1
|
Yap JQ, Nikouee A, Lau JE, Walsh G, Zang QS. Mitochondria at the Heart of Sepsis: Mechanisms, Metabolism, and Sex Differences. Int J Mol Sci 2025; 26:4211. [PMID: 40362448 PMCID: PMC12071423 DOI: 10.3390/ijms26094211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Sepsis is a life-threatening condition that occurs when the body is unable to effectively combat infection, leading to systemic inflammation and multi-organ failure. Interestingly, females exhibit lower sepsis incidence and improved clinical outcomes compared to males. However, the mechanisms underlying these sex-specific differences remain poorly understood. While sex hormones have been a primary focus, emerging evidence suggests that non-hormonal factors also play contributory roles. Despite sex differences in sepsis, clinical management is the same for both males and females, with treatment focused on combating infection using antibiotics and hemodynamic support through fluid therapy. However, even with these interventions, mortality remains high, highlighting the need for more effective and targeted therapeutic strategies. Sepsis-induced cardiomyopathy (SIC) is a key contributor to multi-organ failure and is characterized by left ventricular dilation and impaired cardiac contractility. In this review, we explore sex-specific differences in sepsis and SIC, with a particular focus on mitochondrial metabolism. Mitochondria generate the ATP required for cardiac function through fatty acid and glucose oxidation, and recent studies have revealed distinct metabolic profiles between males and females, which can further differ in the context of sepsis and SIC. Targeting these metabolic pathways could provide new avenues for sepsis treatment.
Collapse
Affiliation(s)
- John Q. Yap
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Azadeh Nikouee
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Jessie E. Lau
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Gabriella Walsh
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| | - Qun Sophia Zang
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA; (J.Q.Y.); (A.N.); (J.E.L.); (G.W.)
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
- Cardiovascular Research Institute, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL 60153, USA
| |
Collapse
|
2
|
Specht SJ, Rohringer S, Hager P, Grasl C, Schmitt AM, Pach VJC, Ehrmann K, Baudis S, Liska R, Kiss H, Schneider KH, Podesser BK, Bergmeister H. Decellularized Extracellular Matrix and Polyurethane Vascular Grafts Have Positive Effects on the Inflammatory and Pro-Thrombotic State of Aged Endothelial Cells. J Biomed Mater Res A 2025; 113:e37830. [PMID: 39610352 DOI: 10.1002/jbm.a.37830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 11/30/2024]
Abstract
In vitro assessment of small-diameter synthetic vascular grafts usually uses standard cell culture conditions with early-passage cells. However, these conduits are mainly implanted in elderly patients and are subject to complex cellular interactions influenced by age and inflammation. Understanding these factors is central to the development of vascular grafts tailored to the specific needs of patients. In this study, the effects of aged endothelial cells subjected to pro- and anti-inflammatory agents and cultivated on a newly developed biodegradable electrospun thermoplastic polyurethane/poly(urethane-urea) blend (TPU/TPUU), on clinically available expanded polytetrafluorethylene (ePTFE), and on decellularized extracellular matrix (dECM) grafts were investigated. Young and aged endothelial cells were exposed to pro- and anti-inflammatory agents and characterized by morphology, migration capacity, and gene expression. In addition, the cells were seeded onto the various graft materials and examined microscopically alongside gene expression analyses. When exposed to pro-inflammatory cytokines, young and aged cells demonstrated signs of endothelial activation. Cells seeded on ePTFE showed reduced attachment and increased expression of pro-inflammatory genes compared with the other materials. dECM and TPU/TPUU substrates provided better support for endothelialization with aged cells under inflammatory conditions compared with ePTFE. Moreover, TPU/TPUU showed positive effects on reducing pro-thrombotic and pro-inflammatory gene expression in endothelial cells. Our results thus emphasize the importance of developing new synthetic graft materials as an alternative for clinically used ePTFE.
Collapse
Affiliation(s)
- Sophie J Specht
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Sabrina Rohringer
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Pia Hager
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christian Grasl
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Anna-Maria Schmitt
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Virginia J C Pach
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Katharina Ehrmann
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Applied Synthetic Chemistry, Technical University of Vienna, Vienna, Austria
| | - Stefan Baudis
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Applied Synthetic Chemistry, Technical University of Vienna, Vienna, Austria
| | - Robert Liska
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Applied Synthetic Chemistry, Technical University of Vienna, Vienna, Austria
| | - Herbert Kiss
- Department of Obstetrics and Gynecology, Division of Obstetrics and Feto-Maternal Medicine, Medical University of Vienna, Vienna, Austria
| | - Karl H Schneider
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Bruno K Podesser
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Helga Bergmeister
- Center for Biomedical Research and Translational Surgery, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
3
|
Williams B, Zou L, Pittet JF, Chao W. Sepsis-Induced Coagulopathy: A Comprehensive Narrative Review of Pathophysiology, Clinical Presentation, Diagnosis, and Management Strategies. Anesth Analg 2024; 138:696-711. [PMID: 38324297 PMCID: PMC10916756 DOI: 10.1213/ane.0000000000006888] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 02/08/2024]
Abstract
Physiological hemostasis is a balance between pro- and anticoagulant pathways, and in sepsis, this equilibrium is disturbed, resulting in systemic thrombin generation, impaired anticoagulant activity, and suppression of fibrinolysis, a condition termed sepsis-induced coagulopathy (SIC). SIC is a common complication, being present in 24% of patients with sepsis and 66% of patients with septic shock, and is often associated with poor clinical outcomes and high mortality. 1 , 2 Recent preclinical and clinical studies have generated new insights into the molecular pathogenesis of SIC. In this article, we analyze the complex pathophysiology of SIC with a focus on the role of procoagulant innate immune signaling in hemostatic activation--tissue factor production, thrombin generation, endotheliopathy, and impaired antithrombotic functions. We also review clinical presentations of SIC, the diagnostic scoring system and laboratory tests, the current standard of care, and clinical trials evaluating the efficacies of anticoagulant therapies.
Collapse
Affiliation(s)
- Brittney Williams
- From the Division of Cardiothoracic Anesthesia, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, Maryland
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| | - Jean-Francois Pittet
- Division of Critical Care, Department of Anesthesiology and Perioperative Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology & Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
4
|
Kang JY, Lee M, Song JH, Choi EJ, Kim DU, Lim SK, Kim N, Chang JY. Lactic Acid Bacteria Strains Used as Starters for Kimchi Fermentation Protect the Disruption of Tight Junctions in the Caco-2 Cell Monolayer Model. J Microbiol Biotechnol 2022; 32:1583-1588. [PMID: 36453076 PMCID: PMC9843746 DOI: 10.4014/jmb.2209.09026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 12/05/2022]
Abstract
In this study, we investigated the effect of lactic acid bacteria (LAB) strains used as starters for kimchi fermentation, namely Lactococcus lactis WiKim0124, Companilactobacillus allii WiKim39, Leuconostoc mesenteroides WiKim0121 Leuconostoc mesenteroides WiKim33, and Leuconostoc mesenteroides WiKim32, on the intestinal epithelial tight junctions (TJs). These LAB strains were not cytotoxic to Caco-2 cells at 500 μg/ml concentration. In addition, hydrogen peroxide (H2O2) decreased Caco-2 viability, but the LAB strains protected the cells against H2O2-induced cytotoxicity. We also found that lipopolysaccharide (LPS) promoted Caco-2 proliferation; however, no specific changes were observed upon treatment with LAB strains and LPS. Our evaluation of the permeability in the Caco-2 monolayer model confirmed its increase by both LPS and H2O2. The LAB strains inhibited the increase in permeability by protecting TJs, which we evaluated by measuring TJ proteins such as zonula occludens-1 and occludin, and analyzing them by western blotting and immunofluorescence staining. Our findings show that LAB strains used for kimchi fermentation can suppress the increase in intestinal permeability due to LPS and H2O2 by protecting TJs. Therefore, these results suggest the possibility of enhancing the functionality of kimchi through its fermentation using functional LAB strains.
Collapse
Affiliation(s)
- Jin Yong Kang
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Moeun Lee
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Jung Hee Song
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Eun Ji Choi
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Da un Kim
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Seul Ki Lim
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Namhee Kim
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea
| | - Ji Yoon Chang
- Research and Development Division, World Institute of Kimchi, Gwangju 61755, Republic of Korea,Corresponding author Phone: +82-62-610-1765 Fax: +82-62-610-1853 E-mail:
| |
Collapse
|
5
|
Dull RO, Hahn RG. The glycocalyx as a permeability barrier: basic science and clinical evidence. Crit Care 2022; 26:273. [PMID: 36096866 PMCID: PMC9469578 DOI: 10.1186/s13054-022-04154-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/15/2022] [Indexed: 11/11/2022] Open
Abstract
Preclinical studies in animals and human clinical trials question whether the endothelial glycocalyx layer is a clinically important permeability barrier. Glycocalyx breakdown products in plasma mostly originate from 99.6–99.8% of the endothelial surface not involved in transendothelial passage of water and proteins. Fragment concentrations correlate poorly with in vivo imaging of glycocalyx thickness, and calculations of expected glycocalyx resistance are incompatible with measured hydraulic conductivity values. Increases in plasma breakdown products in rats did not correlate with vascular permeability. Clinically, three studies in humans show inverse correlations between glycocalyx degradation products and the capillary leakage of albumin and fluid.
Collapse
|
6
|
The Methyltransferase Smyd1 Mediates LPS-Triggered Up-Regulation of IL-6 in Endothelial Cells. Cells 2021; 10:cells10123515. [PMID: 34944023 PMCID: PMC8700543 DOI: 10.3390/cells10123515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/16/2022] Open
Abstract
The lysine methyltransferase Smyd1 with its characteristic catalytic SET-domain is highly enriched in the embryonic heart and skeletal muscles, participating in cardiomyogenesis, sarcomere assembly and chromatin remodeling. Recently, significant Smyd1 levels were discovered in endothelial cells (ECs) that responded to inflammatory cytokines. Based on these biochemical properties, we hypothesized that Smyd1 is involved in inflammation-triggered signaling in ECs and therefore, investigated its role within the LPS-induced signaling cascade. Human endothelial cells (HUVECs and EA.hy926 cells) responded to LPS stimulation with higher intrinsic Smyd1 expression. By transfection with expression vectors containing gene inserts encoding either intact Smyd1, a catalytically inactive Smyd1-mutant or Smyd1-specific siRNAs, we show that Smyd1 contributes to LPS-triggered expression and secretion of IL-6 in EA.hy926 cells. Further molecular analysis revealed this process to be based on two signaling pathways: Smyd1 increased the activity of NF-κB and promoted the trimethylation of lysine-4 of histone-3 (H3K4me3) within the IL-6 promoter, as shown by ChIP-RT-qPCR combined with IL-6-promoter-driven luciferase reporter gene assays. In summary, our experimental analysis revealed that LPS-binding to ECs leads to the up-regulation of Smyd1 expression to transduce the signal for IL-6 up-regulation via activation of the established NF-κB pathway as well as via epigenetic trimethylation of H3K4.
