1
|
Puthiyottil S, Skaria T. Paracrine signaling mediators of vascular endothelial barrier dysfunction in sepsis: implications for therapeutic targeting. Tissue Barriers 2025:2503523. [PMID: 40376886 DOI: 10.1080/21688370.2025.2503523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/25/2025] [Accepted: 04/30/2025] [Indexed: 05/18/2025] Open
Abstract
Vascular endothelial barrier disruption is a critical determinant of morbidity and mortality in sepsis. Whole blood represents a key source of paracrine signaling molecules inducing vascular endothelial barrier disruption in sepsis. This study analyzes whole-genome transcriptome data from sepsis patients' whole blood available in the NCBI GEO database to identify paracrine mediators of vascular endothelial barrier dysfunction, uncovering novel insights that may guide drug repositioning strategies. This study identifies the regulated expression of paracrine signaling molecules TFPI, MMP9, PROS1, JAG1, S1PR1, and S1PR5 which either disrupt or protect vascular endothelial barrier function in sepsis and could serve as potential targets for repositioning existing drugs. Specifically, TFPI (barrier protective), MMP9 (barrier destructive), PROS1 (barrier protective), and JAG1 (barrier destructive) are upregulated, while S1PR1 (barrier protective) and S1PR5 (barrier protective) are downregulated. Our observations highlight the importance of considering both protective and disruptive mediators in the development of therapeutic strategies to restore endothelial barrier integrity in septic patients. Identifying TFPI, MMP9, PROS1, JAG1, S1PR1, and S1PR5 as druggable paracrine regulators of vascular endothelial barrier function in sepsis could pave the way for precision medicine approaches, enabling personalized treatments that target specific mediators of endothelial barrier disruption to improve patient outcomes in sepsis.
Collapse
Affiliation(s)
- Shahid Puthiyottil
- Department of Bioscience and Engineering, National Institute of Technology Calicut, Calicut, India
| | - Tom Skaria
- Department of Bioscience and Engineering, National Institute of Technology Calicut, Calicut, India
| |
Collapse
|
2
|
Li HF, Lin H, Liu HT, Lin TJ, Tseng TL. Activating transcription factor-3 orchestrates the modulation of vascular anti-contractile activity and relaxation by governing the secretion of HDL-bound sphingosine-1-phosphate in perivascular adipose tissue. Br J Pharmacol 2025; 182:1763-1782. [PMID: 39843165 DOI: 10.1111/bph.17433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 11/05/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND AND PURPOSE Perivascular adipose tissues (PVATs) play a critical role in modulating vascular homeostasis and protecting against cardiovascular dysfunction-mediated blood pressure dysregulation. We demonstrated that the activating transcription factor-3 (Atf3) gene in the PVAT is crucial for improving vascular wall tension abnormalities; however, its protective mechanism remains unclear. Herein, we aim to determine whether ATF3 regulates PVAT-derived relaxing factor (PVDRF) biosynthesis and if its secretion contributes to vasorelaxation. EXPERIMENTAL APPROACH This study employed an in vivo animal model using global Atf3-deficient mice, in vitro blood vessel myography, and biochemical analyses to evaluate ATF3-mediated PVDRF release and reactivity in the vasculature. KEY RESULTS Wild-type (WT) mouse thoracic aortic PVAT extracts significantly induced resting tone dilation and attenuated vasoconstrictor-induced contractile responses compared to Atf3-/- mice. Heat-stable PVAT extracts from WT mice caused sustained and reproducible vasodilation without tachyphylaxis in control aortic rings. Biochemical evaluation of PVDRF release revealed that Atf3-/- mice had lower sphingosine-1-phosphate (S1P) and HDL cholesterol (HDL-C) levels than WT mice. Furthermore, PVAT extracts from WT mice induced long-lasting vasorelaxation, which was significantly inhibited by the S1P3 receptor antagonist TY52156 and scavenger receptor class B type 1 receptor antagonist glyburide. CONCLUSION AND IMPLICATIONS ATF3 within the PVAT can modulate vascular function by strengthening sphingosine kinase 1 (sphk1)-S1P-S1P3 receptor lipid signalling and stimulating S1P binding to HDL to form the vasodilator HDL-S1P. ATF3 is an essential modulator for maintaining the physiological function of PVAT, providing a novel target for treatment of obesity-related cardiovascular diseases.
Collapse
Affiliation(s)
- Hsiao-Fen Li
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Heng Lin
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tsung-Jen Lin
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- CardioVascular Research Center, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Tzu-Ling Tseng
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- CardioVascular Research Center, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
- Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
3
|
Poteryaeva ON, Usynin IF. Molecular mechanisms of the regulatory action of high-density lipoproteins on the endothelial function. BIOMEDITSINSKAIA KHIMIIA 2024; 70:206-217. [PMID: 39239895 DOI: 10.18097/pbmc20247004206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Endothelial dysfunction underlies the pathogenesis of many diseases, primarily cardiovascular diseases. Epidemiological studies have shown an inverse dependence between the plasma level of high-density lipoproteins (HDL) and cardiovascular diseases. The results of experimental studies indicate that the antiatherogenic effect of HDL is associated not only with their participation in the reverse transport of excess cholesterol, but also with their regulatory effect on the functions of cells of various organs and tissues, including endothelial cells. The purpose of this review is to consider recent data on the participation of plasma receptors and related intracellular signaling pathways in the mechanism of protective effect of HDL on endothelial cell functions. Understanding the mechanisms of cell function regulation under the influence of HDL is an important step for the development of new ways of pharmacological correction of impaired endothelial functions and creation of effective endothelial protection drugs.
Collapse
Affiliation(s)
- O N Poteryaeva
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| | - I F Usynin
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| |
Collapse
|
4
|
Ziegler AC, Haider RS, Hoffmann C, Gräler MH. S1PR3 agonism and S1P lyase inhibition rescue mice in the severe state of experimental sepsis. Biomed Pharmacother 2024; 174:116575. [PMID: 38599060 DOI: 10.1016/j.biopha.2024.116575] [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: 01/15/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024] Open
Abstract
Sepsis is characterized as life-threatening organ dysfunction caused by a dysregulated host response to an infection. Despite numerous clinical trials that addressed this syndrome, there is still no causative treatment available to dampen its severity. Curtailing the infection at an early stage with anti-infectives is the only effective treatment regime besides intensive care. In search for additional treatment options, we recently discovered the inhibition of the sphingosine 1-phosphate (S1P) lyase and subsequent activation of the S1P receptor type 3 (S1PR3) in pre-conditioning experiments as promising targets for sepsis prevention. Here, we demonstrate that treatment of septic mice with the direct S1P lyase inhibitor C31 or the S1PR3 agonist CYM5541 in the advanced phase of sepsis resulted in a significantly increased survival rate. A single dose of each compound led to a rapid decline of sepsis severity in treated mice and coincided with decreased cytokine release and increased lung barrier function with unaltered bacterial load. The survival benefit of both compounds was completely lost in S1PR3 deficient mice. Treatment of the murine macrophage cell line J774.1 with either C31 or CYM5541 resulted in decreased protein kinase B (Akt) and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) phosphorylation without alteration of the mitogen-activated protein kinase (MAPK) p38 and p44/42 phosphorylation. Thus, activation of S1PR3 in the acute phase of sepsis by direct agonism or S1P lyase inhibition dampened Akt and JNK phosphorylation, resulting in decreased cytokine release, improved lung barrier stability, rapid decline of sepsis severity and better survival in mice.
Collapse
Affiliation(s)
- Anke C Ziegler
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Str. 2. Jena D-07745, Germany
| | - Raphael S Haider
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena D-07745, Germany; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK; Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Midlands NG2 7AG, UK
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena D-07745, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Str. 2. Jena D-07745, Germany; Center for Sepsis Control and Care, Jena University Hospital, Jena 07740, Germany.
| |
Collapse
|
5
|
Velagapudi S, Wang D, Poti F, Feuerborn R, Robert J, Schlumpf E, Yalcinkaya M, Panteloglou G, Potapenko A, Simoni M, Rohrer L, Nofer JR, von Eckardstein A. Sphingosine-1-phosphate receptor 3 regulates the transendothelial transport of high-density lipoproteins and low-density lipoproteins in opposite ways. Cardiovasc Res 2024; 120:476-489. [PMID: 38109696 PMCID: PMC11060483 DOI: 10.1093/cvr/cvad183] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/08/2023] [Accepted: 10/20/2023] [Indexed: 12/20/2023] Open
Abstract
AIMS The entry of lipoproteins from blood into the arterial wall is a rate-limiting step in atherosclerosis. It is controversial whether this happens by filtration or regulated transendothelial transport.Because sphingosine-1-phosphate (S1P) preserves the endothelial barrier, we investigated in vivo and in vitro, whether S1P and its cognate S1P-receptor 3 (S1P3) regulate the transendothelial transport of lipoproteins. METHODS AND RESULTS Compared to apoE-haploinsufficient mice (CTRL), apoE-haploinsufficient mice with additional endothelium-specific knock-in of S1P3 (S1P3-iECKI) showed decreased transport of LDL and Evan's Blue but increased transport of HDL from blood into the peritoneal cave. After 30 weeks of high-fat diet feeding, S1P3-iECKI mice had lower levels of non-HDL-cholesterol and less atherosclerosis than CTRL mice. In vitro stimulation with an S1P3 agonist increased the transport of 125I-HDL but decreased the transport of 125I-LDL through human aortic endothelial cells (HAECs). Conversely, inhibition or knock-down of S1P3 decreased the transport of 125I-HDL but increased the transport of 125I-LDL. Silencing of SCARB1 encoding scavenger receptor B1 (SR-BI) abrogated the stimulation of 125I-HDL transport by the S1P3 agonist. The transendothelial transport of 125I-LDL was decreased by silencing of SCARB1 or ACVLR1 encoding activin-like kinase 1 but not by interference with LDLR. None of the three knock-downs prevented the stimulatory effect of S1P3 inhibition on transendothelial 125I-LDL transport. CONCLUSION S1P3 regulates the transendothelial transport of HDL and LDL oppositely by SR-BI-dependent and SR-BI-independent mechanisms, respectively. This divergence supports a contention that lipoproteins pass the endothelial barrier by specifically regulated mechanisms rather than passive filtration.
Collapse
Affiliation(s)
- Srividya Velagapudi
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Dongdong Wang
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Francesco Poti
- Department of Medicine and Surgery—Unit of Neurosciences, University of Parma, Parma, Italy
- Department of Biomedical, Metabolic and Neural Sciences—Unit of Endocrinology, University of Modena and Reggio Emilia, Modena, Italy
| | - Renata Feuerborn
- Central Laboratory Facility, University Hospital of Münster, Münster, Germany
| | - Jerome Robert
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Eveline Schlumpf
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Mustafa Yalcinkaya
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Grigorios Panteloglou
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Anton Potapenko
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences—Unit of Endocrinology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| | - Jerzy-Roch Nofer
- Central Laboratory Facility, University Hospital of Münster, Münster, Germany
- Institute of Laboratory Medicine, Marien-Hospital Osnabrück, Niels-Stensen-Kliniken, Osnabrück, Germany
| | - Arnold von Eckardstein
- Institute of Clinical Chemistry, University of Zurich and University Hospital of Zurich, Rämistrasse 100, CH-8091 Zürich, Switzerland
| |
Collapse
|
6
|
Panagiotides NG, Poledniczek M, Andreas M, Hülsmann M, Kocher AA, Kopp CW, Piechota-Polanczyk A, Weidenhammer A, Pavo N, Wadowski PP. Myocardial Oedema as a Consequence of Viral Infection and Persistence-A Narrative Review with Focus on COVID-19 and Post COVID Sequelae. Viruses 2024; 16:121. [PMID: 38257821 PMCID: PMC10818479 DOI: 10.3390/v16010121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Microvascular integrity is a critical factor in myocardial fluid homeostasis. The subtle equilibrium between capillary filtration and lymphatic fluid removal is disturbed during pathological processes leading to inflammation, but also in hypoxia or due to alterations in vascular perfusion and coagulability. The degradation of the glycocalyx as the main component of the endothelial filtration barrier as well as pericyte disintegration results in the accumulation of interstitial and intracellular water. Moreover, lymphatic dysfunction evokes an increase in metabolic waste products, cytokines and inflammatory cells in the interstitial space contributing to myocardial oedema formation. This leads to myocardial stiffness and impaired contractility, eventually resulting in cardiomyocyte apoptosis, myocardial remodelling and fibrosis. The following article reviews pathophysiological inflammatory processes leading to myocardial oedema including myocarditis, ischaemia-reperfusion injury and viral infections with a special focus on the pathomechanisms evoked by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In addition, clinical implications including potential long-term effects due to viral persistence (long COVID), as well as treatment options, are discussed.
