1
|
Fang Y, Qiu J, Xu Y, Wu Q, Huo XC, Liu SH. Ophiopogonin D Alleviates Sepsis-Induced Acute Lung Injury Through Improving Microvascular Endothelial Barrier Dysfunction via Inhibition of HIF-1α-VEGF Pathway. Cell Biochem Biophys 2025; 83:2519-2531. [PMID: 39890704 DOI: 10.1007/s12013-024-01661-7] [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] [Accepted: 12/23/2024] [Indexed: 02/03/2025]
Abstract
Pulmonary endothelial barrier dysfunction is a hallmark of sepsis-induced acute lung injury (ALI). Ophiopogonin D (OP-D), isolated from the roots of Ophiopogon japonicus, is involved in regulating inflammation, apoptosis and intestinal permeability. However, the role of OP-D in ALI has not been reported and the related mechanisms remain unclear. In this study, cecal ligation and puncture (CLP) was used to establish a septic ALI model in mice. We found that OP-D effectively alleviated lung pathological damage. Moreover, OP-D decreased pulmonary microvascular permeability, restrained the inflammatory response and apoptosis in murine lung tissues and LPS-exposed PMVECs. Specifically, OP-D exerted the beneficial effects via mediating the inactivation of HIF-1α-VEGF pathway, which was partly abrogated by the overexpression of HIF-1α. Collectively, our findings showed that OP-D protected against sepsis-induced ALI through improving pulmonary microvascular endothelial barrier dysfunction via suppressing HIF-1α-VEGF pathway.
Collapse
Affiliation(s)
- Yi Fang
- Department of Anesthesiology, Affiliated Changsha Central Hospital to Nanhua University, Changsha, 410016, Hunan, PR China
| | - Jun Qiu
- The first-affiliated hospital of Hunan normal university (The second tumor ward, Hunan Provincial People's Hospital), Changsha, 410006, Hunan, PR China
| | - Yu Xu
- Department of Anesthesiology, Affiliated Changsha Central Hospital to Nanhua University, Changsha, 410016, Hunan, PR China
| | - Qing Wu
- Department of Anesthesiology, Affiliated Changsha Central Hospital to Nanhua University, Changsha, 410016, Hunan, PR China
| | - Xing-Chen Huo
- Department of Anesthesiology, Affiliated Changsha Central Hospital to Nanhua University, Changsha, 410016, Hunan, PR China
| | - Song-Hua Liu
- Department of Anesthesiology, Affiliated Changsha Central Hospital to Nanhua University, Changsha, 410016, Hunan, PR China.
| |
Collapse
|
2
|
Chorazy N, Wojnar-Lason K, Gdula AM, Bakker D, Zuurbier CJ, Chlopicki S, Pacia MZ. Sodium-glucose co-transporters (SGLT2) inhibitors prevent lipid droplets formation in vascular inflammation or lipid overload by SGLT2-independent mechanism. Biomed Pharmacother 2025; 185:117967. [PMID: 40088775 DOI: 10.1016/j.biopha.2025.117967] [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: 12/23/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The formation of vascular lipid droplets (LDs) induced by vascular inflammation or lipid overload contributes to vascular pathophysiology in diabetes and cardiometabolic diseases, while sodium-glucose co-transporter 2 inhibitors (SGLT2-I) are beneficial in treating these conditions. Thus, we hypothesized that SGLT2-I would directly modify vascular LDs formation during vascular inflammation or lipid overload, and explored underlying mechanisms. METHODS LDs formation in isolated murine aorta from wild-type or SGLT2-KO animals was induced by either treatment with tumour necrosis factor (TNF) to induce vascular inflammation or using oleic acid (OA) to mimic lipid overload. Vascular LDs and markers of vascular inflammation were monitored through fluorescence microscopy. Pharmacological inhibitors of sodium-hydrogen exchanger 1 (NHE1), endothelial sodium channels (EnNaC), sodium-calcium exchanger (NCX), protein kinase C (PKC), and NOX1/4 were used to test their role in empagliflozin's effects on vascular LDs. RESULTS Empagliflozin, dapagliflozin or ertugliflozin inhibited LDs formation in aorta exposed to TNF or OA. Empagliflozin reduced vascular inflammation (based on ICAM-1) and TNF/OA-induced LDs formation. These effects persisted in SGLT2-KO mice. Inhibition of NHE1, PKC or NOX1/4 recapitulated empagliflozin's effects on TNF-induced vascular inflammation, without additional effects of empagliflozin. However, NHE1 inhibition was not involved in the SGLT2-independent reduction of OA-induced LDs formation by empagliflozin. CONCLUSIONS This is the first report demonstrating that SGLT2-I prevent the formation of LDs in the vasculature. Empagliflozin downregulates LDs formation in vascular inflammation or lipid overload via an SGLT2-independent mechanism. Empagliflozin's protective effects involve the NHE1/PKC/NOX pathway in the TNF response but not in the OA response.
Collapse
Affiliation(s)
- Natalia Chorazy
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, Krakow, Poland; Jagiellonian University, Doctoral School of Exact and Natural Sciences, Lojasiewicza 11, Krakow, Poland
| | - Kamila Wojnar-Lason
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, Krakow, Poland; Jagiellonian University, Medical Collage, Chair of Pharmacology, Grzegorzecka 16, Krakow, Poland; Augusta University, Medical College of Georgia, Vascular Biology Center, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Anna M Gdula
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, Krakow, Poland
| | - Diane Bakker
- Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Department of Anaesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care and Anaesthesiology (L.E.I.C.A.), Department of Anaesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Stefan Chlopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, Krakow, Poland; Jagiellonian University, Medical Collage, Chair of Pharmacology, Grzegorzecka 16, Krakow, Poland
| | - Marta Z Pacia
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, Krakow, Poland.
| |
Collapse
|
3
|
Marchenko VA, Zhilinskaya IN. Endothelial activation and dysfunction caused by influenza A virus ( Alphainfluenzavirus influenzae). Vopr Virusol 2024; 69:465-478. [PMID: 39841412 DOI: 10.36233/0507-4088-264] [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: 09/16/2024] [Indexed: 01/23/2025]
Abstract
Annual epidemics of influenza result in 3-5 million cases of severe illness and more than 600 000 deaths. Severe forms of influenza are usually characterized by vascular endothelial cells damage. Thus, influenza A viruses, including subtypes A(H1N1)pdm09, A(H3N2), as well as highly pathogenic avian influenza viruses, can infect the vascular endothelium, leading to activation and subsequent dysfunction of these cells. In turn, endothelial dysfunction resulting in systemic morphofunctional changes of endothelial cells, which leads to impaired vascular tone, thrombosis and other complications, and is also a risk factor and profoundly implicated in the pathogenesis of many cardiovascular diseases. Thus, endothelial dysfunction is an important aspect of the pathogenesis of severe influenza, which must be considered in the pathogenetic therapy of this infectious disease. The aim of the review is to analyze the causes and specify mechanisms of development of endothelial activation and dysfunction caused by influenza A virus.
Collapse
Affiliation(s)
- V A Marchenko
- North-Western State Medical University Named after I.I. Mechnikov
| | - I N Zhilinskaya
- North-Western State Medical University Named after I.I. Mechnikov
| |
Collapse
|
4
|
Breslin JW. Edema and lymphatic clearance: molecular mechanisms and ongoing challenges. Clin Sci (Lond) 2023; 137:1451-1476. [PMID: 37732545 PMCID: PMC11025659 DOI: 10.1042/cs20220314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/18/2023] [Accepted: 08/31/2023] [Indexed: 09/22/2023]
Abstract
Resolution of edema remains a significant clinical challenge. Conditions such as traumatic shock, sepsis, or diabetes often involve microvascular hyperpermeability, which leads to tissue and organ dysfunction. Lymphatic insufficiency due to genetic causes, surgical removal of lymph nodes, or infections, leads to varying degrees of tissue swelling that impair mobility and immune defenses. Treatment options are limited to management of edema as there are no specific therapeutics that have demonstrated significant success for ameliorating microvascular leakage or impaired lymphatic function. This review examines current knowledge about the physiological, cellular, and molecular mechanisms that control microvascular permeability and lymphatic clearance, the respective processes for interstitial fluid formation and removal. Clinical conditions featuring edema, along with potential future directions are discussed.
Collapse
Affiliation(s)
- Jerome W Breslin
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, FL, U.S.A
| |
Collapse
|
5
|
Shi X, Seidle KA, Simms KJ, Dong F, Chilian WM, Zhang P. Endothelial progenitor cells in the host defense response. Pharmacol Ther 2023; 241:108315. [PMID: 36436689 PMCID: PMC9944665 DOI: 10.1016/j.pharmthera.2022.108315] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Extensive injury of endothelial cells in blood vasculature, especially in the microcirculatory system, frequently occurs in hosts suffering from sepsis and the accompanied systemic inflammation. Pathological factors, including toxic components derived from invading microbes, oxidative stress associated with tissue ischemia/reperfusion, and vessel active mediators generated during the inflammatory response, are known to play important roles in mediating endothelial injury. Collapse of microcirculation and tissue edema developed from the failure of endothelial barrier function in vital organ systems, including the lung, brain, and kidney, are detrimental, which often predict fatal outcomes. The host body possesses a substantial capacity for maintaining vascular homeostasis and repairing endothelial damage. Bone marrow and vascular wall niches house endothelial progenitor cells (EPCs). In response to septic challenges, EPCs in their niche environment are rapidly activated for proliferation and angiogenic differentiation. In the meantime, release of EPCs from their niches into the blood stream and homing of these vascular precursors to tissue sites of injury are markedly increased. The recruited EPCs actively participate in host defense against endothelial injury and repair of damage in blood vasculature via direct differentiation into endothelial cells for re-endothelialization as well as production of vessel active mediators to exert paracrine and autocrine effects on angiogenesis/vasculogenesis. In recent years, investigations on significance of EPCs in host defense and molecular signaling mechanisms underlying regulation of the EPC response have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches for effective prevention and treatment of sepsis-induced vascular injury as well as vital organ system failure.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Kelly A Seidle
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Kevin J Simms
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America
| | - Ping Zhang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown, OH 44272, United States of America.
