1
|
Li X, Wang M, Wolfsgruber M, Klatt OC, Hollmann MW, Preckel B, Zuurbier CJ, Weber NC. Empagliflozin prevents TNF-α induced endothelial dysfunction under flow -the potential involvement of calcium and sodium-hydrogen exchanger. Eur J Pharmacol 2025; 986:177147. [PMID: 39571673 DOI: 10.1016/j.ejphar.2024.177147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Empagliflozin (EMPA) attenuates inflammation-induced ROS generation in static endothelial cells through inhibition of sodium hydrogen exchanger 1 (NHE1) and modulation of ion homeostasis. We hypothesize that EMPA will alleviate TNF-α stimulated endothelial dysfunction under flow conditions, and that this might be mediated by NHE1 and intracellular Ca2+. METHODS Human coronary artery endothelial cells were pre-treated with EMPA or vehicle before starting flow with or without TNF-α. Intracellular Ca2+ was recorded for 5 min at the start of flow. ROS generation and NO bioavailability, Piezo-1, cytokines, adhesion molecules, VE-cadherin and eNOS were detected after 6 h. BAPTA-AM was applied to chelate intracellular Ca2+ and NHE1 was knocked down with specific siRNA. RESULTS Under flow conditions, EMPA inhibited ROS production and [Ca2+] increase in cells exposed to TNF-α (P < 0.05). BAPTA-AM and NHE1 knockdown both reduced ROS generation (P < 0.05), and genetical inhibition of NHE1 led to reduction of intracellular [Ca2+] in HCAECs receiving TNF-α (P < 0.05). Yet, EMPA showed no effect on the increased cytokine production, adhesion molecule expression and phosphorylation of eNOS in endothelial cells exposed to TNF-α. CONCLUSION EMPA mitigates increased ROS production and impaired NO bioavailability in TNF-α stimulated cells under flow. The anti-oxidative effect of EMPA is mediated by the decreased intracellular [Ca2+] following NHE1 inhibition.
Collapse
Affiliation(s)
- Xiaoling Li
- Amsterdam, University Medical Centers, Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam Cardiovascular Science, Meibergdreef 11, 1105 AZ, Amsterdam, the Netherlands; Center for cell lineage and Development, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Mengnan Wang
- Amsterdam, University Medical Centers, Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam Cardiovascular Science, Meibergdreef 11, 1105 AZ, Amsterdam, the Netherlands
| | - Marlene Wolfsgruber
- Amsterdam, University Medical Centers, Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam Cardiovascular Science, Meibergdreef 11, 1105 AZ, Amsterdam, the Netherlands
| | - Olivia C Klatt
- Amsterdam, University Medical Centers, Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam Cardiovascular Science, Meibergdreef 11, 1105 AZ, Amsterdam, the Netherlands
| | - Markus W Hollmann
- Amsterdam, University Medical Centers, Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam Cardiovascular Science, Meibergdreef 11, 1105 AZ, Amsterdam, the Netherlands
| | - Benedikt Preckel
- Amsterdam, University Medical Centers, Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam Cardiovascular Science, Meibergdreef 11, 1105 AZ, Amsterdam, the Netherlands
| | - Coert J Zuurbier
- Amsterdam, University Medical Centers, Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam Cardiovascular Science, Meibergdreef 11, 1105 AZ, Amsterdam, the Netherlands
| | - Nina C Weber
- Amsterdam, University Medical Centers, Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam Cardiovascular Science, Meibergdreef 11, 1105 AZ, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Xue S, Zhou X, Yang ZH, Si XK, Sun X. Stroke-induced damage on the blood-brain barrier. Front Neurol 2023; 14:1248970. [PMID: 37840921 PMCID: PMC10569696 DOI: 10.3389/fneur.2023.1248970] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/08/2023] [Indexed: 10/17/2023] Open
Abstract
The blood-brain barrier (BBB) is a functional phenotype exhibited by the neurovascular unit (NVU). It is maintained and regulated by the interaction between cellular and non-cellular matrix components of the NVU. The BBB plays a vital role in maintaining the dynamic stability of the intracerebral microenvironment as a barrier layer at the critical interface between the blood and neural tissues. The large contact area (approximately 20 m2/1.3 kg brain) and short diffusion distance between neurons and capillaries allow endothelial cells to dominate the regulatory role. The NVU is a structural component of the BBB. Individual cells and components of the NVU work together to maintain BBB stability. One of the hallmarks of acute ischemic stroke is the disruption of the BBB, including impaired function of the tight junction and other molecules, as well as increased BBB permeability, leading to brain edema and a range of clinical symptoms. This review summarizes the cellular composition of the BBB and describes the protein composition of the barrier functional junction complex and the mechanisms regulating acute ischemic stroke-induced BBB disruption.
Collapse
Affiliation(s)
| | | | | | | | - Xin Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Mauro AK, Berdahl DM, Khurshid N, Clemente L, Ampey AC, Shah DM, Bird IM, Boeldt DS. Conjugated linoleic acid improves endothelial Ca2+ signaling by blocking growth factor and cytokine-mediated Cx43 phosphorylation. Mol Cell Endocrinol 2020; 510:110814. [PMID: 32259635 PMCID: PMC7253345 DOI: 10.1016/j.mce.2020.110814] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/13/2020] [Accepted: 03/31/2020] [Indexed: 12/21/2022]
Abstract
Sustained Ca2+ burst signaling is crucial for endothelial vasodilator production and is disrupted by growth factors and cytokines. Conjugated linoleic acid (CLA), a Src inhibitor in certain preparations, is generally regarded as safe during pregnancy by the FDA. Multiple CLA preparations; t10, c12 or c9, t11 CLA, or a 1:1 mixture of the two were administered before growth factor or cytokine treatment. Growth factors and cytokines caused a significant decrease in Ca2+ burst numbers in response to ATP stimulation. Both t10, c12 CLA and the 1:1 mixture rescued VEGF165 or TNFα inhibited Ca2+ bursts and correlated with Src-specific phosphorylation of connexin 43. VEGF165, TNFα, and IL-6 in combination at physiologic concentrations revealed IL-6 amplified the inhibitory effects of lower dose of VEGF165 and TNFα. Again, the 1:1 CLA mixture was most effective at rescue of function. Therefore, CLA formulations may be a promising treatment for endothelial dysfunction in diseases such as preeclampsia.
Collapse
Affiliation(s)
- Amanda K Mauro
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Danielle M Berdahl
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA; Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Nauman Khurshid
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA; Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Luca Clemente
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Amanda C Ampey
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Dinesh M Shah
- Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Ian M Bird
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA; Department of Pediatrics, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA
| | - Derek S Boeldt
- Perinatal Research Laboratories, Department of Obstetrics & Gynecology, University of Wisconsin - Madison, School Medicine and Public Health, Madison, WI, 53715, USA.
| |
Collapse
|
4
|
Colás-Algora N, García-Weber D, Cacho-Navas C, Barroso S, Caballero A, Ribas C, Correas I, Millán J. Compensatory increase of VE-cadherin expression through ETS1 regulates endothelial barrier function in response to TNFα. Cell Mol Life Sci 2020; 77:2125-2140. [PMID: 31396656 PMCID: PMC11105044 DOI: 10.1007/s00018-019-03260-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/24/2019] [Accepted: 07/29/2019] [Indexed: 02/07/2023]
Abstract
VE-cadherin plays a central role in controlling endothelial barrier function, which is transiently disrupted by proinflammatory cytokines such as tumor necrosis factor (TNFα). Here we show that human endothelial cells compensate VE-cadherin degradation in response to TNFα by inducing VE-cadherin de novo synthesis. This compensation increases adherens junction turnover but maintains surface VE-cadherin levels constant. NF-κB inhibition strongly reduced VE-cadherin expression and provoked endothelial barrier collapse. Bacterial lipopolysaccharide and TNFα upregulated the transcription factor ETS1, in vivo and in vitro, in an NF-κB dependent manner. ETS1 gene silencing specifically reduced VE-cadherin protein expression in response to TNFα and exacerbated TNFα-induced barrier disruption. We propose that TNFα induces not only the expression of genes involved in increasing permeability to small molecules and immune cells, but also a homeostatic transcriptional program in which NF-κB- and ETS1-regulated VE-cadherin expression prevents the irreversible damage of endothelial barriers.
Collapse
Affiliation(s)
| | - Diego García-Weber
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain.
- INSERM, U1016, Institut Cochin, Paris, France.
| | | | - Susana Barroso
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain
| | - Alvaro Caballero
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
| | - Catalina Ribas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Isabel Correas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28049, Madrid, Spain.
| |
Collapse
|
5
|
Yang Y, Dong X, Zheng S, Sun J, Ye J, Chen J, Fang Y, Zhao B, Yin Z, Cao P, Luo L. GSTpi regulates VE-cadherin stabilization through promoting S-glutathionylation of Src. Redox Biol 2019; 30:101416. [PMID: 31927409 PMCID: PMC6957793 DOI: 10.1016/j.redox.2019.101416] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/09/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023] Open
Abstract
GSTpi is a Phase II metabolic enzyme which is originally considered as an important facilitator of cellular detoxification. Here, we found that GSTpi stabilized VE-cadherin in endothelial cell membrane through inhibiting VE-cadherin phosphorylation and VE-cadherin/catenin complex dissociation, and consequently maintained endothelial barrier function. Our findings demonstrated a novel mechanism that GSTpi inhibited VE-cadherin phosphorylation through suppressing the activation of Src/VE-cadherin pathway. Mass spectrometry analysis and molecular docking showed that GSTpi enhanced Src S-glutathionylation at Cys185, Cys245, and Cys400 of Src. More important, we found that GSTpi promoted S-glutathionylation of Src was essential for GSTpi to inhibit Src phosphorylation and activation. Furthermore, in vivo experiments indicated that AAV-GSTpi exerted the protective effect on pulmonary vessel permeability in the animal model of acute lung injury. This study revealed a novel regulatory effect of GSTpi on vascular endothelial barrier function and the importance of S-glutathionylation of Src induced by GSTpi in the activation of Src/VE-cadherin pathway. GSTpi regulates endothelial barrier function in response to pro-inflammatory stress. GSTpi inhibits the destabilization of membrane VE-cadherin through suppressing the activation of Src/VE-cadherin pathway. GSTpi selectively inhibits Src phosphorylation by S-glutathionylating novel cysteines of Src. GSTpi exerts the protective effect on pulmonary vessel permeability in the animal model of acute lung injury.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210093, Jiangsu, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Xiaoliang Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Shuangning Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Jinbing Sun
- Changshu No.1 People's Hospital Affiliated to Soochow University, Changshu, 215500, China
| | - Juan Ye
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Jiao Chen
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Yuan Fang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Bing Zhao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, Jiangsu, China.