Collapse
Affiliation(s)
- Noel G. Panagiotides
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Michael Poledniczek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | - Martin Andreas
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Martin Hülsmann
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Alfred A. Kocher
- Department of Cardiac Surgery, Medical University of Vienna, 1090 Vienna, Austria; (M.A.); (A.A.K.)
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Annika Weidenhammer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.G.P.); (M.P.); (M.H.); (A.W.); (N.P.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
7
|
Wu J, Liang Y, Fu P, Feng A, Lu Q, Unwalla HJ, Marciano DP, Black SM, Wang T. Sphingosine-1-Phosphate Receptor 3 Induces Endothelial Barrier Loss via ADAM10-Mediated Vascular Endothelial-Cadherin Cleavage. Int J Mol Sci 2023; 24:16083. [PMID: 38003272 PMCID: PMC10671260 DOI: 10.3390/ijms242216083] [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: 09/27/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Mechanical ventilation (MV) is a life-supporting strategy employed in the Intensive Care Unit (ICU). However, MV-associated mechanical stress exacerbates existing lung inflammation in ICU patients, resulting in limited improvement in mortality and a condition known as Ventilator-Induced Lung Injury (VILI). Sphingosine-1-phosphate (S1P) is a circulating bioactive lipid that maintains endothelial integrity primarily through S1P receptor 1 (S1PR1). During VILI, mechanical stress upregulates endothelial S1PR3 levels. Unlike S1PR1, S1PR3 mediates endothelial barrier disruption through Rho-dependent pathways. However, the specific impact of elevated S1PR3 on lung endothelial function, apart from Rho activation, remains poorly understood. In this study, we investigated the effects of S1PR3 in endothelial pathobiology during VILI using an S1PR3 overexpression adenovirus. S1PR3 overexpression caused cytoskeleton rearrangement, formation of paracellular gaps, and a modified endothelial response towards S1P. It resulted in a shift from S1PR1-dependent barrier enhancement to S1PR3-dependent barrier disruption. Moreover, S1PR3 overexpression induced an ADAM10-dependent cleavage of Vascular Endothelial (VE)-cadherin, which hindered endothelial barrier recovery. S1PR3-induced cleavage of VE-cadherin was at least partially regulated by S1PR3-mediated NFκB activation. Additionally, we employed an S1PR3 inhibitor TY-52156 in a murine model of VILI. TY-52156 effectively attenuated VILI-induced increases in bronchoalveolar lavage cell counts and protein concentration, suppressed the release of pro-inflammatory cytokines, and inhibited lung inflammation as assessed via a histological evaluation. These findings confirm that mechanical stress associated with VILI increases S1PR3 levels, thereby altering the pulmonary endothelial response towards S1P and impairing barrier recovery. Inhibiting S1PR3 is validated as an effective therapeutic strategy for VILI.
Collapse
Affiliation(s)
- Jialin Wu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Hoshang J. Unwalla
- Department of Immunology and Nanomedicine, Florida International University, Miami, FL 33199, USA
| | - David P. Marciano
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Cellular Biology and Pharmacology, Florida International University, Miami, FL 33199, USA
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular Biology and Pharmacology, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular Biology and Pharmacology, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
8
|
Zhao BH, Ruze A, Zhao L, Li QL, Tang J, Xiefukaiti N, Gai MT, Deng AX, Shan XF, Gao XM. The role and mechanisms of microvascular damage in the ischemic myocardium. Cell Mol Life Sci 2023; 80:341. [PMID: 37898977 PMCID: PMC11073328 DOI: 10.1007/s00018-023-04998-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023]
Abstract
Following myocardial ischemic injury, the most effective clinical intervention is timely restoration of blood perfusion to ischemic but viable myocardium to reduce irreversible myocardial necrosis, limit infarct size, and prevent cardiac insufficiency. However, reperfusion itself may exacerbate cell death and myocardial injury, a process commonly referred to as ischemia/reperfusion (I/R) injury, which primarily involves cardiomyocytes and cardiac microvascular endothelial cells (CMECs) and is characterized by myocardial stunning, microvascular damage (MVD), reperfusion arrhythmia, and lethal reperfusion injury. MVD caused by I/R has been a neglected problem compared to myocardial injury. Clinically, the incidence of microvascular angina and/or no-reflow due to ineffective coronary perfusion accounts for 5-50% in patients after acute revascularization. MVD limiting drug diffusion into injured myocardium, is strongly associated with the development of heart failure. CMECs account for > 60% of the cardiac cellular components, and their role in myocardial I/R injury cannot be ignored. There are many studies on microvascular obstruction, but few studies on microvascular leakage, which may be mainly due to the lack of corresponding detection methods. In this review, we summarize the clinical manifestations, related mechanisms of MVD during myocardial I/R, laboratory and clinical examination means, as well as the research progress on potential therapies for MVD in recent years. Better understanding the characteristics and risk factors of MVD in patients after hemodynamic reconstruction is of great significance for managing MVD, preventing heart failure and improving patient prognosis.
Collapse
Affiliation(s)
- Bang-Hao Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Jing Tang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Nilupaer Xiefukaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Min-Tao Gai
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - An-Xia Deng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xue-Feng Shan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, the First Affiliated Hospital of Xinjiang Medical University, Clinical Medical Research Institute of Xinjiang Medical University, 137 Liyushan South Road, Urumqi, 830054, China.
- Xinjiang Key Laboratory of Medical Animal Model Research, Urumqi, China.
| |
Collapse
|
9
|
Alshaikh RA, Zaki RGE, El Din RAS, Ryan KB, Waeber C. Siponimod As a Novel Inhibitor of Retinal Angiogenesis: In Vitro and In Vivo Evidence of Therapeutic Efficacy. J Pharmacol Exp Ther 2023; 386:224-241. [PMID: 37188532 DOI: 10.1124/jpet.122.001529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/13/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) receptors control endothelial cell proliferation, migration, and survival. Evidence of the ability of S1P receptor modulators to influence multiple endothelial cell functions suggests their potential use for antiangiogenic effect. The main purpose of our study was to investigate the potential of siponimod for the inhibition of ocular angiogenesis in vitro and in vivo. We investigated the effects of siponimod on the metabolic activity (thiazolyl blue tetrazolium bromide assay), cell toxicity (lactate dehydrogenase release), basal proliferation and growth factor-induced proliferation (bromodeoxyuridine assay), and migration (transwell migration assay) of human umbilical vein endothelial cells (HUVEC) and retinal microvascular endothelial cells (HRMEC). The effects of siponimod on HRMEC monolayer integrity, barrier function under basal conditions, and tumor necrosis factor alpha (TNF-α)-induced disruption were assessed using the transendothelial electrical resistance and fluorescein isothiocyanate-dextran permeability assays. Siponimod's effect on TNF-α-induced distribution of barrier proteins in HRMEC was investigated using immunofluorescence. Finally, the effect of siponimod on ocular neovascularization in vivo was assessed using suture-induced corneal neovascularization in albino rabbits. Our results show that siponimod did not affect endothelial cell proliferation or metabolic activity but significantly inhibited endothelial cell migration, increased HRMEC barrier integrity, and reduced TNF-α-induced barrier disruption. Siponimod also protected against TNF-α-induced disruption of claudin-5, zonula occludens-1, and vascular endothelial-cadherin in HRMEC. These actions are mainly mediated by sphingosine-1-phosphate receptor 1 modulation. Finally, siponimod prevented the progression of suture-induced corneal neovascularization in albino rabbits. In conclusion, the effects of siponimod on various processes known to be involved in angiogenesis support its therapeutic potential in disorders associated with ocular neovascularization. SIGNIFICANCE STATEMENT: Siponimod is an extensively characterized sphingosine-1-phosphate receptor modulator already approved for the treatment of multiple sclerosis. It inhibited retinal endothelial cell migration, potentiated endothelial barrier function, protected against tumor necrosis factor alpha-induced barrier disruption, and also inhibited suture-induced corneal neovascularization in rabbits. These results support its use for a novel therapeutic indication in the management of ocular neovascular diseases.
Collapse
Affiliation(s)
- Rasha A Alshaikh
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| | - Rania Gamal Eldin Zaki
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| | - Rania A Salah El Din
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| | - Katie B Ryan
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| | - Christian Waeber
- School of Pharmacy (R.A.A., K.B.R., C.W.), SSPC The SFI Research Centre for Pharmaceuticals, School of Pharmacy (K.B.R.), and Department of Pharmacology and Therapeutics (C.W.), University College Cork, Cork, Ireland; Department of Ophthalmology (R.G.E.Z.) and Department of Anatomy and Embryology (R.A.S.E.D.), Faculty of Medicine, Ain Shams University, Cairo, Egypt; and Department of Anatomy and Embryology, Faculty of Medicine, Newgiza University (R.A.S.E.D.)
| |
Collapse
|
10
|
Kloka JA, Friedrichson B, Wülfroth P, Henning R, Zacharowski K. Microvascular Leakage as Therapeutic Target for Ischemia and Reperfusion Injury. Cells 2023; 12:1345. [PMID: 37408180 DOI: 10.3390/cells12101345] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/03/2023] [Accepted: 05/07/2023] [Indexed: 07/07/2023] Open
Abstract
Reperfusion injury is a very common complication of various indicated therapies such as the re-opening of vessels in the myocardium or brain as well as reflow in hemodynamic shutdown (cardiac arrest, severe trauma, aortic cross-clamping). The treatment and prevention of reperfusion injury has therefore been a topic of immense interest in terms of mechanistic understanding, the exploration of interventions in animal models and in the clinical setting in major prospective studies. While a wealth of encouraging results has been obtained in the lab, the translation into clinical success has met with mixed outcomes at best. Considering the still very high medical need, progress continues to be urgently needed. Multi-target approaches rationally linking interference with pathophysiological pathways as well as a renewed focus on aspects of microvascular dysfunction, especially on the role of microvascular leakage, are likely to provide new insights.
Collapse
Affiliation(s)
- Jan Andreas Kloka
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | - Benjamin Friedrichson
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| | | | | | - Kai Zacharowski
- Department of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany
| |
Collapse
|
11
|
Shcheblykin DV, Bolgov AA, Pokrovskii MV, Stepenko JV, Tsuverkalova JM, Shcheblykina OV, Golubinskaya PA, Korokina LV. Endothelial dysfunction: developmental mechanisms and therapeutic strategies. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.80376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Introduction: Every year the importance of the normal functioning of the endothelial layer of the vascular wall in maintaining the health of the body becomes more and more obvious.
The physiological role of the endothelium: The endothelium is a metabolically active organ actively involved in the regulation of hemostasis, modulation of inflammation, maintenance of hemovascular homeostasis, regulation of angiogenesis, vascular tone, and permeability.
Risk factors for the development of endothelial dysfunction: Currently, insufficient bioavailability of nitric oxide is considered the most significant risk factor for endothelial dysfunction.
Mechanisms of development of endothelial dysfunction: The genesis of endothelial dysfunction is a multifactorial process. Among various complex mechanisms, this review examines oxidative stress, inflammation, hyperglycemia, vitamin D deficiency, dyslipidemia, excess visceral fat, hyperhomocysteinemia, hyperuricemia, as well as primary genetic defect of endotheliocytes, as the most common causes in the population underlying the development of endothelial dysfunction.
Markers of endothelial dysfunction in various diseases: This article discusses the main biomarkers of endothelial dysfunction currently used, as well as promising biomarkers in the future for laboratory diagnosis of this pathology.
Therapeutic strategies: Therapeutic approaches to the endothelium in order to prevent or reduce a degree of damage to the vascular wall are briefly described.
Conclusion: Endothelial dysfunction is a typical pathological process involved in the pathogenesis of many diseases. Thus, pharmacological agents with endothelioprotective properties can provide more therapeutic benefits than a drug without such an effect.