| |
Collapse
|
6
|
Raghavan S, Brishti MA, Collier DM, Leo MD. Hypoxia induces purinergic receptor signaling to disrupt endothelial barrier function. Front Physiol 2022; 13:1049698. [PMID: 36479340 PMCID: PMC9720161 DOI: 10.3389/fphys.2022.1049698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/10/2022] [Indexed: 09/15/2023] Open
Abstract
Blood-brain-barrier permeability is regulated by endothelial junctional proteins and is vital in limiting access to and from the blood to the CNS. When stressed, several cells, including endothelial cells, can release nucleotides like ATP and ADP that signal through purinergic receptors on these cells to disrupt BBB permeability. While this process is primarily protective, unrestricted, uncontrolled barrier disruption during injury or inflammation can lead to serious neurological consequences. Purinergic receptors are broadly classified into two families: the P1 adenosine and P2 nucleotide receptors. The P2 receptors are further sub-classified into the P2XR ion channels and the P2YR GPCRs. While ATP mainly activates P2XRs, P2YRs have a broader range of ligand selectivity. The P2Y1R, essential for platelet function, is reportedly ubiquitous in its expression. Prior studies using gene knockout and specific antagonists have shown that these approaches have neuroprotective effects following occlusive stroke. Here we investigated the expression of P2Y1R in primary cultured brain endothelial cells and its relation to the maintenance of BBB function. Results show that following in vitro hypoxia and reoxygenation, P2Y1R expression is upregulated in both control and diabetic cells. At the same time, endothelial junctional markers, ZO-1 and VE-cadherin, were downregulated, and endothelial permeability increased. siRNA knockdown of P2Y1R and MRS 2500 effectively blocked this response. Thus, we show that P2Y1R signaling in endothelial cells leads to the downregulation of endothelial barrier function.
Collapse
Affiliation(s)
| | | | | | - M. Dennis Leo
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
7
|
Basiglio CL, Crocenzi FA, Sánchez Pozzi EJ, Roma MG. Oxidative Stress and Localization Status of Hepatocellular Transporters: Impact on Bile Secretion and Role of Signaling Pathways. Antioxid Redox Signal 2021; 35:808-831. [PMID: 34293961 DOI: 10.1089/ars.2021.0021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Significance: Most hepatopathies are primarily or secondarily cholestatic in nature. Oxidative stress (OS) is a frequent trait among them, and impairs the machinery to generate bile by triggering endocytic internalization of hepatocellular transporters, thus causing cholestasis. This is critical, since it leads to accelerated transporter degradation, which could explain the common post-transcriptional downregulation of transporter expression in human cholestatic diseases. Recent Advances: The mechanisms involved in OS-induced hepatocellular transporter internalization are being revealed. Filamentous actin (F-actin) cytoskeleton disorganization and/or detachment of crosslinking actin proteins that afford transporter stability have been characterized as causal factors. Activation of redox-sensitive signaling pathways leading to changes in phosphorylation status of these structures is involved, including Ca2+-mediated activation of "classical" and "novel" protein kinase C (PKC) isoforms or redox-signaling cascades downstream of NADPH oxidase. Critical Issues: Despite the well-known occurrence of hepatocellular transporter internalization in human hepatopathies, the cholestatic implications of this phenomenon have been overlooked. Accordingly, no specific treatment has been established in the clinical practice for its prevention/reversion. Future Directions: We need to improve our knowledge on the pro-oxidant triggering factors and the multiple signaling pathways that mediate this oxidative injury in each cholestatic hepatopathy, so as to envisage tailor-made therapeutic strategies for each case. Meanwhile, administration of antioxidants or heme oxygenase-1 induction to elevate the hepatocellular levels of the endogenous scavenger bilirubin are promising alternatives that need to be re-evaluated and implemented. They may complement current treatments in cholestasis aimed to enhance transcriptional carrier expression, by providing membrane stability to the newly synthesized carriers. Antioxid. Redox Signal. 35, 808-831.
Collapse
Affiliation(s)
- Cecilia L Basiglio
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Fernando A Crocenzi
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Enrique J Sánchez Pozzi
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| | - Marcelo G Roma
- Instituto de Fisiología Experimental (IFISE), Facultad de Ciencias Bioquímicas y Farmacéuticas, CONICET, U.N.R., Rosario, Argentina
| |
Collapse
|
8
|
Han T, Lai Y, Jiang Y, Liu X, Li D. Influenza A virus infects pulmonary microvascular endothelial cells leading to microvascular leakage and release of pro-inflammatory cytokines. PeerJ 2021; 9:e11892. [PMID: 34414033 PMCID: PMC8344683 DOI: 10.7717/peerj.11892] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/10/2021] [Indexed: 11/20/2022] Open
Abstract
Objective To investigate the replication of influenza A virus A/Puerto Rico/8/34 (H1N1) in pulmonary microvascular endothelial cells and its effect on endothelial barrier function. Methods Human pulmonary microvascular endothelial cells were infected with influenza A/Puerto Rico/8/34 (H1N1) virus. Plaque reduction assay, real-time quantitative PCR, immunofluorescence staining, and western blot were used to elucidate the replication process of virus-infected endothelial cells. In addition, real-time quantitative PCR was used to detect the relative expression levels of mRNA of some inflammatory factors. The endothelial resistance assay was used to determine the permeability of the endothelial monolayer. Excavation and analysis of data from open databases, such as the GeneCards database, DAVID Bioinformatics Resources, STRING search tool, and DGIdb database determined the genes, proteins, and signal pathways related to microvascular leakage caused by the H1N1 virus, and predicted the drugs that could be effective for treatment. Results In vitro experiments showed that the influenza virus can infect endothelial cells, leading to a significant increase in the permeability of pulmonary microvascular endothelial cells and the release of pro-inflammatory cytokines, but does not efficiently replicate in endothelial cells. A total of 107 disease-related target genes were obtained from the Gene-cards database. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis showed that these genes mainly affected the pathways related to "Inflammatory bowel disease" (IBD), "Chagas disease" (American trypanosomiasis), "Influenza A", and also played a key role in anti-inflammation and regulation of immunity. After enrichment analysis, 46 hub genes were screened. A total of 42 FDA-approved drugs corresponding to the hub genes were screened from the DGIdb database, and these could be formulated for topical application. In addition, these drugs can be used to treat other diseases, including cancer, inflammatory diseases, immune system disorders, and cardiovascular diseases. Conclusion H1N1 influenza virus affects the barrier function of endothelial cells indirectly. Combined with bioinformatics tools, we can better understand the possible mechanism of action of influenza A (H1N1) virus causing pulmonary microvascular leakage and provide new clues for the treatment of pulmonary microvascular leakage.
Collapse
Affiliation(s)
- Tiantian Han
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanni Lai
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yong Jiang
- Respiratory Department, Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Xiaohong Liu
- Respiratory Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Danhua Li
- Clinical Lab, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Chi Y, Liu X, Chai J. A narrative review of changes in microvascular permeability after burn. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:719. [PMID: 33987417 PMCID: PMC8106041 DOI: 10.21037/atm-21-1267] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/17/2021] [Indexed: 12/16/2022]
Abstract
OBJECTIVE We aimed to review and discuss some of the latest research results related to post-burn pathophysiological changes and provide some clues for future study. BACKGROUND Burns are one of the most common and serious traumas and consist of a series of pathophysiological changes of thermal injury. Accompanied by thermal damage to skin and soft tissues, inflammatory mediators are released in large quantities. Changes in histamine, bradykinin, and cytokines such as vascular endothelial growth factor (VEGF), metabolic factors such as adenosine triphosphate (ATP), and activated neutrophils all affect the body's vascular permeability. METHODS We searched articles with subject words "microvascular permeability", "burn" "endothelium", and "endothelial barrier" in PubMed in English published from the beginning of database to Dec, 2020. CONCLUSIONS The essence of burn shock is the rapid and extensive fluid transfer in burn and non-burn tissue. After severe burns, the local and systemic vascular permeability increase, causing intravascular fluid extravasation, leading to a progressive decrease in effective circulation volume, an increase in systemic vascular resistance, a decrease in cardiac output, peripheral tissue edema, multiple organ failure, and even death. There are many cells, tissues, mediators and structures involved in the pathophysiological process of the damage to vascular permeability. Ulinastatin is a promising agent for this problem.
Collapse
Affiliation(s)
- Yunfei Chi
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| | - Xiangyu Liu
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| | - Jiake Chai
- Burn Institute, The Fourth Medical Center of the PLA General Hospital, Beijing, China
| |
Collapse
|
10
|
Dolmatova EV, Wang K, Mandavilli R, Griendling KK. The effects of sepsis on endothelium and clinical implications. Cardiovasc Res 2021; 117:60-73. [PMID: 32215570 PMCID: PMC7810126 DOI: 10.1093/cvr/cvaa070] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/03/2020] [Accepted: 03/20/2020] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Sepsis accounts for nearly 700 000 deaths in Europe annually and is caused by an overwhelming host response to infection resulting in organ failure. The endothelium is an active contributor to sepsis and as such represents a major target for therapy. During sepsis, endothelial cells amplify the immune response and activate the coagulation system. They are both a target and source of inflammation and serve as a link between local and systemic immune responses. In response to cytokines produced by immune cells, the endothelium expresses adhesion molecules and produces vasoactive compounds, inflammatory cytokines, and chemoattractants, thus switching from an anticoagulant to procoagulant state. These responses contribute to local control of infection, but systemic activation can lead to microvascular thrombosis, capillary permeability, hypotension, tissue hypoxia, and ultimately tissue damage. This review focuses on the role of the endothelium in leucocyte adhesion and transmigration as well as production of reactive oxygen and nitrogen species, microRNAs and cytokines, formation of signalling microparticles, and disseminated intravascular coagulation. We also discuss alterations in endothelial permeability and apoptosis. Finally, we review the diagnostic potential of endothelial markers and endothelial pathways as therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Elena V Dolmatova
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Keke Wang
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Rohan Mandavilli
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, Atlanta, GA 30322, USA
| |
Collapse
|
11
|
Ignorance of pre-ED healthcare setting is a factor leading to inappropriate initial antibiotic treatment of sepsis in ED and poor outcomes in ICU. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 53:532-541. [PMID: 31917133 DOI: 10.1016/j.jmii.2019.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 11/01/2019] [Accepted: 11/18/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Appropriate initial antibiotic therapy is critical for successfully treating sepsis. In the emergency department (ED), clinicians often rely on septic symptoms to guide empirical therapy. The aim of this study was to investigate whether history of contacting pre-ED healthcare setting is easy to be neglected and whether the patients received more inappropriate initial antibiotic therapy and developed poorer outcomes. METHODS Septic patients (n = 453) admitted from ED to the intensive care unit (ICU) between 2014 and 2017 were retrospectively selected. Appropriate antibiotic treatment or not was determined by checking whether the selected antibiotics can effectively eradicate the bacteria identified. Various indexes were compared between patients with appropriate and inappropriate initial antibiotic treatments, including septic symptoms (qSOFA scores) in ED, septic-severity change in ICU (SOFA-score ratios), and septic outcomes (APACHE II scores, stay length, 30-day survival probability). These indexes were also compared between pre-ED healthcare and pre-ED community patients. RESULTS In comparison with pre-ED community patients, pre-ED healthcare patients received more inappropriate initial antibiotic treatment in ED, showing poorer outcomes in ICU, including septic severity, stay-lengths in ICU and 30-day survival probabilities. Pre-ED settings is more significant than qSOFA scores to predict the inappropriate initial antibiotic treatment. CONCLUSIONS Pre-ED healthcare settings, which are indexes for infection with antibiotic resistant pathogens, are easy to be neglected in the first hour in ED. We suggested that standard operating procedure for getting enough information of pre-ED settings should be incorporated to the 1 h bundle of sepsis guideline.