| | - Peng Cao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China; Laboratory of Cellular and Molecular Biology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, Jiangsu, China; Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Lan Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210093, Jiangsu, China.
| |
Collapse
|
6
|
Zamorano P, Koning T, Oyanadel C, Mardones GA, Ehrenfeld P, Boric MP, González A, Soza A, Sánchez FA. Galectin-8 induces endothelial hyperpermeability through the eNOS pathway involving S-nitrosylation-mediated adherens junction disassembly. Carcinogenesis 2019; 40:313-323. [PMID: 30624618 DOI: 10.1093/carcin/bgz002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 12/04/2018] [Accepted: 01/04/2019] [Indexed: 12/26/2022] Open
Abstract
The permeability of endothelial cells is regulated by the stability of the adherens junctions, which is highly sensitive to kinase-mediated phosphorylation and endothelial nitric oxide synthase (eNOS)-mediated S-nitrosylation of its protein components. Solid tumors can produce a variety of factors that stimulate these signaling pathways leading to endothelial cell hyperpermeability. This generates stromal conditions that facilitate tumoral growth and dissemination. Galectin-8 (Gal-8) is overexpressed in several carcinomas and has a variety of cellular effects that can contribute to tumor pathogenicity, including angiogenesis. Here we explored whether Gal-8 has also a role in endothelial permeability. We show that recombinant Gal-8 activates eNOS, induces S-nitrosylation of p120-catenin (p120) and dissociation of adherens junction, leading to hyperpermeability of the human endothelial cell line EAhy926. This pathway involves focal-adhesion kinase (FAK) activation downstream of eNOS as a requirement for eNOS-mediated p120 S-nitrosylation. This suggests a reciprocal, yet little understood, regulation of phosphorylation and S-nitrosylation events acting upon adherens junction permeability. In addition, glutathione S-transferase (GST)-Gal-8 pull-down experiments and function-blocking β1-integrin antibodies point to β1-integrins as cell surface components involved in Gal-8-induced hyperpermeability. Endogenous Gal-8 secreted from the breast cancer cell line MCF-7 has similar hyperpermeability and signaling effects. Furthermore, the mouse cremaster model system showed that Gal-8 also activates eNOS, induces S-nitrosylation of adherens junction components and is an effective hyperpermeability agent in vivo. These results add endothelial permeability regulation by S-nitrosylation as a new function of Gal-8 that can potentially contribute to the pathogenicity of tumors overexpressing this lectin.
Collapse
Affiliation(s)
- Patricia Zamorano
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Tania Koning
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Claudia Oyanadel
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Gonzalo A Mardones
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Instituto de Fisiología, Valdivia, Chile.,Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile
| | - Pamela Ehrenfeld
- Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile.,Instituto de Anatomía, Histología y Patología, Universidad Austral de Chile, Valdivia, Chile
| | | | - Alfonso González
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Fundación Ciencia y Vida. Santiago, Chile
| | - Andrea Soza
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Fabiola A Sánchez
- Instituto de Inmunología, Universidad Austral de Chile, Valdivia 5110566, Chile.,Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Valdivia, Chile
| |
Collapse
|
7
|
Ludewig P, Winneberger J, Magnus T. The cerebral endothelial cell as a key regulator of inflammatory processes in sterile inflammation. J Neuroimmunol 2018; 326:38-44. [PMID: 30472304 DOI: 10.1016/j.jneuroim.2018.10.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 09/17/2018] [Accepted: 10/24/2018] [Indexed: 11/18/2022]
Abstract
Cerebral endothelial cells accomplish numerous tasks connected to the maintenance of homeostasis of the central nervous system. They create a barrier between the central nervous system and peripheral blood and regulate mechanotransduction, vascular permeability, rheology, thrombogenesis, and leukocyte adhesion. In pathophysiological conditions (e.g., stroke or ischemia-reperfusion injury) the endothelial functions are impaired, leading to increased vascular permeability, vascular inflammation, leukocyte-endothelium interactions, and transendothelial migration, driving CNS inflammation and neuronal destruction. This review describes the current knowledge on the regulatory roles of endothelial cells in neuroinflammatory processes.
Collapse
Affiliation(s)
- Peter Ludewig
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany.
| | - Jack Winneberger
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology at the University Medical Center Hamburg- Eppendorf, Hamburg, Germany
| |
Collapse
|
8
|
Causation by Diesel Exhaust Particles of Endothelial Dysfunctions in Cytotoxicity, Pro-inflammation, Permeability, and Apoptosis Induced by ROS Generation. Cardiovasc Toxicol 2018; 17:384-392. [PMID: 26965709 DOI: 10.1007/s12012-016-9364-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Epidemiological studies suggest that an increase of diesel exhaust particles (DEP) in ambient air corresponds to an increase in hospital-recorded myocardial infarctions within 48 h after exposure. Among the many theories to explain this data are endothelial dysfunction and translocation of DEP into vasculature. The mechanisms for such DEP-induced vascular permeability remain unknown. One of the major mechanisms underlying the effects of DEP is suggested to be oxidative stress. Experiments have shown that DEP induce the generation of reactive oxygen species (ROS), such as superoxide anion and H2O2 in the HUVEC tube cells. Transcription factor Nrf2 is translocated to the cell nucleus, where it activates transcription of the antioxidative enzyme HO-1 and sequentially induces the release of vascular permeability factor VEGF-A. Furthermore, a recent study shows that DEP-induced intracellular ROS may cause the release of pro-inflammatory TNF-α and IL-6, which may induce endothelial permeability as well by promoting VEGF-A secretion independently of HO-1 activation. These results demonstrated that the adherens junction molecule, VE-cadherin, becomes redistributed from the membrane at cell-cell borders to the cytoplasm in response to DEP, separating the plasma membranes of adjacent cells. DEP were occasionally found in endothelial cell cytoplasm and in tube lumen. In addition, the induced ROS is cytotoxic to the endothelial tube-like HUVEC. Acute DEP exposure stimulates ATP depletion, followed by depolarization of their actin cytoskeleton, which sequentially inhibits PI3K/Akt activity and induces endothelial apoptosis. Nevertheless, high-dose DEP augments tube cell apoptosis up to 70 % but disrupts the p53 negative regulator Mdm2. In summary, exposure to DEP affects parameters influencing vasculature permeability and viability, i.e., oxidative stress and its upregulated antioxidative and pro-inflammatory responses, which sequentially induce vascular permeability factor, VEGF-A release and disrupt cell-cell junction integrity. While exposure to a low dose of DEP actin triggers cytoskeleton depolarization, reduces PI3K/Akt activity, and induces a p53/Mdm2 feedback loop, a high dose causes apoptosis by depleting Mdm2. Addition of ROS scavenger N-acetyl cysteine suppresses DEP-induced oxidative stress efficiently and reduces subsequent damages by increasing endogenous glutathione.
Collapse
|
9
|
Buravkova LB, Rudimov EG, Andreeva ER, Grigoriev AI. The ICAM-1 expression level determines the susceptibility of human endothelial cells to simulated microgravity. J Cell Biochem 2017; 119:2875-2885. [PMID: 29080356 DOI: 10.1002/jcb.26465] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/24/2017] [Indexed: 01/13/2023]
Abstract
Microgravity is a principal risk factor hampering human cardiovascular regulation during space flights. Endothelial dysfunction associated with the impaired integrity and uniformity of the monolayer represents a potential trigger for vascular damage. We characterized the expression profile of the multi-step cascade of adhesion molecules (ICAM-1, VCAM-1, E-selectin, VE-cadherin) in umbilical cord endothelial cells (ECs) after 24 h of exposure to simulated microgravity (SMG), pro-inflammatory cytokine TNF-α, and the combination of the two. Random Positioning Machine (RPM)-mediated SMG was used to mimic microgravity effects. SMG stimulated the expression of E-selectin, which is known to be involved in slowing leukocyte rolling. Primary ECs displayed heterogeneity with respect to the proportion of ICAM-1-positive cells. ECs were divided into two groups: pre-activated ECs displaying high proportion of ICAM-1+ -cells (ECs-1) (greater than 50%) and non-activated ECs with low proportion of ICAM-1+ -cells (ECs-2) (less than 25%). Only non-activated ECs-2 responded to SMG by elevating gene transcription and increasing ICAM-1 and VE-cadherin expression. This effect was enhanced after cumulative SMG-TNF-α exposure. ECs-1 displayed an unexpected decrease in number of E-selectin- and ICAM-1-positive ECs and pronounced up-regulation of VCAM1 upon activation of inflammation, which was partially abolished by SMG. Thus, non-activated ECs-2 are quite resistant to the impacts of microgravity and even exhibited an elevation of the VE-cadherin gene and protein expression, thus improving the integrity of the endothelial monolayer. Pre-activation of ECs with inflammatory stimuli may disturb the EC adhesion profile, attenuating its barrier function. These alterations may be among the mechanisms underlying cardiovascular dysregulation in real microgravity conditions.
Collapse
Affiliation(s)
- Ludmila B Buravkova
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Eugene G Rudimov
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Elena R Andreeva
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Anatoly I Grigoriev
- Cell Physiology Laboratory, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
10
|
Abstract
Endothelial cells line blood vessels and provide a dynamic interface between the blood and tissues. They remodel to allow leukocytes, fluid and small molecules to enter tissues during inflammation and infections. Here we compare the signaling networks that contribute to endothelial permeability and leukocyte transendothelial migration, focusing particularly on signals mediated by small GTPases that regulate cell adhesion and the actin cytoskeleton. Rho and Rap GTPase signaling is important for both processes, but they differ in that signals are activated locally under leukocytes, whereas endothelial permeability is a wider event that affects the whole cell. Some molecules play a unique role in one of the two processes, and could therefore be targeted to selectively alter either endothelial permeability or leukocyte transendothelial migration.
Collapse
Affiliation(s)
- Camilla Cerutti
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| | - Anne J Ridley
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
11
|
Yang J, Wang Y, Zeng Z, Qiao L, Zhuang L, Gao Q, Ma D, Huang X. Smad4 deletion in blood vessel endothelial cells promotes ovarian cancer metastasis. Int J Oncol 2017; 50:1693-1700. [PMID: 28393199 DOI: 10.3892/ijo.2017.3957] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 02/23/2017] [Indexed: 11/06/2022] Open
Abstract
SMAD4 is a critical co-smad in signal transduction pathways activated in response to transforming growth factor-β (TGF-β)-related ligands, regulating cell growth and differentiation. The roles played by SMAD4 inactivation in tumors highlighted it as a tumor-suppressor gene. Herein, we report that loss of SMAD4 expression in vascular endothelial cells promotes ovarian cancer invasion. SiRNA transfer of this gene in the HUVEC reduced SMAD4 protein expression and function. Although it reduced the vessel endothelial cell tubule formation in vitro and in vivo, it did not affect the tumor growth significantly in vivo. However, it weakened the barrier integrity in endothelial cells and increased vessel permeability and the ovarian cancer liver metastasis. We documented reduced angiogenesis and increased invasion histologically and by intravital microscopy, and gained mechanistic insight at the messenger and gene level. Finally, we found a negative reciprocal regulation between SMAD4 and FYN. FYN is one of the Src family kinases (SFK), activation of which can cause dissociation of cell-cell junctions and adhesion, resulting in paracellular hypermeability. Upon SMAD4 deletion, we detected high expression levels of FYN in vessel endothelial cells, suggesting the mechanism of the ovarian tumor cells cross the endothelial barrier and transform to an invasive phenotype.