Collapse
|
7
|
Hoshika S, Sun X, Kuranaga E, Umetsu D. Reduction of endocytic activity accelerates cell elimination during tissue remodeling of the Drosophila epidermal epithelium. Development 2020; 147:dev.179648. [PMID: 32156754 DOI: 10.1242/dev.179648] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022]
Abstract
Epithelial tissues undergo cell turnover both during development and for homeostatic maintenance. Cells that are no longer needed are quickly removed without compromising the barrier function of the tissue. During metamorphosis, insects undergo developmentally programmed tissue remodeling. However, the mechanisms that regulate this rapid tissue remodeling are not precisely understood. Here, we show that the temporal dynamics of endocytosis modulate physiological cell properties to prime larval epidermal cells for cell elimination. Endocytic activity gradually reduces as tissue remodeling progresses. This reduced endocytic activity accelerates cell elimination through the regulation of Myosin II subcellular reorganization, junctional E-cadherin levels, and caspase activation. Whereas the increased Myosin II dynamics accelerates cell elimination, E-cadherin plays a protective role against cell elimination. Reduced E-cadherin is involved in the amplification of caspase activation by forming a positive-feedback loop with caspase. These findings reveal the role of endocytosis in preventing cell elimination and in the cell-property switching initiated by the temporal dynamics of endocytic activity to achieve rapid cell elimination during tissue remodeling.
Collapse
Affiliation(s)
- Shinichiro Hoshika
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Xiaofei Sun
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Erina Kuranaga
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Daiki Umetsu
- Laboratory for Histogenetic Dynamics, Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, 980-8578, Japan
| |
Collapse
|
8
|
Mignemi NA, McClatchey PM, Kilchrist KV, Williams IM, Millis BA, Syring KE, Duvall CL, Wasserman DH, McGuinness OP. Rapid changes in the microvascular circulation of skeletal muscle impair insulin delivery during sepsis. Am J Physiol Endocrinol Metab 2019; 316:E1012-E1023. [PMID: 30860883 PMCID: PMC6620574 DOI: 10.1152/ajpendo.00501.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/21/2019] [Accepted: 03/07/2019] [Indexed: 01/07/2023]
Abstract
Sepsis costs the healthcare system $23 billion annually and has a mortality rate between 10 and 40%. An early indication of sepsis is the onset of hyperglycemia, which is the result of sepsis-induced insulin resistance in skeletal muscle. Previous investigations have focused on events in the myocyte (e.g., insulin signaling and glucose transport and subsequent metabolism) as the causes for this insulin-resistant state. However, the delivery of insulin to the skeletal muscle is also an important determinant of insulin action. Skeletal muscle microvascular blood flow, which delivers the insulin to the muscle, is known to be decreased during sepsis. Here we test whether the reduced capillary blood flow to skeletal muscle belies the sepsis-induced insulin resistance by reducing insulin delivery to the myocyte. We hypothesize that decreased capillary flow and consequent decrease in insulin delivery is an early event that precedes gross cardiovascular alterations seen with sepsis. This hypothesis was examined in mice treated with either lipopolysaccharide (LPS) or polymicrobial sepsis followed by intravital microscopy of the skeletal muscle microcirculation. We calculated insulin delivery to the myocyte using two independent methods and found that LPS and sepsis rapidly reduce insulin delivery to the skeletal muscle by ~50%; this was driven by decreases in capillary flow velocity and the number of perfused capillaries. Furthermore, the changes in skeletal muscle microcirculation occur before changes in both cardiac output and arterial blood pressure. These data suggest that a rapid reduction in skeletal muscle insulin delivery contributes to the induction of insulin resistance during sepsis.
Collapse
Affiliation(s)
- Nicholas A Mignemi
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - P Mason McClatchey
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Kameron V Kilchrist
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - Ian M Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Bryan A Millis
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Biophotonics Center, Vanderbilt University , Nashville, Tennessee
| | - Kristen E Syring
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Mouse Metabolic Phenotyping Center , Nashville, Tennessee
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee
- Vanderbilt Mouse Metabolic Phenotyping Center , Nashville, Tennessee
| |
Collapse
|
9
|
Wang L, Mehta S, Ahmed Y, Wallace S, Pape MC, Gill SE. Differential Mechanisms of Septic Human Pulmonary Microvascular Endothelial Cell Barrier Dysfunction Depending on the Presence of Neutrophils. Front Immunol 2018; 9:1743. [PMID: 30116240 PMCID: PMC6082932 DOI: 10.3389/fimmu.2018.01743] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 07/16/2018] [Indexed: 01/06/2023] Open
Abstract
Sepsis is characterized by injury of pulmonary microvascular endothelial cells (PMVEC) leading to barrier dysfunction. Multiple mechanisms promote septic PMVEC barrier dysfunction, including interaction with circulating leukocytes and PMVEC apoptotic death. Our previous work demonstrated a strong correlation between septic neutrophil (PMN)-dependent PMVEC apoptosis and pulmonary microvascular albumin leak in septic mice in vivo; however, this remains uncertain in human PMVEC. Thus, we hypothesize that human PMVEC apoptosis is required for loss of PMVEC barrier function under septic conditions in vitro. To assess this hypothesis, human PMVECs cultured alone or in coculture with PMN were stimulated with PBS or cytomix (equimolar interferon γ, tumor necrosis factor α, and interleukin 1β) in the absence or presence of a pan-caspase inhibitor, Q-VD, or specific caspase inhibitors. PMVEC barrier function was assessed by transendothelial electrical resistance (TEER), as well as fluoroisothiocyanate-labeled dextran and Evans blue-labeled albumin flux across PMVEC monolayers. PMVEC apoptosis was identified by (1) loss of cell membrane polarity (Annexin V), (2) caspase activation (FLICA), and (3) DNA fragmentation [terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)]. Septic stimulation of human PMVECs cultured alone resulted in loss of barrier function (decreased TEER and increased macromolecular flux) associated with increased apoptosis (increased Annexin V, FLICA, and TUNEL staining). In addition, treatment of septic PMVEC cultured alone with Q-VD decreased PMVEC apoptosis and prevented septic PMVEC barrier dysfunction. In septic PMN-PMVEC cocultures, there was greater trans-PMVEC macromolecular flux (both dextran and albumin) vs. PMVEC cultured alone. PMN presence also augmented septic PMVEC caspase activation (FLICA staining) vs. PMVEC cultured alone but did not affect septic PMVEC apoptosis. Importantly, pan-caspase inhibition (Q-VD treatment) completely attenuated septic PMN-dependent PMVEC barrier dysfunction. Moreover, inhibition of caspase 3, 8, or 9 in PMN-PMVEC cocultures also reduced septic PMVEC barrier dysfunction whereas inhibition of caspase 1 had no effect. Our data demonstrate that human PMVEC barrier dysfunction under septic conditions in vitro (cytomix stimulation) is clearly caspase-dependent, but the mechanism differs depending on the presence of PMN. In isolated PMVEC, apoptosis contributes to septic barrier dysfunction, whereas PMN presence enhances caspase-dependent septic PMVEC barrier dysfunction independently of PMVEC apoptosis.
Collapse
Affiliation(s)
- Lefeng Wang
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada
| | - Sanjay Mehta
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada.,Division of Respirology, Western University, London, ON, Canada
| | - Yousuf Ahmed
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada
| | - Shelby Wallace
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - M Cynthia Pape
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada
| | - Sean E Gill
- Centre for Critical Illness Research, Lawson Health Research Institute, London, ON, Canada.,Department of Medicine, Western University, London, ON, Canada.,Division of Respirology, Western University, London, ON, Canada.,Department of Physiology and Pharmacology, Western University, London, ON, Canada
| |
Collapse
|
10
|
Ying L, Alvira CM, Cornfield DN. Developmental differences in focal adhesion kinase expression modulate pulmonary endothelial barrier function in response to inflammation. Am J Physiol Lung Cell Mol Physiol 2018; 315:L66-L77. [PMID: 29597831 PMCID: PMC6087892 DOI: 10.1152/ajplung.00363.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 03/22/2018] [Accepted: 03/23/2018] [Indexed: 01/11/2023] Open
Abstract
Compromised pulmonary endothelial cell (PEC) barrier function characterizes acute respiratory distress syndrome (ARDS), a cause of substantial morbidity and mortality. Survival from ARDS is greater in children compared with adults. Whether developmental differences intrinsic to PEC barrier function contribute to this survival advantage remains unknown. To test the hypothesis that PEC barrier function is more well-preserved in neonatal lungs compared with adult lungs in response to inflammation, we induced lung injury in neonatal and adult mice with systemic lipopolysaccharide (LPS). We assessed PEC barrier function in vivo and in vitro, evaluated changes in the expression of focal adhesion kinase 1 (FAK1) and phosphorylation in response to LPS, and determined the effect of FAK silencing and overexpression on PEC barrier function. We found that LPS induced a greater increase in lung permeability and PEC barrier disruption in the adult mice, despite similar degrees of inflammation and apoptosis. Although baseline expression was similar, LPS increased FAK1 expression in neonatal PEC but increased FAK1 phosphorylation and decreased FAK1 expression in adult PEC. Pharmacologic inhibition of FAK1 accentuated LPS-induced barrier disruption most in adult PEC. Finally, in response to LPS, FAK silencing markedly impaired neonatal PEC barrier function, whereas FAK overexpression preserved adult PEC barrier function. Thus, developmental differences in FAK expression during inflammatory injury serve to preserve neonatal pulmonary endothelial barrier function compared with that of adults and suggest that intrinsic differences in the immature versus pulmonary endothelium, especially relative to FAK1 phosphorylation, may contribute to the improved outcomes of children with ARDS.
Collapse
Affiliation(s)
- Lihua Ying
- Division of Pulmonary Medicine, Department of Pediatrics, Stanford University School of Medicine , Stanford, California
| | - Cristina M Alvira
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of Medicine , Stanford, California
| | - David N Cornfield
- Division of Pulmonary Medicine, Department of Pediatrics, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
11
|
Microvascular dysfunction in patients with diabetes after cardioplegic arrest and cardiopulmonary bypass. Curr Opin Cardiol 2018; 31:618-624. [PMID: 27652811 DOI: 10.1097/hco.0000000000000340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PURPOSE OF REVIEW The purpose of the current review is to describe the changes of microvascular function in patients with diabetes after cardioplegic arrest and cardiopulmonary bypass (CPB) and cardiac surgery. RECENT FINDINGS Cardiac surgery, especially that involving cardioplegia and CPB, is associated with significant changes in vascular reactivity of coronary/peripheral microcirculation, vascular permeability, gene/protein expression, and programmed cell death, as well as with increased morbidity and mortality after surgical procedures. In particular, these changes are more profound in patients with poorly controlled diabetes. SUMMARY Because alterations in vasomotor regulation are critical aspects of mortality and morbidity of cardioplegia/CPB, a better understanding of diabetic regulation of microvascular function may lead to improved postoperative outcomes of patients with diabetes after cardioplegia/CPB and cardiac surgery.