Collapse
|
12
|
Characterization of Biomarker Levels in Crimean-Congo Hemorrhagic Fever and Hantavirus Fever with Renal Syndrome. Viruses 2019; 11:v11080686. [PMID: 31357521 PMCID: PMC6722556 DOI: 10.3390/v11080686] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/20/2019] [Accepted: 07/25/2019] [Indexed: 01/02/2023] Open
Abstract
Hemorrhagic fever with renal syndrome (HFRS) and Crimean-Congo hemorrhagic fever (CCHF) are important viral hemorrhagic fevers (VHF), especially in the Balkan region. Infections with Dobrava or Puumala orthohantavirus and Crimean-Congo hemorrhagic fever orthonairovirus can vary from a mild, nonspecific febrile illness, to a severe disease with a fatal outcome. The pathogenesis of both diseases is poorly understood, but it has been suggested that a host’s immune mechanism might influence the pathogenesis of the diseases and survival. The aim of our study is to characterize cytokine response in patients with VHF in association with the disease progression and viral load. Forty soluble mediators of the immune response, coagulation, and endothelial dysfunction were measured in acute serum samples in 100 HFRS patients and 70 CCHF patients. HFRS and CCHF patients had significantly increased levels of IL-6, IL-12p70, IP-10, INF-γ, TNF-α, GM-CSF, MCP-3, and MIP-1b in comparison to the control group. Interestingly, HFRS patients had higher concentrations of serum MIP-1α, MIP-1β, which promote activation of macrophages and NK cells. HFRS patients had increased concentrations of IFN-γ and TNF-α, while CCHF patients had significantly higher concentrations of IFN-α and IL-8. In both, CCHF and HFRS patients’ viral load significantly correlated with IP-10. Patients with fatal outcome had significantly elevated concentrations of IL-6, IFN-α2 and MIP-1α, while GRO-α, chemokine related to activation of neutrophils and basophils, was downregulated. Our study provided a comprehensive characterization of biomarkers released in the acute stages of CCHF and HFRS.
Collapse
|
13
|
Role of ERK1/2 in TNFα-induced internalization of Abcc2 in rat hepatocyte couplets. Biochem Pharmacol 2019; 164:311-320. [DOI: 10.1016/j.bcp.2019.04.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/22/2019] [Indexed: 01/10/2023]
|
14
|
Roma MG, Barosso IR, Miszczuk GS, Crocenzi FA, Pozzi EJS. Dynamic Localization of Hepatocellular Transporters: Role in Biliary Excretion and Impairment in Cholestasis. Curr Med Chem 2019; 26:1113-1154. [DOI: 10.2174/0929867325666171205153204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 12/25/2022]
Abstract
Bile flow generation is driven by the vectorial transfer of osmotically active compounds from sinusoidal blood into a confined space, the bile canaliculus. Hence, localization of hepatocellular transporters relevant to bile formation is crucial for bile secretion. Hepatocellular transporters are localized either in the plasma membrane or in recycling endosomes, from where they can be relocated to the plasma membrane on demand, or endocytosed when the demand decreases. The balance between endocytic internalization/ exocytic targeting to/from this recycling compartment is therefore the main determinant of the hepatic capability to generate bile, and to dispose endo- and xenobiotics. Furthermore, the exacerbated endocytic internalization is a common pathomechanisms in both experimental and human cholestasis; this results in bile secretory failure and, eventually, posttranslational transporter downregulation by increased degradation. This review summarizes the proposed structural mechanisms accounting for this pathological condition (e.g., alteration of function, localization or expression of F-actin or F-actin/transporter cross-linking proteins, and switch to membrane microdomains where they can be readily endocytosed), and the mediators implicated (e.g., triggering of “cholestatic” signaling transduction pathways). Lastly, we discussed the efficacy to counteract the cholestatic failure induced by transporter internalization of a number of therapeutic experimental approaches based upon the use of compounds that trigger exocytic targetting of canalicular transporters (e.g., cAMP, tauroursodeoxycholate). This therapeutics may complement treatments aimed to transcriptionally improve transporter expression, by affording proper localization and membrane stability to the de novo synthesized transporters.
Collapse
Affiliation(s)
- Marcelo G. Roma
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| | - Ismael R. Barosso
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| | - Gisel S. Miszczuk
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| | - Fernando A. Crocenzi
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| | - Enrique J. Sánchez Pozzi
- Instituto de Fisiologia Experimental (IFISE) - Facultad de Ciencias Bioquimicas y Farmaceuticas (CONICET - U.N.R.), S2002LRL, Rosario, Argentina
| |
Collapse
|
15
|
Song X, Liu Y, Dong L, Wang Y. Stromal-Interacting Molecule 1 (Stim1)/Orai1 Modulates Endothelial Permeability in Ventilator-Induced Lung Injury. Med Sci Monit 2018; 24:9413-9423. [PMID: 30589833 PMCID: PMC6322368 DOI: 10.12659/msm.911268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Increased endothelial permeability is involved in ventilator-induced lung injury (VILI). Stim1/Orai1 mediates store-operated Ca2+ activation, which modulates endothelial permeability. However, the underlying mechanisms of the Stim1/Orai1 pathway in VILI are poorly understood. Material/Methods Wistar rats were exposed to low tidal volume (7 mL/kg) or high tidal volume (40 mL/kg) ventilation. Human Lung Microvascular Endothelial Cells (HULEC) were subjected to 8% or 18% cyclic stretching (CS). BTP2 pretreatment was performed. Lung wet/dry weight ratio, histological changes of lung injury, and bronchoalveolar lavage fluid (BALF) protein were measured. Endothelial permeability and intracellular calcium concentration were evaluated in HULECs. Protein expression was determined by Western blotting. Results High tidal volume mechanical ventilation-induced lung injury (such as severe congestion and hemorrhage) and BTP2 pretreatment protected lungs from injury. The expression of Stim1, Orai1, and PKCα, lung wet/dry weight ratio, and BALF protein level significantly increased in the high tidal volume group compared to the control group and low tidal volume group. Importantly, BTP2 pretreatment alleviated the above-mentioned effects. Compared with exposure to 8% CS, the protein levels of Stim1, Orai1, and PKCα in HULECs significantly increased after exposure to 18% CS for 4 h, whereas BTP2 pretreatment significantly inhibited the increase (P<0.05). BTP2 pretreatment also suppressed increase of endothelial permeability and the intracellular calcium induced by 18% CS (P<0.05). Conclusions When exposed to high tidal volume or large-magnitude CS, Stim1 and Orai1 expression are upregulated, which further activates calcium-sensitive PKCα and results in calcium overload, endothelial hyperpermeability, and, finally, lung injury.
Collapse
Affiliation(s)
- Xiumei Song
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Yang Liu
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Ling Dong
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, Jinan, Shandong, China (mainland)
| | - Yuelan Wang
- Department of Anesthesiology, Qianfoshan Hospital, Shandong University, Jinan, Shandong, China (mainland)
| |
Collapse
|
16
|
Ozer EK, Goktas MT, Kilinc I, Toker A, Bariskaner H, Ugurluoglu C, Iskit AB. Infliximab alleviates the mortality, mesenteric hypoperfusion, aortic dysfunction, and multiple organ damage in septic rats. Can J Physiol Pharmacol 2017; 95:866-872. [DOI: 10.1139/cjpp-2016-0628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Tumor necrosis factor-alpha (TNF-α) is a pivotal mediator that triggers inflammatory process, oxidative stress, and multiple organ injury in sepsis. We investigated the effects of infliximab on survival, mesenteric artery blood flow (MBF), vascular reactivity, and oxidative and inflammatory injuries in cecal ligation and puncture (CLP)-induced sepsis. Wistar rats were divided into Sham, CLP, Sham+infliximab, and CLP+infliximab subgroups. Twenty-four hours before the operations, rats were injected intraperitoneally with infliximab (7 mg/kg) or vehicle (saline; 1 mL/kg). Twenty hours after the operations, MBF and phenylephrine responses of isolated aortic rings were measured. Tissue damages were examined biochemically and histopathologically. Furthermore, survival rates were monitored throughout 96 h. Infliximab improved survival, mesenteric perfusion, and aortic function after CLP. Increases of serum AST, ALT, LDH, BUN, Cr, and inflammatory cytokines (tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6) induced by CLP were blocked by infliximab. Infliximab prevented malondialdehyde elevations in septic liver, lung, spleen, and kidney tissues, as well as glutathione reductions in septic liver, spleen, and kidney tissues. Protective effects of infliximab on multiple organ damage were also observed histopathologically. Infliximab showed protective effects in sepsis due to its improvement effects on mesenteric perfusion, aortic function, and its anti-inflammatory and antioxidative effects.