Collapse
Affiliation(s)
- Jie Yang
- Cancer Biology Research Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ya Wang
- Cancer Biology Research Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhen Zeng
- Cancer Biology Research Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Long Qiao
- Cancer Biology Research Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Liang Zhuang
- Cancer Biology Research Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qinglei Gao
- Cancer Biology Research Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ding Ma
- Cancer Biology Research Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiaoyuan Huang
- Cancer Biology Research Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
12
|
Barabutis N, Verin A, Catravas JD. Regulation of pulmonary endothelial barrier function by kinases. Am J Physiol Lung Cell Mol Physiol 2016; 311:L832-L845. [PMID: 27663990 DOI: 10.1152/ajplung.00233.2016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/15/2016] [Indexed: 12/15/2022] Open
Abstract
The pulmonary endothelium is the target of continuous physiological and pathological stimuli that affect its crucial barrier function. The regulation, defense, and repair of endothelial barrier function require complex biochemical processes. This review examines the role of endothelial phosphorylating enzymes, kinases, a class with profound, interdigitating influences on endothelial permeability and lung function.
Collapse
Affiliation(s)
- Nektarios Barabutis
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia
| | - Alexander Verin
- Vascular Biology Center, Augusta University, Augusta, Georgia; and
| | - John D Catravas
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, .,School of Medical Diagnostic and Translational Sciences, College of Health Sciences, Old Dominion University, Norfolk, Virginia
| |
Collapse
|
13
|
Zhang J, Jiang Z, Bao C, Mei J, Zhu J. Cardiopulmonary bypass increases pulmonary microvascular permeability through the Src kinase pathway: Involvement of caveolin-1 and vascular endothelial cadherin. Mol Med Rep 2016; 13:2918-24. [PMID: 26847917 PMCID: PMC4768976 DOI: 10.3892/mmr.2016.4831] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 01/12/2016] [Indexed: 11/22/2022] Open
Abstract
Changes in pulmonary microvascular permeability following cardiopulmonary bypass (CPB) and the underlying mechanisms have not yet been established. Therefore, the aim of the present study was to elucidate the alterations in pulmonary microvascular permeability following CPB and the underlying mechanism. The pulmonary microvascular permeability was measured using Evans Blue dye (EBD) exclusion, and the neutrophil infiltration and proinflammatory cytokine secretion was investigated. In addition, the activation of Src kinase and the phosphorylation of caveolin-1 and vascular endothelial cadherin (VE-cadherin) was examined. The results revealed that CPB increased pulmonary microvascular leakage, neutrophil count and proinflammatory cytokines in the bronchoalveolar lavage fluid, and activated Src kinase. The administration of PP2, an inhibitor of Src kinase, decreased the activation of Src kinase and attenuated the increase in pulmonary microvascular permeability observed following CPB. Two important proteins associated with vascular permeability, caveolin-1 and VE-cadherin, were significantly activated at 24 h in the lung tissues following CPB, which correlated with the alterations in pulmonary microvascular permeability and Src kinase. PP2 administration inhibited their activation, suggesting that they are downstream factors of Src kinase activation. The data indicated that the Src kinase pathway increased pulmonary microvascular permeability following CPB, and the activation of caveolin-1 and VE-cadherin may be involved. Inhibition of this pathway may provide a potential therapy for acute lung injury following cardiac surgery.
Collapse
Affiliation(s)
- Junwen Zhang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Zhaolei Jiang
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Chunrong Bao
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Ju Mei
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| | - Jiaquan Zhu
- Department of Cardiothoracic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, P.R. China
| |
Collapse
|
14
|
Timmerman I, Daniel AE, Kroon J, van Buul JD. Leukocytes Crossing the Endothelium: A Matter of Communication. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:281-329. [PMID: 26940521 DOI: 10.1016/bs.ircmb.2015.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Leukocytes cross the endothelial vessel wall in a process called transendothelial migration (TEM). The purpose of leukocyte TEM is to clear the causing agents of inflammation in underlying tissues, for example, bacteria and viruses. During TEM, endothelial cells initiate signals that attract and guide leukocytes to sites of tissue damage. Leukocytes react by attaching to these sites and signal their readiness to move back to endothelial cells. Endothelial cells in turn respond by facilitating the passage of leukocytes while retaining overall integrity. In this review, we present recent findings in the field and we have endeavored to synthesize a coherent picture of the intricate interplay between endothelial cells and leukocytes during TEM.
Collapse
Affiliation(s)
- Ilse Timmerman
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Anna E Daniel
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jaap D van Buul
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Shear-dependent attenuation of cellular ROS levels can suppress proinflammatory cytokine injury to human brain microvascular endothelial barrier properties. J Cereb Blood Flow Metab 2015; 35:1648-56. [PMID: 25991069 PMCID: PMC4640321 DOI: 10.1038/jcbfm.2015.102] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/22/2015] [Indexed: 12/21/2022]
Abstract
The regulatory interplay between laminar shear stress and proinflammatory cytokines during homeostatic maintenance of the brain microvascular endothelium is largely undefined. We hypothesized that laminar shear could counteract the injurious actions of proinflammatory cytokines on human brain microvascular endothelial cell (HBMvEC) barrier properties, in-part through suppression of cellular redox signaling. For these investigations, HBMvECs were exposed to either shear stress (8 dynes/cm(2), 24 hours) or cytokines (tumor necrosis factor-α (TNF-α) or interleukin-6 (IL-6), 0 to 100 ng/mL, 6 or 18 hours). Human brain microvascular endothelial cell 'preshearing'±cytokine exposure was also performed. Either cytokine dose-dependently decreased expression and increased phosphorylation (pTyr/pThr) of interendothelial occludin, claudin-5, and vascular endothelial-cadherin; observations directly correlating to endothelial barrier reduction, and in precise contrast to effects seen with shear. We further observed that, relative to unsheared cells, HBMvECs presheared for 24 hours exhibited significantly reduced reactive oxygen species production and barrier permeabilization in response to either TNF-α or IL-6 treatment. Shear also downregulated NADPH oxidase (nicotinamide adenine dinucleotide phosphate-oxidase) activation in HBMvECs, as manifested in the reduced expression and coassociation of gp91phox and p47phox. These findings lead us to conclude that physiologic shear can protect the brain microvascular endothelium from injurious cytokine effects on interendothelial junctions and barrier function by regulating the cellular redox state in-part through NADPH oxidase inhibition.
Collapse
|
16
|
Khakpour S, Wilhelmsen K, Hellman J. Vascular endothelial cell Toll-like receptor pathways in sepsis. Innate Immun 2015; 21:827-46. [DOI: 10.1177/1753425915606525] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 08/11/2015] [Indexed: 12/20/2022] Open
Abstract
The endothelium forms a vast network that dynamically regulates vascular barrier function, coagulation pathways and vasomotor tone. Microvascular endothelial cells are uniquely situated to play key roles during infection and injury, owing to their widespread distribution throughout the body and their constant interaction with circulating blood. While not viewed as classical immune cells, endothelial cells express innate immune receptors, including the Toll-like receptors (TLRs), which activate intracellular inflammatory pathways mediated through NF-κB and the MAP kinases. TLR agonists, including LPS and bacterial lipopeptides, directly upregulate microvascular endothelial cell expression of inflammatory mediators. Intriguingly, TLR activation also modulates microvascular endothelial cell permeability and the expression of coagulation pathway intermediaries. Microvascular thrombi have been hypothesized to trap microorganisms thereby limiting the spread of infection. However, dysregulated activation of endothelial inflammatory pathways is also believed to lead to coagulopathy and increased vascular permeability, which together promote sepsis-induced organ failure. This article reviews vascular endothelial cell innate immune pathways mediated through the TLRs as they pertain to sepsis, highlighting links between TLRs and coagulation and permeability pathways, and their role in healthy and pathologic responses to infection and sepsis.
Collapse
Affiliation(s)
- Samira Khakpour
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
- Biomedical Sciences and Immunology Programs, University of California, San Francisco, CA, USA
| | - Kevin Wilhelmsen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
| | - Judith Hellman
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA, USA
- Biomedical Sciences and Immunology Programs, University of California, San Francisco, CA, USA
| |
Collapse
|
17
|
Tseng CY, Chang JF, Wang JS, Chang YJ, Gordon MK, Chao MW. Protective Effects of N-Acetyl Cysteine against Diesel Exhaust Particles-Induced Intracellular ROS Generates Pro-Inflammatory Cytokines to Mediate the Vascular Permeability of Capillary-Like Endothelial Tubes. PLoS One 2015; 10:e0131911. [PMID: 26148005 PMCID: PMC4492618 DOI: 10.1371/journal.pone.0131911] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/08/2015] [Indexed: 01/01/2023] Open
Abstract
Exposure to diesel exhaust particles (DEP) is associated with pulmonary and cardiovascular diseases. Previous studies using in vitro endothelial tubes as a simplified model of capillaries have found that DEP-induced ROS increase vascular permeability with rearrangement or internalization of adherens junctional VE-cadherin away from the plasma membrane. This allows DEPs to penetrate into the cell and capillary lumen. In addition, pro-inflammatory cytokines are up-regulated and mediate vascular permeability in response to DEP. However, the mechanisms through which these DEP-induced pro-inflammatory cytokines increase vascular permeability remain unknown. Hence, we examined the ability of DEP to induce permeability of human umbilical vein endothelial cell tube cells to investigate these mechanisms. Furthermore, supplementation with NAC reduces ROS production following exposure to DEP. HUVEC tube cells contributed to a pro-inflammatory response to DEP-induced intracellular ROS generation. Endothelial oxidative stress induced the release of TNF-α and IL-6 from tube cells, subsequently stimulating the secretion of VEGF-A independent of HO-1. Our data suggests that DEP-induced intracellular ROS and release of the pro-inflammatory cytokines TNF- α and IL-6, which would contribute to VEGF-A secretion and disrupt cell-cell borders and increase vasculature permeability. Addition of NAC suppresses DEP-induced ROS efficiently and reduces subsequent damages by increasing endogenous glutathione.
