothelial permeability. Growing evidence suggests that, although each protective agonist triggers a unique pattern of signaling pathways, selected common mechanisms contributing to endothelial barrier protection may be shared by different barrier protective agents. Therefore, understanding of basic barrier protective mechanisms in pulmonary endothelium is essential for selection of optimal treatment of pulmonary edema of different etiology. This article focuses on mechanisms of lung vascular permeability, reviews major intracellular signaling cascades involved in endothelial monolayer barrier preservation and summarizes a current knowledge regarding recently identified compounds which either reduce pulmonary endothelial barrier disruption and hyperpermeability, or reverse preexisting lung vascular barrier compromise induced by pathologic insults.
Collapse
Affiliation(s)
- Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA.
| | | | | |
Collapse
|
38
|
Schmidt TT, Tauseef M, Yue L, Bonini MG, Gothert J, Shen TL, Guan JL, Predescu S, Sadikot R, Mehta D. Conditional deletion of FAK in mice endothelium disrupts lung vascular barrier function due to destabilization of RhoA and Rac1 activities. Am J Physiol Lung Cell Mol Physiol 2013; 305:L291-300. [PMID: 23771883 DOI: 10.1152/ajplung.00094.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Loss of lung-fluid homeostasis is the hallmark of acute lung injury (ALI). Association of catenins and actin cytoskeleton with vascular endothelial (VE)-cadherin is generally considered the main mechanism for stabilizing adherens junctions (AJs), thereby preventing disruption of lung vascular barrier function. The present study identifies endothelial focal adhesion kinase (FAK), a nonreceptor tyrosine kinase that canonically regulates focal adhesion turnover, as a novel AJ-stabilizing mechanism. In wild-type mice, induction of ALI by intraperitoneal administration of lipopolysaccharide or cecal ligation and puncture markedly decreased FAK expression in lungs. Using a mouse model in which FAK was conditionally deleted only in endothelial cells (ECs), we show that loss of EC-FAK mimicked key features of ALI (diffuse lung hemorrhage, increased transvascular albumin influx, edema, and neutrophil accumulation in the lung). EC-FAK deletion disrupted AJs due to impairment of the fine balance between the activities of RhoA and Rac1 GTPases. Deletion of EC-FAK facilitated RhoA's interaction with p115-RhoA guanine exchange factor, leading to activation of RhoA. Activated RhoA antagonized Rac1 activity, destabilizing AJs. Inhibition of Rho kinase, a downstream effector of RhoA, reinstated normal endothelial barrier function in FAK-/- ECs and lung vascular integrity in EC-FAK-/- mice. Our findings demonstrate that EC-FAK plays an essential role in maintaining AJs and thereby lung vascular barrier function by establishing the normal balance between RhoA and Rac1 activities.
Collapse
Affiliation(s)
- Tracy Thennes Schmidt
- Dept. of Pharmacology, The Univ. of Illinois, College of Medicine, 835 S. Wolcott Ave., Chicago, IL 60612.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Hardin C, Rajendran K, Manomohan G, Tambe DT, Butler JP, Fredberg JJ, Martinelli R, Carman CV, Krishnan R. Glassy dynamics, cell mechanics, and endothelial permeability. J Phys Chem B 2013; 117:12850-6. [PMID: 23638866 DOI: 10.1021/jp4020965] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
A key feature of all inflammatory processes is disruption of the vascular endothelial barrier. Such disruption is initiated in part through active contraction of the cytoskeleton of the endothelial cell (EC). Because contractile forces are propagated from cell to cell across a great many cell-cell junctions, this contractile process is strongly cooperative and highly nonlocal. We show here that the characteristic length scale of propagation is modulated by agonists and antagonists that impact permeability of the endothelial barrier. In the presence of agonists including thrombin, histamine, and H2O2, force correlation length increases, whereas in the presence of antagonists including sphingosine-1-phosphate, hepatocyte growth factor, and the rho kinase inhibitor, Y27632, force correlation length decreases. Intercellular force chains and force clusters are also evident, both of which are reminiscent of soft glassy materials approaching a glass transition.
