Article Data

  • Views 2398
  • Dowloads 247

Reviews

Open Access Special Issue

New insights in ARDS pathogenesis

  • Paloma González-Rodríguez1
  • Raquel Herrero1,2,3,*,
  • Gema Sánchez1,4
  • Jose Angel Lorente1,2,3,5,6

1Foundation for Biomedical Research of the Getafe University Hospital, 28905 Getafe, Madrid, Spain

2Department of Critical Care Medicine, Getafe University Hospital, 28905 Getafe, Madrid, Spain

3CIBER of respiratory diseases (CIBERES), Carlos III Health Institute, 28029 Madrid, Spain

4Department of Clinical Analysis, Getafe University Hospital, 28905 Getafe, Madrid, Spain

5Clinical Department, European University of Madrid, 28670 Villaviciosa de Odón, Madrid, Spain

6Department of Bioengineering, Carlos III University, 28911 Leganés, Madrid, Spain

DOI: 10.22514/sv.2022.058 Vol.18,Issue 5,September 2022 pp.51-67

Submitted: 15 March 2022 Accepted: 06 July 2022

Published: 08 September 2022

(This article belongs to the Special Issue New Insights in Acute Respiratory Distress Syndrome)

*Corresponding Author(s): Raquel Herrero E-mail: raquelher@hotmail.com

Abstract

Acute respiratory distress syndrome (ARDS) is a life-threatening condition in critically ill patients characterized by hypoxemia and non-compliant lung. In this review, we discuss the pathophysiology of ARDS, including the mechanisms involved in the formation of pulmonary edema, the dysregulated inflammatory and immune responses, the activation of procoagulant events, the cellular communication by extracellular vesicles (EVs) between different types of cells and the interaction of the lung with other organs. Activation of inflammation, coagulation, and cell death processes result in the disruption of the alveolar-capillary membrane and the consequent protein-rich edema formation in the alveoli, in which structural and functional alterations of the alveolar epithelium play an essential role. Inflammation and activated endothelial cells trigger coagulation cascades and platelets that generate a procoagulant state in both the airspace and the intravascular compartment with the formation of fibrin in airspaces and thrombi in the microvasculature that aggravate alveolar injury and gas exchange. The crosstalk between epithelial/endothelial cells, platelets, and immune cells is mediated by EVs, whose role in the pathogenesis of ARDS is not known. Finally, the interaction of the lung with other organs has become an important determinant in the development and resolution of ARDS. Understanding the pathophysiological mechanisms involved in ARDS is crucial to developing new therapeutic strategies.


Keywords

Acute respiratory distress syndrome; Mechanisms; Pulmonary edema; Inflammation; Coagulation; Extracellular vesicles (EVs); Organ interaction


Cite and Share

Paloma González-Rodríguez,Raquel Herrero,Gema Sánchez,Jose Angel Lorente. New insights in ARDS pathogenesis. Signa Vitae. 2022. 18(5);51-67.

References

[1] Ranieri VM, Rubenfeld GD, Thompson BT, Ferguson ND, Caldwell E, Fan E, et al. Acute respiratory distress syndrome: the Berlin definition. JAMA. 2012; 307: 2526–2533.

[2] Matthay MA, Zemans RL, Zimmerman GA, Arabi YM, Beitler JR, Mercat A, et al. Acute respiratory distress syndrome. Nature Reviews Disease Primers. 2019; 5: 18.

[3] Diamond M, Peniston HL, Sanghavi D, Mahapatra S. Acute respiratory distress syndrome. StatPearls Publishing: Treasure Island (FL). 2022.

[4] Zhou F, Yu T, Du R, Fan G, Liu Y, Liu Z, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. The Lancet. 2020; 395: 1054–1062.

[5] Williams GW, Berg NK, Reskallah A, Yuan X, Eltzschig HK. Acute respiratory distress syndrome. Anesthesiology. 2021; 134: 270–282.

[6] Kao K, Hu H, Chang C, Hung C, Chiu L, Li S, et al. Diffuse alveolar damage associated mortality in selected acute respiratory distress syndrome patients with open lung biopsy. Critical Care. 2015; 19: 228.

[7] Lorente JA, Ballén-Barragán A, Herrero R, Esteban A. Acute respiratory distress syndrome: does histology matter? Critical Care. 2015; 19: 337.

[8] Hasan SS, Capstick T, Ahmed R, Kow CS, Mazhar F, Merchant HA, et al. Mortality in COVID-19 patients with acute respiratory distress syndrome and corticosteroids use: a systematic review and meta-analysis. Expert Review of Respiratory Medicine. 2020; 14: 1149–1163.

[9] Bellani G, Laffey JG, Pham T, Fan E, Brochard L, Esteban A, et al. Epidemiology, patterns of care, and mortality for patients with acute respiratory distress syndrome in intensive care units in 50 countries. JAMA. 2016; 315: 788.

[10] Gumbert SD, Kork F, Jackson ML, Vanga N, Ghebremichael SJ, Wang CY, et al. Perioperative acute kidney injury. Anesthesiology. 2020; 132: 180–204.