Collapse
|
12
|
Zoppi N, Chiarelli N, Ritelli M, Colombi M. Multifaced Roles of the αvβ3 Integrin in Ehlers-Danlos and Arterial Tortuosity Syndromes' Dermal Fibroblasts. Int J Mol Sci 2018; 19:ijms19040982. [PMID: 29587413 PMCID: PMC5979373 DOI: 10.3390/ijms19040982] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/19/2018] [Accepted: 03/24/2018] [Indexed: 02/07/2023] Open
Abstract
The αvβ3 integrin, an endothelial cells’ receptor-binding fibronectin (FN) in the extracellular matrix (ECM) of blood vessels, regulates ECM remodeling during migration, invasion, angiogenesis, wound healing and inflammation, and is also involved in the epithelial mesenchymal transition. In vitro-grown human control fibroblasts organize a fibrillar network of FN, which is preferentially bound on the entire cell surface to its canonical α5β1 integrin receptor, whereas the αvβ3 integrin is present only in rare patches in focal contacts. We report on the preferential recruitment of the αvβ3 integrin, due to the lack of FN–ECM and its canonical integrin receptor, in dermal fibroblasts from Ehlers–Danlos syndromes (EDS) and arterial tortuosity syndrome (ATS), which are rare multisystem connective tissue disorders. We review our previous findings that unraveled different biological mechanisms elicited by the αvβ3 integrin in fibroblasts derived from patients affected with classical (cEDS), vascular (vEDS), hypermobile EDS (hEDS), hypermobility spectrum disorders (HSD), and ATS. In cEDS and vEDS, respectively, due to defective type V and type III collagens, αvβ3 rescues patients’ fibroblasts from anoikis through a paxillin-p60Src-mediated cross-talk with the EGF receptor. In hEDS and HSD, without a defined molecular basis, the αvβ3 integrin transduces to the ILK-Snail1-axis inducing a fibroblast-to-myofibroblast-transition. In ATS cells, the deficiency of the dehydroascorbic acid transporter GLUT10 leads to redox imbalance, ECM disarray together with the activation of a non-canonical αvβ3 integrin-TGFBRII signaling, involving p125FAK/p60Src/p38MAPK. The characterization of these different biological functions triggered by αvβ3 provides insights into the multifaced nature of this integrin, at least in cultured dermal fibroblasts, offering future perspectives for research in this field.
Collapse
Affiliation(s)
- Nicoletta Zoppi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Nicola Chiarelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Marco Ritelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Marina Colombi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
13
|
Aloin Suppresses Lipopolysaccharide-Induced Inflammatory Response and Apoptosis by Inhibiting the Activation of NF-κB. Molecules 2018; 23:molecules23030517. [PMID: 29495390 PMCID: PMC6017010 DOI: 10.3390/molecules23030517] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/09/2018] [Accepted: 02/17/2018] [Indexed: 12/30/2022] Open
Abstract
Numerous herbal-derived natural products are excellent anti-inflammatory agents. Several studies have reported that aloin, the major anthraquinone glycoside obtained from the Aloe species, exhibits anti-inflammatory activity. However, the molecular mechanism of this activity is not well understood. In this report, we found that aloin suppresses lipopolysaccharide-induced pro-inflammatory cytokine secretion and nitric oxide production, and downregulates the expression of tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). Aloin inhibits the phosphorylation and acetylation of the NF-κB p65 subunit by suppressing the upstream kinases p38 and Msk1, preventing LPS-induced p65 translocation to the nucleus. We have also shown that aloin inhibits LPS-induced caspase-3 activation and apoptotic cell death. Collectively, these findings suggest that aloin effectively suppresses the inflammatory response, primarily through the inhibition of NF-κB signaling.
Collapse
|
14
|
Hattori Y, Hattori K, Suzuki T, Matsuda N. Recent advances in the pathophysiology and molecular basis of sepsis-associated organ dysfunction: Novel therapeutic implications and challenges. Pharmacol Ther 2017; 177:56-66. [DOI: 10.1016/j.pharmthera.2017.02.040] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
15
|
Yan M, Zhang X, Chen A, Gu W, Liu J, Ren X, Zhang J, Wu X, Place AT, Minshall RD, Liu G. Endothelial cell SHP-2 negatively regulates neutrophil adhesion and promotes transmigration by enhancing ICAM-1-VE-cadherin interaction. FASEB J 2017; 31:4759-4769. [PMID: 28701303 DOI: 10.1096/fj.201700280r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 06/27/2017] [Indexed: 12/16/2022]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) mediates the firm adhesion of leukocytes to endothelial cells and initiates subsequent signaling that promotes their transendothelial migration (TEM). Vascular endothelial (VE)-cadherin plays a critical role in endothelial cell-cell adhesion, thereby controlling endothelial permeability and leukocyte transmigration. This study aimed to determine the molecular signaling events that originate from the ICAM-1-mediated firm adhesion of neutrophils that regulate VE-cadherin's role as a negative regulator of leukocyte transmigration. We observed that ICAM-1 interacts with Src homology domain 2-containing phosphatase-2 (SHP-2), and SHP-2 down-regulation via silencing of small interfering RNA in endothelial cells enhanced neutrophil adhesion to endothelial cells but inhibited neutrophil transmigration. We also found that VE-cadherin associated with the ICAM-1-SHP-2 complex. Moreover, whereas the activation of ICAM-1 leads to VE-cadherin dissociation from ICAM-1 and VE-cadherin association with actin, SHP-2 down-regulation prevented ICAM-1-VE-cadherin association and promoted VE-cadherin-actin association. Furthermore, SHP-2 down-regulation in vivo promoted LPS-induced neutrophil recruitment in mouse lung but delayed neutrophil extravasation. These results suggest that SHP-2-via association with ICAM-1-mediates ICAM-1-induced Src activation and modulates VE-cadherin switching association with ICAM-1 or actin, thereby negatively regulating neutrophil adhesion to endothelial cells and enhancing their TEM.-Yan, M., Zhang, X., Chen, A., Gu, W., Liu, J., Ren, X., Zhang, J., Wu, X., Place, A. T., Minshall, R. D., Liu, G. Endothelial cell SHP-2 negatively regulates neutrophil adhesion and promotes transmigration by enhancing ICAM-1-VE-cadherin interaction.
Collapse
Affiliation(s)
- Meiping Yan
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xinhua Zhang
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ao Chen
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wei Gu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jie Liu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaojiao Ren
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jianping Zhang
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaoxiong Wu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aaron T Place
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard D Minshall
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA.,Department of Anesthesiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Guoquan Liu
- Department of Basic Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China;
| |
Collapse
|
16
|
Inhibition of Murine Pulmonary Microvascular Endothelial Cell Apoptosis Promotes Recovery of Barrier Function under Septic Conditions. Mediators Inflamm 2017; 2017:3415380. [PMID: 28250575 PMCID: PMC5303866 DOI: 10.1155/2017/3415380] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/25/2016] [Accepted: 12/20/2016] [Indexed: 12/13/2022] Open
Abstract
Sepsis is characterized by injury of the pulmonary microvasculature and the pulmonary microvascular endothelial cells (PMVEC), leading to barrier dysfunction and acute respiratory distress syndrome (ARDS). Our recent work identified a strong correlation between PMVEC apoptosis and microvascular leak in septic mice in vivo, but the specific role of apoptosis in septic PMVEC barrier dysfunction remains unclear. Thus, we hypothesize that PMVEC apoptosis is likely required for PMVEC barrier dysfunction under septic conditions in vitro. Septic stimulation (mixture of tumour necrosis factor α, interleukin 1β, and interferon γ [cytomix]) of isolated murine PMVEC resulted in a significant loss of barrier function as early as 4 h after stimulation, which persisted until 24 h. PMVEC apoptosis, as reflected by caspase activation, DNA fragmentation, and loss of membrane polarity, was first apparent at 8 h after cytomix. Pretreatment of PMVEC with the pan-caspase inhibitor Q-VD significantly decreased septic PMVEC apoptosis and was associated with reestablishment of PMVEC barrier function at 16 and 24 h after stimulation but had no effect on septic PMVEC barrier dysfunction over the first 8 h. Collectively, our data suggest that early septic murine PMVEC barrier dysfunction driven by proinflammatory cytokines is not mediated through apoptosis, but PMVEC apoptosis contributes to late septic PMVEC barrier dysfunction.
Collapse
|
17
|
Galkin II, Pletjushkina OY, Zinovkin RA, Zakharova VV, Chernyak BV, Popova EN. Mitochondria-Targeted Antioxidant SkQR1 Reduces TNF-Induced Endothelial Permeability in vitro. BIOCHEMISTRY (MOSCOW) 2017; 81:1188-1197. [PMID: 27908243 DOI: 10.1134/s0006297916100163] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Prolonged or excessive increase in the circulatory level of proinflammatory tumor necrosis factor (TNF) leads to abnormal activation and subsequent damage to endothelium. TNF at high concentrations causes apoptosis of endothelial cells. Previously, using mitochondria-targeted antioxidants of SkQ family, we have shown that apoptosis of endothelial cells is dependent on the production of reactive oxygen species (ROS) in mitochondria (mito-ROS). Now we have found that TNF at low concentrations does not cause cell death but activates caspase-3 and caspase-dependent increase in endothelial permeability in vitro. This effect is probably due to the cleavage of β-catenin - an adherent junction protein localized in the cytoplasm. We have also shown that extracellular matrix metalloprotease 9 (MMP9) VE-cadherin shedding plays a major role in the TNF-induced endothelial permeability. The mechanisms of the caspase-3 and MMP9 activation are probably not related to each other since caspase inhibition did not affect VE-cadherin cleavage and MMP9 inhibition had no effect on the caspase-3 activation. Mitochondria-targeted antioxidant SkQR1 inhibited TNF-induced increase in endothelial permeability. SkQR1 also inhibited caspase-3 activation, β-catenin cleavage, and MMP9-dependent VE-cadherin shedding. The data suggest that mito-ROS are involved in the increase in endothelial permeability due to the activation of both caspase-dependent cleavage of intracellular proteins and of MMP9-dependent cleavage of the transmembrane cell-to-cell contact proteins.
Collapse
Affiliation(s)
- I I Galkin
- Lomonosov Moscow State University, Belozersky Institute of Physico-Chemical Biology, Moscow, 119991, Russia.
| | | | | | | | | | | |
Collapse
|
18
|
Teng X, Qin L, Le Borgne R, Toyama Y. Remodeling of adhesion and modulation of mechanical tensile forces during apoptosis in Drosophila epithelium. Development 2016; 144:95-105. [PMID: 27888195 DOI: 10.1242/dev.139865] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 11/10/2016] [Indexed: 02/01/2023]
Abstract
Apoptosis is a mechanism of eliminating damaged or unnecessary cells during development and tissue homeostasis. During apoptosis within a tissue, the adhesions between dying and neighboring non-dying cells need to be remodeled so that the apoptotic cell is expelled. In parallel, contraction of actomyosin cables formed in apoptotic and neighboring cells drives cell extrusion. To date, the coordination between the dynamics of cell adhesion and the progressive changes in tissue tension around an apoptotic cell is not fully understood. Live imaging of histoblast expansion, which is a coordinated tissue replacement process during Drosophila metamorphosis, shows remodeling of adherens junctions (AJs) between apoptotic and non-dying cells, with a reduction in the levels of AJ components, including E-cadherin. Concurrently, surrounding tissue tension is transiently released. Contraction of a supra-cellular actomyosin cable, which forms in neighboring cells, brings neighboring cells together and further reshapes tissue tension toward the completion of extrusion. We propose a model in which modulation of tissue tension represents a mechanism of apoptotic cell extrusion.