Collapse
Affiliation(s)
- Corey Hardin
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital , Boston, Massachusetts
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Ritt M, Guan JL, Sivaramakrishnan S. Visualizing and manipulating focal adhesion kinase regulation in live cells. J Biol Chem 2013; 288:8875-86. [PMID: 23393139 DOI: 10.1074/jbc.m112.421164] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Focal Adhesion Kinase (FAK) is essential for cell migration and plays an important role in tumor metastasis. However, the complex intermolecular and intramolecular interactions that regulate FAK activity at the focal adhesion remain unresolved. We have engineered a toolbox of FRET sensors that retain all of the individual FAK domains but modulate a key intramolecular regulatory interaction between the band 4.1/ezrin/radixin/moesin (FERM) and kinase domains of FAK. We demonstrate systematic control and quantitative measurement of the FERM-kinase interaction at focal adhesions, which in turn allows us to control cell migration. Using these sensors, we find that Tyr-397 phosphorylation, rather than kinase activity of FAK, is the key determinant of cell migration. Our sensors directly demonstrate, for the first time, a pH-dependent change in a protein-protein interaction at a macromolecular structure in live cells. The FERM-kinase interaction at focal adhesions is enhanced at acidic pH, with a concomitant decrease in Tyr-397 phosphorylation, providing a potential mechanism for enhanced migration of cancer cells.
Collapse
Affiliation(s)
- Michael Ritt
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | |
Collapse
|
41
|
Goddard LM, Iruela-Arispe ML. Cellular and molecular regulation of vascular permeability. Thromb Haemost 2013; 109:407-15. [PMID: 23389236 DOI: 10.1160/th12-09-0678] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 01/30/2013] [Indexed: 02/07/2023]
Abstract
Vascular permeability is a highly coordinated process that integrates vesicular trafficking, complex junctional rearrangements, and refined cytoskeletal dynamics. In response to the extracellular environment, these three cellular activities have been previously assumed to work in parallel to regulate the passage of solutes between the blood and tissues. New developments in the area of vascular permeability, however have highlighted the interdependence between trans- and para-cellular pathways, the cross-communication between adherens and tight junctions, and the instructional role of pericytes on endothelial expression of barrier-related genes. Additionally, significant effort has been placed in understanding the molecular underpinings that contribute to barrier restoration following acute permeability events and in clarifying the importance of context-dependent signaling initiated by permeability mediators. Finally, recent findings have uncovered an unpredicted role for transcription factors in the coordination of vascular permeability and clarified how junctional complexes can transmit signals to the nucleus to control barrier function. The goal of this review is to provide a concise and updated view of vascular permeability, discuss the most recent advances in molecular and cellular regulation, and introduce integrated information on the central mechanisms involved in trans-endothelial transport.