Collapse
|
12
|
Al-Rashdi AA, Sabt BI, Al-Mujaini AS. Effect of fingolimod therapy on quantitative macular changes among patients with relapsing-remitting multiple sclerosis: a four-year follow-up study from Oman. BMC Ophthalmol 2022; 22:470. [PMID: 36471269 PMCID: PMC9720917 DOI: 10.1186/s12886-022-02701-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/23/2022] [Indexed: 12/09/2022] Open
Abstract
PURPOSE Fingolimod (FTY-720) is an immunomodulatory oral agent approved for the treatment of relapsing-remitting multiple sclerosis (RRMS); however, several clinical trials have shown that some recipients may develop macular oedema (ME) as an adverse reaction. As there are no studies assessing the long-term (> 1 year) effect of fingolimod on the macula, this study aimed to evaluate the quantitative effect of fingolimod therapy on central macular thickness (CMT) and total macular volume (TMV) over a four-year period. METHODS This retrospective longitudinal cohort study was performed between January 2014 and December 2018. A total of 21 patients with RRMS receiving fingolimod therapy were recruited and followed-up over 4 years to assess CMT and TMV changes measured using spectral domain optical coherence tomography. A paired sample t-test was used to compare mean CMT and TMV values calculated at baseline prior to the initiation of fingolimod therapy with those observed at three, six, 12, 24, 36 and 48 months of treatment. RESULTS None of the patients developed ME over the four-year study period. In addition, there was no significant difference in baseline mean CMT values and those observed at a four-year follow-up. Although mean TMV values remained constant initially, there was a significant decrease towards the end of the study period. CONCLUSIONS Long-term fingolimod therapy did not result in significant CFT changes. While there was a reduction in TMV towards the end of the study, this is likely due to the degenerative effect of the disease itself on the nerve fibres of the retina.
Collapse
Affiliation(s)
| | - Buthaina I. Sabt
- grid.412855.f0000 0004 0442 8821Department of Ophthalmology, Sultan Qaboos University Hospital, Muscat, Oman
| | - Abdullah S. Al-Mujaini
- grid.412846.d0000 0001 0726 9430Department of Ophthalmology, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| |
Collapse
|
13
|
Yazbeck P, Cullere X, Bennett P, Yajnik V, Wang H, Kawada K, Davis V, Parikh A, Kuo A, Mysore V, Hla T, Milstone D, Mayadas TN. DOCK4 Regulation of Rho GTPases Mediates Pulmonary Vascular Barrier Function. Arterioscler Thromb Vasc Biol 2022; 42:886-902. [PMID: 35477279 PMCID: PMC9233130 DOI: 10.1161/atvbaha.122.317565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 04/12/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND The vascular endothelium maintains tissue-fluid homeostasis by controlling the passage of large molecules and fluid between the blood and interstitial space. The interaction of catenins and the actin cytoskeleton with VE-cadherin (vascular endothelial cadherin) is the primary mechanism for stabilizing AJs (adherens junctions), thereby preventing lung vascular barrier disruption. Members of the Rho (Ras homology) family of GTPases and conventional GEFs (guanine exchange factors) of these GTPases have been demonstrated to play important roles in regulating endothelial permeability. Here, we evaluated the role of DOCK4 (dedicator of cytokinesis 4)-an unconventional Rho family GTPase GEF in vascular function. METHODS We generated mice deficient in DOCK4' used DOCK4 silencing and reconstitution approaches in human pulmonary artery endothelial cells' used assays to evaluate protein localization, endothelial cell permeability, and small GTPase activation. RESULTS Our data show that DOCK4-deficient mice are viable. However, these mice have hemorrhage selectively in the lung, incomplete smooth muscle cell coverage in pulmonary vessels, increased basal microvascular permeability, and impaired response to S1P (sphingosine-1-phosphate)-induced reversal of thrombin-induced permeability. Consistent with this, DOCK4 rapidly translocates to the cell periphery and associates with the detergent-insoluble fraction following S1P treatment, and its absence prevents S1P-induced Rac-1 activation and enhancement of barrier function. Moreover, DOCK4-silenced pulmonary artery endothelial cells exhibit enhanced basal permeability in vitro that is associated with enhanced Rho GTPase activation. CONCLUSIONS Our findings indicate that DOCK4 maintains AJs necessary for lung vascular barrier function by establishing the normal balance between RhoA (Ras homolog family member A) and Rac-1-mediated actin cytoskeleton remodeling, a previously unappreciated function for the atypical GEF family of molecules. Our studies also identify S1P as a potential upstream regulator of DOCK4 activity.
Collapse
Affiliation(s)
- Pascal Yazbeck
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Xavier Cullere
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Paul Bennett
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Vijay Yajnik
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02445
| | - Huan Wang
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Kenji Kawada
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02445
| | - Vanessa Davis
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Asit Parikh
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02445
| | - Andrew Kuo
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, MA 20115
| | - Vijayashree Mysore
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Timothy Hla
- Vascular Biology Program, Boston Children’s Hospital and Harvard Medical School, Boston, MA 20115
| | - David Milstone
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| | - Tanya N. Mayadas
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115
| |
Collapse
|
14
|
Nuñez-Borque E, Fernandez-Bravo S, Yuste-Montalvo A, Esteban V. Pathophysiological, Cellular, and Molecular Events of the Vascular System in Anaphylaxis. Front Immunol 2022; 13:836222. [PMID: 35371072 PMCID: PMC8965328 DOI: 10.3389/fimmu.2022.836222] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/07/2022] [Indexed: 01/10/2023] Open
Abstract
Anaphylaxis is a systemic hypersensitivity reaction that can be life threatening. Mechanistically, it results from the immune activation and release of a variety of mediators that give rise to the signs and symptoms of this pathological event. For years, most of the research in anaphylaxis has focused on the contribution of the immune component. However, approaches that shed light on the participation of other cellular and molecular agents are necessary. Among them, the vascular niche receives the various signals (e.g., histamine) that elicit the range of anaphylactic events. Cardiovascular manifestations such as increased vascular permeability, vasodilation, hypotension, vasoconstriction, and cardiac alterations are crucial in the pathophysiology of anaphylaxis and are highly involved to the development of the most severe cases. Specifically, the endothelium, vascular smooth muscle cells, and their molecular signaling outcomes play an essential role downstream of the immune reaction. Therefore, in this review, we synthesized the vascular changes observed during anaphylaxis as well as its cellular and molecular components. As the risk of anaphylaxis exists both in clinical procedures and in routine life, increasing our knowledge of the vascular physiology and their molecular mechanism will enable us to improve the clinical management and how to treat or prevent anaphylaxis. Key Message Anaphylaxis, the most severe allergic reaction, involves a variety of immune and non-immune molecular signals that give rise to its pathophysiological manifestations. Importantly, the vascular system is engaged in processes relevant to anaphylactic events such as increased vascular permeability, vasodilation, hypotension, vasoconstriction, and decreased cardiac output. The novelty of this review focuses on the fact that new studies will greatly improve the understanding of anaphylaxis when viewed from a vascular molecular angle and specifically from the endothelium. This knowledge will improve therapeutic options to treat or prevent anaphylaxis.
Collapse
Affiliation(s)
- Emilio Nuñez-Borque
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Sergio Fernandez-Bravo
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alma Yuste-Montalvo
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, Instituto en Investigación Sanitaria - Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain.,Faculty of Medicine and Biomedicine, Alfonso X El Sabio University, Madrid, Spain
| |
Collapse
|
15
|
Nikdoust F, Pazoki M, Mohammadtaghizadeh M, Aghaali MK, Amrovani M. Exosomes: Potential Player in Endothelial Dysfunction in Cardiovascular Disease. Cardiovasc Toxicol 2022; 22:225-235. [PMID: 34669097 PMCID: PMC8527819 DOI: 10.1007/s12012-021-09700-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/24/2021] [Indexed: 02/08/2023]
Abstract
Exosomes are spherical bilayer membrane vesicles with an average diameter of 40-100 nm. These particles perform a wide range of biological activities due to their contents, including proteins, nucleic acids, lipids, lncRNA, and miRNA. Exosomes are involved in inflammation induction, oxidative stress and apoptosis, which can be effective in endothelial dysfunction. Due to the induction of mentioned processes in the endothelial cells, the intercellular connections are destroyed, cell permeability increases and finally cell efficiency decreases and functional defects occur. Cardiovascular disease (CVDs) are of consequences of endothelial dysfunction. Thus by identifying the exosome signaling pathways, which induce inflammation, oxidative stress, and apoptosis, endothelial dysfunction and subsequently CVDs can be reduced; exosomes can be used for appropriate target therapy.
Collapse
Affiliation(s)
- Farahnaz Nikdoust
- Department of Cardiology, Shariati Hospital, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Pazoki
- Department of Cardiology, Rasoul Akram General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mahsa Karimzadeh Aghaali
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Amrovani
- High Institute for Education and Research in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
16
|
Zhang H, Kim H, Park BW, Noh M, Kim Y, Park J, Park JH, Kim JJ, Sim WS, Ban K, Park HJ, Kwon YG. CU06-1004 enhances vascular integrity and improves cardiac remodeling by suppressing edema and inflammation in myocardial ischemia-reperfusion injury. Exp Mol Med 2022; 54:23-34. [PMID: 34997212 PMCID: PMC8814060 DOI: 10.1038/s12276-021-00720-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury accelerates the cardiomyocytes (CMs) death by oxidative stress, and thereby deteriorates cardiac function. There has been a paradigm shift in the therapeutic perspective more towards the prevention or amelioration of damage caused by reperfusion. Cardiac microvascular endothelial cells (CMECs) are more vulnerable to reperfusion injury and play the crucial roles more than CMs in the pathological process of early I/R injury. In this study, we investigate that CU06-1004, as a vascular leakage blocker, can improve cardiac function by inhibiting CMEC's hyperpermeability and subsequently reducing the neutrophil's plugging and infiltration in infarcted hearts. CU06-1004 was delivered intravenously 5 min before reperfusion and the rats were randomly divided into three groups: (1) vehicle, (2) low-CU06-1004 (1 mg/kg, twice at 24 h intervals), and (3) high-CU06-1004 (5 mg/kg, once before reperfusion). CU06-1004 treatment reduced necrotic size and cardiac edema by enhancing vascular integrity, as demonstrated by the presence of intact junction proteins on CMECs and surrounding pericytes in early I/R injury. It also decreased the expression of vascular cell adhesion molecule 1 (VCAM-1) on CMECs, resulting in reduced infiltration of neutrophils and macrophages. Echocardiography showed that the CU06-1004 treatment significantly improved cardiac function compared with the vehicle group. Interestingly, single high-dose treatment with CU06-1004 provided a greater functional improvement than repetitive low-dose treatment until 8 weeks post I/R. These findings demonstrate that CU06-1004 enhances vascular integrity and improves cardiac function by preventing lethal myocardial I/R injury. It can provide a promising therapeutic option, as potential adjunctive therapy to current reperfusion strategies.
Collapse
Affiliation(s)
- Haiying Zhang
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749 Republic of Korea ,R&D Department, Curacle Co. Ltd, Seongnam-si, Republic of Korea
| | - Hyeok Kim
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Bong Woo Park
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Minyoung Noh
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749 Republic of Korea
| | - Yeomyeong Kim
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749 Republic of Korea
| | - Jeongeun Park
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749 Republic of Korea
| | - Jae-Hyun Park
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Jin-Ju Kim
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Woo-Sup Sim
- grid.411947.e0000 0004 0470 4224Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591 Republic of Korea
| | - Kiwon Ban
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, City University of Hong Kong, Kowloon Tong, 999077 Hong Kong
| | - Hun-Jun Park
- Department of Medical Life Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea. .,Division of Cardiology, Department of Internal Medicine, The Catholic University of Korea, Banpo-daero 222, Seocho-gu, Seoul, 137701, Republic of Korea.
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 120-749, Republic of Korea.
| |
Collapse
|
17
|
HDL and Endothelial Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:27-47. [DOI: 10.1007/978-981-19-1592-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
18
|
van Leeuwen ALI, Borgdorff MP, Dekker NAM, van den Brom CE. Therapeutically Targeting Microvascular Leakage in Experimental Hemorrhagic SHOCK: A Systematic Review and Meta-Analysis. Shock 2021; 56:890-900. [PMID: 33927137 DOI: 10.1097/shk.0000000000001796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND Microvascular leakage is proposed as main contributor to disturbed microcirculatory perfusion following hemorrhagic shock and fluid resuscitation, leading to organ dysfunction and unfavorable outcome. Currently, no drugs are available to reduce or prevent microvascular leakage in clinical practice. We therefore aimed to provide an overview of therapeutic agents targeting microvascular leakage following experimental hemorrhagic shock and fluid resuscitation. METHODS PubMed, EMBASE.com, and Cochrane Library were searched in January 2021 for preclinical studies of hemorrhagic shock using any therapeutic agent on top of standard fluid resuscitation. Primary outcome was vascular leakage, defined as edema, macromolecule extravasation, or glycocalyx degradation. Drugs were classified by targeting pathways and subgroup analyses were performed per organ. RESULTS Forty-five studies, published between 1973 and 2020, fulfilled eligibility criteria. The included studies tested 54 different therapeutics mainly in pulmonary and intestinal vascular beds. Most studies induced trauma besides hemorrhagic shock. Forty-four therapeutics (81%) were found effective to reduce microvascular leakage, edema formation, or glycocalyx degradation in at least one organ. Targeting oxidative stress and apoptosis was the predominantly effective strategy (SMD: -2.18, CI [-3.21, -1.16], P < 0.0001). Vasoactive agents were found noneffective in reducing microvascular leakage (SMD: -0.86, CI [-3.07, 1.36], P = 0.45). CONCLUSION Pharmacological modulation of pathways involved in cell metabolism, inflammation, endothelial barrier regulation, sex hormones and especially oxidative stress and apoptosis were effective in reducing microvascular leakage in experimental hemorrhagic shock with fluid resuscitation. Future studies should investigate whether targeting these pathways can restore microcirculatory perfusion and reduce organ injury following hemorrhagic shock. SYSTEMATIC REVIEW REGISTRATION NUMBER CRD42018095432.