Collapse
Affiliation(s)
- Xiang Teng
- Department of Biological Sciences, National University of Singapore, Singapore 117543.,Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Lei Qin
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Roland Le Borgne
- CNRS, UMR 6290, Institute of Genetics and Development of Rennes, F-35043 Rennes, France.,Université Rennes 1, Faculté de Médecine, F35043 Rennes, France
| | - Yusuke Toyama
- Department of Biological Sciences, National University of Singapore, Singapore 117543 .,Mechanobiology Institute, National University of Singapore, Singapore 117411.,Temasek Life Sciences Laboratory, Singapore 117604
| |
Collapse
|
19
|
Feng J, Liu Y, Singh AK, Ehsan A, Sellke N, Liang J, Sellke FW. Effects of diabetes and cardiopulmonary bypass on expression of adherens junction proteins in human peripheral tissue. Surgery 2016; 161:823-829. [PMID: 27838104 DOI: 10.1016/j.surg.2016.08.057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 08/17/2016] [Accepted: 08/23/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND We investigated the changes in adherens junction proteins, such as vascular endothelial-cadherin and β-catenin, of skeletal muscle and vessels in patients with or without diabetes in the setting of cardiopulmonary bypass and cardiac operation. METHODS Skeletal muscle tissue samples were harvested pre- and post-cardiopulmonary bypass from nondiabetic (hemoglobin A1c: 5.4 ± 0.1), controlled diabetic (hemoglobin A1c: 6.3 ± 0.1), and uncontrolled diabetic patients (hemoglobin A1c: 9.6 ± 0.3) undergoing coronary artery bypass grafting operation (n = 8 per group). The expression/phosphorylation of adherens junction proteins vascular endothelial-cadherin and β-catenin were assessed by immunoblotting and immuno-histochemistry. Endothelial function of skeletal muscle arterioles was determined by videomicroscopy in response to the vasodilator substance P. RESULTS The protein expression of total vascular endothelial-cadherin was not changed at baseline or between pre-and post-cardiopulmonary bypass among groups. The pre-cardiopulmonary bypass level of phospho-vascular endothelial-cadherin was found to be significantly increased in the uncontrolled diabetic patients group compared with the nondiabetic or controlled diabetic groups (P < .05). The post-cardiopulmonary bypass levels of phospho-vascular endothelial-cadherin were significantly increased compared with pre-cardiopulmonary bypass in all groups (P < .05 each), and this increase was greater in the uncontrolled diabetic patients group than that of the nondiabetic or controlled diabetic groups (P < .05). Expression of basal β-catenin protein in the uncontrolled diabetic group was decreased compared with nondiabetic or controlled diabetic groups (P < .05). There were significant decreases in the β-catenin protein expression between pre- and post-cardiopulmonary bypass in all 3 groups (P < .05 each), and this decrease was greater in the uncontrolled diabetic patients group than the nondiabetic group (P < .05). There were decreases in the relaxation response of skeletal muscle arterioles to substance P after cardiopulmonary bypass in all 3 groups (P < .05), and this alteration was more pronounced in the uncontrolled diabetic patients (P < .05). CONCLUSION Uncontrolled diabetes causes inactivation and reduction in the expression of endothelial adherens junction proteins in the arterioles of skeletal muscle early after cardiopulmonary bypass. The enhanced phosphorylation of vascular endothelial-cadherin and degradation of β-catenin indicate deterioration of these proteins and damage of the cell-cell endothelial junctions, specifically in the diabetic peripheral vessels. These alterations may contribute to the increases in peripheral vascular permeability and endothelial dysfunction.
Collapse
Affiliation(s)
- Jun Feng
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Yuhong Liu
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Arun K Singh
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Afshin Ehsan
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Nicholas Sellke
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Justin Liang
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
20
|
Yi L, Huang X, Guo F, Zhou Z, Dou Y, Huan J. Yes-associated protein (YAP) signaling regulates lipopolysaccharide-induced tissue factor expression in human endothelial cells. Surgery 2016; 159:1436-48. [PMID: 26791271 DOI: 10.1016/j.surg.2015.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND Sepsis-induced acute lung injury (ALI) is characterized by fibrin deposition, which indicates the local activation of coagulation. Tissue factor (TF), expressed in the pulmonary microvasculature, acts as a critical initiator of blood coagulation and ALI in sepsis. The molecular mechanism of lipopolysaccharide (LPS)-induced TF expression in endothelial cells (ECs), however, has not been determined. In this study, we implicate the Rho-associated protein kinase (ROCK)/Yes associated protein (YAP)/early growth response (Egr-1) signaling pathway in LPS-induced TF expression in vitro and in sepsis-induced ALI in vivo. METHODS Human umbilical vein ECs incubated with LPS were pretreated with or without the ROCK inhibitor Y-27632, a YAP small, interfering RNA (siRNA) and an Egr-1 siRNA. ROCK, YAP and Egr-1 signaling-induced protein expression was investigated by Western blot. The LPS-induced activation of YAP was analyzed by an immunofluorescent assay. Furthermore, we intratracheally injected YAP siRNA to assess septic ALI in mice by hematoxylin and eosin staining. RESULTS LPS rapidly induced ROCK activation and increased TF expression in ECs. LPS caused YAP shuttling into the nuclei of ECs and combined with Egr-1 via the activation of ROCK. Furthermore, the LPS-mediated TF expression increase was prevented by ROCK inactivation, YAP knockdown and Egr-1 depletion, suggesting that LPS-induced TF expression is closely associated with the ROCK/YAP/Egr-1 signaling pathway in ECs. Finally, an intratracheal injection of YAP siRNA relieved lung injury in septic mice. CONCLUSION This study not only suggests that ROCK/YAP/Egr-1 signaling regulates TF expression after stimulation with LPS in ECs, but it also indicates that LPS-induced activation of YAP signaling plays an important role in septic ALI in mice. Our findings provide a new insight into the pathogenic mechanism of TF expression, which is closely linked to septic ALI, and YAP signaling is considered to be a novel target for therapeutic intervention under septic conditions.
Collapse
Affiliation(s)
- Lei Yi
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoqin Huang
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Guo
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zengding Zhou
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Dou
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingning Huan
- Department of Burn and Plastic Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
21
|
Wang C, Armstrong SM, Sugiyama MG, Tabuchi A, Krauszman A, Kuebler WM, Mullen B, Advani S, Advani A, Lee WL. Influenza-Induced Priming and Leak of Human Lung Microvascular Endothelium upon Exposure to Staphylococcus aureus. Am J Respir Cell Mol Biol 2015; 53:459-70. [PMID: 25693001 DOI: 10.1165/rcmb.2014-0373oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A major cause of death after influenza virus infection is lung injury due to a bacterial superinfection, yet the mechanism is unknown. Death has been attributed to virus-induced immunosuppression and bacterial overgrowth, but this hypothesis is based on data from the preantibiotic era and animal models that omit antimicrobial therapy. Because of diagnostic uncertainty, most patients with influenza receive antibiotics, making bacterial overgrowth unlikely. Respiratory failure after superinfection presents as acute respiratory distress syndrome, a disorder characterized by lung microvascular leak and edema. The objective of this study was to determine whether the influenza virus sensitizes the lung endothelium to leak upon exposure to circulating bacterial-derived molecular patterns from Staphylococcus aureus. In vitro as well as in vivo models of influenza followed by S. aureus superinfection were used. Molecular mechanisms were explored using molecular biology, knockout mice, and human autopsy specimens. Influenza virus infection sensitized human lung endothelium to leak when challenged with S. aureus, even at low doses of influenza and even when the pathogens were given days apart. Influenza virus increased endothelial expression of TNFR1 both in vitro and in intact lungs, a finding corroborated by human autopsy specimens of patients with influenza. Leak was recapitulated with protein A, a TNFR1 ligand, and sequential infection caused protein A-dependent loss of IκB, cleavage of caspases 8 and 3, and lung endothelial apoptosis. Mice infected sequentially with influenza virus and S. aureus developed significantly increased lung edema that was protein A and TNFR1 dependent. Influenza virus primes the lung endothelium to leak, predisposing patients to acute respiratory distress syndrome upon exposure to S. aureus.
Collapse
Affiliation(s)
- Changsen Wang
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
| | - Susan M Armstrong
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada.,2 Institute of Medical Science
| | - Michael G Sugiyama
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada.,3 Department of Laboratory Medicine and Pathobiology
| | - Arata Tabuchi
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
| | - Adrienn Krauszman
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
| | - Wolfgang M Kuebler
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
| | - Brendan Mullen
- 4 Department of Pathology, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Suzanne Advani
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew Advani
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada.,5 Department of Medicine, University of Toronto, Toronto, Ontario, Canada; and
| | - Warren L Lee
- 1 Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada.,2 Institute of Medical Science.,3 Department of Laboratory Medicine and Pathobiology.,6 Interdepartmental Division of Critical Care and.,5 Department of Medicine, University of Toronto, Toronto, Ontario, Canada; and
| |
Collapse
|
22
|
Kefir induces apoptosis and inhibits cell proliferation in human acute erythroleukemia. Med Oncol 2015; 33:7. [DOI: 10.1007/s12032-015-0722-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 12/14/2015] [Indexed: 02/01/2023]
|
23
|
Chen J, Wang J, Su C, Qian W, Sun L, Sun H, Chen J, Zhang H, Zhang J. Urinary trypsin inhibitor attenuates LPS-induced endothelial barrier dysfunction by upregulation of vascular endothelial-cadherin expression. Inflamm Res 2015; 65:213-24. [PMID: 26681130 DOI: 10.1007/s00011-015-0907-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 11/07/2015] [Accepted: 11/16/2015] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Urinary trypsin inhibitor (UTI) decreases inflammatory cytokine levels and mortality in experimental animal models of inflammation. Here, we observed the effect of UTI on lipopolysaccharide (LPS)-induced hyperpermeability in human umbilical vein endothelial cells (HUVECs) and explored the role of vascular endothelial-cadherin (VE-cadherin) in its effect. METHODS The effect of UTI on endothelial barrier hyperpermeability was detected by an electrical cell-substrate impedance sensing (ECIS) system and a transwell chamber system. The expression of VE-cadherin in HUVECs was examined by real-time PCR and western blot. RESULTS We demonstrated that the alleviation of LPS-induced barrier dysfunction could be achieved by pretreatment with 3000 U/mL of UTI. VE-cadherin monoclonal antibody (mAb) could inhibit the protective effects. UTI maintained VE-cadherin expression by increasing protein stability at both the transcriptional and post-transcriptional levels. Meanwhile, VE-cadherin expression on the cell surface increased when the cells were pretreated with UTI. Furthermore, pretreatment with UTI decreased the phosphorylation of VE-cadherin at Tyr658 but not Tyr731. CONCLUSIONS Our data show that prophylactic UTI maintains the endothelial barrier function, increases VE-cadherin expression, and inhibits the phosphorylation of VE-cadherin at Tyr658 under inflammatory conditions. It suggests a scientific and potential clinical therapeutic importance of UTI in treatment of inflammatory disorders.
Collapse
Affiliation(s)
- Jie Chen
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Jun Wang
- The Laboratory of Neurotoxicology, School of Public Health, Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Chenglei Su
- Department of Emergency, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, 221000, People's Republic of China
| | - Wenyi Qian
- The Laboratory of Neurotoxicology, School of Public Health, Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Li Sun
- Department of Basic Medical College, Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Hao Sun
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Junjie Chen
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Huazhong Zhang
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China
| | - Jinsong Zhang
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, People's Republic of China.
| |
Collapse
|
24
|
Feng J, Cohn WE, Parnis SM, Sodha NR, Clements RT, Sellke N, Frazier OH, Sellke FW. New continuous-flow total artificial heart and vascular permeability. J Surg Res 2015; 199:296-305. [PMID: 26188957 PMCID: PMC4636951 DOI: 10.1016/j.jss.2015.06.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 06/03/2015] [Accepted: 06/12/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND We tested the short-term effects of completely nonpulsatile versus pulsatile circulation after ventricular excision and replacement with total implantable pumps in an animal model on peripheral vascular permeability. METHODS Ten calves underwent cardiac replacement with two HeartMate III continuous-flow rotary pumps. In five calves, the pump speed was rapidly modulated to impart a low-frequency pulse pressure in the physiologic range (10-25 mm Hg) at a rate of 40 pulses per minute (PP). The remaining five calves were supported with a pulseless systemic circulation and no modulation of pump speed (NP). Skeletal muscle biopsies were obtained before cardiac replacement (baseline) and on postoperative days (PODs) 1, 7, and 14. Skeletal muscle-tissue water content was measured, and morphologic alterations of skeletal muscle were assessed. VE-cadherin, phospho-VE-cadherin, and CD31 were analyzed by immunohistochemistry. RESULTS There were no significant changes in tissue water content and skeletal muscle morphology within group or between groups at baseline, PODs 1, 7, and 14, respectively. There were no significant alterations in the expression and/or distribution of VE-cadherin, phospho-VE-cadherin, and CD31 in skeletal muscle vasculature at baseline, PODs 1, 7, and 14 within each group or between the two groups, respectively. Although continuous-flow total artificial heart (CFTAH) with or without a pulse pressure caused slight increase in tissue water content and histologic damage scores at PODs 7 and 14, it failed to reach statistical significance. CONCLUSIONS There was no significant adherens-junction protein degradation and phosphorylation in calf skeletal muscle microvasculature after CFTAH implantation, suggesting that short term of CFTAH with or without pulse pressure did not cause peripheral endothelial injury and did not increase the peripheral microvascular permeability.