Collapse
Affiliation(s)
- Erdem Kamil Ozer
- Department of Pharmacology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Mustafa Tugrul Goktas
- Department of Pharmacology, Faculty of Medicine, Yildirim Beyazit University, Ankara, Turkey
| | - Ibrahim Kilinc
- Department of Biochemistry, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Aysun Toker
- Department of Biochemistry, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Hulagu Bariskaner
- Department of Pharmacology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Ceyhan Ugurluoglu
- Department of Pathology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Alper Bektas Iskit
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
17
|
PKC mediated phosphorylation of TIMAP regulates PP1c activity and endothelial barrier function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:431-439. [DOI: 10.1016/j.bbamcr.2016.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/10/2016] [Accepted: 12/04/2016] [Indexed: 12/17/2022]
|
18
|
Abdullah Z, Bayraktutan U. Suppression of PKC-α attenuates TNF-α-evoked cerebral barrier breakdown via regulations of MMP-2 and plasminogen-plasmin system. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1354-66. [PMID: 27094771 DOI: 10.1016/j.bbadis.2016.03.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/08/2016] [Accepted: 03/30/2016] [Indexed: 10/21/2022]
Abstract
Ischaemic stroke, accompanied by neuroinflammation, impairs blood-brain barrier integrity through a complex mechanism involving both protein kinase C (PKC) and urokinase. Using an in vitro model of human blood-brain barrier (BBB) composed of brain microvascular endothelial cells (HBMEC) and astrocytes, this study assessed the putative roles of these elements in BBB damage evoked by enhanced availability of pro-inflammatory cytokine, TNF-α. Treatment of HBMEC with TNF-α significantly increased the mRNA and protein expressions of all plasminogen-plasmin system (PPS) components, namely tissue plasminogen activator, urokinase, urokinase plasminogen activator receptor and plasminogen activator inhibitor-1 and also the activities of urokinase, total PKC and extracellular MMP-2. Inhibition of urokinase by amiloride abated the effects of TNF-α on BBB integrity and MMP-2 activity without affecting that of total PKC. Conversely, pharmacological inhibition of conventional PKC isoforms dramatically suppressed TNF-α-induced overactivation of urokinase. Knockdown of PKC-α gene via specific siRNA in HBMEC suppressed the stimulatory effects of TNF-α on protein expression of all PPS components, MMP-2 activity, DNA fragmentation rates and pro-apoptotic caspase-3/7 activities. Establishment of co-cultures with BMEC transfected with PKC-α siRNA attenuated the disruptive effects of TNF-α on BBB integrity and function. This was partly due to elevations observed in expression of a tight junction protein, claudin-5 and partly to prevention of stress fibre formation. In conclusion, specific inhibition of PKC-α in cerebral conditions associated with exaggerated release of pro-inflammatory cytokines, notably TNF-α may be of considerable therapeutic value and help maintain endothelial cell viability, appropriate cytoskeletal structure and basement membrane.
Collapse
Affiliation(s)
- Zuraidah Abdullah
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham NG5 1PB, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, Clinical Sciences Building, School of Medicine, Hucknall Road, Nottingham NG5 1PB, UK.
| |
Collapse
|
19
|
Chai J, Long B, Liu X, Li Y, Han N, Zhao P, Chen W. Effects of sevoflurane on tight junction protein expression and PKC-α translocation after pulmonary ischemia-reperfusion injury. Exp Mol Med 2015; 47:e167. [PMID: 26045255 PMCID: PMC4491722 DOI: 10.1038/emm.2015.27] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 01/07/2015] [Accepted: 01/09/2015] [Indexed: 12/18/2022] Open
Abstract
Pulmonary dysfunction caused by ischemia-reperfusion injury is the leading cause of mortality in lung transplantation. We aimed to investigate the effects of sevoflurane pretreatment on lung permeability, tight junction protein occludin and zona occludens 1 (ZO-1) expression, and translocation of protein kinase C (PKC)-α after ischemia–reperfusion. A lung ischemia-reperfusion injury model was established in 96 male Wistar rats following the modified Eppinger method. The rats were divided into four groups with 24 rats in each group: a control (group C), an ischemia-reperfusion group (IR group), a sevoflurane control group (sev-C group), and a sevoflurane ischemia-reperfusion group (sev–IR group). There were three time points in each group: ischemic occlusion for 45 min, reperfusion for 60 min and reperfusion for 120 min; and there were six rats per time point. For the 120-min reperfusion group, six extra rats underwent bronchoalveolar lavage. Mean arterial pressure (MAP) and pulse oxygen saturation (SpO2) were recorded at each time point. The wet/dry weight ratio and lung permeability index (LPI) were measured. Quantitative RT-PCR and Western blot were used to measure pulmonary occludin and ZO-1, and Western blot was used to measure cytosolic and membranous PKC-α in the lung. Lung permeability was significantly increased after ischemia–reperfusion. Sevoflurane pretreatment promoted pulmonary expression of occludin and ZO-1 after reperfusion and inhibited the translocation of PKC-α. In conclusion, sevoflurane pretreatment alleviated lung permeability by upregulating occludin and ZO-1 after ischemia–reperfusion. Sevoflurane pretreatment inhibited the translocation and activation of PKC-α, which also contributed to the lung-protective effect of sevoflurane.
Collapse
Affiliation(s)
- Jun Chai
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Bo Long
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Xiaomei Liu
- Central Laboratory of Shengjing Hospital, China Medical University, Shenyang City, China
| | - Yan Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Ning Han
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Weimin Chen
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang City, China
| |
Collapse
|
20
|
Carbone F, Montecucco F. Inflammation in arterial diseases. IUBMB Life 2015; 67:18-28. [PMID: 25631520 DOI: 10.1002/iub.1344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/28/2014] [Indexed: 12/26/2022]
Abstract
The pathophysiology of some inflammatory arterial diseases (such as vasculitis, atherogenesis and aneurysms) has been widely investigated in the last decades. Among different soluble molecules, proinflammatory cytokines (such as TNF-α, IL-1 and IL-6) were shown to trigger critical pathways regulating these arterial diseases. Together with these cytokines, chemokines were also associated with endothelial dysfunction and intima injury in arterial diseases. Recently, autoantibodies have been also described to pathophysiologically influence not only autoimmune vasculitis but also atherogenesis and more in general vascular inflammation. These soluble mediators actively trigger inflammatory functions of leukocytes and vascular cells. For instance, B and T lymphocytes, macrophages and neutrophils were shown to actively participate in inflammatory processes within the arterial wall in vasculitis, atherogenesis and aneurysms. The aim of this narrative review is to provide an overview of pathophysiology and treatments targeting arterial inflammation in these diseases.
Collapse
Affiliation(s)
- Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa School of Medicine, IRCCS Azienda Ospedaliera Universitaria San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy; Division of Cardiology, Foundation for Medical Researches, Department of Medical Specialties, University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
21
|
Elevated levels of plasma TNF-α are associated with microvascular endothelial dysfunction in patients with sepsis through activating the NF-κB and p38 mitogen-activated protein kinase in endothelial cells. Shock 2014; 41:275-81. [PMID: 24430552 DOI: 10.1097/shk.0000000000000116] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Inflammatory responses can induce microvascular and endothelial dysfunction, which is associated with the development of sepsis. This study is aimed at examining the concentrations of plasma tissue factor (TF), von Willebrand factor (vWF), and tumor necrosis factor-α (TNF-α) in patients with sepsis and at determining how septic plasma (SP) regulates TF and vWF expression and p38 mitogen activated protein kinase (p38 MAPK)/nuclear factor-κB (NF-κB) pathways in human endothelial cells. The concentrations of plasma TF, vWF, and TNF-α in 22 septic patients and eight healthy controls (HCs) were examined by enzyme-linked immunosorbent assay, and their potential association with disease severity was analyzed. Human umbilical vein endothelial cells (HUVECs) were treated with SP from patients or normal plasma (NP) from the HCs, and the levels of TF and vWF were measured. The SP-induced ERK, p38 MAPK, and NF-κB activation was characterized by Western blot and immunofluorescent assays. The SP-induced HUVEC apoptosis was detected by flow cytometry. The concentrations of plasma TF, vWF, and TNF-α in the patients were significantly higher than that in the HCs and were positively correlated with the Acute Physiology and Chronic Health Evaluation II scores in the patients. Furthermore, treatment with SP, but not NP, induced TF and vWF production in HUVECs in a dose- and time-dependent manner, which was associated with sequential activation of the p38 MAPK and NF-κB pathways. Septic plasma induced HUVEC apoptosis, which was inhibited by activating the NF-κB pathway. The sepsis-related inflammatory factors promoted endothelial cell activation, dysfunction, and apoptosis through activation of the p38 MAPK pathway that was regulated by NF-κB signaling.
Collapse
|
22
|
Weidert E, Pohler SE, Gomez EW, Dong C. Actinomyosin contraction, phosphorylation of VE-cadherin, and actin remodeling enable melanoma-induced endothelial cell-cell junction disassembly. PLoS One 2014; 9:e108092. [PMID: 25225982 PMCID: PMC4167543 DOI: 10.1371/journal.pone.0108092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/25/2014] [Indexed: 02/07/2023] Open
Abstract
During melanoma cell extravasation through the vascular endothelium, melanoma cells interact with endothelial cells through secretion of cytokines and by adhesion between proteins displayed on opposing cell surfaces. How these tumor cell associated signals together regulate the dynamics of intracellular signaling pathways within endothelial cells leading to endothelial cell-cell junction disruption is not well understood. Here, we used a combination of experimental and computational approaches to examine the individual and combined effects of activation of the vascular cell adhesion molecule (VCAM)-1, interleukin (IL)-8, and IL-1β signaling pathways on the integrity of vascular junctions. Our simulations predict a multifaceted interplay of signaling resulting from individual activation of VCAM-1, IL-8 and IL-1β pathways that is neither synergistic nor additive compared to all inputs turned on simultaneously. Furthermore, we show that the levels of phosphorylated proteins associated with actinomyosin contractility and junction disassembly peak prior to those related to actin remodeling. The results of this work provide insight into the dynamics of tumor-mediated endothelial junction disassembly and suggest that targeting proteins downstream of several interaction pathways may be the most effective therapeutic approach to reduce melanoma extravasation.