Collapse
Affiliation(s)
- Chia-Yi Tseng
- Department of Biomedical Engineering, College of Engineering, Chung Yuan Christian University, Zhongli district, Taoyuan city, Taiwan
- Center of Nanotechnology, Chung Yuan Christian University, Zhongli district, Taoyuan city, Taiwan
| | - Jing-Fen Chang
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Zhongli district, Taoyuan city, Taiwan
| | - Jhih-Syuan Wang
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Zhongli district, Taoyuan city, Taiwan
| | - Yu-Jung Chang
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Zhongli district, Taoyuan city, Taiwan
| | - Marion K. Gordon
- Joint program of Toxicology, Rutgers University, Piscataway, New Jersey, United States of America
| | - Ming-Wei Chao
- Center of Nanotechnology, Chung Yuan Christian University, Zhongli district, Taoyuan city, Taiwan
- Department of Bioscience Technology, College of Science, Chung Yuan Christian University, Zhongli district, Taoyuan city, Taiwan
- * E-mail:
| |
Collapse
|
18
|
Baumgartner W, Weth A, Gutberlet J, Harms G, Groschner K. Localization of VE-cadherin in plasmalemmal cholesterol rich microdomains and the effects of cholesterol depletion on VE-cadherin mediated cell–cell adhesion. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:1725-32. [DOI: 10.1016/j.bbalip.2014.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 08/01/2014] [Accepted: 08/25/2014] [Indexed: 10/24/2022]
|
19
|
Marcos-Ramiro B, García-Weber D, Millán J. TNF-induced endothelial barrier disruption: beyond actin and Rho. Thromb Haemost 2014; 112:1088-102. [PMID: 25078148 DOI: 10.1160/th14-04-0299] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 06/16/2014] [Indexed: 11/05/2022]
Abstract
The decrease of endothelial barrier function is central to the long-term inflammatory response. A pathological alteration of the ability of endothelial cells to modulate the passage of cells and solutes across the vessel underlies the development of inflammatory diseases such as atherosclerosis and multiple sclerosis. The inflammatory cytokine tumour necrosis factor (TNF) mediates changes in the barrier properties of the endothelium. TNF activates different Rho GTPases, increases filamentous actin and remodels endothelial cell morphology. However, inhibition of actin-mediated remodelling is insufficient to prevent endothelial barrier disruption in response to TNF, suggesting that additional molecular mechanisms are involved. Here we discuss, first, the pivotal role of Rac-mediated generation of reactive oxygen species (ROS) to regulate the integrity of endothelial cell-cell junctions and, second, the ability of endothelial adhesion receptors such as ICAM-1, VCAM-1 and PECAM-1, involved in leukocyte transendothelial migration, to control endothelial permeability to small molecules, often through ROS generation. These adhesion receptors regulate endothelial barrier function in ways both dependent on and independent of their engagement by immune cells, and orchestrate the crosstalk between leukocyte transendothelial migration and endothelial permeability during inflammation.
Collapse
Affiliation(s)
| | | | - J Millán
- Jaime Millán, Centro de Biología Molecular Severo Ochoa, C/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain, Tel.: +34 911964713, Fax: +34 911964420, E-mail:
| |
Collapse
|
20
|
Galectin-1 induces vascular permeability through the neuropilin-1/vascular endothelial growth factor receptor-1 complex. Angiogenesis 2014; 17:839-49. [PMID: 24719187 DOI: 10.1007/s10456-014-9431-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 04/04/2014] [Indexed: 10/25/2022]
Abstract
Galectin-1 (Gal-1) is a β-galactoside-binding lectin that regulates endothelial cell migration, proliferation, and adhesion. However, the effect of Gal-1 on vascular permeability and the underlying mechanisms are unclear. We found that high Gal-1 expression was associated with elevated tumor vascular permeability in specimens of oral squamous cell carcinoma. Using transendothelial passage of FITC-dextran and a Miles assay, we demonstrated that Gal-1 increased vascular permeability extracellularly through its carbohydrate recognition domain. Mechanism dissection revealed that the neuropilin (NRP)-1/vascular endothelial growth factor receptor- (VEGFR)-1 complex was required for Gal-1-regulated vascular permeability. Activation of VEGFR-1 triggered activation of Akt which led to a reduction in vascular endothelial-cadherin at cell-cell junctions and resulted in cytoskeletal rearrangement. Both inhibition of Gal-1 secreted from cancer cells and administration of an anti-Gal-1 antibody in the tumor microenvironment suppressed tumor growth and vascular permeability in xenograft models. In conclusion, our results demonstrate a novel function of Gal-1 of increasing vascular permeability through the NRP-1/VEGFR1 and Akt signaling pathway and indicate that targeting Gal-1 by an anti-Gal-1 antibody is a feasible therapy for vascular hyperpermeability and cancer.
Collapse
|
21
|
Seynhaeve ALB, Rens JAP, Schipper D, Eggermont AMM, Ten Hagen TLM. Exposing endothelial cells to tumor necrosis factor-α and peripheral blood mononuclear cells damage endothelial integrity via interleukin-1ß by degradation of vascular endothelial-cadherin. Surgery 2013; 155:545-53. [PMID: 24439748 DOI: 10.1016/j.surg.2013.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 10/11/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND AND PURPOSE We demonstrated previously that the administration of tumor necrosis factor alpha (TNF-α) for the treatment of solid tumors enhanced the response to chemotherapy by augmenting intratumoral drug accumulation. TNF-α changes the integrity of the endothelial cell monolayer in combination with interferon gamma (IFN-γ), which is further enhanced by the addition of peripheral blood mononuclear cells (PBMCs). The improved effect of PBMCs was mostly induced by the endogenous production of interleukin-1beta (IL-1ß) after TNF-α stimulation. In the current study, we demonstrate that exposing endothelial cells to TNF-α and PBMCs mediates the loss of vascular endothelial (VE)-cadherin, an important adherens junction protein for maintaining endothelial integrity, through endogenous IL-1ß. This loss increases permeability of the endothelial layer, thereby explaining the augmented passage of chemotherapeutics into the tumor. METHODS Human umbilical vein endothelial cells were exposed to TNF-α, IFN-γ, PBMCs, or IL-1ß, and the effects on the endothelial integrity were assessed by morphological changes and permeability changes with the use of fluorescein isothiocyanate-labeled bovine serum albumin flux. The loss of VE-cadherin was assessed using immunofluorescence, western blotting, and polymerase chain reaction. RESULTS Incubating endothelial cells with TNF-α, IFN-γ, and PBMCs increased cell elongation, gap formation, and subsequently the permeability of fluorescein isothiocyanate-labeled bovine serum albumin compared with control or TNF-α and IFN-γ-treated cells (P < .05). When PBMCs were replaced with IL-1ß, identical changes were observed. These changes in integrity were associated with a loss of VE-cadherin at the membrane. CONCLUSION We conclude that VE-cadherin is lost at the membrane when endothelial cells are exposed to TNF-α, IFN-γ, and PBMCs, which results in loss of integrity. IL-1ß can mimic the effects of PBMCs, indicating a dominant role of endogenously produced IL-1ß in this process.
Collapse
Affiliation(s)
- Ann L B Seynhaeve
- Laboratory of Experimental Surgical Oncology, Department of Surgical Oncology, Erasmus MC, Rotterdam, The Netherlands.
| | - Joost A P Rens
- Laboratory of Experimental Surgical Oncology, Department of Surgical Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Debby Schipper
- Laboratory of Experimental Surgical Oncology, Department of Surgical Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - Alexander M M Eggermont
- Laboratory of Experimental Surgical Oncology, Department of Surgical Oncology, Erasmus MC, Rotterdam, The Netherlands; Institut Gustave Roussy, Villejuif, Paris-Sud, France
| | - Timo L M Ten Hagen
- Laboratory of Experimental Surgical Oncology, Department of Surgical Oncology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Abstract
The Src family kinases (SFKs) c-Src and Yes mediate vascular leakage in response to various stimuli including lipopolysaccharide (LPS) and vascular endothelial growth factor (VEGF). Here, we define an opposing function of another SFK, Lyn, which in contrast to other SFKs, strengthens endothelial junctions and thereby restrains the increase in vascular permeability. Mice lacking Lyn displayed increased mortality in LPS-induced endotoxemia and increased vascular permeability in response to LPS or VEGF challenge compared with wild-type littermates. Lyn knockout mice repopulated with wild-type bone marrow-derived cells have higher vascular permeability than wild-type mice, suggesting a role of endothelial Lyn in the maintenance of the vascular barrier. Small interfering RNA-mediated down-regulation of Lyn disrupted endothelial barrier integrity, whereas expression of a constitutively active mutant of Lyn enhanced the barrier. However, down-regulation of Lyn did not affect LPS-induced endothelial permeability. We demonstrate that Lyn association with focal adhesion kinase (FAK) and phosphorylation of FAK at tyrosine residues 576/577 and 925 were required for Lyn-dependent stabilization of endothelial adherens junctions. Thus, in contrast to c-Src and Yes, which increase vascular permeability in response to stimuli, Lyn stabilizes endothelial junctions through phosphorylation of FAK. Therefore, therapeutics activating Lyn kinase may strengthen the endothelial barrier junction and hence have anti-inflammatory potential.
Collapse
|
23
|
Flynn KM, Michaud M, Canosa S, Madri JA. CD44 regulates vascular endothelial barrier integrity via a PECAM-1 dependent mechanism. Angiogenesis 2013; 16:689-705. [PMID: 23504212 DOI: 10.1007/s10456-013-9346-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 03/08/2013] [Indexed: 01/13/2023]
Abstract
Vascular integrity is a critical parameter in normal growth and development. Loss of appropriate vascular barrier function is present in various immune- and injury-mediated pathological conditions. CD44 is an adhesion molecule expressed by multiple cell types, including endothelial cells (EC). The goal of the present study was to examine how loss of CD44 affected vascular permeability. Using C57BL/6 WT and CD44-KO mice, we found no significant permeability to Evan's Blue in either strain at baseline. However, there was significantly increased histamine-induced permeability in CD44-deficient mice compared to WT counterparts. Similar results were observed in vitro, where CD44-deficient endothelial monolayers were also impermeable to 40kD-FITC dextran in the absence of vasoactive challenge, but exhibited enhanced and prolonged permeability following histamine. However, CD44-KO monolayers have reduced baseline barrier strength by electrical resistance, which correlated with increased permeability, at baseline, to smaller molecular weight 4-kD FITC-dextran, suggesting weakly formed endothelial junctions. The CD44-KO EC displayed several characteristics consistent with impaired barrier function/dysfunctional EC junctions, including differential expression, phosphorylation, and localization of endothelial junction proteins, increased matrix metalloprotease expression, and altered cellular morphology. Reduced platelet endothelial cell adhesion molecule-1 (PECAM-1) expression by CD44-KO EC in vivo and in vitro was also observed. Reconstitution of murine CD44 or PECAM-1 restored these defects to near WT status, suggesting CD44 regulates vascular permeability and integrity through a PECAM-1 dependent mechanism.