Collapse
Affiliation(s)
- Lauren M Goddard
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA
| | | |
Collapse
|
42
|
Abbasi T, Garcia JGN. Sphingolipids in lung endothelial biology and regulation of vascular integrity. Handb Exp Pharmacol 2013:201-26. [PMID: 23563658 DOI: 10.1007/978-3-7091-1511-4_10] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Of the multiple and diverse homeostatic events that involve the lung vascular endothelium, participation in preserving vascular integrity and therefore organ function is paramount. We were the first to show that the lipid growth factor and angiogenic factor, sphingosine-1-phosphate, is a critical agonist involved in regulation of human lung vascular barrier function (Garcia et al. J Clin Invest, 2011). Utilizing both in vitro models and preclinical murine, rat, and canine models of acute and chronic inflammatory lung injury, we have shown that S1Ps, as well as multiple S1P analogues such as FTY720 and ftysiponate, serve as protective agents limiting the disruption of the vascular EC monolayer in the pulmonary microcirculation and attenuate parenchymal accumulation of inflammatory cells and high protein containing extravasated fluid, thereby reducing interstitial and alveolar edema. The vasculo-protective mechanism of these therapeutic effects occurs via ligation of specific G-protein-coupled receptors and an intricate interplay of S1P with other factors (such as MAPKS, ROCKs, Rho, Rac1) with rearrangement of the endothelial cytoskeleton to form strong cortical actin rings in the cell periphery and enhanced cell-to-cell and cell-to-matrix tethering dynamics. This cascade leads to reinforcement of focal adhesions and paracellular junctional complexes via cadherin, paxillin, catenins, and zona occludens. S1P through its interaction with Rac and Rho influences the cytoskeletal rearrangement indicated in the later stages of angiogenesis as a stabilizing force, preventing excessive vascular permeability. These properties translate into a therapeutic potential for acute and chronic inflammatory lung injuries. S1P has potential for providing a paradigm shift in the approach to disruption of critical endothelial gatekeeper function, loss of lung vascular integrity, and increased vascular permeability, defining features of acute lung injury (ALI), and may prove to exhibit an intrinsically protective role in the pulmonary vasculature ameliorating agonist- or sepsis-induced pulmonary injury and vascular leakage.
Collapse
Affiliation(s)
- Taimur Abbasi
- Department of Medicine, The University of Illinois, Chicago, IL 60612, USA
| | | |
Collapse
|
43
|
Parsons SA, Sharma R, Roccamatisi DL, Zhang H, Petri B, Kubes P, Colarusso P, Patel KD. Endothelial paxillin and focal adhesion kinase (FAK) play a critical role in neutrophil transmigration. Eur J Immunol 2012; 42:436-46. [PMID: 22095445 DOI: 10.1002/eji.201041303] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During an inflammatory response, endothelial cells undergo morphological changes to allow for the passage of neutrophils from the blood vessel to the site of injury or infection. Although endothelial cell junctions and the cytoskeleton undergo reorganization during inflammation, little is known about another class of cellular structures, the focal adhesions. In this study, we examined several focal adhesion proteins during an inflammatory response. We found that there was selective loss of paxillin and focal adhesion kinase (FAK) from focal adhesions in proximity to transmigrating neutrophils; in contrast the levels of the focal adhesion proteins β1-integrin and vinculin were unaffected. Paxillin was lost from focal adhesions during neutrophil transmigration both under static and flow conditions. Down-regulating endothelial paxillin with siRNA blocked neutrophil transmigration while having no effect on rolling or adhesion. As paxillin dynamics are regulated partly by FAK, the role of FAK in neutrophil transmigration was examined using two complementary methods. siRNA was used to down-regulate total FAK protein while dominant-negative, kinase-deficient FAK was expressed to block FAK signaling. Disruption of the FAK protein or FAK signaling decreased neutrophil transmigration. Collectively, these findings reveal a novel role for endothelial focal adhesion proteins paxillin and FAK in regulating neutrophil transmigration.