Collapse
Affiliation(s)
- Eric Weidert
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Steven E. Pohler
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Esther W. Gomez
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (EWG); (CD)
| | - Cheng Dong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (EWG); (CD)
| |
Collapse
|
23
|
Amado-Azevedo J, Valent ET, Van Nieuw Amerongen GP. Regulation of the endothelial barrier function: a filum granum of cellular forces, Rho-GTPase signaling and microenvironment. Cell Tissue Res 2014; 355:557-76. [PMID: 24633925 DOI: 10.1007/s00441-014-1828-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/24/2014] [Indexed: 12/20/2022]
Abstract
Although the endothelium is an extremely thin single-cell layer, it performs exceedingly well in preventing blood fluids from leaking into the surrounding tissues. However, specific pathological conditions can affect this cell layer, compromising the integrity of the barrier. Vascular leakage is a hallmark of many cardiovascular diseases and despite its medical importance, no specialized therapies are available to prevent it or reduce it. Small guanosine triphosphatases (GTPases) of the Rho family are known to be key regulators of various aspects of cell behavior and studies have shown that they can exert both positive and negative effects on endothelial barrier integrity. Moreover, extracellular matrix stiffness has now been implicated in the regulation of Rho-GTPase signaling, which has a direct impact on the integrity of endothelial junctions. However, knowledge about both the precise mechanism of this regulation and the individual contribution of the specific regulatory proteins remains fragmentary. In this review, we discuss recent findings concerning the balanced activities of Rho-GTPases and, in particular, aspects of the regulation of the endothelial barrier. We highlight the role of Rho-GTPases in the intimate relationships between biomechanical forces, microenvironmental influences and endothelial intercellular junctions, which are all interwoven in a beautiful filigree-like fashion.
Collapse
Affiliation(s)
- Joana Amado-Azevedo
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Van den Boechorststraat 7, 1081BT, Amsterdam, The Netherlands
| | | | | |
Collapse
|
24
|
Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal 2014; 20:1126-67. [PMID: 23991888 PMCID: PMC3929010 DOI: 10.1089/ars.2012.5149] [Citation(s) in RCA: 3131] [Impact Index Per Article: 284.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Abstract Reactive oxygen species (ROS) are key signaling molecules that play an important role in the progression of inflammatory disorders. An enhanced ROS generation by polymorphonuclear neutrophils (PMNs) at the site of inflammation causes endothelial dysfunction and tissue injury. The vascular endothelium plays an important role in passage of macromolecules and inflammatory cells from the blood to tissue. Under the inflammatory conditions, oxidative stress produced by PMNs leads to the opening of inter-endothelial junctions and promotes the migration of inflammatory cells across the endothelial barrier. The migrated inflammatory cells not only help in the clearance of pathogens and foreign particles but also lead to tissue injury. The current review compiles the past and current research in the area of inflammation with particular emphasis on oxidative stress-mediated signaling mechanisms that are involved in inflammation and tissue injury.
Collapse
Affiliation(s)
- Manish Mittal
- 1 Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois
| | | | | | | | | |
Collapse
|
25
|
Abstract
The acute respiratory distress syndrome (ARDS) is a major public health problem and a leading source of morbidity in intensive care units. Lung tissue in patients with ARDS is characterized by inflammation, with exuberant neutrophil infiltration, activation, and degranulation that is thought to initiate tissue injury through the release of proteases and oxygen radicals. Treatment of ARDS is supportive primarily because the underlying pathophysiology is poorly understood. This gap in knowledge must be addressed to identify urgently needed therapies. Recent research efforts in anti-inflammatory drug development have focused on identifying common control points in multiple signaling pathways. The protein kinase C (PKC) serine-threonine kinases are master regulators of proinflammatory signaling hubs, making them attractive therapeutic targets. Pharmacological inhibition of broad-spectrum PKC activity and, more importantly, of specific PKC isoforms (as well as deletion of PKCs in mice) exerts protective effects in various experimental models of lung injury. Furthermore, PKC isoforms have been implicated in inflammatory processes that may be involved in the pathophysiologic changes that result in ARDS, including activation of innate immune and endothelial cells, neutrophil trafficking to the lung, regulation of alveolar epithelial barrier functions, and control of neutrophil proinflammatory and prosurvival signaling. This review focuses on the mechanistic involvement of PKC isoforms in the pathogenesis of ARDS and highlights the potential of developing new therapeutic paradigms based on the selective inhibition (or activation) of specific PKC isoforms.
Collapse
|
26
|
Continuous blood purification ameliorates endothelial hyperpermeability in SAP patients with MODS by regulating tight junction proteins via ROCK. Int J Artif Organs 2013; 36:700-9. [PMID: 23918271 DOI: 10.5301/ijao.5000216] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2013] [Indexed: 01/31/2023]
Abstract
BACKGROUND Excessive activation of inflammatory mediator cascade during severe acute pancreatitis (SAP) is a major cause of multiple organ dysfunction and is associated with a high mortality. Recently, more and more studies have shown that continuous blood purification (CBP) could improve the prognosis of patients with multiple organ dysfunction syndrome (MODS), but the exact mechanism is still unclear. Many researchers have found that the disruption of tight junction barrier was an important factor for endothelial hyperpermeability, which played a key role in the pathogenesis of MODS. Previously, we found CBP could attenuate endothelial hyperpermeability in SAP patients with lung injury through regulating cytoskeleton reorganization mediated by RhoA/ROCK. However, the effect of CBP on the change of tight junction proteins in SAP patients with MODS was still unknown. This study aimed to investigate the role of tight junctions in endothelial hyperpermeability in SAP patients with MODS using an in vitro model, and the effect of CBP on tight junction barrier.
METHODS Before CBP and after CBP, blood samples were collected to observe hepatic and renal function, and arterial blood gas, while the APACHE II score was calculated to evaluate the severity of patients. To test whether RhoA/ROCK signaling pathway was involved, human umbilical vein endothelial cells (HUVECs) were exposed to serum samples taken from patients at specific time points during CBP, or preincubated with ROCK inhibitor, Y-27632, followed by treatment with serum. Then, the changes in endothelial cell permeability and the expression and distribution of tight junction proteins occludin and claudin-1 were observed.
RESULTS Compared with before CBP, the APACHE II score, serum creatinine and alanine aminotransferase decreased significantly, while PaO2/FiO2 increased significantly after CBP. Meanwhile, endothelial permeability induced by serum from patients significantly increased, while the expression of tight junction proteins occludin and claudin-1 significantly decreased, and severe disruption of occludin and claudin-1 was found in these cells. However, pretreated with Rho-kinase inhibitor, Y-27632 could lessen all of these abnormalities, and in a dose-dependent way. Endothelial hyperpermeability, the abnormal expression and distribution of occludin and claudin-1 were attenuated in HUVECs treated with serum from patients after CBP treatment.
CONCLUSIONS The abnormality of tight junctions mediated by ROCK was an important mechanism for endothelial hyperpermeability induced by serum from SAP patients with MODS. CBP could ameliorate the disorganization and redistribution of tight junction proteins, hence improve the endothelial permeability.
Collapse
|
27
|
Armstrong SM, Mubareka S, Lee WL. The lung microvascular endothelium as a therapeutic target in severe influenza. Antiviral Res 2013; 99:113-8. [PMID: 23685311 DOI: 10.1016/j.antiviral.2013.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 05/03/2013] [Accepted: 05/07/2013] [Indexed: 12/27/2022]
Abstract
Severe infections with influenza virus are characterized by acute respiratory distress syndrome (ARDS), a life-threatening disorder in which the alveolocapillary membrane in the lung becomes leaky. This leads to alveolar flooding, hypoxemia and respiratory failure. Recent data suggest that influenza virus can exert both direct and indirect effects on the lung endothelium, activating it and inducing microvascular leak. These findings raise the possibility that enhancing lung endothelial barrier integrity or modulating lung endothelial activation may prove therapeutically useful for severe influenza. In this paper, we review evidence that lung endothelial activation and vascular leak are a "final common pathway" in severe influenza, as has been reported in bacterial sepsis, and that enhancing endothelial barrier function may improve the outcome of illness. We describe a number of experimental therapies that have shown promise in preventing or reversing increased vascular leak in animal models of sepsis or influenza.
Collapse
|
28
|
Bin Tarif A, Lasecka L, Holzer B, Baron MD. Ganjam virus/Nairobi sheep disease virus induces a pro-inflammatory response in infected sheep. Vet Res 2012; 43:71. [PMID: 23083136 PMCID: PMC3507801 DOI: 10.1186/1297-9716-43-71] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 10/01/2012] [Indexed: 11/10/2022] Open
Abstract
Partly due to climate change, and partly due to changes of human habitat occupation, the impact of tick-borne viruses is increasing. Nairobi sheep disease virus (NSDV) and Ganjam virus (GV) are two names for the same virus, which causes disease in sheep and goats and is currently known to be circulating in India and East Africa. The virus is transmitted by ixodid ticks and causes a severe hemorrhagic disease. We have developed a real-time PCR assay for the virus genome and validated it in a pilot study of the pathogenicity induced by two different isolates of NSDV/GV. One isolate was highly adapted to tissue culture, grew in most cell lines tested, and was essentially apathogenic in sheep. The second isolate appeared to be poorly adapted to cell culture and retained pathogenicity in sheep. The real-time PCR assay for virus easily detected 4 copies or less of the viral genome, and allowed a quantitative measure of the virus in whole blood. Measurement of the changes in cytokine mRNAs showed similar changes to those observed in humans infected by the closely related virus Crimean Congo hemorrhagic fever virus.