Collapse
Affiliation(s)
- Kelly M Flynn
- Department of Pathology, Yale University School of Medicine, 310 Cedar Street LH115, P.O. Box 208023, New Haven, CT 06520-8023, USA
| | | | | | | |
Collapse
|
24
|
Carra S, Foglia E, Cermenati S, Bresciani E, Giampietro C, Lora Lamia C, Dejana E, Beltrame M, Cotelli F. Ve-ptp modulates vascular integrity by promoting adherens junction maturation. PLoS One 2012; 7:e51245. [PMID: 23251467 PMCID: PMC3522677 DOI: 10.1371/journal.pone.0051245] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 11/01/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Endothelial cell junctions control blood vessel permeability. Altered permeability can be associated with vascular fragility that leads to vessel weakness and haemorrhage formation. In vivo studies on the function of genes involved in the maintenance of vascular integrity are essential to better understand the molecular basis of diseases linked to permeability defects. Ve-ptp (Vascular Endothelial-Protein Tyrosine Phosphatase) is a transmembrane protein present at endothelial adherens junctions (AJs). METHODOLOGY/PRINCIPAL FINDINGS We investigated the role of Ve-ptp in AJ maturation/stability and in the modulation of endothelial permeability using zebrafish (Danio rerio). Whole-mount in situ hybridizations revealed zve-ptp expression exclusively in the developing vascular system. Generation of altered zve-ptp transcripts, induced separately by two different splicing morpholinos, resulted in permeability defects closely linked to vascular wall fragility. The ultrastructural analysis revealed a statistically significant reduction of junction complexes and the presence of immature AJs in zve-ptp morphants but not in control embryos. CONCLUSIONS/SIGNIFICANCE Here we show the first in vivo evidence of a potentially critical role played by Ve-ptp in AJ maturation, an important event for permeability modulation and for the development of a functional vascular system.
Collapse
Affiliation(s)
- Silvia Carra
- Dipartimento di Biologia, Universitàdegli Studi di Milano, Milan, Italy
- Dipartimento di Bioscienze, Universitàdegli Studi di Milano, Milan, Italy
| | - Efrem Foglia
- Dipartimento di Biologia, Universitàdegli Studi di Milano, Milan, Italy
- Dipartimento di Bioscienze, Universitàdegli Studi di Milano, Milan, Italy
| | - Solei Cermenati
- Dipartimento di Bioscienze, Universitàdegli Studi di Milano, Milan, Italy
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Universitàdegli Studi di Milano, Milan, Italy
| | - Erica Bresciani
- Dipartimento di Biologia, Universitàdegli Studi di Milano, Milan, Italy
| | | | - Carla Lora Lamia
- Dipartimento di Biologia, Universitàdegli Studi di Milano, Milan, Italy
- Dipartimento di Bioscienze, Universitàdegli Studi di Milano, Milan, Italy
| | - Elisabetta Dejana
- Dipartimento di Bioscienze, Universitàdegli Studi di Milano, Milan, Italy
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Universitàdegli Studi di Milano, Milan, Italy
- FIRC Institute of Molecular Oncology, Milan, Italy
| | - Monica Beltrame
- Dipartimento di Bioscienze, Universitàdegli Studi di Milano, Milan, Italy
- Dipartimento di Scienze Biomolecolari e Biotecnologie, Universitàdegli Studi di Milano, Milan, Italy
| | - Franco Cotelli
- Dipartimento di Biologia, Universitàdegli Studi di Milano, Milan, Italy
- Dipartimento di Bioscienze, Universitàdegli Studi di Milano, Milan, Italy
- * E-mail:
| |
Collapse
|
25
|
Naikawadi RP, Cheng N, Vogel SM, Qian F, Wu D, Malik AB, Ye RD. A critical role for phosphatidylinositol (3,4,5)-trisphosphate-dependent Rac exchanger 1 in endothelial junction disruption and vascular hyperpermeability. Circ Res 2012; 111:1517-27. [PMID: 22965143 DOI: 10.1161/circresaha.112.273078] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RATIONALE The small GTPase Rac is critical to vascular endothelial functions, yet its regulation in endothelial cells remains unclear. Understanding the upstream pathway may delineate Rac activation mechanisms and its role in maintaining vascular endothelial barrier integrity. OBJECTIVE By investigating phosphatidylinositol (3,4,5)-trisphosphate-dependent Rac exchanger 1 (P-Rex1), one of the Rac-specific guanine nucleotide exchange factors previously known for G protein-coupled receptor signaling, we sought to determine whether Rac-guanine nucleotide exchange factor is nodal for signal integration and potential target for drug intervention. METHODS AND RESULTS Using gene deletion and small interference RNA silencing approach, we investigated the role of P-Rex1 in human lung microvascular endothelial cells. Tumor necrosis factor α (TNF-α) exposure led to disruption of endothelial junctions, and silencing P-Rex1 protected junction integrity. TNF-α stimulated Rac activation and reactive oxygen species production in a P-Rex1-dependent manner. Removal of P-Rex1 significantly reduced intercellular adhesion molecule-1 expression, polymorphonuclear leukocyte transendothelial migration, and leukocyte sequestration in TNF-α-challenged mouse lungs. The P-Rex1 knockout mice were also refractory to lung vascular hyperpermeability and edema in a lipopolysaccharide-induced sepsis model. CONCLUSIONS These results demonstrate for the first time that P-Rex1 expressed in endothelial cells is activated downstream of TNF-α, which is not a G protein-coupled receptor agonist. Our data identify P-Rex1 as a critical mediator of vascular barrier disruption. Targeting P-Rex1 may effectively protect against TNF-α- and lipopolysaccharide-induced endothelial junction disruption and vascular hyperpermeability.
Collapse
Affiliation(s)
- Ram P Naikawadi
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Metabotropic glutamate receptor 5 mediates phosphorylation of vascular endothelial cadherin and nuclear localization of β-catenin in response to homocysteine. Vascul Pharmacol 2012; 56:159-67. [PMID: 22285407 DOI: 10.1016/j.vph.2012.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 12/31/2011] [Accepted: 01/16/2012] [Indexed: 11/22/2022]
Abstract
Elevated plasma homocysteine (Hcy) is an independent risk factor for vascular disease and stroke in part by causing generalized endothelial dysfunction. A receptor that is sensitive to Hcy and its intracellular signaling systems has not been identified. β-catenin is a pleiotropic regulator of transcription and cell function. Using a brain microvascular endothelial cell line (bEnd.3), we tested the hypothesis that Hcy causes receptor-dependent nuclear translocation of β-catenin. Hcy increased phosphorylation of Y731 on vascular endothelial cadherin (VE-cadherin), a site involved in coupling β-catenin to VE-cadherin. This was blocked by inhibition of either metabotropic glutamate receptor 5 (mGluR5) or ionotropic glutamate receptor (NMDAr) and by shRNA knockdown of mGluR5. Expression of these receptors was confirmed by flow cytometry, immunohistochemistry, and western blotting. Directed pharmacology with specific agonists elucidated a signaling cascade where Hcy activates mGluR5 which activates NMDAr with subsequent PKC activation and uncoupling of the VE-cadherin/β-catenin complex. Moreover, Hcy caused a shift in localization of β-catenin from membrane-bound VE-cadherin to the cell nucleus, where it bound DNA, including a regulatory region of the gene for claudin-5, leading to reduced expression of claudin-5. Nuclear localization, DNA binding of β-catenin, and reduced claudin-5 expression were blocked by inhibition of mGluR5. Knockdown of mGluR5 expression with shRNA also rescued claudin-5 expression from the effects of Hcy treatment. These data uniquely identify mGluR5 as a master switch that drives β-catenin nuclear localization in vascular endothelium and regulates cell-cell coupling in response to elevated Hcy levels. These studies dissect a pharmacological opportunity for developing new therapeutic strategies in HHcy.
Collapse
|
27
|
Zhang RY, Li L, Wu J, Tang YQ. Vascular endothelial-cadherin: a possible link between endocytosis and ectodomain shedding. J Allergy Clin Immunol 2012; 129:266; author reply 266-7. [PMID: 21982111 DOI: 10.1016/j.jaci.2011.08.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 08/03/2011] [Indexed: 11/28/2022]
|
28
|
Hernandez NM, Casselbrant A, Joshi M, Johansson BR, Sumitran-Holgersson S. Antibodies to kidney endothelial cells contribute to a "leaky" glomerular barrier in patients with chronic kidney diseases. Am J Physiol Renal Physiol 2011; 302:F884-94. [PMID: 22189942 DOI: 10.1152/ajprenal.00250.2011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anti-endothelial cell antibodies (AECA) have been reported to cause endothelial dysfunction, but their clinical importance for tissue-specific endothelial cells is not clear. We hypothesized that AECA reactive with human kidney endothelial cells (HKEC) may cause renal endothelial dysfunction in patients with chronic kidney diseases. We report that a higher fraction (56%) of end-stage renal disease (ESRD) patients than healthy controls (5%) have AECA reactive against kidney endothelial cells (P <0.001). The presence of antibodies was associated with female gender (P < 0.001), systolic hypertension (P < 0.01), and elevated TNF-α (P < 0.05). These antibodies markedly decrease expression of both adherens and tight junction proteins VE-cadherin, claudin-1, and zonula occludens-1 and provoked a rapid increase in cytosolic free Ca(2+) and rearrangement of actin filaments in HKEC compared with controls. This was followed by an enhancement in protein flux and phosphorylation of VE-cadherin, events associated with augmented endothelial cell permeability. Additionally, kidney biopsies from ESRD patients with AECA but not controls demonstrated a marked decrease in adherens and tight junctions in glomerular endothelium, confirming our in vitro data. In summary, our data demonstrate a causal link between AECA and their capacity to induce alterations in glomerular vascular permeability.