Collapse
Affiliation(s)
- Sean A Parsons
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Mascall KS, Small GR, Gibson G, Nixon GF. Sphingosine-1-phosphate-induced release of TIMP-2 from vascular smooth muscle cells inhibits angiogenesis. J Cell Sci 2012; 125:2267-75. [PMID: 22344262 DOI: 10.1242/jcs.099044] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Following myocardial infarction, angiogenesis occurs as a result of thrombus formation, which permits reperfusion of damaged myocardium. Sphingosine 1-phosphate (S1P) is a naturally occurring lipid mediator released from platelets and is found in high concentrations at sites of thrombosis. S1P might therefore be involved in regulating angiogenesis following myocardial infarction and might influence reperfusion. The aims of this study were to determine the effects of S1P in human coronary arterial cell angiogenesis and delineate the subsequent mechanisms. An in vitro model of angiogenesis was developed using a co-culture of human coronary artery endothelial cells, human coronary smooth muscle cells and human fibroblasts. In this model, S1P inhibited angiogenesis and this was dependent on the presence of smooth muscle cells. The mechanism of the inhibitory effect was through S1P-induced release of a soluble mediator from smooth muscle cells. This mediator was identified as tissue inhibitor of metalloproteinase-2 (TIMP-2). Release of TIMP-2 was dependent on S1P-induced activation of Rho kinase and directly contributed to incomplete formation of endothelial cell adherens junctions. This was observed as a diffuse localisation of VE-cadherin, leading to decreased tubulogenesis. A similar inhibitory response to S1P was demonstrated in an ex vivo human arterial model of angiogenesis. In summary, S1P-induced inhibition of angiogenesis in human artery endothelial cells is mediated by TIMP-2 from vascular smooth muscle cells. This reduces the integrity of intercellular junctions between nascent endothelial cells. S1P might therefore inhibit the angiogenic response following myocardial infarction.
Collapse
Affiliation(s)
- Keith S Mascall
- School of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | | | | |
Collapse
|
45
|
Shigemizu D, Hu Z, Hung JH, Huang CL, Wang Y, DeLisi C. Using functional signatures to identify repositioned drugs for breast, myelogenous leukemia and prostate cancer. PLoS Comput Biol 2012; 8:e1002347. [PMID: 22346740 PMCID: PMC3276504 DOI: 10.1371/journal.pcbi.1002347] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 11/25/2011] [Indexed: 01/21/2023] Open
Abstract
The cost and time to develop a drug continues to be a major barrier to widespread distribution of medication. Although the genomic revolution appears to have had little impact on this problem, and might even have exacerbated it because of the flood of additional and usually ineffective leads, the emergence of high throughput resources promises the possibility of rapid, reliable and systematic identification of approved drugs for originally unintended uses. In this paper we develop and apply a method for identifying such repositioned drug candidates against breast cancer, myelogenous leukemia and prostate cancer by looking for inverse correlations between the most perturbed gene expression levels in human cancer tissue and the most perturbed expression levels induced by bioactive compounds. The method uses variable gene signatures to identify bioactive compounds that modulate a given disease. This is in contrast to previous methods that use small and fixed signatures. This strategy is based on the observation that diseases stem from failed/modified cellular functions, irrespective of the particular genes that contribute to the function, i.e., this strategy targets the functional signatures for a given cancer. This function-based strategy broadens the search space for the effective drugs with an impressive hit rate. Among the 79, 94 and 88 candidate drugs for breast cancer, myelogenous leukemia and prostate cancer, 32%, 13% and 17% respectively are either FDA-approved/in-clinical-trial drugs, or drugs with suggestive literature evidences, with an FDR of 0.01. These findings indicate that the method presented here could lead to a substantial increase in efficiency in drug discovery and development, and has potential application for the personalized medicine. The effective drug of a given disease is aimed to bring abnormal functions associated with disease back to the normal state. Using expression profile as the surrogate marker of the cellular function, we introduce a novel procedure to identify candidate therapeutics by searching for those bioactive compounds that either down-regulate abnormally over-expressed genes, or up-regulate those that are abnormally under-expressed. We show that the approach detects a pool of plausible candidates as repositioning/new drugs. In contrast to previous studies, our approach uses a variable big number of genes and/or gene combinations as a representation of functional signatures to identify bioactive compounds that modulate a given disease, irrespective of the particular genes that contribute to the cellular functions; therefore it covers potential drugs with heterogeneous properties. The method may also have potential application for the personalized medicine.