Collapse
Affiliation(s)
- Abid Bin Tarif
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey, GU24 0NF, United Kingdom.
| | | | | | | |
Collapse
|
29
|
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
|
30
|
Grinnell KL, Harrington EO. Interplay between FAK, PKCδ, and p190RhoGAP in the regulation of endothelial barrier function. Microvasc Res 2012; 83:12-21. [PMID: 21549132 PMCID: PMC3175025 DOI: 10.1016/j.mvr.2011.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 04/15/2011] [Accepted: 04/16/2011] [Indexed: 11/16/2022]
Abstract
Disruption of either intercellular or extracellular junctions involved in maintaining endothelial barrier function can result in increased endothelial permeability. Increased endothelial permeability, in turn, allows for the unregulated movement of fluid and solutes out of the vasculature and into the surrounding connective tissue, contributing to a number of disease states, including stroke and pulmonary edema (Ermert et al., 1995; Lee and Slutsky, 2010; van Hinsbergh, 1997; Waller et al., 1996; Warboys et al., 2010). Thus, a better understanding of the molecular mechanisms by which endothelial cell junction integrity is controlled is necessary for development of therapies aimed at treating such conditions. In this review, we will discuss the functions of three signaling molecules known to be involved in regulation of endothelial permeability: focal adhesion kinase (FAK), protein kinase C delta (PKCδ), and p190RhoGAP (p190). We will discuss the independent functions of each protein, as well as the interplay that exists between them and the effects of such interactions on endothelial function.
Collapse
Affiliation(s)
- Katie L Grinnell
- Vascular Research Laboratory, Providence VA Medical Center, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02908, USA
| | | |
Collapse
|
31
|
Abstract
Mast cells (MCs) promote a wide range of localized and systemic inflammatory responses. Their involvement in immediate as well as chronic inflammatory reactions at both local and distal sites points to an extraordinarily powerful immunoregulatory capacity with spatial and temporal versatility. MCs are preferentially found in close proximity to both vascular and lymphatic vessels. On activation, they undergo a biphasic secretory response involving the rapid release of prestored vasoactive mediators followed by de novo synthesized products. Many actions of MCs are related to their capacity to regulate vascular flow and permeability and to the recruitment of various inflammatory cells from the vasculature into inflammatory sites. These mediators often work in an additive fashion and achieve their inflammatory effects locally by directly acting on the vascular and lymphatic endothelia, but they also can affect distal sites. Along these lines, the lymphatic and endothelial vasculatures of the host act as a conduit for the dissemination of MC signals during inflammation. The central role of the MC-endothelial cell axis to immune homeostasis is emphasized by the fact that some of the most effective current treatments for inflammatory disorders are directed at interfering with this interaction.
Collapse
|
32
|
Yang G, Lucas R, Caldwell R, Yao L, Romero MJ, Caldwell RW. Novel mechanisms of endothelial dysfunction in diabetes. J Cardiovasc Dis Res 2011; 1:59-63. [PMID: 20877687 PMCID: PMC2945199 DOI: 10.4103/0975-3583.64432] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Diabetes mellitus is a major risk factor for cardiovascular morbidity and mortality. This condition increases the risk of developing coronary, cerebrovascular, and peripheral arterial disease fourfold. Endothelial dysfunction is a major contributor to the pathogenesis of vascular disease in diabetes mellitus patients and has recently received increased attention. In this review article, some recent developments that could improve the knowledge of diabetes-induced endothelial dysfunction are discussed.
Collapse
Affiliation(s)
- Guang Yang
- Department of Vascular Biology Center, Medical College of Georgia, Augusta, GA, USA
| | | | | | | | | | | |
Collapse
|
33
|
Ding RQ, Tsao J, Chai H, Mochly-Rosen D, Zhou W. Therapeutic potential for protein kinase C inhibitor in vascular restenosis. J Cardiovasc Pharmacol Ther 2010; 16:160-7. [PMID: 21183728 DOI: 10.1177/1074248410382106] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Vascular restenosis, an overreaction of biological response to injury, is initialized by thrombosis and inflammation. This response is characterized by increased smooth muscle cell migration and proliferation. Available pharmacological treatments include anticoagulants, antiplatelet agents, immunosuppressants, and antiproliferation agents. Protein kinase C (PKC), a large family of serine/threonine kinases, has been shown to participate in various pathological stages of restenosis. Consequently, PKC inhibitors are expected to exert a wide range of pharmacological activities therapeutically beneficial for restenosis. In this review, the roles of PKC isozymes in platelets, leukocytes, endothelial cells, and smooth muscle cells are discussed, with emphasis given to smooth muscle cells. We will describe cellular and animal studies assessing prevention of restenosis with PKC inhibitors, particularly targeting -α, -β, -δ, and -ζ isozymes. The delivery strategy, efficacy, and safety of such PKC regulators will also be discussed.
Collapse
Affiliation(s)
- Richard Qinxue Ding
- Division of Vascular and Endovascular Surgery, Department of Surgery, Stanford University, Stanford, CA 94350, USA
| | | | | | | | | |
Collapse
|
34
|
Cui GY, Gao XM, Qi SH, Gillani A, Gao L, Shen X, Zhang YD. The action of thrombin in intracerebral hemorrhage induced brain damage is mediated via PKCα/PKCδ signaling. Brain Res 2010; 1398:86-93. [PMID: 21172324 DOI: 10.1016/j.brainres.2010.11.095] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 11/25/2010] [Accepted: 11/30/2010] [Indexed: 10/18/2022]
Abstract
The present study investigates the role of protein kinase C alpha/delta (PKCα/PKCδ) in brain injury induced by intracerebral hemorrhage (ICH) by utilizing a rat model that received intracerebral injections of autologous blood and thrombin (TM). The activation and expression of PKC and PKCδ were analyzed by Western blot and immunohistochemistry. A PKC inhibitor, dihydrochloride (H7), was administrated intraperitoneally after injury to evaluate the effect of inhibition of PKC on ICH and TM induced brain damage. Our data indicate that both ICH and TM increased the expression of PKCα/PKCδ in the brain tissue, and PKCα expression peaked at 6h, while PKCδ expression reached its maximum value at 72h post-injury. Administration of H7 significantly reduced the inflammatory cells infiltrate, permeability of brain-blood barrier (BBB), brain edema, and neuronal death. We conclude that both PKCα and PKCδ play important roles in ICH and TM-induced brain injury, and dihydrochloride (H7) can attenuate brain damage after ICH.
Collapse
Affiliation(s)
- Gui-Yun Cui
- Department of Neurology, Nanjing Brain Hospital, Nanjing Medical University, No. 264 Guangzhou Road, Nanjing 210029, PR China
| | | | | | | | | | | | | |
Collapse
|
35
|
Cirrhotic patients in the medical intensive care unit: Early prognosis and long-term survival*. Crit Care Med 2010; 38:2108-16. [DOI: 10.1097/ccm.0b013e3181f3dea9] [Citation(s) in RCA: 167] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
36
|
Role of src-suppressed C kinase substrate in rat pulmonary microvascular endothelial hyperpermeability stimulated by inflammatory cytokines. Inflamm Res 2010; 59:949-58. [PMID: 20454828 DOI: 10.1007/s00011-010-0207-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Revised: 04/04/2010] [Accepted: 04/21/2010] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE The aim of the study was to investigate the role of src-suppressed C kinase substrate (SSeCKS) in the modulation of rat pulmonary microvascular endothelial cells (RPMVEC) permeability elicited by interleukin (IL)-1β and tumor necrosis factor (TNF)-α. METHODS The gene expression of SSeCKS was analyzed by reverse transcription-polymerase chain reaction. Immunoblotting was used to determine the SSeCKS protein expression and the activation of the protein kinase C (PKC) signaling pathway. A RPMVEC monolayer was constructed to determine changes of transendothelial electrical resistance (TER) and FITC-dextran flux (P (d)) across the monolayer. SSeCKS-specific small interfering RNA was transfected into RPMVEC. RESULTS IL-1β and TNF-α activated the PKC signaling pathway in RPMVEC, and up-regulated the gene and protein expression of SSeCKS. Depletion of endogenous SSeCKS in RPMVEC significantly attenuated cytokine-induced decrease in TER and increase in P (d), but not to the basal levels. PKC inhibitors also significantly decreased cytokine-induced hyperpermeability and SSeCKS expression. CONCLUSIONS SSeCKS is involved in the endothelial hyperpermeability induced by IL-1β and TNF-α in inflammatory process.
Collapse
|
37
|
Protein kinase Calpha: disease regulator and therapeutic target. Trends Pharmacol Sci 2009; 31:8-14. [PMID: 19969380 PMCID: PMC2809215 DOI: 10.1016/j.tips.2009.10.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 10/16/2009] [Accepted: 10/19/2009] [Indexed: 12/28/2022]
Abstract
Protein kinase Cα (PKCα) is a member of the AGC (which includes PKD, PKG and PKC) family of serine/threonine protein kinases that is widely expressed in mammalian tissues. It is closely related in structure, function and regulation to other members of the protein kinase C family, but has specific functions within the tissues in which it is expressed. There is substantial recent evidence, from gene knockout studies in particular, that PKCα activity regulates cardiac contractility, atherogenesis, cancer and arterial thrombosis. Selective targeting of PKCα therefore has potential therapeutic value in a wide variety of disease states, although will be technically complicated by the ubiquitous expression and multiple functions of the molecule.
Collapse
|
38
|
Rodríguez C, Alcudia JF, Martínez-González J, Guadall A, Raposo B, Sánchez-Gómez S, Badimon L. Statins normalize vascular lysyl oxidase down-regulation induced by proatherogenic risk factors. Cardiovasc Res 2009; 83:595-603. [PMID: 19406911 DOI: 10.1093/cvr/cvp136] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Statins are lipid-lowering drugs widely used in the management of vascular diseases. Clinical and experimental evidence suggest that statins improve endothelial function by both cholesterol-lowering-dependent and -independent mechanisms. We have previously shown that endothelial dysfunction induced by risk factors and proinflammatory cytokines is associated with down-regulation of lysyl oxidase (LOX), a key enzyme modulating extracellular matrix maturation and vascular integrity. Our aim was to analyse whether statins could normalize LOX expression impaired by proatherogenic risk factors. METHODS AND RESULTS We observed that pharmacological concentrations of statins (atorvastatin and simvastatin) modulated LOX transcriptional activity, counteracting the down-regulation of LOX (at the mRNA, protein, and activity level) caused by tumour necrosis factor-alpha (TNFalpha) in porcine, bovine, and human aortic endothelial cells. Geranylgeraniol but not farnesol reversed this effect, suggesting the involvement of geranylgeranylated proteins. In accordance, inhibitors of RhoA/Rho kinase also counteracted LOX down-regulation caused by TNFalpha, and over-expression of a RhoA dominant-negative mutant mimicked statin effects. Statins were also able to counteract the decrease in LOX expression produced by atherogenic concentrations of LDL by a similar mechanism and to partially prevent the increase in endothelial permeability elicited by these lipoproteins. Finally, in the in vivo porcine model of hypercholesterolaemia, we observed that statins abrogated the reduction of vascular LOX expression triggered by high plasma levels of LDL. CONCLUSION These data indicate that statins normalize vascular LOX expression altered by atherogenic risk factors through a RhoA/Rho kinase-dependent mechanism. Thus, modulation of LOX by statins could contribute to vascular protection and to the cardiovascular risk reduction achieved by this therapy.