Collapse
Affiliation(s)
- Jun Feng
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island
| | - William E Cohn
- Cardiovascular Research Laboratories, Department of Surgery, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas
| | - Steven M Parnis
- Cardiovascular Research Laboratories, Department of Surgery, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas
| | - Neel R Sodha
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Richard T Clements
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Nicholas Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island
| | - O Howard Frazier
- Cardiovascular Research Laboratories, Department of Surgery, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island.
| |
Collapse
|
25
|
Abstract
AbstractThe bovine mammary gland is a dynamic and complex organ composed of various cell types that work together for the purpose of milk synthesis and secretion. A layer of endothelial cells establishes the blood–milk barrier, which exists to facilitate the exchange of solutes and macromolecules necessary for optimal milk production. During bacterial challenge, however, endothelial cells divert some of their lactation function to protect the underlying tissue from damage by initiating inflammation. At the onset of inflammation, endothelial cells tightly regulate the movement of plasma components and leukocytes into affected tissue. Unfortunately, endothelial dysfunction as a result of exacerbated or sustained inflammation can negatively affect both barrier integrity and the health of surrounding extravascular tissue. The objective of this review is to highlight the role of endothelial cells in supporting milk production and regulating optimal inflammatory responses. The consequences of endothelial dysfunction and sustained inflammation on milk synthesis and secretion are discussed. Given the important role of endothelial cells in orchestrating the inflammatory response, a better understanding of endothelial function during mastitis may support development of targeted therapies to protect bovine mammary tissue and mammary endothelium.
Collapse
|
26
|
Feng J, Liu Y, Sabe AA, Sadek AA, Singh AK, Sodha NR, Sellke FW. Differential impairment of adherens-junction expression/phosphorylation after cardioplegia in diabetic versus non-diabetic patients. Eur J Cardiothorac Surg 2015; 49:937-43. [PMID: 26069241 DOI: 10.1093/ejcts/ezv202] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 05/04/2015] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES Previous animal studies have demonstrated that endothelial adherens-junction molecules are significantly altered in animal myocardium and microvasculature after cardioplegia and cardiopulmonary bypass (CP/CPB). We investigated the effects of diabetes on expression/phosphorylation/localization of vascular endothelial (VE)-cadherin, β- and γ-catenin in human atrial myocardium and coronary vasculature in the setting of CP/CPB. METHODS Right atrial tissue was harvested pre- and post-CP/CPB from non-diabetic (ND) [haemoglobin A1c (HbA1c): 5.4 ± 0.15], controlled (CDM) (HbA1c: 6.3 ± 0.14) and uncontrolled diabetic (UDM) (HbA1c: 9.9 ± 0.72) patients (n = 10/group). Expression/phosphorylation/localization of VE-cadherin, β- and γ-catenin were assessed by immunoblotting, immunoprecipitation and immunohistochemistry. In vitro atrial microvascular reactivity was assessed by videomicroscopy in response to the endothelium-dependent vasodilator adenosine 5'-diphosphate (ADP). RESULTS There were no significant differences in VE-cadherin protein expression between pre- and post-CP/CPB among groups. There were significant decreases in VE-cadherin densities in vessels of the UDM group versus the ND group at baseline or post-CP/CPB, respectively (P < 0.05). The level of basal phosphorylated VE-cadherin tends to be higher in the UDM compared with the ND group (P < 0.05). CP/CPB induced more phosphorylation of VE-cadherin in all groups (versus pre-CP/CPB; P < 0.05, respectively) and this effect was more pronounced in the UDM group (P < 0.05 versus ND or CDM). The protein levels of both catenins (β and γ) were lower in post-CP/CPB in UDM than ND patients (P < 0.05). There were significant decreases in vasodilatory response to endothelial-dependent vasodilator ADP after CP/CPB (P < 0.05). This alteration was more pronounced in UDM patients (P < 0.05). CONCLUSIONS These data suggest that poorly controlled diabetes down-regulates endothelial adherens-junction protein activation/expression/localization in the setting of CP/CPB. The increased tyrosine phosphorylation and deterioration of VE-cadherin indicate the damage of the cell-cell endothelial junctions in the diabetic vessels undergoing CP/CPB and cardiac surgery. These alterations may lead to increase in vascular permeability and endothelial dysfunction and affect outcomes in diabetic patients after cardiac surgery.
Collapse
Affiliation(s)
- Jun Feng
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Yuhong Liu
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Ashraf A Sabe
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Ahmed A Sadek
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Arun K Singh
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Neel R Sodha
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
27
|
Kása A, Csortos C, Verin AD. Cytoskeletal mechanisms regulating vascular endothelial barrier function in response to acute lung injury. Tissue Barriers 2015; 3:e974448. [PMID: 25838980 DOI: 10.4161/21688370.2014.974448] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/04/2014] [Indexed: 01/11/2023] Open
Abstract
Endothelial cells (EC) form a semi-permeable barrier between the interior space of blood vessels and the underlying tissues. In acute lung injury (ALI) the EC barrier is weakened leading to increased vascular permeability. It is widely accepted that EC barrier integrity is critically dependent upon intact cytoskeletal structure and cell junctions. Edemagenic agonists, like thrombin or endotoxin lipopolysaccharide (LPS), induced cytoskeletal rearrangement, and EC contractile responses leading to disruption of intercellular contacts and EC permeability increase. The highly clinically-relevant cytoskeletal mechanisms of EC barrier dysfunction are currently under intense investigation and will be described and discussed in the current review.
Collapse
Key Words
- AJ, adherens junction
- ALI, Acute Lung Injury
- ARDS, Acute Respiratory Distress Syndrome
- CPI-17, PKC potentiated inhibitory protein of 17 kDa
- CaD, caldesmon
- EC, endothelial cells
- GJ, gap junction
- HSP-27, small heat shock actin-capping protein of 27 kDa
- IL, interleukin
- LPS, lipopolysaccharide
- MLC, myosin light chain
- MLCK, Ca2+/calmodulin (CaM) dependent MLC kinase
- MLCP, myosin light chain phosphatase
- MT, microtubules
- MYPT1, myosin phosphatase targeting subunit 1
- PKA, protein kinase A
- PKC, protein kinase C
- SM, smooth muscle
- TJ, tight junction
- TLR4, toll-like receptor 4
- TNFα, tumor necrosis factor α
- acute lung injury
- barrier function
- cytoskeleton
- endothelial junctions
- pulmonary endothelium
- thrombin
Collapse
Affiliation(s)
- Anita Kása
- Vascular Biology Center; Georgia Regents University ; Augusta, GA USA
| | - Csilla Csortos
- Department of Medical Chemistry; Faculty of Medicine; University of Debrecen ; Debrecen, Hungary
| | - Alexander D Verin
- Vascular Biology Center; Georgia Regents University ; Augusta, GA USA ; Division of Pulmonary; Medicine Medical College of Georgia; Georgia Regents University; Augusta, GA USA
| |
Collapse
|
28
|
Li L, Hu J, He T, Zhang Q, Yang X, Lan X, Zhang D, Mei H, Chen B, Huang Y. P38/MAPK contributes to endothelial barrier dysfunction via MAP4 phosphorylation-dependent microtubule disassembly in inflammation-induced acute lung injury. Sci Rep 2015; 5:8895. [PMID: 25746230 PMCID: PMC4352893 DOI: 10.1038/srep08895] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 02/10/2015] [Indexed: 02/07/2023] Open
Abstract
Excessive activation of inflammation and the accompanying lung vascular endothelial barrier disruption are primary pathogenic features of acute lung injury (ALI). Microtubule-associated protein 4 (MAP4), a tubulin assembly-promoting protein, is important for maintaining the microtubule (MT) cytoskeleton and cell-cell junctional structures. However, both the involvement and exact mechanism of MAP4 in the development of endothelial barrier disruption in ALI remains unknown. In this study, lipopolysaccharide (LPS) and tumour necrosis factor-α (TNF-α) were applied to human pulmonary microvascular endothelial cells (HPMECs) to mimic the endothelial damage during inflammation in vitro. We demonstrated that the MAP4 (Ser696 and Ser787) phosphorylation increased concomitantly with the p38/MAPK pathway activation by the LPS and TNF-α stimulation of HPMECs, which induced MT disassembly followed by hyperpermeability. Moreover, the application of taxol, the overexpression of a MAP4 (Ala) mutant, or the application of the p38/MAPK inhibitor SB203580 inhibited the MT disruption and the intracellular junction dysfunction. In contrast, MKK6 (Glu), which constitutively activated p38/MAPK, resulted in microtubule depolymerisation and, subsequently, hyperpermeability. Our findings reveal a novel role of MAP4 in endothelial barrier dysfunction.
Collapse
Affiliation(s)
- Lingfei Li
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jiongyu Hu
- Endocrinology Department, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Ting He
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xu Yang
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xiaodong Lan
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Dongxia Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hao Mei
- Department of Biostatistics in the School of Public Health, Yale University
| | - Bing Chen
- Endocrinology Department, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
29
|
Attenuated Blood-Brain Barrier Dysfunction by XQ-1H Following Ischemic Stroke in Hyperlipidemic Rats. Mol Neurobiol 2014; 52:162-75. [DOI: 10.1007/s12035-014-8851-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 07/31/2014] [Indexed: 01/16/2023]
|
30
|
Lee TH, Chang J, Kim BM. Saikosaponin C inhibits lipopolysaccharide-induced apoptosis by suppressing caspase-3 activation and subsequent degradation of focal adhesion kinase in human umbilical vein endothelial cells. Biochem Biophys Res Commun 2014; 445:615-21. [PMID: 24565837 DOI: 10.1016/j.bbrc.2014.02.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 02/11/2014] [Indexed: 01/05/2023]
Abstract
Bacterial lipopolysaccharide (LPS) is an important mediator of inflammation and a potent inducer of endothelial cell damage and apoptosis. In this study, we investigated the protective effects of saikosaponin C (SSc), one of the active ingredients produced by the traditional Chinese herb, Radix Bupleuri, against LPS-induced apoptosis in human umbilical endothelial cells (HUVECs). LPS triggered caspase-3 activation, which was found to be important in LPS-induced HUVEC apoptosis. Inhibition of caspase-3 also inhibited LPS-induced degradation of focal adhesion kinase (FAK), indicating that caspase-3 is important in LPS-mediated FAK degradation as well as in apoptosis in HUVECs. SSc significantly inhibited LPS-induced apoptotic cell death in HUVECs through the selective suppression of caspase-3. SSc was also shown to rescue LPS-induced FAK degradation and other cell adhesion signals. Furthermore, the protective effects of SSc against LPS-induced apoptosis were abolished upon pretreatment with a FAK inhibitor, highlighting the importance of FAK in SSc activity. Taken together, these results show that SSc efficiently inhibited LPS-induced apoptotic cell death via inhibition of caspase-3 activation and caspase-3-mediated-FAK degradation. Therefore, SSc represents a promising therapeutic candidate for the treatment of vascular endothelial cell injury and cellular dysfunction.