[11] de Oliveira RP, Teixeira C, Rosa RG. Acute respiratory distress syndrome: how do patients fare after the intensive care unit? Brazilian Journal of Intensive Therapy. 2019; 31: 555–560.

[12] Mart MF, Ware LB. The long-lasting effects of the acute respiratory distress syndrome. Expert Review of Respiratory Medicine. 2020; 14: 577–586.

[13] Matthay MA, Zemans RL. The acute respiratory distress syndrome: pathogenesis and treatment. Annual Review of Pathology: Mechanisms of Disease. 2011; 6: 147–163.

[14] Zemans RL, Matthay MA. Bench-to-bedside review: the role of the alveolar epithelium in the resolution of pulmonary edema in acute lung injury. Critical Care. 2004; 8: 469–477.

[15] Bachofen M, Weibel ER. Structural alterations of lung parenchyma in the adult respiratory distress syndrome. Clinics in Chest Medicine. 1982; 3: 35–56.

[16] Matthay MA, Zemans RL, Zimmerman GA, Arabi YM, Beitler JR, Mercat A, et al. Acute respiratory distress syndrome. Nature Reviews Disease Primers. 2019; 5: 18.

[17] Holter JF, Weiland JE, Pacht ER, Gadek JE, Davis WB. Protein permeability in the adult respiratory distress syndrome. Loss of size selectivity of the alveolar epithelium. Journal of Clinical Investigation. 1986; 78: 1513–1522.

[18] Ware LB, Fremont RD, Bastarache JA, Calfee CS, Matthay MA. Determining the aetiology of pulmonary oedema by the oedema fluid-to-plasma protein ratio. European Respiratory Journal. 2010; 35: 331–337.

[19] Herrero R, Sanchez G, Lorente JA. New insights into the mechanisms of pulmonary edema in acute lung injury. Annals of Translational Medicine. 2018; 6: 32–32.

[20] Matthay MA. Resolution of pulmonary edema. Thirty years of progress. American Journal of Respiratory and Critical Care Medicine. 2014; 189: 1301–1308.

[21] Gwoździńska P, Buchbinder BA, Mayer K, Herold S, Morty RE, Seeger W, et al. Hypercapnia impairs ENaC cell surface stability by promoting phosphorylation, polyubiquitination and endocytosis of β-ENac in a human alveolar epithelial cell line. Frontiers in Immunology. 2017; 8: 591.

[22] Vadász I, Sznajder JI. Gas exchange disturbances regulate alveolar fluid clearance during acute lung injury. Frontiers in Immunology. 2017; 8: 757.

[23] Herrero R, Tanino M, Smith LS, Kajikawa O, Wong VA, Mongovin S, et al. The Fas/FasL pathway impairs the alveolar fluid clearance in mouse lungs. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2013; 305: L377–L388.

[24] Herrero R, Kajikawa O, Matute-Bello G, Wang Y, Hagimoto N, Mongovin S, et al. The biological activity of FasL in human and mouse lungs is determined by the structure of its stalk region. Journal of Clinical Investigation. 2011; 121: 1174–1190.

[25] Herrero R, Prados L, Ferruelo A, Puig F, Pandolfi R, Guillamat-Prats R, et al. Fas activation alters tight junction proteins in acute lung injury. Thorax. 2019; 74: 69–82.

[26] Fujishima S. Pathophysiology and biomarkers of acute respiratory distress syndrome. Journal of Intensive Care. 2014; 2: 32.

[27] Xiang M, Fan J. Pattern recognition receptor-dependent mechanisms of acute lung injury. Molecular Medicine. 2010; 16: 69–82.

[28] Mazzon E, Cuzzocrea S. Role of TNF-α in lung tight junction alteration in mouse model of acute lung inflammation. Respiratory Research. 2007; 8: 75.

[29] Lin X, Barravecchia M, Kothari P, Young JL, Dean DA. Β1-Na+, K+-ATPase gene therapy upregulates tight junctions to rescue lipopolysaccharide-induced acute lung injury. Gene Therapy. 2016; 23: 489–499.

[30] Davey A, McAuley DF, O’Kane CM. Matrix metalloproteinases in acute lung injury: mediators of injury and drivers of repair. European Respiratory Journal. 2011; 38: 959–970.

[31] Mammoto A, Mammoto T, Kanapathipillai M, Wing Yung C, Jiang E, Jiang A, et al. Control of lung vascular permeability and endotoxin-induced pulmonary oedema by changes in extracellular matrix mechanics. Nature Communications. 2013; 4: 1759.

[32] Watson WH, Ritzenthaler JD, Roman J. Lung extracellular matrix and redox regulation. Redox Biology. 2016; 8: 305–315.

[33] Pugin J, Verghese G, Widmer MC, Matthay MA. The alveolar space is the site of intense inflammatory and profibrotic reactions in the early phase of acute respiratory distress syndrome. Critical Care Medicine. 1999; 27: 304–312.

[34] Pittet J, Griffiths MJD, Geiser T, Kaminski N, Dalton SL, Huang X, et al. TGF-β is a critical mediator of acute lung injury. Journal of Clinical Investigation. 2001; 107: 1537–1544.