Collapse
Affiliation(s)
- Cristina Rodríguez
- Centro de Investigación Cardiovascular, Antoni M Claret 167, Barcelona 08025, Spain.
| | | | | | | | | | | | | |
Collapse
|
39
|
Johnson A. TNF-induced activation of pulmonary microvessel endothelial cells: a role for GSK3beta. Am J Physiol Lung Cell Mol Physiol 2009; 296:L700-9. [PMID: 19218353 DOI: 10.1152/ajplung.90566.2008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The hypothesis tested was PKCalpha mediates the phosphorylation of glycogen synthetase kinase 3beta (GSK3beta) and that the GSK3beta inhibition modulates the response to tumor necrosis factor-alpha (TNF) in rat pulmonary microvessel endothelial cells (PMEC). PMEC were treated with TNF for 4.0 h (100 ng/ml) or vehicle. First, to assess the role of PKCalpha in the phosphorylation of GSK3beta (i.e., an indicator of GSK3beta inhibition), PMEC were pretreated with 1) nonsense-RNA-PKCalpha, 2) siRNA-PKCalpha, and 3) the PKC inhibitor Gö6983. In the nonsense RNA-PKCalpha+TNF and TNF groups, there was increased phosphorylated GSK3beta-Ser9 that did not occur in the Gö6983+TNF group. In the TNF groups, there was a significant correlation between PKCalpha protein and phosphorylated GSK3beta-Ser9 that did not occur in the groups without TNF. Second, to assess the role of GSK3beta in beta-catenin activity, PMEC were pretreated with 1) wild-type (w) GSK3beta plasmid to enhance GSK3beta activity, 2) kinase dead (kd)-GSK3beta plasmid, and 3) the GSK3beta inhibitor SB-216763. In the TNF group, there was increased unphosphorylated beta-catenin-Ser37/33 compared with the control group. In the GSK3beta-inhibited groups (i.e., SB-216763 and kdGSK3beta) +/- TNF, the unphosphorylated beta-catenin-Ser37/33 was similar to the TNF group. In the GSK3beta-enhanced group +/- TNF, the unphosphorylated beta-catenin-Ser37/33 was similar to the control. Finally, PMEC were also treated with TOPflash, a beta-catenin-dependent promoter luciferase reporter, or the mutant construct FOPflash, 2 days before treatment with TNF. In the TNF group, there was an increased TOPflash/FOPflash activity ratio compared with the control group. In the GSK3beta-inhibited groups (i.e., SB-216763 and kdGSK3beta) +/- TNF, the TOPflash/FOPflash activity ratio was similar to the TNF group. In the GSK3beta-enhanced group +/- TNF, the TOPflash/FOPflash activity ratio was similar to the control. The data indicate that TNF induces endothelial activation that is modulated by a PKCalpha-dependent inhibition of GSK3beta.
Collapse
Affiliation(s)
- Arnold Johnson
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA.
| |
Collapse
|
40
|
Arias HR, Richards VE, Ng D, Ghafoori ME, Le V, Mousa SA. Role of non-neuronal nicotinic acetylcholine receptors in angiogenesis. Int J Biochem Cell Biol 2009; 41:1441-51. [PMID: 19401144 DOI: 10.1016/j.biocel.2009.01.013] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 01/17/2009] [Indexed: 12/16/2022]
Abstract
Angiogenesis is a critical physiological process for cell survival and development. Endothelial cells, necessary for the course of angiogenesis, express several non-neuronal nicotinic acetylcholine receptors (AChRs). The most important functional non-neuronal AChRs are homomeric alpha7 AChRs and several heteromeric AChRs formed by a combination of alpha3, alpha5, beta2, and beta4 subunits, including alpha3beta4-containing AChRs. In endothelial cells, alpha7 AChR stimulation indirectly triggers the activation of the integrin alphavbeta3 receptor and an intracellular MAP kinase (ERK) pathway that mediates angiogenesis. Non-selective cholinergic agonists such as nicotine have been shown to induce angiogenesis, enhancing tumor progression. Moreover, alpha7 AChR selective antagonists such as alpha-bungarotoxin and methyllycaconitine as well as the non-specific antagonist mecamylamine have been shown to inhibit endothelial cell proliferation and ultimately blood vessel formation. Exploitation of such pharmacologic properties can lead to the discovery of new specific cholinergic antagonists as anti-cancer therapies. Conversely, the pro-angiogenic effect elicited by specific agonists can be used to treat diseases that respond to revascularization such as diabetic ischemia and atherosclerosis, as well as to accelerate wound healing. In this mini-review we discuss the pharmacological evidence supporting the importance of non-neuronal AChRs in angiogenesis. We also explore potential intracellular mechanisms by which alpha7 AChR activation mediates this vital cellular process.
Collapse
Affiliation(s)
- Hugo R Arias
- Department of Pharmaceutical Sciences, College of Pharmacy, Midwestern University, 19555 N. 59th Avenue, Glendale, AZ 85308-3550, USA.
| | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Gaudreault N, Perrin RM, Guo M, Clanton CP, Wu MH, Yuan SY. Counter regulatory effects of PKCbetaII and PKCdelta on coronary endothelial permeability. Arterioscler Thromb Vasc Biol 2008; 28:1527-33. [PMID: 18497307 PMCID: PMC2626185 DOI: 10.1161/atvbaha.108.166975] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The aim of this study was to examine the endothelial distribution and activity of selected PKC isoforms in coronary vessels with respect to their functional impact on endothelial permeability under the experimental conditions relevant to diabetes. METHODS AND RESULTS En face immunohistochemistry demonstrated a significant increase of PKC(betaII) and decrease of PKCdelta expression in coronary arterial endothelium of Zucker diabetic rats. To test whether changes in PKC expression alter endothelial barrier properties, we measured the transcellular electric resistance in human coronary microvascular endothelial monolayers and found that either PKC(betaII) overexpression or PKCdelta inhibition disrupted the cell-cell adhesive barrier. Three-dimensional fluorescence microscopy revealed that hyperpermeability was caused by altered PKC activity in association with distinct translocation of PKC(betaII) to the cell-cell junction and PKCdelta localization to the cytosol. Further analyses in fractionated endothelial lysates confirmed the differential redistribution of these isozymes. Additionally, FRET analysis of PKC subcellular dynamics demonstrated a high PKC(betaII) activity at the cell surface and junction, whereas PKCdelta activity is concentrated in intracellular membrane organelles. CONCLUSIONS Taken together, these data suggest that PKC(betaII) and PKCdelta counter-regulate coronary endothelial barrier properties by targeting distinctive subcellular sites. Imbalanced PKC(betaII)/PKCdelta expression and activity may contribute to endothelial hyperpermeability and coronary dysfunction in diabetes.
Collapse
Affiliation(s)
- Nathalie Gaudreault
- Department of Surgery, University of California Davis School of Medicine, 4625 2nd Avenue, Room 3006, Sacramento, CA 95817, USA
| | | | | | | | | | | |
Collapse
|
43
|
Sumagin R, Lomakina E, Sarelius IH. Leukocyte-endothelial cell interactions are linked to vascular permeability via ICAM-1-mediated signaling. Am J Physiol Heart Circ Physiol 2008; 295:H969-H977. [PMID: 18641276 DOI: 10.1152/ajpheart.00400.2008] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Two key characteristics of the inflammatory response are the recruitment of leukocytes to inflamed tissue as well as changes in vessel permeability. We explored the relationship between these two processes using intravital confocal microscopy in cremasters of anesthetized (65 mg/kg Nembutal ip) mice. We provide direct evidence that intercellular adhesion molecule-1 (ICAM-1) links leukocyte-endothelial cell interactions and changes in solute permeability (Ps). Importantly, we show that arterioles, not just venules, respond to proinflammatory stimuli, thus contributing to microvascular exchange. We identified two independent, ICAM-1-mediated pathways regulating Ps. Under control conditions in wild-type (WT) mice, there is a constitutive PKC-dependent pathway (Ps = 1.0 +/- 0.10 and 2.2 +/- 0.46 x 10(-6) cm/s in arterioles and venules, respectively), which was significantly reduced in ICAM-1 knockout (KO) mice (Ps = 0.54 +/- 0.07 and 0.77 +/- 0.11 x 10(-6) cm/s). The PKC inhibitor bisindolylmaleimid l (1 micromol/l in 0.01% DMSO) decreased P(s) in WT mice to levels similar to those in ICAM-1 KO mice. Likewise, a PKC activator (phorbol-12-myristate-acetate; 1 micromol/l in 0.01% DMSO) successfully restored Ps in ICAM-1 KO vessels to be not different from that of the WT controls. On the other hand, during TNF-alpha-induced inflammation, Ps in WT mice was significantly increased (2-fold in venules and 2.5-fold in arterioles) in a Src-dependent and PKC-independent manner. The blockade of Src (PP2; 2 micromol/l in 0.01% DMSO) but not PKC significantly reduced the TNF-alpha-dependent increase in Ps. We conclude that ICAM-1 plays an essential role in the regulation of Ps in microvessels and that there are two separate (constitutive and inducible) signaling pathways that regulate permeability under normal and inflamed conditions.