Collapse
Affiliation(s)
- Anoek L I van Leeuwen
- Department of Anesthesiology, Amsterdam UMC, VU University, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Department of Physiology, Amsterdam UMC, VU University, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Marieke P Borgdorff
- Department of Anesthesiology, Amsterdam UMC, VU University, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Nicole A M Dekker
- Department of Anesthesiology, Amsterdam UMC, VU University, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Department of Physiology, Amsterdam UMC, VU University, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | - Charissa E van den Brom
- Department of Anesthesiology, Amsterdam UMC, VU University, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Department of Physiology, Amsterdam UMC, VU University, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
- Department of Intensive Care, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Akbari E, Spychalski GB, Menyhert MM, Rangharajan KK, Tinapple JW, Prakash S, Song JW. Endothelial barrier function is co-regulated at vessel bifurcations by fluid forces and sphingosine-1-phosphate. BIOMATERIALS AND BIOSYSTEMS 2021; 3:100020. [PMID: 35317095 PMCID: PMC8936769 DOI: 10.1016/j.bbiosy.2021.100020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/12/2021] [Accepted: 05/29/2021] [Indexed: 12/31/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid mediator of endothelial barrier function. Prior studies have implicated mechanical stimulation due to intravascular laminar shear stress in co-regulating S1P signaling in endothelial cells (ECs). Yet, vascular networks in vivo consist of vessel bifurcations, and this geometry generates hemodynamic forces at the bifurcation point distinct from laminar shear stress. However, the role of these forces at vessel bifurcations in regulating S1P-dependent endothelial barrier function is not known. In this study, we implemented a microfluidic platform that recapitulates the flow dynamics of vessel bifurcations with in situ quantification of the permeability of microvessel analogues. Co-application of S1P with impinging bifurcated fluid flow, which is characterized by approximately zero shear stress and 38 dyn•cm-2 stagnation pressure at the vessel bifurcation point, promotes vessel stabilization. Similarly, co-treatment of S1P with 3 dyn•cm-2 laminar shear stress is also protective of endothelial barrier function. Moreover, it is shown that vessel stabilization due to bifurcated fluid flow and laminar shear stress is dependent on S1P receptor 1 or 2 signaling. Collectively, these findings demonstrate the endothelium-protective function of fluid forces at vessel bifurcations and their involvement in coordinating S1P-dependent regulation of vessel permeability.
Collapse
Affiliation(s)
- Ehsan Akbari
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Griffin B. Spychalski
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Miles M. Menyhert
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Kaushik K. Rangharajan
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Joseph W. Tinapple
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States, 43210
| | - Shaurya Prakash
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States, 43210
| | - Jonathan W. Song
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States, 43210
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States, 43210
| |
Collapse
|
20
|
Dubrovskyi O, Hasten E, Dudek SM, Flavin MT, Chan LLY. Development of an Image-Based HCS-Compatible Method for Endothelial Barrier Function Assessment. SLAS DISCOVERY 2021; 26:1079-1090. [PMID: 34269109 DOI: 10.1177/24725552211030900] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The recent renascence of phenotypic drug discovery (PDD) is catalyzed by its ability to identify first-in-class drugs and deliver results when the exact molecular mechanism is partially obscure. Acute respiratory distress syndrome (ARDS) is a severe, life-threatening condition with a high mortality rate that has increased in frequency due to the COVID-19 pandemic. Despite decades of laboratory and clinical study, no efficient pharmacological therapy for ARDS has been found. An increase in endothelial permeability is the primary event in ARDS onset, causing the development of pulmonary edema that leads to respiratory failure. Currently, the detailed molecular mechanisms regulating endothelial permeability are poorly understood. Therefore, the use of the PDD approach in the search for efficient ARDS treatment can be more productive than classic target-based drug discovery (TDD), but its use requires a new cell-based assay compatible with high-throughput (HTS) and high-content (HCS) screening. Here we report the development of a new plate-based image cytometry method to measure endothelial barrier function. The incorporation of image cytometry in combination with digital image analysis substantially decreases assay variability and increases the signal window. This new method simultaneously allows for rapid measurement of cell monolayer permeability and cytological analysis. The time-course of permeability increase in human pulmonary artery endothelial cells (HPAECs) in response to the thrombin and tumor necrosis factor α treatment correlates with previously published data obtained by transendothelial resistance (TER) measurements. Furthermore, the proposed image cytometry method can be easily adapted for HTS/HCS applications.
Collapse
Affiliation(s)
- Oleksii Dubrovskyi
- UICentre, College of Pharmacy, University of Illinois in Chicago, Chicago, IL, USA
| | - Erica Hasten
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC, Lawrence, MA, USA
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep, and Allergy, College of Medicine, University of Illinois in Chicago, Chicago, IL, USA
| | - Michael T Flavin
- UICentre, College of Pharmacy, University of Illinois in Chicago, Chicago, IL, USA
| | - Leo Li-Ying Chan
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC, Lawrence, MA, USA
| |
Collapse
|
21
|
Jujic A, Matthes F, Vanherle L, Petzka H, Orho-Melander M, Nilsson PM, Magnusson M, Meissner A. Plasma S1P (Sphingosine-1-Phosphate) Links to Hypertension and Biomarkers of Inflammation and Cardiovascular Disease: Findings From a Translational Investigation. Hypertension 2021; 78:195-209. [PMID: 33993723 DOI: 10.1161/hypertensionaha.120.17379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Amra Jujic
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
- Wallenberg Centre for Molecular Medicine (A.J., F.M., L.V., M.M., A.M.), Lund University, Malmö, Sweden
- Lund University Diabetes Centre (A.J.), Lund University, Malmö, Sweden
| | - Frank Matthes
- Wallenberg Centre for Molecular Medicine (A.J., F.M., L.V., M.M., A.M.), Lund University, Malmö, Sweden
- Department of Experimental Medical Sciences (F.M., L.V., A.M.), Lund University, Malmö, Sweden
| | - Lotte Vanherle
- Wallenberg Centre for Molecular Medicine (A.J., F.M., L.V., M.M., A.M.), Lund University, Malmö, Sweden
- Department of Experimental Medical Sciences (F.M., L.V., A.M.), Lund University, Malmö, Sweden
| | - Henning Petzka
- Department of Mathematics, Lund Technical University, Sweden (H.P.)
| | - Marju Orho-Melander
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
| | - Peter M Nilsson
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
- Department of Internal Medicine, Clinical Research Unit, Malmö, Sweden (P.M.N.)
| | - Martin Magnusson
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
- Wallenberg Centre for Molecular Medicine (A.J., F.M., L.V., M.M., A.M.), Lund University, Malmö, Sweden
- Hypertension in Africa Research Team, North West University Potchefstroom, South Africa (M.M.)
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden (M.M.)
| | - Anja Meissner
- Department of Clinical Sciences (A.J., M.O.-M., P.M.N., M.M.), Lund University, Malmö, Sweden
- Department of Experimental Medical Sciences (F.M., L.V., A.M.), Lund University, Malmö, Sweden
| |
Collapse
|
22
|
Ziegler AC, Gräler MH. Barrier maintenance by S1P during inflammation and sepsis. Tissue Barriers 2021; 9:1940069. [PMID: 34152926 DOI: 10.1080/21688370.2021.1940069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a multifaceted lipid signaling molecule that activates five specific G protein-coupled S1P receptors. Despite the fact that S1P is known as one of the strongest barrier-enhancing molecules for two decades, no medical application is available yet. The reason for this lack of translation into clinical practice may be the complex regulatory network of S1P signaling, metabolism and transportation.In this review, we will provide an overview about the physiology and the network of S1P signaling with the focus on endothelial barrier maintenance in inflammation. We briefly describe the physiological role of S1P and the underlying S1P signaling in barrier maintenance, outline differences of S1P signaling and metabolism in inflammatory diseases, discuss potential targets and compounds for medical intervention, and summarize our current knowledge regarding the role of S1P in the maintenance of specialized barriers like the blood-brain barrier and the placenta.
Collapse
Affiliation(s)
- Anke C Ziegler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
23
|
Pepe G, Cotugno M, Marracino F, Giova S, Capocci L, Forte M, Stanzione R, Bianchi F, Marchitti S, Di Pardo A, Sciarretta S, Rubattu S, Maglione V. Differential Expression of Sphingolipid Metabolizing Enzymes in Spontaneously Hypertensive Rats: A Possible Substrate for Susceptibility to Brain and Kidney Damage. Int J Mol Sci 2021; 22:ijms22073796. [PMID: 33917593 PMCID: PMC8038804 DOI: 10.3390/ijms22073796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023] Open
Abstract
Alterations in the metabolism of sphingolipids, a class of biologically active molecules in cell membranes with direct effect on vascular homeostasis, are increasingly recognized as important determinant in different vascular disorders. However, it is not clear whether sphingolipids are implicated in the pathogenesis of hypertension-related cerebrovascular and renal damage. In this study, we evaluated the existence of possible abnormalities related to the sphingolipid metabolism in the brain and kidneys of two well validated spontaneously hypertensive rat strains, the stroke-prone (SHRSP) and the stroke-resistant (SHRSR) models, as compared to the normotensive Wistar Kyoto (WKY) rat strain. Our results showed a global alteration in the metabolism of sphingolipids in both cerebral and renal tissues of both hypertensive strains as compared to the normotensive rat. However, few defects, such as reduced expression of enzymes involved in the metabolism/catabolism of sphingosine-1-phosphate and in the de novo biosynthetic pathways, were exclusively detected in the SHRSP. Although further studies are necessary to fully understand the significance of these findings, they suggest that defects in specific lipid molecules and/or their related metabolic pathways may likely contribute to the pathogenesis of hypertensive target organ damage and may eventually serve as future therapeutic targets to reduce the vascular consequences of hypertension.
Collapse
Affiliation(s)
- Giuseppe Pepe
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Maria Cotugno
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Federico Marracino
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Susy Giova
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Luca Capocci
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Maurizio Forte
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Rosita Stanzione
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Franca Bianchi
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Simona Marchitti
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
| | - Alba Di Pardo
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
- Correspondence: (A.D.P.); (S.R.); (V.M.)
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina 04100, Italy;
| | - Speranza Rubattu
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University, Rome 00185, Italy
- Correspondence: (A.D.P.); (S.R.); (V.M.)
| | - Vittorio Maglione
- IRCCS Neuromed, Pozzilli 86077, Italy; (G.P.); (M.C.); (F.M.); (S.G.); (L.C.); (M.F.); (R.S.); (F.B.); (S.M.)
- Correspondence: (A.D.P.); (S.R.); (V.M.)
| |
Collapse
|
24
|
Park KS, Schecterson L, Gumbiner BM. Enhanced endothelial barrier function by monoclonal antibody activation of vascular endothelial cadherin. Am J Physiol Heart Circ Physiol 2021; 320:H1403-H1410. [PMID: 33577432 DOI: 10.1152/ajpheart.00002.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Excessive vascular permeability occurs in inflammatory disease processes. Vascular endothelial cadherin (VE-cadherin) is an adhesion protein that controls vascular permeability. We identified monoclonal antibodies (mAbs) to human VE-cadherin that activate cell adhesion and inhibit the increased permeability of endothelial cell monolayers induced by thrombin receptor activator peptide-6 (TRAP-6). Two mAbs, 8A12c and 3A5a, reduce permeability, whereas an inhibitory mAb, 2E11d, enhances permeability. Activating mAbs also reduce permeability induced by tumor necrosis factor-α (TNF-α) and vascular endothelial cell growth factor (VEGF). The activating mAbs also stabilize the organization of the adherens junctions that are disrupted by TRAP-6, VEGF, or TNF-α. The activating mAbs act directly on the adhesive function of VE-cadherin because they did not block the accumulation of actin filaments stimulated by TRAP-6 and enhance physical cell-cell adhesion of VE-cadherin-expressing tissue culture cells. Therefore, VE-cadherin function can be regulated at the cell surface to control endothelial permeability.NEW & NOTEWORTHY Excessive vascular permeability is a serious complication of many inflammatory disease conditions. We have developed monoclonal antibodies that inhibit increases in endothelial monolayer permeability induced by several signaling factors by activating VE-cadherin mediated adhesion and stabilizing cell junctions. These antibodies and/or the mechanisms they reveal may lead to important therapeutics to treat vascular leakiness and inflammation.