Collapse
Affiliation(s)
- Nidia Maritza Hernandez
- Dept. of Transplantation Surgery, Laboratory for Transplantation and Regenerative Medicine, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska Science Park, Medicinaregatan 8A, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
29
|
A kinetic study of the cytokine/chemokines levels and disruption of blood-brain barrier in infant rats after pneumococcal meningitis. J Neuroimmunol 2011; 233:12-7. [DOI: 10.1016/j.jneuroim.2010.10.035] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2010] [Revised: 10/23/2010] [Accepted: 10/29/2010] [Indexed: 11/21/2022]
|
30
|
|
31
|
Cardoso FL, Brites D, Brito MA. Looking at the blood-brain barrier: molecular anatomy and possible investigation approaches. ACTA ACUST UNITED AC 2010; 64:328-63. [PMID: 20685221 DOI: 10.1016/j.brainresrev.2010.05.003] [Citation(s) in RCA: 412] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 05/18/2010] [Accepted: 05/19/2010] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic and complex interface between blood and the central nervous system that strictly controls the exchanges between the blood and brain compartments, therefore playing a key role in brain homeostasis and providing protection against many toxic compounds and pathogens. In this review, the unique properties of brain microvascular endothelial cells and intercellular junctions are examined. The specific interactions between endothelial cells and basement membrane as well as neighboring perivascular pericytes, glial cells and neurons, which altogether constitute the neurovascular unit and play an essential role in both health and function of the central nervous system, are also explored. Some relevant pathways across the endothelium, as well as mechanisms involved in the regulation of BBB permeability, and the emerging role of the BBB as a signaling interface are addressed as well. Furthermore, we summarize some of the experimental approaches that can be used to monitor BBB properties and function in a variety of conditions and have allowed recent advances in BBB knowledge. Elucidation of the molecular anatomy and dynamics of the BBB is an essential step for the development of new strategies directed to maintain or restore BBB integrity and barrier function and ultimately preserve the delicate interstitial brain environment.
Collapse
Affiliation(s)
- Filipa Lourenço Cardoso
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | | | | |
Collapse
|
32
|
Spitzer AL, Chuang KI, Victorino GP, Kasravi B, Curran B, Lee D, Harris HW. Chylomicrons combined with endotoxin moderate microvascular permeability. Innate Immun 2010; 17:283-92. [PMID: 20423922 DOI: 10.1177/1753425910369849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Triglyceride-rich lipoprotein-bound endotoxin (CM-LPS) inhibits the host innate immune response to sepsis by attenuating the hepatocellular response to pro-inflammatory cytokine stimulation. This 'cytokine tolerance' in hepatocytes is a transient, receptor-dependent process that correlates with internalization of CM-LPS via low density lipoprotein (LDL) receptors. Since endothelial cells are integral to the immune response and similarly express LDL receptors, we hypothesized that CM-LPS could be internalized and ultimately attenuate the deleterious effects of pro-inflammatory molecules like tumor necrosis factor-α (TNF-α) and platelet activating factor (PAF) on endothelial permeability. Here, we show that CM-LPS complexes induce cytokine tolerance in endothelial cells. In rats, TNF-α increased hydraulic conductivity 2.5-fold over baseline and PAF increased it 5-fold; but, pretreatment with CM-LPS or an attenuated analog (CM-LPS*) inhibited these changes. Nuclear/cytoplasmic levels of p65 were reduced after TNF-α-stimulation in endothelial cell monolayers pretreated with CM-LPS, a finding consistent with inhibition of nuclear factor (NF)-κB translocation. Also consistent with inhibition was stabilized intercellular adhesion, as illustrated with antibody to VE-cadherin using confocal microscopy. These results provide additional support for the integral role of lipoproteins in the innate immune response to infection and lend further credence to developing lipid-based therapy for Gram-negative sepsis.
Collapse
Affiliation(s)
- Austin L Spitzer
- University of California Surgical Research Laboratory at San Francisco General Hospital, University of California at San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143-0104, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Heupel WM, Efthymiadis A, Schlegel N, Müller T, Baumer Y, Baumgartner W, Drenckhahn D, Waschke J. Endothelial barrier stabilization by a cyclic tandem peptide targeting VE-cadherin transinteraction in vitro and in vivo. J Cell Sci 2009; 122:1616-25. [DOI: 10.1242/jcs.040212] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Inflammatory stimuli result in vascular leakage with potentially life threatening consequences. As a key barrier component, loss of vascular endothelial (VE-) cadherin-mediated adhesion often precedes endothelial breakdown. This study aimed to stabilize VE-cadherin transinteraction and endothelial barrier function using peptides targeting the VE-cadherin adhesive interface. After modelling the transinteracting VE-cadherin structure, an inhibiting single peptide (SP) against a VE-cadherin binding pocket was selected, which specifically blocked VE-cadherin transinteraction as analyzed by single molecule atomic force microscopy (AFM). The tandem peptide (TP) consisting of two SP sequences in tandem was designed to strengthen VE-cadherin adhesion by simultaneously binding and cross-bridging two interacting cadherin molecules. Indeed, in AFM experiments TP specifically rendered VE-cadherin transinteraction resistant against an inhibitory monoclonal antibody. Moreover, TP reduced VE-cadherin lateral mobility and enhanced binding of VE-cadherin-coated microbeads to cultured endothelial cells, but acted independently of the actin cytoskeleton. TP also stabilized endothelial barrier properties against the Ca2+ ionophore A23187 and the inhibitory antibody. Finally, TP abolished endothelial permeability increase induced by tumour necrosis factor-α in microperfused venules in vivo. Stabilization of VE-cadherin adhesion by cross-bridging peptides may therefore be a novel therapeutic approach for the treatment of vascular hyperpermeability.
Collapse
Affiliation(s)
- Wolfgang-Moritz Heupel
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, D-97070 Würzburg, Germany
| | - Athina Efthymiadis
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, D-97070 Würzburg, Germany
| | - Nicolas Schlegel
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, D-97070 Würzburg, Germany
| | - Thomas Müller
- University of Würzburg, Department of Botany I, Julius-von-Sachs-Platz 2, D-97082 Würzburg, Germany
| | - Yvonne Baumer
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, D-97070 Würzburg, Germany
| | - Werner Baumgartner
- RWTH Aachen, Institute of Biology II, Kopernikusstr. 16, D-52056 Aachen, Germany
| | - Detlev Drenckhahn
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, D-97070 Würzburg, Germany
| | - Jens Waschke
- University of Würzburg, Institute of Anatomy and Cell Biology, Koellikerstr. 6, D-97070 Würzburg, Germany
| |
Collapse
|
34
|
Nagababu E, Usatyuk PV, Enika D, Natarajan V, Rifkind JM. Vascular endothelial barrier dysfunction mediated by amyloid-beta proteins. J Alzheimers Dis 2009; 17:845-54. [PMID: 19542618 PMCID: PMC2852470 DOI: 10.3233/jad-2009-1104] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neuronal inflammation is very common in Alzheimer's disease (AD). This inflammation can be caused by infiltration of neutrophils across the blood brain barrier. Endothelial permeability changes are required for the infiltration of high molecular weight components to the brain. Deposition of toxic amyloid-beta (A beta) fibrils in the cerebral vasculature, as well as in brain neurons, has been implicated in the development of AD. This study investigates the effect of A beta fibrils on the permeability of the endothelium and the mechanism for the observed permeability changes. A beta(1-40) and A beta(1-42) fibrils, but not monomers, were found to increase permeability of bovine pulmonary arterial endothelial cells in a dose- and time dependent manner as detected by transendothelial electrical resistance. This increase in permeability is only partially (25%) inhibited by catalase and is not associated with an increase in cytosolic Ca+2 or tyrosine phosphorylation. These results indicate that hydrogen peroxide is not the primary mediator for the permeability changes. Treatment of cells with both amyloid fibrils resulted in stress fiber formation, disruption and aggregation of actin filaments, and cellular gap formation. The results of this study reveal that A beta increases the permeability of endothelium by inducing change in the cytoskeleton network.
Collapse
Affiliation(s)
- Enika Nagababu
- Molecular Dynamics Section, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224
| | - Peter V. Usatyuk
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Divya Enika
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Viswanathan Natarajan
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Joseph M. Rifkind
- Molecular Dynamics Section, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224
| |
Collapse
|
35
|
|
36
|
Oroszlán M, Bieri M, Ligeti N, Farkas A, Koestner SC, Meier B, Mohacsi PJ. Proliferation Signal Inhibitor–induced Decrease of Vascular Endothelial Cadherin Expression and Increase of Endothelial Permeability In Vitro Are Prevented by an Anti-oxidant. J Heart Lung Transplant 2008; 27:1311-8. [DOI: 10.1016/j.healun.2008.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 08/19/2008] [Accepted: 08/26/2008] [Indexed: 10/21/2022] Open
|
37
|
Sandoval KE, Witt KA. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis 2008; 32:200-19. [PMID: 18790057 DOI: 10.1016/j.nbd.2008.08.005] [Citation(s) in RCA: 765] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 07/29/2008] [Accepted: 08/10/2008] [Indexed: 01/01/2023] Open
Abstract
The blood-brain barrier (BBB) is formed by the endothelial cells of cerebral microvessels, providing a dynamic interface between the peripheral circulation and the central nervous system. The tight junctions (TJs) between the endothelial cells serve to restrict blood-borne substances from entering the brain. Under ischemic stroke conditions decreased BBB TJ integrity results in increased paracellular permeability, directly contributing to cerebral vasogenic edema, hemorrhagic transformation, and increased mortality. This loss of TJ integrity occurs in a phasic manner, which is contingent on several interdependent mechanisms (ionic dysregulation, inflammation, oxidative and nitrosative stress, enzymatic activity, and angiogenesis). Understanding the inter-relation of these mechanisms is critical for the development of new therapies. This review focuses on those aspects of ischemic stroke impacting BBB TJ integrity and the principle regulatory pathways, respective to the phases of paracellular permeability.
Collapse
Affiliation(s)
- Karin E Sandoval
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL 62026, USA
| | | |
Collapse
|
38
|
Chen L, Borozan I, Milkiewicz P, Sun J, Meng X, Coltescu C, Edwards AM, Ostrowski MA, Guindi M, Heathcote EJ, McGilvray ID. Gene expression profiling of early primary biliary cirrhosis: possible insights into the mechanism of action of ursodeoxycholic acid. Liver Int 2008; 28:997-1010. [PMID: 18422935 DOI: 10.1111/j.1478-3231.2008.01744.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
OBJECTIVES Primary biliary cirrhosis (PBC) is a poorly understood disease, both in terms of its pathogenesis and the mechanism of action of its most common treatment, ursodeoxycholic acid (UDCA). We used gene expression profiling to compare liver tissue from treatment-naïve and UDCA-treated patients in order to outline some of the molecular changes associated with PBC and its treatment. PATIENTS AND EXPERIMENTAL DESIGN: Liver biopsy specimens from non-cirrhotic, treatment-naïve (n=11) patients were compared with biopsies from UDCA-treated patients (n=20) and with 10 normal, healthy female controls. Gene expression was determined using a 19K cDNA microarray. In order to determine whether the observed changes in gene expression levels were specific to PBC or generic to liver damage overall, PBC samples were also compared with chronically diseased [48 hepatitis C virus (HCV), 18 hepatitis B virus (HBV)] and acutely stressed liver tissue (25 liver biopsies taken after reperfusion of liver transplant grafts). RESULTS We found a gene signature specific to PBC (P<or=0.012), containing biologically plausible genes (221 genes with adjusted P</=0.05). Differences in the expression of selected genes were confirmed using real-time polymerase chain reaction. When gene expression from non-cirrhotic UDCA-treated (n=20) and UDCA-naïve liver tissue was compared, we found a striking downregulation of a number of genes involved in protein biosynthetic pathways. CONCLUSIONS These studies highlight the genes associated with both treatment-naïve and UDCA-treated PBC, and suggest that the effects of UDCA are mediated, at least in part, via a modulation of protein biosynthesic pathways.