Collapse
Affiliation(s)
- Ronen Sumagin
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA
| | | | | |
Collapse
|
44
|
Tinsley JH, Hunter FA, Childs EW. PKC and MLCK-dependent, cytokine-induced rat coronary endothelial dysfunction. J Surg Res 2008; 152:76-83. [PMID: 18621396 DOI: 10.1016/j.jss.2008.02.022] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 01/22/2008] [Accepted: 02/11/2008] [Indexed: 01/01/2023]
Abstract
BACKGROUND Heart disease is one of the leading causes of death in the United States, killing nearly one million people every year. Inflammatory mediators or cytokines are released following myocardial infarction and ischemia/reperfusion injury. These cytokines, of which interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha (TNF-alpha) are among the most important, propagate the activation of a multitude of signaling pathways, such as the protein kinase C (PKC) and myosin light chain kinase (MLCK) pathways, which lead to deleterious changes in the structure and function of the coronary microvascular endothelium. METHODS The effects of cytokines on rat heart microvascular endothelial cell monolayer integrity, PKC activity, and adherens junction protein alteration were examined. Further, an in vivo rat coronary ischemia/reperfusion injury model was used to determine vascular leakage and TNF-alpha release. RESULTS Administration of the above mentioned cytokines to cell monolayers resulted in significant increases in PKC activation, gap formation, and hyperpermeability across the monolayer and beta-catenin phosphorylation/reorganization. Inhibition of conventional PKC and MLCK attenuated permeability increases. Ischemia/reperfusion injury to the left ventricle resulted in TNF-alpha release as well as conventional PKC- and MLCK-dependent protein extravasation from the circulation to the heart tissue. CONCLUSION These results identify the conventional PKC and MLCK pathways as important factors in coronary endothelial dysfunction elicited by IR injury and cytokine release. Further examination of these molecular signaling cascades has the potential of identifying targets for therapeutic intervention following ischemic events in the heart.
Collapse
Affiliation(s)
- John H Tinsley
- Department of Internal Medicine, Scott and White Hospital, Temple, Texas 76504, USA.
| | | | | |
Collapse
|
45
|
|
46
|
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]
|
47
|
Klinger JR, Murray JD, Casserly B, Alvarez DF, King JA, An SS, Choudhary G, Owusu-Sarfo AN, Warburton R, Harrington EO. Rottlerin causes pulmonary edema in vivo: a possible role for PKCδ. J Appl Physiol (1985) 2007; 103:2084-94. [PMID: 17901241 DOI: 10.1152/japplphysiol.00695.2007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the present study, we assessed the effects of chemical inhibitors shown to be selective for protein kinase C (PKC) isoforms on lung barrier function both in vitro and in vivo. Rottlerin, a purported inhibitor of PKCδ, but not other chemical inhibitors, dose dependently promoted barrier dysfunction in lung endothelial cells in vitro. This barrier dysfunction correlated with structural changes in focal adhesions and stress fibers, which were consistent with functional changes in cell stiffness. To determine whether the effects noted in vitro correlated with changes in intact lungs, we tested the effects of rottlerin in the formation of pulmonary edema in rats using both ex vivo and in vivo models. Isolated, perfused lungs demonstrated a significant increase in filtration coefficients on exposure to rottlerin, compared with vehicle-treated lungs, an effect that correlated with increased extravasation of Evan's blue dye (EBD)-conjugated albumin. Additionally, compared with vehicle, the ratio of the wet lung weights to dry lung weights was significantly greater on exposure of animals to rottlerin; rottlerin also produced a dose-dependent increase in EBD extravasation into the lungs. These effects on lung edema occurred without any increase in right ventricular pressures. Microscopic assessment of edema in the ex vivo lungs demonstrated perivascular cuffing, with no evidence of septal capillary leak, in rottlerin-exposed lungs. Taken together, rottlerin increases barrier dysfunction in pulmonary endothelial cell monolayers and causes pulmonary edema in rats; results suggestive of an important role for PKCδ in maintaining lung endothelial barrier function.
Collapse
Affiliation(s)
- James R Klinger
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, and Department of Medicine, Brown Medical School, Providence, RI 02908, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Woods ME, Olano JP. Host defenses to Rickettsia rickettsii infection contribute to increased microvascular permeability in human cerebral endothelial cells. J Clin Immunol 2007; 28:174-85. [PMID: 17957455 DOI: 10.1007/s10875-007-9140-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2007] [Accepted: 09/19/2007] [Indexed: 12/07/2022]
Abstract
Rickettsiae are arthropod-borne intracellular bacterial pathogens that primarily infect the microvascular endothelium leading to systemic spread of the organisms and the major pathophysiological effect, increased microvascular permeability, and edema in vital organs such as the lung and brain. Much work has been done on mechanisms of immunity to rickettsiae, as well as the responses of endothelial cells to rickettsial invasion. However, to date, no one has described the mechanisms of increased microvascular permeability during acute rickettsiosis. We sought to establish an in vitro model of human endothelial-target rickettsial infection using the etiological agent of Rocky Mountain spotted fever, Rickettsia rickettsii, and human cerebral microvascular endothelial cells. Endothelial cells infected with R. rickettsii exhibited a dose-dependent decrease in trans-endothelial electrical resistance, which translates into increased monolayer permeability. Additionally, we showed that the addition of pro-inflammatory stimuli essential to rickettsial immunity dramatically enhanced this effect. This increase in permeability correlates with dissociation of adherens junctions between endothelial cells and is not dependent on the presence of nitric oxide. Taken together, these results demonstrate for the first time that increased microvascular permeability associated with rickettsial infection is partly attributable to intracellular rickettsiae and partly attributable to the immune defenses that have evolved to protect the host from rickettsial spread.
Collapse
Affiliation(s)
- Michael E Woods
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd. Rt 0428, Galveston, TX, USA
| | | |
Collapse
|
49
|
Rodríguez C, Alcudia JF, Martínez-González J, Raposo B, Navarro MA, Badimon L. Lysyl oxidase (LOX) down-regulation by TNFalpha: a new mechanism underlying TNFalpha-induced endothelial dysfunction. Atherosclerosis 2007; 196:558-64. [PMID: 17673218 DOI: 10.1016/j.atherosclerosis.2007.06.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 06/06/2007] [Accepted: 06/15/2007] [Indexed: 10/23/2022]
Abstract
OBJECTIVE TNFalpha is a pro-inflammatory cytokine that induces endothelial dysfunction and promotes atherosclerosis progression. Down-regulation of lysyl oxidase (LOX), a key enzyme in extracellular matrix maturation, by pro-atherogenic risk factors such as LDL and homocysteine, is associated with an impairment of endothelial barrier function. Our hypothesis is that the inflammatory cytokine TNFalpha could also modulate LOX expression/function in endothelial cells. METHODS The study was carried out in human umbilical vein endothelial cells (HUVEC), porcine aortic endothelial cells (PAEC) and bovine aortic endothelial cells (BAEC). LOX mRNA levels were analysed by real-time PCR and LOX activity was assessed by a high sensitive fluorescent assay. Promoter activity was determined by transient transfection using a luciferase reporter system. RESULTS TNFalpha decreases LOX mRNA levels in endothelial cells in a dose- and time-dependent manner. The effect of TNFalpha was observed at low concentrations (0.1-1 ng/mL) and was maximal at 2.5 ng/mL (after 21 h). In transfection assays, TNFalpha reduced LOX transcriptional activity to a similar extent than LOX mRNA. Furthermore, TNFalpha decreases endothelial LOX enzymatic activity. By using both TNF receptor (TNFR) agonist and blocking antibodies we determined the involvement of TNFR2 on LOX down-regulation. Moreover, while TNFR-associated factor-2 (TRAF-2) did not mediate signalling events leading to LOX inhibition, PKC inhibitors counteracted the TNFalpha-induced decrease of LOX mRNA levels. Finally, TNFalpha administration significantly reduced vascular LOX expression in rat aorta. CONCLUSIONS Endothelial dysfunction induced by TNFalpha is associated with a decrease of LOX expression/activity. Thus, LOX seems to be involved in the impairment of endothelial function triggered by different pathological conditions.
Collapse
MESH Headings
- Animals
- Aorta/cytology
- Apoptosis/drug effects
- Cattle
- Cells, Cultured
- Down-Regulation
- Endothelium, Vascular/cytology
- Endothelium, Vascular/physiopathology
- Humans
- Male
- Protein Kinase C/physiology
- Protein-Lysine 6-Oxidase/biosynthesis
- Rats
- Receptors, Tumor Necrosis Factor, Type I/agonists
- Receptors, Tumor Necrosis Factor, Type I/physiology
- Receptors, Tumor Necrosis Factor, Type II/agonists
- Receptors, Tumor Necrosis Factor, Type II/physiology
- Sus scrofa
- Tumor Necrosis Factor-alpha/physiology
- Umbilical Veins/cytology
Collapse
Affiliation(s)
- C Rodríguez
- Centro de Investigación Cardiovascular, CSIC-ICCC, Hospital de Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
50
|
Doucet A, Favre G, Deschênes G. Molecular mechanism of edema formation in nephrotic syndrome: therapeutic implications. Pediatr Nephrol 2007; 22:1983-90. [PMID: 17554565 PMCID: PMC2064946 DOI: 10.1007/s00467-007-0521-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 04/24/2007] [Accepted: 04/25/2007] [Indexed: 02/07/2023]
Abstract
Sodium retention and edema are common features of nephrotic syndrome that are classically attributed to hypovolemia and activation of the renin-angiotensin-aldosterone system. However, numbers of clinical and experimental findings argue against this underfill theory. In this review we analyze data from the literature in both nephrotic patients and experimental models of nephrotic syndrome that converge to demonstrate that sodium retention is not related to the renin-angiotensin-aldosterone status and that fluid leakage from capillary to the interstitium does not result from an imbalance of Starling forces, but from changes of the intrinsic properties of the capillary endothelial filtration barrier. We also discuss how most recent findings on the cellular and molecular mechanisms of sodium retention has allowed the development of an efficient treatment of edema in nephrotic patients.
Collapse
Affiliation(s)
- Alain Doucet
- Laboratoire de Physiologie et Génomique Rénales, CNRS/UPMC UMR 7134, Institut des Cordeliers, 15 rue de l'Ecole de Médecine, 75270, Paris, France.
| | | | | |
Collapse
|