Collapse
Affiliation(s)
- Ki-Sook Park
- Department of Biomedical Science and Technology/East-West Medical Research Institute, Kyung Hee University, Seoul, South Korea.,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Leslayann Schecterson
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Barry M Gumbiner
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington.,Department of Pediatrics, University of Washington, Seattle, Washington.,Department of Biochemistry, University of Washington, Seattle, Washington
| |
Collapse
|
25
|
Obesity and impaired barrier function after shock: A biomimetic in vitro model using microfluidics. J Trauma Acute Care Surg 2021; 89:544-550. [PMID: 32467464 DOI: 10.1097/ta.0000000000002804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Impaired microvascular perfusion in the obese patient has been linked to chronic adverse health consequences. The impact on acute illnesses including trauma, sepsis, and hemorrhagic shock (HS) is uncertain. Studies have shown that endothelial glycocalyx and vascular endothelial derangements are causally linked to perfusion abnormalities. Trauma and HS are also associated with impaired microvascular perfusion in which glycocalyx injury and endothelial dysfunction are sentinel events. We postulate that obesity may impact the adverse consequences of HS on the vascular barrier. This was studied in vivo in a biomimetic model of HS using microfluidic technology. METHODS Human umbilical vein endothelial cell monolayers were established in a microfluidic device. Cells were exposed to standard or biomimetic shock conditions (hypoxia plus epinephrine) followed by perfusion from plasma obtained from obese or nonobese subjects. Endothelial glycocalyx and endothelial cellular injury were then determined. RESULTS Plasma from nonobese patients completely reversed glycocalyx and endothelial vascular barrier injury. Plasma from obese patients was only partially protective and was associated with differences in adipokines and other substances in the plasma of these patients. CONCLUSION Our study supports that obesity impairs HS resuscitation. This may be due to microrheological differences between nonobese and obese individuals and may contribute to the poorer outcome in this patient population.
Collapse
|
26
|
Fu G, Dong Y, Zhang X, Hu K. Metabolomic profiles and pathways of praziquantel in crucian carp. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2020; 80:103466. [PMID: 32798734 DOI: 10.1016/j.etap.2020.103466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 06/11/2023]
Abstract
Praziquantel (PZQ) is a drug commonly used to treat some parasitic infections in animals. This study aimed to apply a reliable and simple method to identify important biological metabolites relevant to PZQ in crucian carp (Carassius auratus) to decipher the metabolic pathways and provide a basis for developing new anti-parasite drugs. The experimental group of crucian carp was administered oral PZQ at a dose of 10 mg kg-1 via a stomach feed tube. All biological blood samples were analysed using liquid chromatography electrospray ionization/quadrupole time-of-flight mass spectrometry (LC ESI/Q-TOF MS). MetPA analysis was used to identify relevant pathways for PZQ in crucian carp. Thirty-five potential metabolic pathways were revealed by MetPA network software. Furthermore, the chemical structures of the related metabolites and pathways were identified by comparison with data obtained from free online databases. Forty-four significantly differentially abundant endogenous metabolites were found in the PZQ-treated crucian carp. The changes in metabolomic profiles and pathways induced by PZQ played a role in inhibiting pathogenesis.
Collapse
Affiliation(s)
- Guihong Fu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai 201306, China; Hunan Engineering Research Center for Utilization of Characteristics of Aquatic Resources, Hunan Agricultural University, Changsha 410128, China
| | - Yaping Dong
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai 201306, China
| | - Xiaoming Zhang
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai 201306, China
| | - Kun Hu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, 999 Hucheng Huan Road, Shanghai 201306, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai 201306, China.
| |
Collapse
|
27
|
Herland A, Maoz BM, FitzGerald EA, Grevesse T, Vidoudez C, Sheehy SP, Budnik N, Dauth S, Mannix R, Budnik B, Parker KK, Ingber DE. Proteomic and Metabolomic Characterization of Human Neurovascular Unit Cells in Response to Methamphetamine. ACTA ACUST UNITED AC 2020; 4:e1900230. [PMID: 32744807 DOI: 10.1002/adbi.201900230] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 07/02/2020] [Indexed: 01/31/2023]
Abstract
The functional state of the neurovascular unit (NVU), composed of the blood-brain barrier and the perivasculature that forms a dynamic interface between the blood and the central nervous system (CNS), plays a central role in the control of brain homeostasis and is strongly affected by CNS drugs. Human primary brain microvascular endothelium, astrocyte, pericyte, and neural cell cultures are often used to study NVU barrier functions as well as drug transport and efficacy; however, the proteomic and metabolomic responses of these different cell types are not well characterized. Culturing each cell type separately, using deep coverage proteomic analysis and characterization of the secreted metabolome, as well as measurements of mitochondrial activity, the responses of these cells under baseline conditions and when exposed to the NVU-impairing stimulant methamphetamine (Meth) are analyzed. These studies define the previously unknown metabolic and proteomic profiles of human brain pericytes and lead to improved characterization of the phenotype of each of the NVU cell types as well as cell-specific metabolic and proteomic responses to Meth.
Collapse
Affiliation(s)
- Anna Herland
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Division of Micro and Nanosystems, KTH Royal Institute of Technology, Stockholm, 10044, Sweden.,AIMES, Center for the Advancement of Integrated Engineering and Medical Sciences, Department of Neuroscience, Karolinska Institute, Stockholm, 17177, Sweden
| | - Ben M Maoz
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA.,Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, 6997801, Israel.,Department of Biomedical Engineering, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Edward A FitzGerald
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Thomas Grevesse
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Charles Vidoudez
- Small Molecule Mass Spectrometry Facility, Harvard University, Cambridge, MA, 02138, USA
| | - Sean P Sheehy
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Nikita Budnik
- Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Stephanie Dauth
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Robert Mannix
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA
| | - Bogdan Budnik
- Mass Spectrometry and Proteomics Resource Laboratory, Harvard University, Cambridge, MA, 02138, USA
| | - Kevin Kit Parker
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Disease Biophysics Group, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, 02115, USA.,Vascular Biology Program and Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
28
|
Wafa D, Koch N, Kovács J, Kerék M, Proia RL, Tigyi GJ, Benyó Z, Miklós Z. Opposing Roles of S1P 3 Receptors in Myocardial Function. Cells 2020; 9:cells9081770. [PMID: 32722120 PMCID: PMC7466142 DOI: 10.3390/cells9081770] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/12/2020] [Accepted: 07/22/2020] [Indexed: 01/09/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a lysophospholipid mediator with diverse biological function mediated by S1P1–5 receptors. Whereas S1P was shown to protect the heart against ischemia/reperfusion (I/R) injury, other studies highlighted its vasoconstrictor effects. We aimed to separate the beneficial and potentially deleterious cardiac effects of S1P during I/R and identify the signaling pathways involved. Wild type (WT), S1P2-KO and S1P3-KO Langendorff-perfused murine hearts were exposed to intravascular S1P, I/R, or both. S1P induced a 45% decrease of coronary flow (CF) in WT-hearts. The presence of S1P-chaperon albumin did not modify this effect. CF reduction diminished in S1P3-KO but not in S1P2-KO hearts, indicating that in our model S1P3 mediates coronary vasoconstriction. In I/R experiments, S1P3 deficiency had no influence on postischemic CF but diminished functional recovery and increased infarct size, indicating a cardioprotective effect of S1P3. Preischemic S1P exposure resulted in a substantial reduction of postischemic CF and cardiac performance and increased the infarcted area. Although S1P3 deficiency increased postischemic CF, this failed to improve cardiac performance. These results indicate a dual role of S1P3 involving a direct protective action on the myocardium and a cardiosuppressive effect due to coronary vasoconstriction. In acute coronary syndrome when S1P may be released abundantly, intravascular and myocardial S1P production might have competing influences on myocardial function via activation of S1P3 receptors.
Collapse
Affiliation(s)
- Dina Wafa
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (N.K.); (J.K.); (M.K.); (G.J.T.); (Z.B.)
- Correspondence: (D.W.); (Z.M.)
| | - Nóra Koch
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (N.K.); (J.K.); (M.K.); (G.J.T.); (Z.B.)
| | - Janka Kovács
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (N.K.); (J.K.); (M.K.); (G.J.T.); (Z.B.)
| | - Margit Kerék
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (N.K.); (J.K.); (M.K.); (G.J.T.); (Z.B.)
| | - Richard L. Proia
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institues of Health, Bethesda, MD 20892, USA;
| | - Gábor J. Tigyi
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (N.K.); (J.K.); (M.K.); (G.J.T.); (Z.B.)
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (N.K.); (J.K.); (M.K.); (G.J.T.); (Z.B.)
| | - Zsuzsanna Miklós
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (N.K.); (J.K.); (M.K.); (G.J.T.); (Z.B.)
- Correspondence: (D.W.); (Z.M.)
| |
Collapse
|
29
|
Di Pardo A, Pepe G, Capocci L, Marracino F, Amico E, Del Vecchio L, Giova S, Jeong SK, Park BM, Park BD, Maglione V. Treatment with K6PC-5, a selective stimulator of SPHK1, ameliorates intestinal homeostasis in an animal model of Huntington's disease. Neurobiol Dis 2020; 143:105009. [PMID: 32634578 DOI: 10.1016/j.nbd.2020.105009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 02/08/2023] Open
Abstract
Emerging evidence indicates that Huntington's disease (HD) may be described as multi-organ pathology. In this context, we and others have contributed to demonstrate that the disease is characterized by an impairment of the homeostasis of gastro-intestinal (GI) tract. Sphingolipids represent a class of molecules involved in the regulation and maintenance of different tissues and organs including GI system. In this study, we investigated whether the alteration of Sphingosine-1-phosphate (S1P) metabolism, previously described in human HD brains and animal models, is also detectable peripherally in R6/2 HD mice. Our findings indicate, for the first time, that sphingolipid metabolism is perturbed early in the disease in the intestinal tract of HD mice and, its modulation by K6PC-5, a selective activator of S1P synthesis, preserved intestinal integrity and homeostasis. These results further support the evidence that modulation of sphingolipid pathways may represent a potential therapeutic option in HD and suggest that it has also the potential to counteract the peripheral disturbances which may usually complicate the management of the disease and affect patient's quality of life.
Collapse
Affiliation(s)
| | - G Pepe
- IRCCS Neuromed, Pozzilli, Italy
| | | | | | - E Amico
- IRCCS Neuromed, Pozzilli, Italy
| | - L Del Vecchio
- IRCCS Neuromed, Pozzilli, Italy; Unità complessa di radiodiagnostica (U.O.C.) POS, University of Foggia, Foggia, Italy
| | - S Giova
- IRCCS Neuromed, Pozzilli, Italy
| | - S K Jeong
- Department of Cosmetic Science, Seowon University, Cheongju, Republic of Korea
| | - B M Park
- NeoPharm USA Inc., Engelwood Cliffs, NJ, USA
| | - B D Park
- Dr. Raymond Laboratories, Inc, Englewood cliffs, NJ, USA
| | | |
Collapse
|
30
|
Probst CK, Montesi SB, Medoff BD, Shea BS, Knipe RS. Vascular permeability in the fibrotic lung. Eur Respir J 2020; 56:13993003.00100-2019. [PMID: 32265308 PMCID: PMC9977144 DOI: 10.1183/13993003.00100-2019] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 03/26/2020] [Indexed: 12/26/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is thought to result from aberrant tissue repair processes in response to chronic or repetitive lung injury. The origin and nature of the injury, as well as its cellular and molecular targets, are likely heterogeneous, which complicates accurate pre-clinical modelling of the disease and makes therapeutic targeting a challenge. Efforts are underway to identify central pathways in fibrogenesis which may allow targeting of aberrant repair processes regardless of the initial injury stimulus. Dysregulated endothelial permeability and vascular leak have long been studied for their role in acute lung injury and repair. Evidence that these processes are of importance to the pathogenesis of fibrotic lung disease is growing. Endothelial permeability is increased in non-fibrosing lung diseases, but it resolves in a self-limited fashion in conditions such as bacterial pneumonia and acute respiratory distress syndrome. In progressive fibrosing diseases such as IPF, permeability appears to persist, however, and may also predict mortality. In this hypothesis-generating review, we summarise available data on the role of endothelial permeability in IPF and focus on the deleterious consequences of sustained endothelial hyperpermeability in response to and during pulmonary inflammation and fibrosis. We propose that persistent permeability and vascular leak in the lung have the potential to establish and amplify the pro-fibrotic environment. Therapeutic interventions aimed at recognising and "plugging" the leak may therefore be of significant benefit for preventing the transition from lung injury to fibrosis and should be areas for future research.