Collapse
Affiliation(s)
- Daichi Shigemizu
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Zhenjun Hu
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Jui-Hung Hung
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Chia-Ling Huang
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
| | - Yajie Wang
- Laboratory of Clinical Medical Research, Department of Clinical Laboratory Diagnostics, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Charles DeLisi
- Bioinformatics Program, Boston University, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
46
|
DiPaolo BC, Margulies SS. Rho kinase signaling pathways during stretch in primary alveolar epithelia. Am J Physiol Lung Cell Mol Physiol 2012; 302:L992-1002. [PMID: 22287611 DOI: 10.1152/ajplung.00175.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Alveolar epithelial cells (AECs) maintain integrity of the blood-gas barrier with actin-anchored intercellular tight junctions. Stretched type I-like AECs undergo magnitude- and frequency-dependent actin cytoskeletal remodeling into perijunctional actin rings. On the basis of published studies in human pulmonary artery endothelial cells (HPAECs), we hypothesize that RhoA activity, Rho kinase (ROCK) activity, and phosphorylation of myosin light chain II (MLC2) increase in stretched type I-like AECs in a manner that is dependent on stretch magnitude, and that RhoA, ROCK, or MLC2 activity inhibition will attenuate stretch-induced actin remodeling and preserve barrier properties. Primary type I-like AEC monolayers were stretched biaxially to create a change in surface area (ΔSA) of 12%, 25%, or 37% in a cyclic manner at 0.25 Hz for up to 60 min or left unstretched. Type I-like AECs were also treated with Rho pathway inhibitors (ML-7, Y-27632, or blebbistatin) and stained for F-actin or treated with the myosin phosphatase inhibitor calyculin-A and quantified for monolayer permeability. Counter to our hypothesis, ROCK activity and MLC2 phosphorylation decreased in type I-like AECs stretched to 25% and 37% ΔSA and did not change in monolayers stretched to 12% ΔSA. Furthermore, RhoA activity decreased in type I-like AECs stretched to 37% ΔSA. In contrast, MLC2 phosphorylation in HPAECs increased when HPAECs were stretched to 12% ΔSA but then decreased when they were stretched to 37% ΔSA, similar to type I-like AECs. Perijunctional actin rings were observed in unstretched type I-like AECs treated with the Rho pathway inhibitor blebbistatin. Myosin phosphatase inhibition increased MLC2 phosphorylation in stretched type I-like AECs but had no effect on monolayer permeability. In summary, stretch alters RhoA activity, ROCK activity, and MLC2 phosphorylation in a manner dependent on stretch magnitude and cell type.
Collapse
Affiliation(s)
- Brian C DiPaolo
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd St., Philadelphia, PA 19104-6321, USA
| | | |
Collapse
|
47
|
George B, Verma R, Soofi AA, Garg P, Zhang J, Park TJ, Giardino L, Ryzhova L, Johnstone DB, Wong H, Nihalani D, Salant DJ, Hanks SK, Curran T, Rastaldi MP, Holzman LB. Crk1/2-dependent signaling is necessary for podocyte foot process spreading in mouse models of glomerular disease. J Clin Invest 2012; 122:674-92. [PMID: 22251701 DOI: 10.1172/jci60070] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 12/06/2011] [Indexed: 01/03/2023] Open
Abstract
The morphology of healthy podocyte foot processes is necessary for maintaining the characteristics of the kidney filtration barrier. In most forms of glomerular disease, abnormal filter barrier function results when podocytes undergo foot process spreading and retraction by remodeling their cytoskeletal architecture and intercellular junctions during a process known as effacement. The cell adhesion protein nephrin is necessary for establishing the morphology of the kidney podocyte in development by transducing from the specialized podocyte intercellular junction phosphorylation-mediated signals that regulate cytoskeletal dynamics. The present studies extend our understanding of nephrin function by showing that nephrin activation in cultured podocytes induced actin dynamics necessary for lamellipodial protrusion. This process required a PI3K-, Cas-, and Crk1/2-dependent signaling mechanism distinct from the previously described nephrin-Nck1/2 pathway necessary for assembly and polymerization of actin filaments. Our present findings also support the hypothesis that mechanisms governing lamellipodial protrusion in culture are similar to those used in vivo during foot process effacement in a subset of glomerular diseases. In mice, podocyte-specific deletion of Crk1/2 prevented foot process effacement in one model of podocyte injury and attenuated foot process effacement and associated proteinuria in a delayed fashion in a second model. In humans, focal adhesion kinase and Cas phosphorylation - markers of focal adhesion complex-mediated Crk-dependent signaling - was induced in minimal change disease and membranous nephropathy, but not focal segmental glomerulosclerosis. Together, these observations suggest that activation of a Cas-Crk1/2-dependent complex is necessary for foot process effacement observed in distinct subsets of human glomerular diseases.