Collapse
Affiliation(s)
- Tae Ho Lee
- Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jihoon Chang
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Byeong Mo Kim
- Division of Gerontology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
31
|
Measuring apoptosis by microscopy and flow cytometry. Methods 2013; 61:90-7. [DOI: 10.1016/j.ymeth.2013.01.008] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 01/16/2013] [Accepted: 01/25/2013] [Indexed: 01/01/2023] Open
|
32
|
Herwig MC, Tsokos M, Hermanns MI, Kirkpatrick CJ, Müller AM. Vascular endothelial cadherin expression in lung specimens of patients with sepsis-induced acute respiratory distress syndrome and endothelial cell cultures. Pathobiology 2013; 80:245-51. [PMID: 23635392 DOI: 10.1159/000347062] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 01/14/2013] [Indexed: 11/19/2022] Open
Abstract
AIMS Vascular endothelial (VE) cadherin is a cell adhesion molecule localized at endothelial cell (EC) junctions. As a major component of endothelial adherens junctions, its main function is the maintenance and regulation of EC integrity. In the acute respiratory distress syndrome (ARDS), increased vascular permeability is a major mechanism in pulmonary edema and lung dysfunction. In this study, VE-cadherin expression was investigated in ARDS lungs and control tissue as well as in an ARDS cell culture model. METHODS Lung specimens of patients with ARDS due to Gram-negative sepsis (n = 20; control lung tissue: n = 41) and cell cultures of human pulmonary microvascular ECs and human umbilical vein ECs stimulated with LPS, TNF-α and IFN-γ were stained with a VE-cadherin antibody. Staining intensity was semiquantitatively evaluated by conventional light and immunofluorescence microscopy. RESULTS VE-cadherin expression was statistically significantly reduced in the endothelium of all vessel types in ARDS lungs compared to control tissue. Cell cultures showing disrupted cellular borders confirmed these results. CONCLUSION Reduced expression of VE-cadherin has to be considered as a major mechanism of increased vessel permeability in ARDS. The previously described vessel-type-specific expression pattern of VE-cadherin in the human lung is not influenced by ARDS.
Collapse
Affiliation(s)
- Martina C Herwig
- Division of Ophthalmic Pathology, Department of Ophthalmology, University of Bonn, DE-53127 Bonn, Germany
| | | | | | | | | |
Collapse
|
33
|
Shie JH, Liu HT, Kuo HC. Protein expression profiling in interstitial cystitis/painful bladder syndrome: A pilot study of proteins associated with inflammation, apoptosis, and angiogenesis. UROLOGICAL SCIENCE 2012. [DOI: 10.1016/j.urols.2012.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
34
|
Ji RC. Macrophages are important mediators of either tumor- or inflammation-induced lymphangiogenesis. Cell Mol Life Sci 2012; 69:897-914. [PMID: 21984600 PMCID: PMC11114502 DOI: 10.1007/s00018-011-0848-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/26/2011] [Accepted: 09/22/2011] [Indexed: 01/13/2023]
Abstract
The lymphatic system provides important functions for tissue fluid homeostasis and immune response. Lymphangiogenesis, the formation of new lymphatics, comprises a series of complex cellular events in vitro or in vivo, e.g., proliferation, differentiation, and sprouting. Recent evidence has implied that macrophages act as a direct structural contributor to lymphatic endothelial walls or secret VEGF-C/-D and VEGF-A to initiate lymphangiogenesis in inflamed or tumor tissues. Bone marrow-derived macrophages are versatile cells that express different functional programs in response to exposure to microenvironmental signals, and can be identified by specific expression of a number of proteins, F4/80, CD11b, and CD68. Several causative factors, e.g., NF-κB, IL-1β, TNF-α, SDF-1, M-CSF, especially TonEBP/VEGF-C signaling, may be actively involved in macrophage-induced lymphangiogenesis. Alteration of macrophage phenotype and function has a profound effect on the development and progression of inflammation and malignancy, and macrophage depletion for controlling lymphangiogenesis may provide a novel approach for prevention and treatment of lymphatic-associated diseases.
Collapse
Affiliation(s)
- Rui-Cheng Ji
- Department of Human Anatomy, Oita University Faculty of Medicine, Oita 879-5593, Japan.
| |
Collapse
|
35
|
Omori E, Matsumoto K, Ninomiya-Tsuji J. Non-canonical β-catenin degradation mediates reactive oxygen species-induced epidermal cell death. Oncogene 2011; 30:3336-44. [PMID: 21383695 PMCID: PMC3131442 DOI: 10.1038/onc.2011.49] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 10/26/2010] [Accepted: 11/10/2010] [Indexed: 01/03/2023]
Abstract
β-Catenin is constantly degraded through the ubiquitin-proteasomal pathway. In this study, we report that a different type of β-catenin degradation is causally involved in epidermal cell death. We observed that reactive oxygen species (ROS) caused β-catenin degradation in the epidermal cells through a caspase-dependent mechanism, which results in disruption of cell adhesion. Disruption of cell adhesion increased ROS and activated caspases. Upregulation of the intact β-catenin blocked ROS accumulation and caspase activation. These results indicate that a feed-forward loop consisting of ROS, caspases activation and β-catenin degradation induces epidermal cell death.
Collapse
Affiliation(s)
- Emily Omori
- Department of Environmental and Molecular Toxicology, North Carolina State University, Raleigh, NC 27695-7633 USA
| | - Kunihiro Matsumoto
- Department of Molecular Biology, Graduate School of Science, Nagoya University, Nagoya, 464-8602 JAPAN
| | - Jun Ninomiya-Tsuji
- Department of Environmental and Molecular Toxicology, North Carolina State University, Raleigh, NC 27695-7633 USA
| |
Collapse
|
36
|
Baguley BC, Siemann DW. Temporal aspects of the action of ASA404 (vadimezan; DMXAA). Expert Opin Investig Drugs 2011; 19:1413-25. [PMID: 20964495 DOI: 10.1517/13543784.2010.529128] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
IMPORTANCE OF THE FIELD Tumor vascular disrupting agents (tumor VDAs) act by selective induction of tumor vascular failure. While their action is distinct from that of antiangiogenic agents, their clinical potential is likely to reside in improving the efficacy of combination therapy. AREAS COVERED IN THIS REVIEW This review describes the preclinical development, clinical trial and mode of action of ASA404, a flavonoid class tumor VDA. This class has a unique dual action, simultaneously disrupting vascular endothelial function and stimulating innate tumor immunity. This review covers the early development of ASA404, through to Phase III trial. WHAT THE READER WILL GAIN The reader will gain insight into the sequence of ASA404-induced changes in tumor tissue. Early events include increased vascular permeability, increased endothelial apoptosis and decreased blood flow, while later effects include the induction of serotonin, tumor necrosis factor, other cytokines and chemokines, and nitric oxide. This cascade of events induces sustained reduction of tumor blood flow, induction of tumor hypoxia and increased inflammatory responses. The reader will also gain an appreciation of how the potentiation of radiation and chemotherapeutic effects by ASA404 in murine tumors shaped the development of combination clinical trials. TAKE HOME MESSAGE Although there are species differences in ASA404 activity, many features of its action in mice translate to human studies. The future of ASA404 as an effective clinical agent will rely on the development of an appreciation of its ability to optimize the complex interaction between tumor vasculature and tumor immunity during therapy.
Collapse
Affiliation(s)
- Bruce C Baguley
- The University of Auckland, Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, Private Bag 92019, Auckland, New Zealand.
| | | |
Collapse
|
37
|
Multiple organ failure in sepsis: prognosis and role of systemic inflammatory response. Curr Opin Crit Care 2011; 17:153-9. [DOI: 10.1097/mcc.0b013e328344b446] [Citation(s) in RCA: 176] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
38
|
Wang Y, Bakota E, Chang BH, Entman M, Hartgerink JD, Danesh FR. Peptide nanofibers preconditioned with stem cell secretome are renoprotective. J Am Soc Nephrol 2011; 22:704-17. [PMID: 21415151 PMCID: PMC3065226 DOI: 10.1681/asn.2010040403] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Accepted: 12/06/2010] [Indexed: 12/31/2022] Open
Abstract
Stem cells may contribute to renal recovery following acute kidney injury, and this may occur through their secretion of cytokines, chemokines, and growth factors. Here, we developed an acellular, nanofiber-based preparation of self-assembled peptides to deliver the secretome of embryonic stem cells (ESCs). Using an integrated in vitro and in vivo approach, we found that nanofibers preconditioned with ESCs could reverse cell hyperpermeability and apoptosis in vitro and protect against lipopolysaccharide-induced acute kidney injury in vivo. The renoprotective effect of preconditioned nanofibers associated with an attenuation of Rho kinase activation. We also observed that the combined presence of follistatin, adiponectin, and secretory leukoprotease during preconditioning was essential to the renoprotective properties of the nanofibers. In summary, we developed a designer-peptide nanofiber that can serve as a delivery platform for the beneficial effects of stem cells without the problems of teratoma formation or limited cell engraftment and viability.
Collapse
Affiliation(s)
- Yin Wang
- Departments of Medicine/Nephrology
| | | | | | - Mark Entman
- Cardiovascular Sciences, Baylor College of Medicine, Houston, Texas; and
| | | | | |
Collapse
|
39
|
Real-time impedance analysis of host cell response to meningococcal infection. J Microbiol Methods 2010; 84:101-8. [PMID: 21078346 DOI: 10.1016/j.mimet.2010.11.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 10/28/2010] [Accepted: 11/07/2010] [Indexed: 11/21/2022]
Abstract
Measuring cell proliferation and cell death during bacterial infection involves performing end-point assays that represent the response at a single time point. A new technology from Roche Applied Science and ACEA Biosciences allows continuous monitoring of cells in real-time using specialized cell culture microplates containing micro-electrodes. The xCELLigence system enables continuous measurement and quantification of cell adhesion, proliferation, spreading, cell death and detachment, thus creating a picture of cell function during bacterial infection. Furthermore, lag and log phases can be determined to estimate optimal times to infect cells. In this study we used this system to provide valuable insights into cell function in response to several virulence factors of the meningitis causing pathogen Neisseria meningitidis, including the lipopolysaccharide (LPS), the polysaccharide capsule and the outer membrane protein Opc. We observed that prolonged time of infection with pathogenic Neisseria strains led to morphological changes including cell rounding and loss of cell-cell contact, thus resulting in changed electrical impedance as monitored in real-time. Furthermore, cell function in response to 14 strains of apathogenic Neisseria spp. (N. lactamica and N. mucosa) was analyzed. In contrast, infection with apathogenic N. lactamica isolates did not change electrical impedance monitored for 48 h. Together our data show that this system can be used as a rapid monitoring tool for cellular function in response to bacterial infection and combines high data acquisition rates with ease of handling.