Collapse
Affiliation(s)
- Clemens K. Probst
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sydney B. Montesi
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Benjamin D. Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Barry S. Shea
- Division of Pulmonary and Critical Care Medicine, Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Rachel S. Knipe
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
31
|
Panta CR, Ruisanchez É, Móré D, Dancs PT, Balogh A, Fülöp Á, Kerék M, Proia RL, Offermanns S, Tigyi GJ, Benyó Z. Sphingosine-1-Phosphate Enhances α 1-Adrenergic Vasoconstriction via S1P2-G 12/13-ROCK Mediated Signaling. Int J Mol Sci 2019; 20:ijms20246361. [PMID: 31861195 PMCID: PMC6941080 DOI: 10.3390/ijms20246361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 01/21/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) has been implicated recently in the physiology and pathology of the cardiovascular system including regulation of vascular tone. Pilot experiments showed that the vasoconstrictor effect of S1P was enhanced markedly in the presence of phenylephrine (PE). Based on this observation, we hypothesized that S1P might modulate α1-adrenergic vasoactivity. In murine aortas, a 20-minute exposure to S1P but not to its vehicle increased the Emax and decreased the EC50 of PE-induced contractions indicating a hyperreactivity to α1-adrenergic stimulation. The potentiating effect of S1P disappeared in S1P2 but not in S1P3 receptor-deficient vessels. In addition, smooth muscle specific conditional deletion of G12/13 proteins or pharmacological inhibition of the Rho-associated protein kinase (ROCK) by Y-27632 or fasudil abolished the effect of S1P on α1-adrenergic vasoconstriction. Unexpectedly, PE-induced contractions remained enhanced markedly as late as three hours after S1P-exposure in wild-type (WT) and S1P3 KO but not in S1P2 KO vessels. In conclusion, the S1P–S1P2–G12/13–ROCK signaling pathway appears to have a major influence on α1-adrenergic vasoactivity. This cooperativity might lead to sustained vasoconstriction when increased sympathetic tone is accompanied by increased S1P production as it occurs during acute coronary syndrome and stroke.
Collapse
Affiliation(s)
- Cecília R. Panta
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Correspondence: (C.R.P.); (Z.B.)
| | - Éva Ruisanchez
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Dorottya Móré
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Péter T. Dancs
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Andrea Balogh
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Ágnes Fülöp
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Margit Kerék
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
| | - Richard L. Proia
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), Bethesda, MD 20892, USA;
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany;
| | - Gábor J. Tigyi
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary (D.M.); (P.T.D.); (A.B.); (M.K.); (G.J.T.)
- Correspondence: (C.R.P.); (Z.B.)
| |
Collapse
|
32
|
Diarte-Añazco EMG, Méndez-Lara KA, Pérez A, Alonso N, Blanco-Vaca F, Julve J. Novel Insights into the Role of HDL-Associated Sphingosine-1-Phosphate in Cardiometabolic Diseases. Int J Mol Sci 2019; 20:ijms20246273. [PMID: 31842389 PMCID: PMC6940915 DOI: 10.3390/ijms20246273] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023] Open
Abstract
Sphingolipids are key signaling molecules involved in the regulation of cell physiology. These species are found in tissues and in circulation. Although they only constitute a small fraction in lipid composition of circulating lipoproteins, their concentration in plasma and distribution among plasma lipoproteins appears distorted under adverse cardiometabolic conditions such as diabetes mellitus. Sphingosine-1-phosphate (S1P), one of their main representatives, is involved in regulating cardiomyocyte homeostasis in different models of experimental cardiomyopathy. Cardiomyopathy is a common complication of diabetes mellitus and represents a main risk factor for heart failure. Notably, plasma concentration of S1P, particularly high-density lipoprotein (HDL)-bound S1P, may be decreased in patients with diabetes mellitus, and hence, inversely related to cardiac alterations. Despite this, little attention has been given to the circulating levels of either total S1P or HDL-bound S1P as potential biomarkers of diabetic cardiomyopathy. Thus, this review will focus on the potential role of HDL-bound S1P as a circulating biomarker in the diagnosis of main cardiometabolic complications frequently associated with systemic metabolic syndromes with impaired insulin signaling. Given the bioactive nature of these molecules, we also evaluated its potential of HDL-bound S1P-raising strategies for the treatment of cardiometabolic disease.
Collapse
Affiliation(s)
- Elena M. G. Diarte-Añazco
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
| | - Karen Alejandra Méndez-Lara
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| | - Antonio Pérez
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei d’Endocrinologia, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
| | - Núria Alonso
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei d’Endocrinologia, Hospital Universitari Germans Trias i Pujol, Badalona, 08916 Barcelona, Spain
| | - Francisco Blanco-Vaca
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Servei de Bioquímica, Hospital de la Santa Creu i Sant Pau, IIB Sant Pau, 08041 Barcelona, Spain
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| | - Josep Julve
- Institut de Recerca de l’Hospital de la Santa Creu i Sant Pau, and Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain;
- Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra (Barcelona), Spain;
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain;
- Correspondence: (K.A.M.-L.); (F.B.-V.); (J.J.)
| |
Collapse
|
33
|
Single-cell transcriptomics of the human retinal pigment epithelium and choroid in health and macular degeneration. Proc Natl Acad Sci U S A 2019; 116:24100-24107. [PMID: 31712411 PMCID: PMC6883845 DOI: 10.1073/pnas.1914143116] [Citation(s) in RCA: 254] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The retinal pigment epithelium and the choroid are complex tissues whose dysfunction can lead to irreversible visual loss. In this study, single-cell RNA sequencing of both of these tissues was performed to characterize gene expression patterns specific to the retinal pigment epithelium and all major choroidal cell populations. Unique gene expression signatures of arterial, venous, and choriocapillaris vascular beds within the choroid were identified. RGCC, a gene that responds to complement and has been shown to induce endothelial apoptosis, was specifically expressed in choriocapillaris endothelial cells. This study provides potential insight into the molecular mechanisms of choroidal vascular disease and its contribution to age-related macular degeneration. The human retinal pigment epithelium (RPE) and choroid are complex tissues that provide crucial support to the retina. Disease affecting either of these supportive tissues can lead to irreversible blindness in the setting of age-related macular degeneration. In this study, single-cell RNA sequencing was performed on macular and peripheral regions of RPE-choroid from 7 human donor eyes in 2 independent experiments. In the first experiment, total RPE/choroid preparations were evaluated and expression profiles specific to RPE and major choroidal cell populations were identified. As choroidal endothelial cells represent a minority of the total RPE/choroidal cell population but are strongly implicated in age-related macular degeneration (AMD) pathogenesis, a second single-cell RNA-sequencing experiment was performed using endothelial cells enriched by magnetic separation. In this second study, we identified gene expression signatures along the choroidal vascular tree, classifying the transcriptome of human choriocapillaris, arterial, and venous endothelial cells. We found that the choriocapillaris highly and specifically expresses the regulator of cell cycle gene (RGCC), a gene that responds to complement activation and induces apoptosis in endothelial cells. In addition, RGCC was the most up-regulated choriocapillaris gene in a donor diagnosed with AMD. These results provide a characterization of the human RPE and choriocapillaris transcriptome, offering potential insight into the mechanisms of choriocapillaris response to complement injury and choroidal vascular disease in age-related macular degeneration.
Collapse
|
34
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
35
|
Igawa S, Choi JE, Wang Z, Chang YL, Wu CC, Werbel T, Ishida-Yamamoto A, Nardo AD. Human Keratinocytes Use Sphingosine 1-Phosphate and its Receptors to Communicate Staphylococcus aureus Invasion and Activate Host Defense. J Invest Dermatol 2019; 139:1743-1752.e5. [PMID: 30807768 PMCID: PMC7682680 DOI: 10.1016/j.jid.2019.02.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/16/2019] [Accepted: 02/01/2019] [Indexed: 02/06/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator generated when a cell membrane or its components are damaged by various factors. S1P regulates diverse cell activities via S1P receptors (S1PRs). Keratinocytes express S1PR1-5. Although it is known that S1PRs control keratinocyte differentiation, apoptosis, and wound healing, S1PR functions in keratinocyte infections have not been fully elucidated. We propose that the S1P-S1PR axis in keratinocytes works as a biosensor for bacterial invasion. Indeed, in human impetigo infection, we found high epidermal expression of S1PR1 and S1PR2 in the skin. Furthermore, in normal human epidermal keratinocytes in vitro, treatment with Staphylococcus aureus bacterial supernatant not only induced S1P production but also increased the transcription of S1PR2, confirming our in vivo observation, as well as increased the levels of TNFA, IL36G, IL6, and IL8 mRNAs. However, direct treatment of normal human epidermal keratinocytes with S1P increased the expressions of IL36G, TNFA, and IL8, but not IL6. In both S1P- and S. aureus bacterial supernatant-treated normal human epidermal keratinocytes, S1PR1 knockdown reduced IL36G, TNFA, and IL8 transcription, and the S1PR2 antagonist JTE013 blocked the secretion of these cytokines. Overall, we have proven that during infections, keratinocytes communicate damage by using S1P release and tight control of S1PR1 and 2.
Collapse
Affiliation(s)
- Satomi Igawa
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Jae Eun Choi
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Yu-Ling Chang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Chia Chi Wu
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | - Tyler Werbel
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA
| | | | - Anna Di Nardo
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, USA,Corresponding author: Anna Di Nardo, Department of Dermatology, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0869, La Jolla, CA 92093, Tel: 858-822-6712, Fax: 858-822-6985, , ORCiD: https://orcid.org/0000-0002-5575-9968
| |
Collapse
|
36
|
Simmons S, Erfinanda L, Bartz C, Kuebler WM. Novel mechanisms regulating endothelial barrier function in the pulmonary microcirculation. J Physiol 2018; 597:997-1021. [PMID: 30015354 DOI: 10.1113/jp276245] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
The pulmonary epithelial and vascular endothelial cell layers provide two sequential physical and immunological barriers that together form a semi-permeable interface and prevent alveolar and interstitial oedema formation. In this review, we focus specifically on the continuous endothelium of the pulmonary microvascular bed that warrants strict control of the exchange of gases, fluid, solutes and circulating cells between the plasma and the interstitial space. The present review provides an overview of emerging molecular mechanisms that permit constant transcellular exchange between the vascular and interstitial compartment, and cause, prevent or reverse lung endothelial barrier failure under experimental conditions, yet with a clinical perspective. Based on recent findings and at times seemingly conflicting results we discuss emerging paradigms of permeability regulation by altered ion transport as well as shifts in the homeostasis of sphingolipids, angiopoietins and prostaglandins.
Collapse
Affiliation(s)
- Szandor Simmons
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Lasti Erfinanda
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Bartz
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada.,Departments of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
37
|
Mehaffey JH, Charles EJ, Narahari AK, Schubert S, Laubach VE, Teman NR, Lynch KR, Kron IL, Sharma AK. Increasing circulating sphingosine-1-phosphate attenuates lung injury during ex vivo lung perfusion. J Thorac Cardiovasc Surg 2018; 156:910-917. [PMID: 29609890 PMCID: PMC6056006 DOI: 10.1016/j.jtcvs.2018.02.090] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/30/2018] [Accepted: 02/07/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND Sphingosine-1-phosphate regulates endothelial barrier integrity and promotes cell survival and proliferation. We hypothesized that upregulation of sphingosine-1-phosphate during ex vivo lung perfusion would attenuate acute lung injury and improve graft function. METHODS C57BL/6 mice (n = 4-8/group) were euthanized, followed by 1 hour of warm ischemia and 1 hour of cold preservation in a model of donation after cardiac death. Subsequently, mice underwent 1 hour of ex vivo lung perfusion with 1 of 4 different perfusion solutions: Steen solution (Steen, control arm), Steen with added sphingosine-1-phosphate (Steen + sphingosine-1-phosphate), Steen plus a selective sphingosine kinase 2 inhibitor (Steen + sphingosine kinase inhibitor), or Steen plus both additives (Steen + sphingosine-1-phosphate + sphingosine kinase inhibitor). During ex vivo lung perfusion, lung compliance and pulmonary artery pressure were continuously measured. Pulmonary vascular permeability was assessed with injection of Evans Blue dye. RESULTS The combination of 1 hour of warm ischemia, followed by 1 hour of cold ischemia created significant lung injury compared with lungs that were immediately harvested after circulatory death and put on ex vivo lung perfusion. Addition of sphingosine-1-phosphate or sphingosine kinase inhibitor alone did not significantly improve lung function during ex vivo lung perfusion compared with Steen without additives. However, group Steen + sphingosine-1-phosphate + sphingosine kinase inhibitor resulted in significantly increased compliance (110% ± 13.9% vs 57.7% ± 6.6%, P < .0001) and decreased pulmonary vascular permeability (33.1 ± 11.9 μg/g vs 75.8 ± 11.4 μg/g tissue, P = .04) compared with Steen alone. CONCLUSIONS Targeted drug therapy with a combination of sphingosine-1-phosphate + sphingosine kinase inhibitor during ex vivo lung perfusion improves lung function in a murine donation after cardiac death model. Elevation of circulating sphingosine-1-phosphate via specific pharmacologic modalities during ex vivo lung perfusion may provide endothelial protection in marginal donor lungs leading to successful lung rehabilitation for transplantation.