Collapse
Affiliation(s)
- Limin Chen
- Banting and Best Department of Medical Research, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Fürst R, Bubik MF, Bihari P, Mayer BA, Khandoga AG, Hoffmann F, Rehberg M, Krombach F, Zahler S, Vollmar AM. Atrial natriuretic peptide protects against histamine-induced endothelial barrier dysfunction in vivo. Mol Pharmacol 2008; 74:1-8. [PMID: 18413663 DOI: 10.1124/mol.108.045773] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Endothelial barrier dysfunction is a hallmark of many severe pathologies, including sepsis or atherosclerosis. The cardiovascular hormone atrial natriuretic peptide (ANP) has increasingly been suggested to counteract endothelial leakage. Surprisingly, the precise in vivo relevance of these observations has never been evaluated. Thus, we aimed to clarify this issue and, moreover, to identify the permeability-controlling subcellular systems that are targeted by ANP. Histamine was used as important pro-inflammatory, permeability-increasing stimulus. Measurements of fluorescein isothiocyanate (FITC)-dextran extravasation from venules of the mouse cremaster muscle and rat hematocrit values were performed to judge changes of endothelial permeability in vivo. It is noteworthy that ANP strongly reduced the histamine-evoked endothelial barrier dysfunction in vivo. In vitro, ANP blocked the breakdown of transendothelial electrical resistance (TEER) induced by histamine. Moreover, as judged by immunocytochemistry and Western blot analysis, ANP inhibited changes of vascular endothelial (VE)-cadherin, beta-catenin, and p120(ctn) morphology; VE-cadherin and myosin light chain 2 (MLC2) phosphorylation; and F-actin stress fiber formation. These changes seem to be predominantly mediated by the natriuretic peptide receptor (NPR)-A, but not by NPR-C. In summary, we revealed ANP as a potent endothelial barrier protecting agent in vivo and identified adherens junctions and the contractile apparatus as subcellular systems targeted by ANP. Thus, our study highlights ANP as an interesting pharmacological compound opening new therapeutic options for preventing endothelial leakage.
Collapse
Affiliation(s)
- Robert Fürst
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Butenandtstr. 5-13, 81377 Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hu G, Place AT, Minshall RD. Regulation of endothelial permeability by Src kinase signaling: vascular leakage versus transcellular transport of drugs and macromolecules. Chem Biol Interact 2008; 171:177-89. [PMID: 17897637 PMCID: PMC3001132 DOI: 10.1016/j.cbi.2007.08.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 06/01/2007] [Accepted: 08/02/2007] [Indexed: 12/17/2022]
Abstract
An important function of the endothelium is to regulate the transport of liquid and solutes across the semi-permeable vascular endothelial barrier. Two cellular pathways have been identified controlling endothelial barrier function. The normally restrictive paracellular pathway, which can become "leaky" during inflammation when gaps are induced between endothelial cells at the level of adherens and tight junctional complexes, and the transcellular pathway, which transports plasma proteins the size of albumin via transcytosis in vesicle carriers originating from cell surface caveolae. During non-inflammatory conditions, caveolae-mediated transport may be the primary mechanism of vascular permeability regulation of fluid phase molecules as well as lipids, hormones, and peptides that bind avidly to albumin. Src family protein tyrosine kinases have been implicated in the upstream signaling pathways that lead to endothelial hyperpermeability through both the paracellular and transcellular pathways. Endothelial barrier dysfunction not only affects vascular homeostasis and cell metabolism, but also governs drug delivery to underlying cells and tissues. In this review of the field, we discuss the current understanding of Src signaling in regulating paracellular and transcellular endothelial permeability pathways and effects on endogenous macromolecule and drug delivery.
Collapse
Affiliation(s)
- Guochang Hu
- Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois at Chicago, Chicago, IL 60612, United States.
| | | | | |
Collapse
|
41
|
|
42
|
Ushio-Fukai M, Frey RS, Fukai T, Malik AB. Chapter 8 Reactive Oxygen Species and Endothelial Permeability. CURRENT TOPICS IN MEMBRANES 2008. [DOI: 10.1016/s1063-5823(08)00208-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
43
|
Petreaca ML, Yao M, Liu Y, DeFea K, Martins-Green M. Transactivation of vascular endothelial growth factor receptor-2 by interleukin-8 (IL-8/CXCL8) is required for IL-8/CXCL8-induced endothelial permeability. Mol Biol Cell 2007; 18:5014-23. [PMID: 17928406 PMCID: PMC2096609 DOI: 10.1091/mbc.e07-01-0004] [Citation(s) in RCA: 168] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Revised: 09/28/2007] [Accepted: 10/02/2007] [Indexed: 12/30/2022] Open
Abstract
Interleukin-8 (IL-8/CXCL8) is a chemokine that increases endothelial permeability during early stages of angiogenesis. However, the mechanisms involved in IL-8/CXCL8-induced permeability are poorly understood. Here, we show that permeability induced by this chemokine requires the activation of vascular endothelial growth factor receptor-2 (VEGFR2/fetal liver kinase 1/KDR). IL-8/CXCL8 stimulates VEGFR2 phosphorylation in a VEGF-independent manner, suggesting VEGFR2 transactivation. We investigated the possible contribution of physical interactions between VEGFR2 and the IL-8/CXCL8 receptors leading to VEGFR2 transactivation. Both IL-8 receptors interact with VEGFR2 after IL-8/CXCL8 treatment, and the time course of complex formation is comparable with that of VEGFR2 phosphorylation. Src kinases are involved upstream of receptor complex formation and VEGFR2 transactivation during IL-8/CXCL8-induced permeability. An inhibitor of Src kinases blocked IL-8/CXCL8-induced VEGFR2 phosphorylation, receptor complex formation, and endothelial permeability. Furthermore, inhibition of the VEGFR abolishes RhoA activation by IL-8/CXCL8, and gap formation, suggesting a mechanism whereby VEGFR2 transactivation mediates IL-8/CXCL8-induced permeability. This study points to VEGFR2 transactivation as an important signaling pathway used by chemokines such as IL-8/CXCL8, and it may lead to the development of new therapies that can be used in conditions involving increases in endothelial permeability or angiogenesis, particularly in pathological situations associated with both IL-8/CXCL8 and VEGF.
Collapse
Affiliation(s)
- Melissa L. Petreaca
- *Graduate Program in Cell, Molecular, and Developmental Biology
- Department of Cell Biology and Neuroscience, and
| | - Min Yao
- Department of Cell Biology and Neuroscience, and
| | - Yan Liu
- Department of Cell Biology and Neuroscience, and
| | - Kathryn DeFea
- *Graduate Program in Cell, Molecular, and Developmental Biology
- Division of Biomedical Sciences, University of California, Riverside, Riverside, CA 92521
| | - Manuela Martins-Green
- *Graduate Program in Cell, Molecular, and Developmental Biology
- Department of Cell Biology and Neuroscience, and
| |
Collapse
|
44
|
Paye JMD, Akers RM, Huckle WR, Forsten-Williams K. Autocrine production of insulin-like growth factor-I (IGF-I) affects paracellular transport across epithelial cells in vitro. ACTA ACUST UNITED AC 2007; 14:85-98. [PMID: 17668352 DOI: 10.1080/15419060701463116] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Autocrine production of growth factors can have significant effects on cell activity. We report for the first time that autocrine production of insulin-like growth factor-I (IGF-I) alters paracellular transport across bovine mammary epithelial cells in vitro. Paracellular transport was assessed by measuring phenol red transport across mammary alveolar cells-large T antigen (MAC-T cells) derived from parental mammary epithelial cells, cultured on porous membranes and compared with two different transfected MAC-T cell lines that constitutively secrete IGF-I. Phenol red transport was essentially blocked in parental cell culture after six days, while IGF-I secreting cells provided essentially no barrier. Surprisingly, neither co-culture studies between parental and IGF-I-secreting cells nor addition of exogenous IGF-I or IGF-binding protein-3 reversed the phenol red transport properties. IGF-I-secreting cells did however express lower levels of the junction components occludin and E-cadherin than parental cells, suggesting that localized autocrine IGF-I activity might lead to increased permeability via changes in both the tight and adherens junction protein levels.
Collapse
Affiliation(s)
- Julie M D Paye
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, Blacksburg 24061, Virginia, USA
| | | | | | | |
Collapse
|
45
|
Wallez Y, Huber P. Endothelial adherens and tight junctions in vascular homeostasis, inflammation and angiogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2007; 1778:794-809. [PMID: 17961505 DOI: 10.1016/j.bbamem.2007.09.003] [Citation(s) in RCA: 347] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Revised: 09/03/2007] [Accepted: 09/05/2007] [Indexed: 11/25/2022]
Abstract
Endothelial cells lining the vessel wall are connected by adherens, tight and gap junctions. These junctional complexes are related to those found at epithelial junctions but with notable changes in terms of specific molecules and organization. Endothelial junctional proteins play important roles in tissue integrity but also in vascular permeability, leukocyte extravasation and angiogenesis. In this review, we will focus on specific mechanisms of endothelial tight and adherens junctions.