Collapse
|
40
|
Hattori Y, Takano KI, Teramae H, Yamamoto S, Yokoo H, Matsuda N. Insights into sepsis therapeutic design based on the apoptotic death pathway. J Pharmacol Sci 2010; 114:354-65. [PMID: 21081836 DOI: 10.1254/jphs.10r04cr] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Sepsis remains the leading cause of death in critically ill patients. A major problem contributing to sepsis-related high mortality is the lack of effective medical treatment. Thus, the key goal in critical care medicine is to develop novel therapeutic strategies that will impact favorably on septic patient outcome. While it is generally accepted that sepsis is an inflammatory state resulting from the systemic response to infection, apoptosis is implicated to be an important mechanism of the death of lymphocytes, gastrointestinal and lung epithelial cells, and vascular endothelial cells associated with the development of multiple organ failure in sepsis. The pivotal role of cell apoptosis is now highlighted by multiple studies demonstrating that prevention of cell apoptosis can improve survival in clinically relevant animal models of sepsis. In this review article, we address the scientific rationale for remedying apoptotic cell death in sepsis and propose that therapeutic efforts aimed at blocking cell signaling pathways leading to apoptosis may represent an attractive target for sepsis therapy.
Collapse
Affiliation(s)
- Yuichi Hattori
- Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | | | | | | | | | | |
Collapse
|
41
|
Neisseria meningitidis induces brain microvascular endothelial cell detachment from the matrix and cleavage of occludin: a role for MMP-8. PLoS Pathog 2010; 6:e1000874. [PMID: 20442866 PMCID: PMC2861698 DOI: 10.1371/journal.ppat.1000874] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 03/24/2010] [Indexed: 02/06/2023] Open
Abstract
Disruption of the blood-brain barrier (BBB) is a hallmark event in the pathophysiology of bacterial meningitis. Several inflammatory mediators, such as tumor necrosis factor alpha (TNF-α), nitric oxide and matrix metalloproteinases (MMPs), contribute to this disruption. Here we show that infection of human brain microvascular endothelial cells (HBMEC) with Neisseria meningitidis induced an increase of permeability at prolonged time of infection. This was paralleled by an increase in MMP-8 activity in supernatants collected from infected cells. A detailed analysis revealed that MMP-8 was involved in the proteolytic cleavage of the tight junction protein occludin, resulting in its disappearance from the cell periphery and cleavage to a lower-sized 50-kDa protein in infected HBMEC. Abrogation of MMP-8 activity by specific inhibitors as well as transfection with MMP-8 siRNA abolished production of the cleavage fragment and occludin remained attached to the cell periphery. In addition, MMP-8 affected cell adherence to the underlying matrix. A similar temporal relationship was observed for MMP activity and cell detachment. Injury of the HBMEC monolayer suggested the requirement of direct cell contact because no detachment was observed when bacteria were placed above a transwell membrane or when bacterial supernatant was directly added to cells. Inhibition of MMP-8 partially prevented detachment of infected HBMEC and restored BBB permeability. Together, we established that MMP-8 activity plays a crucial role in disassembly of cell junction components and cell adhesion during meningococcal infection. A crucial step in the pathogenesis of bacterial meningitis is the disturbance of cerebral microvascular endothelial function, resulting in blood-brain barrier (BBB) breakdown. Matrix metalloproteinases (MMPs) have been implicated in BBB damage in bacterial meningitis in several studies. MMPs are a family of zinc-dependent endopeptidases that catalyze the proteolysis of extracellular matrix proteins, but can also cleave a range of other molecules, including cell adhesion molecules. In this study we showed that brain endothelial cells produced MMPs—in particular MMP-8—upon infection with Neisseria meningitidis, a bacterium that causes meningitis and septic shock. We found that MMP-8 was then involved in disruption of the tight junction protein occludin. In addition to the effect of MMP-8 on the tight junction component, MMP-8 activity also accounted for brain endothelial cell detachment that occurred during prolonged time of infection with N. meningitidis. When we inhibited MMP-8 activity, occludin disruption was completely abolished and cell detachment could be partially prevented, which resulted in restored BBB permeability. Our data reveal a molecular mechanism of cellular dysfunction during meningococcal meningitis that enhances our understanding how MMPs affect cerebral endothelial function and that can aid in our understanding and prevention of this disease.
Collapse
|
42
|
Childs EW, Tharakan B, Nurudeen S, Delmas TL, Hellman J, Christie T, Hunter FA, Smythe WR. Cyclosporine A--protection against microvascular hyperpermeability is calcineurin independent. Am J Surg 2010; 199:542-8. [PMID: 20359571 PMCID: PMC2864542 DOI: 10.1016/j.amjsurg.2009.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 11/20/2009] [Accepted: 11/20/2009] [Indexed: 11/18/2022]
Abstract
BACKGROUND Mitochondria-mediated apoptotic signaling contributes to microvascular hyperpermeability. We hypothesized that cyclosporine A (CsA), which protects mitochondrial transition pores, would attenuate hyperpermeability independent of its calcineurin inhibitory property. METHODS Hyperpermeability was induced in microvascular endothelial cell monolayers using proapoptotic BAK or active caspase-3 after CsA or a specific calcineurin inhibitor, calcineurin autoinhibitory peptide (CIP), treatment. Permeability was measured based on fluorescein isothiocyanate-albumin flux across the monolayers. Mitochondrial transmembrane potential (MTP) was determined using 5,5',6,6'-tetrachoro-1,1',3,3'-tetraethylbenzimidazolyl carbocyanine iodide. Mitochondrial release of cytochrome c was measured using an enzyme-linked immunosorbent assay and caspase-3 activity fluorometrically. RESULTS CsA-attenuated (10 nmol/L) but not CIP-attenuated (100 mumol/L) BAK induced hyperpermeability (P < .05), CsA- but not CIP-attenuated BAK induced a decrease in MTP and an increase in cytochrome c levels and caspase-3 activity (P < .05). CsA and CIP were ineffective against caspase-3-induced hyperpermeability. CONCLUSIONS CsA attenuated hyperpermeability by protecting MTP, thus preventing mitochondria-mediated apoptotic signaling. The protective effect of CsA is independent of calcineurin inhibition.
Collapse
Affiliation(s)
- Ed W Childs
- Department of Surgery, Texas A&M Health Science Center College of Medicine and Scott & White Memorial Hospital, 2401 South 31st St., Temple, TX 76508, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Tikhmyanova N, Little JL, Golemis EA. CAS proteins in normal and pathological cell growth control. Cell Mol Life Sci 2010; 67:1025-48. [PMID: 19937461 PMCID: PMC2836406 DOI: 10.1007/s00018-009-0213-1] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 11/03/2009] [Accepted: 11/09/2009] [Indexed: 12/20/2022]
Abstract
Proteins of the CAS (Crk-associated substrate) family (BCAR1/p130Cas, NEDD9/HEF1/Cas-L, EFS/SIN and CASS4/HEPL) are integral players in normal and pathological cell biology. CAS proteins act as scaffolds to regulate protein complexes controlling migration and chemotaxis, apoptosis, cell cycle, and differentiation, and have more recently been linked to a role in progenitor cell function. Reflecting these complex functions, over-expression of CAS proteins has now been strongly linked to poor prognosis and increased metastasis in cancer, as well as resistance to first-line chemotherapeutics in multiple tumor types including breast and lung cancers, glioblastoma, and melanoma. Further, CAS proteins have also been linked to additional pathological conditions including inflammatory disorders, Alzheimer's and Parkinson's disease, as well as developmental defects. This review will explore the roles of the CAS proteins in normal and pathological states in the context of the many mechanistic insights into CAS protein function that have emerged in the past decade.
Collapse
Affiliation(s)
- Nadezhda Tikhmyanova
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
- Department of Biochemistry, Drexel University Medical School, Philadelphia, PA 19102 USA
| | - Joy L. Little
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
| | - Erica A. Golemis
- Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111 USA
| |
Collapse
|
44
|
Liu ZM, Zhu SM, Qin XJ, Cheng ZD, Liu MY, Zhang HM, Liu DX. Silencing of C5a receptor gene with siRNA for protection from Gram-negative bacterial lipopolysaccharide-induced vascular permeability. Mol Immunol 2010; 47:1325-33. [PMID: 20138669 DOI: 10.1016/j.molimm.2009.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 11/01/2009] [Accepted: 11/03/2009] [Indexed: 02/05/2023]
Abstract
Endothelial barrier dysfunction leading to increased permeability and vascular leakage is an underlying cause of several pathological conditions. Whereas these changes have been shown to be associated with activation of the complement system, leading to the release of C5a and interaction of C5a-C5a receptor (C5aR), the role of C5aR in endothelial cells remain(s) ill-defined. Here, we report an essential role of C5aR in endothelial cell injury and vascular permeability through silencing of the C5aR gene using siRNA. In the cultured mouse dermal microvascular endothelial cells (MEMECs) monolayer transfected with C5aR-siRNA, endotoxin-induced cell injury by evaluated as transendothelial flux, cell detachment, and cytoskeletal disorganization was inhibited. Upregulation of vascular cell adhesion molecule-1 (VCAM-1) was also suppressed. Studies exploring the underlying mechanism of siRNA-mediated suppression in VCAM-1 expression were related to reduction of NF-kappaB activation and nuclear localization of both p50 and p65. The effect was associated with inhibition in activation of protein kinase Cdelta(PKC-delta) and induction of PKC-mediated mitogen-activated protein kinase phosphatases-1 (MKP-1) leading to the increased activity of p42/p44 mitogen-activated protein (MAP) kinase cascade. In the model of mice administrated with C5aR-siRNA, endotoxin-induced plasma leakage was inhibited in local abdominal skin. Systemic administration of endotoxin to mice resulted in increased microvascular permeability in multiple organs was reduced. These studies demonstrate that the C5aR responsible for vascular endothelial cell injury and plasma permeability is an important factor, and that blockade of C5aR may be useful therapeutic targets for the prevention of vascular permeability in pathogenic condition.
Collapse
Affiliation(s)
- Zi-ming Liu
- West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | | | | | | | | | | | | |
Collapse
|
45
|
Shioiri T, Muroi M, Hatao F, Nishida M, Ogawa T, Mimura Y, Seto Y, Kaminishi M, Tanamoto KI. Caspase-3 is activated and rapidly released from human umbilical vein endothelial cells in response to lipopolysaccharide. Biochim Biophys Acta Mol Basis Dis 2009; 1792:1011-8. [PMID: 19559790 DOI: 10.1016/j.bbadis.2009.06.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 06/16/2009] [Accepted: 06/17/2009] [Indexed: 12/25/2022]
Abstract
Endothelial cell injury/dysfunction is considered to play a critical role in the pathogenesis of severe sepsis and septic shock. Although it is considered that endothelial cell apoptosis is involved in endothelial injury/dysfunction, physiological involvement remains ambiguous since the induction of apoptosis requires the inhibition of endogenous apoptosis inhibitors. Here we show that caspase-3 activation, a biological indicator of apoptosis, is observed in response to lipopolysaccharide (LPS) stimulation even under the influence of endogenous apoptosis inhibitors, and that activated caspase-3 is rapidly released from human umbilical vein endothelial cells (HUVEC). In the presence of cycloheximide (CHX), an increase in intracellular caspase-3/7 activity in response to LPS was not detected in HUVEC up to 24 h following stimulation even in the presence of LPS-binding protein (LBP), soluble CD14 and soluble MD-2, whereas the decrease in cell viability and increase in release of the cellular enzyme lactate dehydrogenase (LDH) were observed in a soluble CD14/LBP-dependent manner. On the other hand, even in the absence of CHX, a significant increase in caspase-3/7 activity and a cleaved caspase-3 fragment with a slight increase in LDH release was observed in culture supernatants in response to LPS. This increase in caspase-3/7 activity was observed even when LDH release was undetected. These results indicate that caspase-3 is activated by LPS under physiological conditions and suggest that HUVEC escape from cell death by rapidly releasing activated caspase-3 into extracellular space. Failure of this escape mechanism may result in endothelial injury/dysfunction.