Collapse
Affiliation(s)
- J Hunter Mehaffey
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Eric J Charles
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Adishesh K Narahari
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Sarah Schubert
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Victor E Laubach
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Nicholas R Teman
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Kevin R Lynch
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Va
| | - Irving L Kron
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va
| | - Ashish K Sharma
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Va.
| |
Collapse
|
38
|
McGarrity S, Anuforo Ó, Halldórsson H, Bergmann A, Halldórsson S, Palsson S, Henriksen HH, Johansson PI, Rolfsson Ó. Metabolic systems analysis of LPS induced endothelial dysfunction applied to sepsis patient stratification. Sci Rep 2018; 8:6811. [PMID: 29717213 PMCID: PMC5931560 DOI: 10.1038/s41598-018-25015-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/13/2018] [Indexed: 12/24/2022] Open
Abstract
Endothelial dysfunction contributes to sepsis outcome. Metabolic phenotypes associated with endothelial dysfunction are not well characterised in part due to difficulties in assessing endothelial metabolism in situ. Here, we describe the construction of iEC2812, a genome scale metabolic reconstruction of endothelial cells and its application to describe metabolic changes that occur following endothelial dysfunction. Metabolic gene expression analysis of three endothelial subtypes using iEC2812 suggested their similar metabolism in culture. To mimic endothelial dysfunction, an in vitro sepsis endothelial cell culture model was established and the metabotypes associated with increased endothelial permeability and glycocalyx loss after inflammatory stimuli were quantitatively defined through metabolomics. These data and transcriptomic data were then used to parametrize iEC2812 and investigate the metabotypes of endothelial dysfunction. Glycan production and increased fatty acid metabolism accompany increased glycocalyx shedding and endothelial permeability after inflammatory stimulation. iEC2812 was then used to analyse sepsis patient plasma metabolome profiles and predict changes to endothelial derived biomarkers. These analyses revealed increased changes in glycan metabolism in sepsis non-survivors corresponding to metabolism of endothelial dysfunction in culture. The results show concordance between endothelial health and sepsis survival in particular between endothelial cell metabolism and the plasma metabolome in patients with sepsis.
Collapse
Affiliation(s)
- Sarah McGarrity
- Center for Systems Biology, University of Iceland, Sturlugata 8, Reykjavik, Iceland
| | - Ósk Anuforo
- Center for Systems Biology, University of Iceland, Sturlugata 8, Reykjavik, Iceland
| | - Haraldur Halldórsson
- Medical Department, University of Iceland, Sturlugata 8, Reykjavik, Iceland
- Landspitali, Læknagarður, Hringbraut, Reykjavik, Iceland
| | - Andreas Bergmann
- Center for Systems Biology, University of Iceland, Sturlugata 8, Reykjavik, Iceland
| | | | - Sirus Palsson
- Center for Systems Biology, University of Iceland, Sturlugata 8, Reykjavik, Iceland
| | | | - Pär Ingemar Johansson
- Center for Systems Biology, University of Iceland, Sturlugata 8, Reykjavik, Iceland
- Rigshospitalet, Blegdamsvej 9, 2100, Kobenhavn O, Denmark
| | - Óttar Rolfsson
- Center for Systems Biology, University of Iceland, Sturlugata 8, Reykjavik, Iceland.
- Medical Department, University of Iceland, Sturlugata 8, Reykjavik, Iceland.
| |
Collapse
|
39
|
Radeva MY, Waschke J. Mind the gap: mechanisms regulating the endothelial barrier. Acta Physiol (Oxf) 2018; 222. [PMID: 28231640 DOI: 10.1111/apha.12860] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/21/2016] [Accepted: 02/16/2017] [Indexed: 12/11/2022]
Abstract
The endothelial barrier consists of intercellular contacts localized in the cleft between endothelial cells, which is covered by the glycocalyx in a sievelike manner. Both types of barrier-forming junctions, i.e. the adherens junction (AJ) serving mechanical anchorage and mechanotransduction and the tight junction (TJ) sealing the intercellular space to limit paracellular permeability, are tethered to the actin cytoskeleton. Under resting conditions, the endothelium thereby builds a selective layer controlling the exchange of fluid and solutes with the surrounding tissue. However, in the situation of an inflammatory response such as in anaphylaxis or sepsis intercellular contacts disintegrate in post-capillary venules leading to intercellular gap formation. The resulting oedema can cause shock and multi-organ failure. Therefore, maintenance as well as coordinated opening and closure of interendothelial junctions is tightly regulated. The two principle underlying mechanisms comprise spatiotemporal activity control of the small GTPases Rac1 and RhoA and the balance of the phosphorylation state of AJ proteins. In the resting state, junctional Rac1 and RhoA activity is enhanced by junctional components, actin-binding proteins, cAMP signalling and extracellular cues such as sphingosine-1-phosphate (S1P) and angiopoietin-1 (Ang-1). In addition, phosphorylation of AJ components is prevented by junction-associated phosphatases including vascular endothelial protein tyrosine phosphatase (VE-PTP). In contrast, inflammatory mediators inhibiting cAMP/Rac1 signalling cause strong activation of RhoA and induce AJ phosphorylation finally leading to endocytosis and cleavage of VE-cadherin. This results in dissolution of TJs the outcome of which is endothelial barrier breakdown.
Collapse
Affiliation(s)
- M. Y. Radeva
- Institute of Anatomy and Cell Biology; Ludwig-Maximilians-Universität München; Munich Germany
| | - J. Waschke
- Institute of Anatomy and Cell Biology; Ludwig-Maximilians-Universität München; Munich Germany
| |
Collapse
|
40
|
Pulkoski-Gross MJ, Uys JD, Orr-Gandy KA, Coant N, Bialkowska AB, Szulc ZM, Bai A, Bielawska A, Townsend DM, Hannun YA, Obeid LM, Snider AJ. Novel sphingosine kinase-1 inhibitor, LCL351, reduces immune responses in murine DSS-induced colitis. Prostaglandins Other Lipid Mediat 2017; 130:47-56. [PMID: 28377281 PMCID: PMC5509055 DOI: 10.1016/j.prostaglandins.2017.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/25/2017] [Accepted: 03/28/2017] [Indexed: 02/07/2023]
Abstract
Sphingosine-1-phosphate (S1P) is a biologically active sphingolipid metabolite which has been implicated in many diseases including cancer and inflammatory diseases. Recently, sphingosine kinase 1 (SK1), one of the isozymes which generates S1P, has been implicated in the development and progression of inflammatory bowel disease (IBD). Based on our previous work, we set out to determine the efficacy of a novel SK1 selective inhibitor, LCL351, in a murine model of IBD. LCL351 selectively inhibits SK1 both in vitro and in cells. LCL351, which accumulates in relevant tissues such as colon, did not have any adverse side effects in vivo. In mice challenged with dextran sodium sulfate (DSS), a murine model for IBD, LCL351 treatment protected from blood loss and splenomegaly. Additionally, LCL351 treatment reduced the expression of pro-inflammatory markers, and reduced neutrophil infiltration in colon tissue. Our results suggest inflammation associated with IBD can be targeted pharmacologically through the inhibition and degradation of SK1. Furthermore, our data also identifies desirable properties of SK1 inhibitors.
Collapse
Affiliation(s)
- Michael J Pulkoski-Gross
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and the, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Joachim D Uys
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - K Alexa Orr-Gandy
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Nicolas Coant
- Department of Medicine and the, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Agnieszka B Bialkowska
- Department of Medicine and the, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Zdzislaw M Szulc
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Aiping Bai
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Alicja Bielawska
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Danyelle M Townsend
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Yusuf A Hannun
- Department of Medicine and the, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Lina M Obeid
- Department of Medicine and the, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Northport Veterans Affairs Medical Center, Northport, NY, USA
| | - Ashley J Snider
- Department of Medicine and the, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Northport Veterans Affairs Medical Center, Northport, NY, USA.
| |
Collapse
|
41
|
Dyckman AJ. Modulators of Sphingosine-1-phosphate Pathway Biology: Recent Advances of Sphingosine-1-phosphate Receptor 1 (S1P 1) Agonists and Future Perspectives. J Med Chem 2017; 60:5267-5289. [PMID: 28291340 DOI: 10.1021/acs.jmedchem.6b01575] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The sphingoid base derived class of lipids (sphingolipids) is a family of interconverting molecules that play key roles in numerous structural and signaling processes. The biosynthetic pathway of the sphingolipids affords many opportunities for therapeutic intervention: targeting the ligands directly, targeting the various proteins involved in the interconversion of the ligands, or targeting the receptors that respond to the ligands. The focus of this article is on the most advanced of the sphingosine-related therapeutics, agonists of sphingosine-1-phosphate receptor 1 (S1P1). The diverse structural classes of S1P1 agonists will be discussed and the status of compounds of clinical relevance will be detailed. An examination of how potential safety concerns are being navigated with compounds currently under clinical evaluation is followed by a discussion of the novel methods being explored to identify next-generation S1P1 agonists with improved safety profiles. Finally, therapeutic opportunities for sphingosine-related targets outside of S1P1 are touched upon.
Collapse
Affiliation(s)
- Alaric J Dyckman
- Research and Development, Bristol-Myers Squibb Company , P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
42
|
van der Weyden L, Arends MJ, Campbell AD, Bald T, Wardle-Jones H, Griggs N, Velasco-Herrera MDC, Tüting T, Sansom OJ, Karp NA, Clare S, Gleeson D, Ryder E, Galli A, Tuck E, Cambridge EL, Voet T, Macaulay IC, Wong K, Spiegel S, Speak AO, Adams DJ. Genome-wide in vivo screen identifies novel host regulators of metastatic colonization. Nature 2017; 541:233-236. [PMID: 28052056 PMCID: PMC5603286 DOI: 10.1038/nature20792] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/15/2016] [Indexed: 12/17/2022]
Abstract
Metastasis is the leading cause of death for cancer patients. This multi-stage process requires tumour cells to survive in the circulation, extravasate at distant sites, then proliferate; it involves contributions from both the tumour cell and tumour microenvironment ('host', which includes stromal cells and the immune system). Studies suggest the early steps of the metastatic process are relatively efficient, with the post-extravasation regulation of tumour growth ('colonization') being critical in determining metastatic outcome. Here we show the results of screening 810 mutant mouse lines using an in vivo assay to identify microenvironmental regulators of metastatic colonization. We identify 23 genes that, when disrupted in mouse, modify the ability of tumour cells to establish metastatic foci, with 19 of these genes not previously demonstrated to play a role in host control of metastasis. The largest reduction in pulmonary metastasis was observed in sphingosine-1-phosphate (S1P) transporter spinster homologue 2 (Spns2)-deficient mice. We demonstrate a novel outcome of S1P-mediated regulation of lymphocyte trafficking, whereby deletion of Spns2, either globally or in a lymphatic endothelial-specific manner, creates a circulating lymphopenia and a higher percentage of effector T cells and natural killer (NK) cells present in the lung. This allows for potent tumour cell killing, and an overall decreased metastatic burden.