Collapse
Affiliation(s)
- Yann Wallez
- Vascular Pathophysiology Laboratory, Inserm U882 38054 Grenoble, France
| | | |
Collapse
|
46
|
Paul R, Angele B, Popp B, Klein M, Riedel E, Pfister HW, Koedel U. Differential regulation of blood–brain barrier permeability in brain trauma and pneumococcal meningitis—role of Src kinases. Exp Neurol 2007; 203:158-67. [PMID: 17010340 DOI: 10.1016/j.expneurol.2006.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2006] [Revised: 07/26/2006] [Accepted: 08/01/2006] [Indexed: 11/27/2022]
Abstract
Increased vascular permeability causing vasogenic brain edema is characteristic for many acute neurological diseases such as stroke, brain trauma, and meningitis. Src family kinases, especially c-Src, play an important role in regulating blood-brain barrier permeability in response to VEGF, but also mediate leukocyte function and cytokine signalling. Here we demonstrate that pharmacological inhibition of Src or c-Src deficiency does not influence cerebrospinal fluid (CSF) pleocytosis, brain edema formation, and bacterial outgrowth during experimental pneumococcal meningitis despite the increased cerebral expression of inflammatory chemokines, such as IL-6, CCL-9, CXCL-1, CXCL-2 and G-CSF as determined by protein array analysis. In contrast, inhibition of Src significantly reduced brain edema formation, lesion volume, and clinical worsening in cold-induced brain injury without decreasing cytokine/chemokine expression. While brain trauma was associated with increased cerebral VEGF formation, VEGF levels significantly declined during pneumococcal meningitis. Therefore, we conclude that in brain trauma blood-brain barrier tightness is regulated by the VEGF/Src pathway whereas c-Src does not influence brain edema formation and leukocyte function during bacterial meningitis.
Collapse
Affiliation(s)
- Robert Paul
- Department of Neurology, Klinikum Grosshadern, Ludwig-Maximilians University, Marchioninistr. 15, D-81377 Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
47
|
Menon C, Ghartey A, Canter R, Feldman M, Fraker DL. Tumor necrosis factor-alpha damages tumor blood vessel integrity by targeting VE-cadherin. Ann Surg 2006; 244:781-91. [PMID: 17060772 PMCID: PMC1856603 DOI: 10.1097/01.sla.0000231723.81218.72] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Isolated limb perfusion using high-dose human tumor necrosis factor-alpha with melphalan is effective therapy for bulky extremity in-transit melanoma and sarcoma. OBJECTIVE While it is widely accepted that melphalan is a DNA alkylating agent, the mechanism of selective antitumor effect of tumor necrosis factor-alpha is unclear. METHODS AND RESULTS Electron microscopic analyses of human melanoma biopsies, pre- and post-melphalan perfusion, showed that the addition of tumor necrosis factor-alpha caused gapping between endothelial cells by 3 to 6 hours post-treatment followed by vascular erythrostasis in treated tumors. In human melanoma xenografts raised in mice, tumor necrosis factor-alpha selectively increased tumor vascular permeability by 3 hours and decreased tumor blood flow by 6 hours post-treatment relative to treated normal tissue. In an in vitro tumor endothelial cell model, tumor necrosis factor-alpha caused vascular endothelial adherens junction protein, VE-cadherin, to relocalize within the cell membrane away from cell-cell junctions leading to gapping between endothelial cells by 3 to 6 hours post-treatment. Phosphotyrosinylation was a prerequisite for movement of VE-cadherin away from endothelial cell junctions and for gapping between endothelial cells. Clinical isolated limb perfusion tumor specimens, at 3 hours postperfusion, showed a discontinuous and irregular pattern of VE-cadherin expression at endothelial cell junctions when compared with normal (skin) or pretreatment tumor tissue. CONCLUSIONS Together, the data suggest that tumor necrosis factor-alpha selectively damages the integrity of tumor vasculature by disrupting VE-cadherin complexes at vascular endothelial cell junctions leading to gapping between endothelial cells, causing increased vascular leak and erythrostasis in tumors. VE-cadherin appears to be a potentially good target for selective antitumor therapy.
Collapse
MESH Headings
- Animals
- Antigens, CD/drug effects
- Antigens, CD/metabolism
- Biopsy
- Blood Flow Velocity
- Cadherins/drug effects
- Cadherins/metabolism
- Cell Line, Tumor
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/ultrastructure
- Flow Cytometry
- Humans
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/metabolism
- Mice
- Mice, Nude
- Microscopy, Electron
- Neoplasm Transplantation
- Skin/blood supply
- Skin/physiopathology
- Skin/ultrastructure
- Skin Neoplasms/blood supply
- Skin Neoplasms/drug therapy
- Skin Neoplasms/metabolism
- Transplantation, Heterologous
- Treatment Outcome
- Tumor Necrosis Factor-alpha/therapeutic use
Collapse
Affiliation(s)
- Chandrakala Menon
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
48
|
Angelini DJ, Hyun SW, Grigoryev DN, Garg P, Gong P, Singh IS, Passaniti A, Hasday JD, Goldblum SE. TNF-alpha increases tyrosine phosphorylation of vascular endothelial cadherin and opens the paracellular pathway through fyn activation in human lung endothelia. Am J Physiol Lung Cell Mol Physiol 2006; 291:L1232-45. [PMID: 16891393 DOI: 10.1152/ajplung.00109.2006] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Tumor necrosis factor (TNF)-alpha is a key mediator of sepsis-associated multiorgan failure, including the acute respiratory distress syndrome. We examined the role of protein tyrosine phosphorylation in TNF-alpha-induced pulmonary vascular permeability. Postconfluent human lung microvascular and pulmonary artery endothelial cell (EC) monolayers exposed to human recombinant TNF-alpha displayed a dose- and time-dependent increase in transendothelial [(14)C]albumin flux in the absence of EC injury. TNF-alpha also increased tyrosine phosphorylation of EC proteins, and several substrates were identified as the zonula adherens proteins vascular endothelial (VE)-cadherin, and beta-catenin, gamma-catenin, and p120 catenin (p120(ctn)). Prior protein tyrosine kinase (PTK) inhibition protected against the TNF-alpha effect. TNF-alpha activated multiple PTKs, including src family PTKs. Prior PTK inhibition with the src-selective agents PP1 and PP2 each protected against approximately 60% of the TNF-alpha-induced increment in [(14)C]albumin flux. PP2 also blocked TNF-alpha-induced tyrosine phosphorylation of VE-cadherin, gamma-catenin, and p120(ctn). To identify which src family kinase(s) was required for TNF-alpha-induced vascular permeability, small interfering RNA (siRNA) targeting each of the three src family PTKs expressed in human EC, c-src, fyn, and yes, were introduced into the barrier function assay. Only fyn siRNA protected against the TNF-alpha effect, whereas the c-src and yes siRNAs did not. These combined data suggest that TNF-alpha regulates the pulmonary vascular endothelial paracellular pathway, in part, through fyn activation.
Collapse
Affiliation(s)
- Daniel J Angelini
- Division of Infectious Disease and Pulmonary, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Rao RS, Howard CA, Teague TK. Pulmonary endothelial permeability is increased by fluid from packed red blood cell units but not by fluid from clinically-available washed units. ACTA ACUST UNITED AC 2006; 60:851-8. [PMID: 16612308 DOI: 10.1097/01.ta.0000195727.38068.7d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Massive transfusions are a risk factor for acute respiratory distress syndrome (ARDS) in severely injured patients. Neutrophil priming has been proposed to be an integral part of the early inflammatory response to trauma. To complement that work, we studied another major cell type involved in inflammation: the endothelial cell. Our hypothesis was that soluble factors from units of leukoreduced packed red blood cells (PRBC) directly increase pulmonary endothelial permeability. We also determined whether fluid from clinically-available washed PRBC units affects endothelial permeability. METHODS As a measure of permeability, transendothelial electrical resistance (TER) was determined across monolayers of a human pulmonary microvascular endothelial cell line after addition of full-strength, diluted, and washed PRBC fluid. Monolayers were stained with phalloidin to assess intercellular space. Storage solution Adsol-1 was tested alone to determine additive component effects on TER. RESULTS PRBC fluid decreased TER and increased intercellular space, both of which indicate an increase in endothelial monolayer permeability. PRBC fluid diluted to 2% and washed PRBC fluid did not decrease TER and thereby did not change endothelial permeability. Likewise, Adsol-1 did not duplicate the dramatic decrease in TER seen with the PRBC fluid. CONCLUSIONS Fluid from stored PRBC units contains a soluble, transferable factor that directly increases endothelial permeability. Fluid from washed PRBC units, currently available for patients with immunoglobulin A allergies, does not. This study complements previous work of others that demonstrated that neutrophil priming by PRBC fluid is abrogated by washing. Now that two cell types have been shown to respond more favorably to washed PRBC in vitro, clinical studies should be initiated to investigate whether use of washed PRBC reduces ARDS following transfusions in trauma patients.
Collapse
Affiliation(s)
- Rohini S Rao
- Department of Surgery, University of Oklahoma College of Medicine, Tulsa, 74135, USA
| | | | | |
Collapse
|
50
|
Zhang Y, Zhao S, Gu Y, Lewis DF, Alexander JS, Wang Y. Effects of peroxynitrite and superoxide radicals on endothelial monolayer permeability: potential role of peroxynitrite in preeclampsia. ACTA ACUST UNITED AC 2006; 12:586-92. [PMID: 16325748 DOI: 10.1016/j.jsgi.2005.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2005] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Increased endothelial permeability is associated with increased oxidative stress in the maternal vasculature in women with preeclampsia. This study was to determine if oxidative stress elicited by peroxynitrite could lead to an increase in endothelial permeability. METHODS Endothelial oxidative stress was produced by adding 3-morpholinosydnonimine (SIN-1, a peroxynitrite generator) to the cell culture. Confluent endothelial cells (ECs) grown in cell culture inserts were treated with SIN-1 at a concentration of 0.5 mM alone or in combination with MnTMPyP (a peroxynitrite scavenger) or superoxide dismutase (SOD). EC permeability was determined by measuring EC electrical resistance (ER) and horseradish peroxide (HRP) leakage. Data are presented as means +/- SE and analyzed by analysis of variance (ANOVA). Junctional protein expression and distribution for vascular endothelial (VE)-cadherin, occludin, and phosphorylated focal adhesion kinase (FAK) at tyrosine 397 [pY397] were examined by fluorescent staining of ECs. RESULTS First, ER was significantly reduced and HRP leakage was significantly increased in ECs treated with SIN-1 compared to those in control cells, ER: 26.97 +/- 1.41 versus 42.27 +/- 0.40 Omega.cm2, P <.01; HRP: 0.26 +/- 0.07 versus 0.02 +/- 0.01 OD 470 nm, P <.01, respectively. Second, cells treated with SIN-1 showed formation of gaps and disorganized VE-cadherin and occludin distribution at cell contact regions. FAK[pY397] expression was completely lost in cells treated with SIN-1. Finally, these functional and morphologic changes in ECs induced by SIN-1 were blocked in cells pretreated with MnTMPyP and SOD. CONCLUSIONS Disorganization of junctional proteins and dephosphorylation of FAK[pY397] may account for the increased endothelial permeability induced by oxidative stress associated with preeclampsia.
Collapse
Affiliation(s)
- Yanping Zhang
- Department of Obstetrics and Gynecology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | | | | | | | | | |
Collapse
|