Collapse
Affiliation(s)
- Toshikazu Shioiri
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo 202-8585, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Karahashi H, Michelsen KS, Arditi M. Lipopolysaccharide-induced apoptosis in transformed bovine brain endothelial cells and human dermal microvessel endothelial cells: the role of JNK. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:7280-6. [PMID: 19454725 PMCID: PMC3057198 DOI: 10.4049/jimmunol.0801376] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Stimulation of transformed bovine brain endothelial cells (TBBEC) with LPS leads to apoptosis while human microvessel endothelial cells (HMEC) need the presence of cycloheximide (CHX) with LPS to induce apoptosis. To investigate the molecular mechanism of LPS-induced apoptosis in HMEC or TBBEC, we analyzed the involvement of MAPK and PI3K in TBBEC and HMEC. LPS-induced apoptosis in TBBEC was hallmarked by the activation of caspase 3, caspase 6, and caspase 8 after the stimulation of LPS, followed by poly(ADP-ribose) polymerase cleavage and lactate dehydrogenase release. We also observed DNA cleavage determined by TUNEL staining in TBBEC treated with LPS. Herbimycin A, a tyrosine kinase inhibitor, and SP600125, a JNK inhibitor, suppressed the activation of caspases and lactate dehydrogenase release. Moreover, a PI3K inhibitor (LY294002) suppressed activation of caspases and combined treatment with both SP600125 and LY294002 completely inhibited the activation of caspases. These results suggest that the JNK signaling pathway through the tyrosine kinase and PI3K pathways is involved in the induction of apoptosis in LPS-treated TBBEC. On the other hand, we observed sustained JNK activation in HMEC treated with LPS and CHX, and neither ERK1/2 nor AKT were activated. The addition of SP600125 suppressed phosphorylation of JNK and the activation of caspase 3 in HMEC treated with LPS and CHX. These results suggest that JNK plays an important role in the induction of apoptosis in endothelial cells.
Collapse
Affiliation(s)
- Hisae Karahashi
- Division of Pediatrics Infectious Diseases and Immunology and Immunobiology Research Institute, Cedars-Sinai Medical Center, David Geffen School of Medicine at the University of California Los Angeles, USA
| | - Kathrin S. Michelsen
- Division of Pediatrics Infectious Diseases and Immunology and Immunobiology Research Institute, Cedars-Sinai Medical Center, David Geffen School of Medicine at the University of California Los Angeles, USA
| | - Moshe Arditi
- Division of Pediatrics Infectious Diseases and Immunology and Immunobiology Research Institute, Cedars-Sinai Medical Center, David Geffen School of Medicine at the University of California Los Angeles, USA
| |
Collapse
|
47
|
Moore R, Hawley A, Sigler R, Farris D, Wrobleski S, Ramacciotti E, Myers D. Tissue inhibitor of metalloproteinase-1 is an early marker of acute endothelial dysfunction in a rodent model of venous oxidative injury. Ann Vasc Surg 2009; 23:498-505. [PMID: 19467832 DOI: 10.1016/j.avsg.2009.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 03/06/2009] [Accepted: 03/11/2009] [Indexed: 11/28/2022]
Abstract
Although there are extensive research data regarding arterial endothelial dysfunction, the effects of venous endothelial dysfunction are not well characterized. Matrix metalloproteinases (MMPs) have a defined role in vascular remodeling. MMPs are endopeptidases that are capable of degrading extracellular matrix proteins. We hypothesize that tissue inhibitor of metalloproteinase-1 (TIMP-1) can serve as an indicator of acute venous endothelial dysfunction in a rat model of oxidative injury. The experimental groups evaluated were as follows: rats not undergoing oxidative injury (controls), rats that received rose bengal but no laser (shams), and rats that received both rose bengal and laser illumination, resulting in an oxidative injury. Animals were evaluated at baseline (control, shams) and at 1 hr and 1 day post-oxidative injury. mRNA expression was determined by gene array technology and real-time polymerase chain reaction, plasma and vein wall TIMP-1 protein concentrations were determined by enzyme-linked immunosorbent assay, and vein wall morphometrics (cells/five high-power fields) were performed. B-cell lymphoma 2-like gene expression was upregulated at both 1 hr and 1 day post-injury. TIMP-1 protein and mRNA expression were significantly increased post-oxidative injury. One hour postinjury, vein wall polymorphonuclear leukocytes were present in significant numbers. Our results support the hypothesis that increased expression of TIMP-1 in venous endothelium and plasma may serve as an early indicator of endothelial dysfunction.
Collapse
Affiliation(s)
- Rashida Moore
- Section of Vascular Surgery, Jobst Vascular Research Laboratories, University of Michigan, Ann Arbor, MI 48109-0654, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Cyclosporine A prevents vascular hyperpermeability after hemorrhagic shock by inhibiting apoptotic signaling. ACTA ACUST UNITED AC 2009; 66:1033-9. [PMID: 19359911 DOI: 10.1097/ta.0b013e31816c905f] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Hemorrhagic shock (HS) is associated with the activation of caspase-dependent or -independent apoptotic signaling pathways, disruption of endothelial cell adherens junctions, and vascular hyperpermeability. Recent studies have suggested that the vascular hyperpermeability observed after HS is associated with activation of the intrinsic apoptotic signaling cascade resulting in caspase-mediated cleavage of endothelial cell adherens proteins and subsequent cell-cell detachment. We hypothesized that cyclosporine A (CsA) would attenuate vascular hyperpermeability after HS by protecting mitochondrial transition pores and thereby preventing the activation of caspase-mediated apoptotic signaling. The objective of this study was to determine the effect of CsA on, HS-induced hyperpermeability, mitochondrial membrane depolarization, mitochondrial release of cytochrome c, and caspase 3 activation. METHODS HS was induced in Sprague-Dawley rats by withdrawing blood to reduce the mean arterial pressure to 40 mm Hg for 60 minutes. CsA (10 microL/mL) was given 10 minutes before the shock period. The mesenteric postcapillary venules of the proximal ileum were monitored for permeability changes using intravital microscopy. The changes in mitochondrial transmembrane potential were determined using the cationic dye JC-1. Mitochondrial release of cytochrome c in to the cytosol was detected using ELISA. Caspase-3 activity was measured using a fluorometric assay. RESULTS HS induced vascular hyperpermeability, release of cytochrome c, and activation of caspase-3 (p < 0.05). CsA (10 microL/mL) attenuated HS-induced hyperpermeability (p < 0.05) and prevented HS-induced decrease in mitochondrial transmembrane potential. CsA treatment decreased the HS-induced rise in cytosolic cytochrome c levels and caspase-3 activity (p < 0.05). CONCLUSIONS These findings demonstrate that CsA protects mitochondrial permeability transition pores to prevent HS-induced release of cytochrome c and caspase-3 activation.
Collapse
|
49
|
Kessler T, Müller HAJ. Cleavage of Armadillo/beta-catenin by the caspase DrICE in Drosophila apoptotic epithelial cells. BMC DEVELOPMENTAL BIOLOGY 2009; 9:15. [PMID: 19232093 PMCID: PMC2657781 DOI: 10.1186/1471-213x-9-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Accepted: 02/20/2009] [Indexed: 11/24/2022]
Abstract
BACKGROUND During apoptosis cells become profoundly restructured through concerted cleavage of cellular proteins by caspases. In epithelial tissues, apoptotic cells loose their apical/basal polarity and are extruded from the epithelium. We used the Drosophila embryo as a system to investigate the regulation of components of the zonula adherens during apoptosis. Since Armadillo/beta-catenin (Arm) is a major regulator of cadherin-mediated adhesion, we analyzed the mechanisms of Arm proteolysis in apoptosis. RESULTS We define early and late apoptotic stages and find that early in apoptosis Dalpha-catenin remains relatively stable, while Arm and DE-cadherin protein levels are strongly reduced. Arm is cleaved by caspases in embryo extracts and we provide evidence that the caspase-3 homolog drICE cleaves Arm in vitro and in vivo. Cleavage by drICE creates a stable protein fragment that remains associated with the plasma membrane early in apoptosis. To further understand the role of caspase-mediated cleavage of Arm, we examined potential caspase cleavage sites and found that drICE cleaves Arm at a unique DQVD motif in the N-terminal domain of the protein. Mutation of the drICE cleavage site in Arm results in a protein that is not cleaved in vitro and in vivo. Furthermore we provide evidence that cleavage of Arm plays a role in the removal of DE-cadherin from the plasma membrane during apoptosis. CONCLUSION This study defines the specificity of caspase cleavage of Arm in Drosophila apoptotic cells. Our data suggest that N-terminal truncation of Arm by caspases is evolutionarily conserved and thus might provide a principal mechanism involved in the disassembly of adherens junctions during apoptosis.
Collapse
Affiliation(s)
- Thomas Kessler
- Institut für Genetik, Heinrich Heine Universität, Düsseldorf, Germany
- Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - H Arno J Müller
- Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
50
|
Ngo ML, Mahdi F, Kolte D, Shariat-Madar Z. Upregulation of prolylcarboxypeptidase (PRCP) in lipopolysaccharide (LPS) treated endothelium promotes inflammation. JOURNAL OF INFLAMMATION-LONDON 2009; 6:3. [PMID: 19171072 PMCID: PMC2639534 DOI: 10.1186/1476-9255-6-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2008] [Accepted: 01/27/2009] [Indexed: 02/06/2023]
Abstract
BACKGROUND Prolylcarboxypeptidase (Prcp) gene, along with altered PRCP and kallikrein levels, have been implicated in inflammation pathogenesis. PRCP regulates angiotensin 1-7 (Ang 1-7) - and bradykinin (BK) - stimulated nitric oxide production in endothelial cells. The mechanism through which kallikrein expression is altered during infection is not fully understood. Investigations were performed to determine the association between PRCP and kallikrein levels as a function of the upregulation of PRCP expression and the link between PRCP and inflammation risk in lipopolysaccharide (LPS)-induced endothelium activation. METHODS The Prcp transcript expression in LPS-induced human umbilical vein endothelial cells (HUVEC) activation was determined by RT-PCR for mRNA. PRCP-dependent kallikrein pathway was determined either by Enzyme Linked ImmunoSorbent Assay (ELISA) or by biochemical assay. RESULTS We report that PRCP is critical to the maintenance of the endothelial cells, and its upregulation contributes to the risk of developing inflammation. Significant elevation in kallikrein was seen on LPS-treated HUVECs. The conversion of PK to kallikrein was blocked by the inhibitor of PRCP, suggesting that PRCP might be a risk factor for inflammation. CONCLUSION The increased PRCP lead to a sustained production of bradykinin in endothelium following LPS treatment. This amplification may be an additional mechanism whereby PRCP promotes a sustained inflammatory response. A better appreciation of the role of PRCP in endothelium may contribute to a better understanding of inflammatory vascular disorders and to the development of a novel treatment.
Collapse
Affiliation(s)
- My-Linh Ngo
- School of Pharmacy, Department of Pharmacology, University of Mississippi, Oxford, MS, USA.
| | | | | | | |
Collapse
|