Collapse
Affiliation(s)
| | - Mark J Arends
- University of Edinburgh Division of Pathology, Edinburgh Cancer Research UK Cancer Centre, Institute of Genetics &Molecular Medicine, Edinburgh EH4 2XR, UK
| | | | - Tobias Bald
- Department of Dermatology, University Hospital Magdeburg, Magdeburg 39120, Germany
- Department of Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston 4006, Australia
| | - Hannah Wardle-Jones
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Nicola Griggs
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | | | - Thomas Tüting
- Department of Dermatology, University Hospital Magdeburg, Magdeburg 39120, Germany
| | - Owen J Sansom
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Natasha A Karp
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Diane Gleeson
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Edward Ryder
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Antonella Galli
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Elizabeth Tuck
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Emma L Cambridge
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Thierry Voet
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
- Department of Human Genetics, University of Leuven (KU Leuven), Leuven, 3000, Belgium
| | - Iain C Macaulay
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Kim Wong
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298-0614, USA
| | - Anneliese O Speak
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| | - David J Adams
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Cambridge CB10 1SA, UK
| |
Collapse
|
43
|
Bohannon JK, Sherwood ER. Editorial: Feeling the burn: sphingolipids and infection risk after thermal injury. J Leukoc Biol 2016; 100:1227-1228. [PMID: 27909176 DOI: 10.1189/jlb.4ce0716-299r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 07/27/2016] [Accepted: 08/01/2016] [Indexed: 11/24/2022] Open
Affiliation(s)
- Julia K Bohannon
- Departments of Anesthesiology and Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA; and
| | - Edward R Sherwood
- Departments of Anesthesiology and Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee, USA; and .,Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
44
|
Wong YL, Lautenschläger I, Zitta K, Schildhauer C, Parczany K, Röcken C, Steinfath M, Weiler N, Albrecht M. Adverse effects of hydroxyethyl starch (HES 130/0.4) on intestinal barrier integrity and metabolic function are abrogated by supplementation with Albumin. J Transl Med 2016; 14:60. [PMID: 26920368 PMCID: PMC4769564 DOI: 10.1186/s12967-016-0810-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 02/07/2016] [Indexed: 12/29/2022] Open
Abstract
Background Volume resuscitation with hydroxyethyl starch (HES) is controversially discussed and we recently showed that HES perfusion impairs endothelial and epithelial intestinal barrier integrity. Here we investigated whether Albumin containing HES solutions are superior to HES alone in maintaining intestinal barrier function. Methods An isolated perfused model of the mouse small intestine was used to investigate the effects of: (i) 3 % Albumin (Alb), (ii) 3 % HES or (iii) 1.5 % HES/1.5 % Albumin (HES/Alb). Intestinal morphology, cell damage, metabolic functions, fluid shifts and endothelial/epithelial barrier permeability were evaluated. Potentially involved signaling mechanisms (Erk1/2, Akt and Stat5 phosphorylation) were screened. Results HES induced histomorphological damage (p < 0.01 vs. Alb), by trend elevated the amount of luminal intestinal fatty acid binding protein and reduced galactose uptake (p < 0.001 vs. Alb). Luminal and lymphatic flow rates were increased (p < 0.001 vs. Alb), while vascular flow was decreased (p < 0.001 vs. Alb) during HES perfusion. HES also increased the vascular to luminal FITC-dextran transfer (p < 0.001 vs. Alb), pointing towards a fluid shift from the vascular to the luminal and lymphatic compartments during HES perfusion. Addition of Alb (HES/Alb) reversed all adverse effects of HES (p < 0.05 vs. HES), restored barrier integrity (p < 0.05 vs. HES) and improved metabolic function of the intestine (p < 0.001 vs. HES; p < 0.05 vs. Alb). Mechanistically, HES/Alb perfusion resulted in an increased phosphorylation of Erk1/2 and Akt kinases (p < 0.001 vs. HES), while Stat5 remained unchanged. Conclusions Albumin supplementation abrogates the adverse effects of HES in the intestine and underlying mechanism may function via phosphorylation of Erk1/2 and Akt. Albumin containing HES solutions are superior to HES alone and may improve the suitability of HES in the clinic. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0810-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuk Lung Wong
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Schwanenweg 21, 24105, Kiel, Germany.
| | - Ingmar Lautenschläger
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Schwanenweg 21, 24105, Kiel, Germany.
| | - Karina Zitta
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Schwanenweg 21, 24105, Kiel, Germany.
| | - Christin Schildhauer
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Schwanenweg 21, 24105, Kiel, Germany.
| | - Kerstin Parczany
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Schwanenweg 21, 24105, Kiel, Germany.
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany.
| | - Markus Steinfath
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Schwanenweg 21, 24105, Kiel, Germany.
| | - Norbert Weiler
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Schwanenweg 21, 24105, Kiel, Germany.
| | - Martin Albrecht
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Schleswig-Holstein, Schwanenweg 21, 24105, Kiel, Germany.
| |
Collapse
|
45
|
Jernigan PL, Makley AT, Hoehn RS, Edwards MJ, Pritts TA. The role of sphingolipids in endothelial barrier function. Biol Chem 2016; 396:681-91. [PMID: 25867999 DOI: 10.1515/hsz-2014-0305] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/08/2015] [Indexed: 12/31/2022]
Abstract
Sphingolipids are a ubiquitous family of essential lipids with an increasingly understood role as biologically active mediators in numerous physiologic and pathologic processes. Two particular sphingolipid species, sphingosine-1-phosphate and ceramide, and their metabolites interact both directly and indirectly with endothelial cells to regulate vascular permeability. Sphingosine-1-phosphate generally augments endothelial integrity while ceramide tends to promote vascular leak, and a tight balance between the two is necessary to maintain normal physiologic function. The mechanisms by which sphingolipids regulate endothelial barrier function are complex and occur through multiple different pathways, and disruptions or imbalances in these pathways have been implicated in a number of specific disease processes. With improved understanding of sphingolipid biology, endothelial function, and the interactions between the two, several targets for therapeutic intervention have emerged and there is immense potential for further advancement in this field.
Collapse
|
46
|
Reiss CS. Innate Immunity in Viral Encephalitis. NEUROTROPIC VIRAL INFECTIONS 2016. [PMCID: PMC7153449 DOI: 10.1007/978-3-319-33189-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Carol Shoshkes Reiss
- Departments of Biology and Neural Science, New York University, New York, New York USA
| |
Collapse
|
47
|
Su JB. Vascular endothelial dysfunction and pharmacological treatment. World J Cardiol 2015; 7:719-741. [PMID: 26635921 PMCID: PMC4660468 DOI: 10.4330/wjc.v7.i11.719] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/23/2015] [Accepted: 09/18/2015] [Indexed: 02/06/2023] Open
Abstract
The endothelium exerts multiple actions involving regulation of vascular permeability and tone, coagulation and fibrinolysis, inflammatory and immunological reactions and cell growth. Alterations of one or more such actions may cause vascular endothelial dysfunction. Different risk factors such as hypercholesterolemia, homocystinemia, hyperglycemia, hypertension, smoking, inflammation, and aging contribute to the development of endothelial dysfunction. Mechanisms underlying endothelial dysfunction are multiple, including impaired endothelium-derived vasodilators, enhanced endothelium-derived vasoconstrictors, over production of reactive oxygen species and reactive nitrogen species, activation of inflammatory and immune reactions, and imbalance of coagulation and fibrinolysis. Endothelial dysfunction occurs in many cardiovascular diseases, which involves different mechanisms, depending on specific risk factors affecting the disease. Among these mechanisms, a reduction in nitric oxide (NO) bioavailability plays a central role in the development of endothelial dysfunction because NO exerts diverse physiological actions, including vasodilation, anti-inflammation, antiplatelet, antiproliferation and antimigration. Experimental and clinical studies have demonstrated that a variety of currently used or investigational drugs, such as angiotensin-converting enzyme inhibitors, angiotensin AT1 receptors blockers, angiotensin-(1-7), antioxidants, beta-blockers, calcium channel blockers, endothelial NO synthase enhancers, phosphodiesterase 5 inhibitors, sphingosine-1-phosphate and statins, exert endothelial protective effects. Due to the difference in mechanisms of action, these drugs need to be used according to specific mechanisms underlying endothelial dysfunction of the disease.
Collapse
|
48
|
Levkau B. HDL-S1P: cardiovascular functions, disease-associated alterations, and therapeutic applications. Front Pharmacol 2015; 6:243. [PMID: 26539121 PMCID: PMC4611146 DOI: 10.3389/fphar.2015.00243] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 10/08/2015] [Indexed: 12/17/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid contained in High-density lipoproteins (HDL) and has drawn considerable attention in the lipoprotein field as numerous studies have demonstrated its contribution to several functions inherent to HDL. Some of them are partly and some entirely due to the S1P contained in HDL (HDL-S1P). Despite the presence of over 1000 different lipids in HDL, S1P stands out as it possesses its own cell surface receptors through which it exercises key physiological functions. Most of the S1P in human plasma is associated with HDL, and the amount of HDL-S1P influences the quality and quantity of HDL-dependent functions. The main binding partner of S1P in HDL is apolipoprotein M but others may also exist particularly under conditions of acute S1P elevations. HDL not only exercise functions through their S1P content but have also an impact on genuine S1P signaling by influencing S1P bioactivity and receptor presentation. HDL-S1P content is altered in human diseases such as atherosclerosis, coronary artery disease, myocardial infarction, renal insufficiency and diabetes mellitus. Low HDL-S1P has also been linked to impaired HDL functions associated with these disorders. Although the pathophysiological and molecular reasons for such disease-associated shifts in HDL-S1P are little understood, there have been successful approaches to circumvent their adverse implications by pharmacologically increasing HDL-S1P as means to improve HDL function. This mini-review will cover the current understanding of the contribution of HDL-S1P to physiological HDL function, its alteration in disease and ways for its restoration to correct HDL dysfunction.
Collapse
Affiliation(s)
- Bodo Levkau
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen , Essen, Germany
| |
Collapse
|
49
|
Pigment Epithelium-Derived Factor Induces Endothelial Barrier Dysfunction via p38/MAPK Phosphorylation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:791825. [PMID: 26504830 PMCID: PMC4609513 DOI: 10.1155/2015/791825] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/12/2015] [Indexed: 01/18/2023]
Abstract
Endothelial barrier dysfunction, which is a serious problem that occurs in various inflammatory conditions, permits extravasation of serum components into the surrounding tissues, leading to edema formation and organ failure. Pigment epithelium-derived factor (PEDF), which is a major endogenous antagonist, has been implicated in diverse biological process, but its role in endothelial barrier dysfunction has not been defined. To assess the role of PEDF in the vasculature, we evaluated the effects of exogenous PEDF using human umbilical vein endothelial cells (HUVECs) in vitro. Our results demonstrated that exogenous PEDF activated p38/MAPK signalling pathway in a dose- and time-dependent manner and induced vascular hyperpermeability as measured by the markedly increased FITC-dextran leakage and the decreased transendothelial electrical resistance (TER) across the monolayer cells, which was accompanied by microtubules (MTs) disassembly and F-actin rearrangement. However, the aforementioned alterations can be arrested by the application of low concentration of p38/MAPK inhibitor SB203580. These results reveal a novel role for PEDF as a potential vasoactive substance in inducing hyperpermeability. Furthermore, our results suggest that PEDF and p38/MAPK may serve as therapeutic targets for maintaining vascular integrity.
Collapse
|
50
|
Książek M, Chacińska M, Chabowski A, Baranowski M. Sources, metabolism, and regulation of circulating sphingosine-1-phosphate. J Lipid Res 2015; 56:1271-81. [PMID: 26014962 PMCID: PMC4479332 DOI: 10.1194/jlr.r059543] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/12/2015] [Indexed: 12/16/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that acts either as an intracellular messenger or as a ligand for its membrane receptors. S1P is a normal constituent of blood, where it is found both in plasma and blood cells. Compared with other cell types, sphingolipid metabolism in erythrocytes and platelets has unique features that allow the erythrocytes and platelets to accumulate S1P. In plasma, S1P is bound mainly to HDLs and albumin. Of note, metabolism and biological activity of S1P is to a large extent affected by the type of its carrier. Plasma S1P is characterized by a short half-life, indicating rapid clearance by degradative enzymes and the presence of high-capacity sources involved in maintaining its high concentration. These sources include blood cells, vascular endothelium, and hepatocytes. However, the extent to which each of these contributes to the plasma pool of S1P is a matter of debate. Circulating S1P plays a significant physiological role. It was found to be the key regulator of lymphocyte trafficking, endothelial barrier function, and vascular tone. The purpose of this review is to summarize the present state of knowledge on the metabolism, transport, and origin of plasma S1P, and to discuss the mechanisms regulating its homeostasis in blood.
Collapse
Affiliation(s)
- Monika Książek
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | - Marta Chacińska
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | - Adrian Chabowski
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| | - Marcin Baranowski
- Department of Physiology, Medical University of Białystok, Białystok, Poland
| |
Collapse
|