Collapse
Affiliation(s)
- Britta George
- Renal-Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Usatyuk PV, Natarajan V. Hydroxyalkenals and oxidized phospholipids modulation of endothelial cytoskeleton, focal adhesion and adherens junction proteins in regulating endothelial barrier function. Microvasc Res 2012; 83:45-55. [PMID: 21570987 PMCID: PMC3196796 DOI: 10.1016/j.mvr.2011.04.012] [Citation(s) in RCA: 223] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 04/27/2011] [Accepted: 04/28/2011] [Indexed: 10/18/2022]
Abstract
Lipid peroxidation of polyunsaturated fatty acids generates bioactive aldehydes, which exhibit pro- and anti-inflammatory effects in cells and tissues. Accumulating evidence indicates that 4-hydroxynonenal (4-HNE), a major aldehyde derived from lipid peroxidation of n-6 polyunsaturated fatty acids trigger signals that modulates focal adhesion and adherens junction proteins thereby inducing endothelial barrier dysfunction. Similarly, oxidized phospholipids (Ox-PLs) generated by lipid peroxidation of phospholipids with polyunsaturated fatty acids have been implicated in atherogenesis, inflammation and gene expression. Interestingly, physiological concentration of Ox-PLs is anti-inflammatory and protect against endotoxin- and ventilator-associated acute lung injury. Thus, excess generation of bioactive hydroxyalkenals and Ox-PLs during oxidative stress contributes to pathophysiology of various diseases by modulating signaling pathways that regulate pro- and anti-inflammatory responses and barrier regulation. This review summarizes the role of 4-HNE and Ox-PLs affecting cell signaling pathways and endothelial barrier dysfunction through modulation of the activities of proteins/enzymes by Michael adducts formation, enhancing the level of protein tyrosine phosphorylation of the target proteins, and by reorganization of cytoskeletal, focal adhesion, and adherens junction proteins. A better understanding of molecular mechanisms of hydroxyalkenals- and Ox-PLs-mediated pro-and anti-inflammatory responses and barrier function may lead to development of novel therapies to ameliorate oxidative stress related cardio-pulmonary disorders.
Collapse
Affiliation(s)
- Peter V. Usatyuk
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
- Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, IL 60612
| | - Viswanathan Natarajan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612
- Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612
- Institute for Personalized Respiratory Medicine, University of Illinois at Chicago, Chicago, IL 60612
| |
Collapse
|
49
|
Schuchardt M, Tölle M, Prüfer J, van der Giet M. Pharmacological relevance and potential of sphingosine 1-phosphate in the vascular system. Br J Pharmacol 2011; 163:1140-62. [PMID: 21309759 DOI: 10.1111/j.1476-5381.2011.01260.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) was identified as a crucial molecule for regulating immune responses, inflammatory processes as well as influencing the cardiovascular system. S1P mediates differentiation, proliferation and migration during vascular development and homoeostasis. S1P is a naturally occurring lipid metabolite and is present in human blood in nanomolar concentrations. S1P is not only involved in physiological but also in pathophysiological processes. Therefore, this complex signalling system is potentially interesting for pharmacological intervention. Modulation of the system might influence inflammatory, angiogenic or vasoregulatory processes. S1P activates G-protein coupled receptors, namely S1P(1-5) , whereas only S1P(1-3) is present in vascular cells. S1P can also act as an intracellular signalling molecule. This review highlights the pharmacological potential of S1P signalling in the vascular system by giving an overview of S1P-mediated processes in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). After a short summary of S1P metabolism and signalling pathways, the role of S1P in EC and VSMC proliferation and migration, the cause of relaxation and constriction of arterial blood vessels, the protective functions on endothelial apoptosis, as well as the regulatory function in leukocyte adhesion and inflammatory responses are summarized. This is followed by a detailed description of currently known pharmacological agonists and antagonists as new tools for mediating S1P signalling in the vasculature. The variety of effects influenced by S1P provides plenty of therapeutic targets currently under investigation for potential pharmacological intervention.
Collapse
Affiliation(s)
- Mirjam Schuchardt
- Charité- Universitätsmedizin Berlin, CharitéCentrum 10, Department of Nephrology, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany
| | | | | | | |
Collapse
|
50
|
Kielczewski JL, Li Calzi S, Shaw LC, Cai J, Qi X, Ruan Q, Wu L, Liu L, Hu P, Chan-Ling T, Mames RN, Firth S, Baxter RC, Turowski P, Busik JV, Boulton ME, Grant MB. Free insulin-like growth factor binding protein-3 (IGFBP-3) reduces retinal vascular permeability in association with a reduction of acid sphingomyelinase (ASMase). Invest Ophthalmol Vis Sci 2011; 52:8278-86. [PMID: 21931131 DOI: 10.1167/iovs.11-8167] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
PURPOSE To examine the effect of free insulin-like growth factor (IGF) binding protein-3 (IGFBP-3), independent of the effect of insulin-like growth factors, in modulating retinal vascular permeability. METHODS We assessed the ability of a form of IGFBP-3 that does not bind IGF-1 (IGFBP-3NB), to regulate the blood retinal barrier (BRB) using two distinct experimental mouse models, laser-induced retinal vessel injury and vascular endothelial growth factor (VEGF)-induced retinal vascular permeability. Additionally, in vitro studies were conducted. In the animal models, BRB permeability was quantified by intravenous injection of fluorescein labeled serum albumin followed by digital confocal image analysis of retinal flat-mounts. Claudin-5 and vascular endothelial-cadherin (VE-cadherin) localization at interendothelial junctions was studied by immunofluorescence. In vitro changes in transendothelial electrical resistance (TEER) and flux of fluorescent dextran in bovine retinal endothelial monolayers (BREC) were measured after IGFBP-3NB treatment. Acid (ASMase) and neutral (NSMase) sphingomyelinase mRNA levels and activity were measured in mouse retinas. RESULTS Four days postinjury, laser-injured mouse retinas injected with IGFBP-3NB plasmid demonstrated reduced vascular permeability compared with retinas of laser-injured mouse retinas injected with control plasmid. IGFBP-3NB administration resulted in a significant decrease in laser injury-associated increases in ASMase and NSMase mRNA and activity when compared with laser alone treated mice. In vivo, intravitreal injection of IGFBP-3NB reduced vascular leakage associated with intravitreal VEGF injection. IGFBP-3NB partially restored VEGF-induced in vivo permeability and dissociation of claudin-5 and VE-cadherin at junctional complexes. When IGFBP-3NB was applied basally to bovine retinal endothelial cells (BREC) in vitro, TEER increased and macromolecular flux decreased. CONCLUSIONS Intravitreal administration of IGFBP-3NB preserves junctional integrity in the presence of VEGF or laser injury by reducing BRB permeability in part by modulating sphingomyelinase levels.
Collapse
Affiliation(s)
- Jennifer L Kielczewski
- Departments of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida 32610-0267, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|