[35] Eaton DC, Helms MN, Koval M, Bao HF, Jain L. The contribution of epithelial sodium channels to alveolar function in health and disease. Annual Review of Physiology. 2009; 71: 403–423.

[36] Drakopanagiotakis F, Xifteri A, Polychronopoulos V, Bouros D. Apoptosis in lung injury and fibrosis. European Respiratory Journal. 2008; 32: 1631–1638.

[37] Moss M, Gillespie MK, Ackerson L, Moore FA, Moore EE, Parsons PE. Endothelial cell activity varies in patients at risk for the adult respiratory distress syndrome. Critical Care Medicine. 1996; 24: 1782–1786.

[38] Cheng KT, Xiong S, Ye Z, Hong Z, Di A, Tsang KM, et al. Caspase-11—mediated endothelial pyroptosis underlies endotoxemia-induced lung injury. Journal of Clinical Investigation. 2017; 127: 4124–4135.

[39] Vassiliou AG, Kotanidou A, Dimopoulou I, Orfanos SE. Endothelial damage in acute respiratory distress syndrome. International Journal of Molecular Sciences. 2020; 21: 8793.

[40] Lijnen H, Suffredini A, Pepper M, Steinberg K, Martin T, Pugin J, et al. Increased angiostatin levels in bronchoalveolar lavage fluids from ARDS patients and from human volunteers after lung instillation of endotoxin. Thrombosis and Haemostasis. 2002; 87: 966–971.

[41] Fahy RJ, Lichtenberger F, McKeegan CB, Nuovo GJ, Marsh CB, Wewers MD. The acute respiratory distress syndrome. American Journal of Respiratory Cell and Molecular Biology. 2003; 28: 499–503.

[42] Wang Y, Wang H, Zhang C, Zhang C, Yang H, Gao R, et al. Lung fluid biomarkers for acute respiratory distress syndrome: a systematic review and meta-analysis. Critical Care. 2019; 23: 43.

[43] Leksa V, Godar S, Schiller HB, Fuertbauer E, Muhammad A, Slezakova K, et al. TGF-β-induced apoptosis in endothelial cells mediated by M6P/IGFII-R and mini-plasminogen. Journal of Cell Science. 2005; 118: 4577–4586.

[44] Polunovsky VA, Wendt CH, Ingbar DH, Peterson MS, Bitterman PB. Induction of endothelial cell apoptosis by TNF α: modulation by inhibitors of protein synthesis. Experimental Cell Research. 1994; 214: 584–594.

[45] Lee YH, Kayyali US, Sousa AM, Rajan T, Lechleider RJ, Day RM. Transforming growth factor-β1 effects on endothelial monolayer permeability involve focal adhesion kinase/Src. American Journal of Respiratory Cell and Molecular Biology. 2007; 37: 485–493.

[46] Bruewer M, Luegering A, Kucharzik T, Parkos CA, Madara JL, Hopkins AM, et al. Proinflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. The Journal of Immunology. 2003; 171: 6164–6172.

[47] Li Q, Zhang Q, Wang M, Zhao S, Ma J, Luo N, et al. Interferon-gamma and tumor necrosis factor-alpha disrupt epithelial barrier function by altering lipid composition in membrane microdomains of tight junction. Clinical Immunology 2008; 126: 67–80.

[48] Ganter MT, Roux J, Miyazawa B, Howard M, Frank JA, Su G, et al. Interleukin-1β causes acute lung injury via αvβ5 and αvβ6 integrin-dependent mechanisms. 2008; 102: 804–812.

[49] Millar FR, Summers C, Griffiths MJ, Toshner MR, Proudfoot AG. The pulmonary endothelium in acute respiratory distress syndrome: insights and therapeutic opportunities. Thorax. 2016; 71: 462–473.

[50] Imai Y, Kuba K, Penninger JM. The renin-angiotensin system in acute respiratory distress syndrome. Drug Discovery Today: Disease Mechanisms. 2006; 3: 225–229.

[51] Revercomb L, Hanmandlu A, Wareing N, Akkanti B, Karmouty-Quintana H. Mechanisms of pulmonary hypertension in acute respiratory distress syndrome (ARDS). Frontiers in Molecular Biosciences. 2021; 7: 624093.

[52] Kuba K, Yamaguchi T, Penninger JM. Angiotensin-converting enzyme 2 (ACE2) in the pathogenesis of ARDS in COVID-19. Frontiers in Immunology. 2021; 12: 732690.

[53] Yanagi S, Tsubouchi H, Miura A, Matsumoto N, Nakazato M. Breakdown of epithelial barrier integrity and overdrive activation of alveolar epithelial cells in the pathogenesis of acute respiratory distress syndrome and lung fibrosis. BioMed Research International. 2015; 2015: 1–12.

[54] Quílez ME, López-Aguilar J, Blanch L. Organ crosstalk during acute lung injury, acute respiratory distress syndrome, and mechanical ventilation. Current Opinion in Critical Care. 2012; 18: 23–28.

[55] Iwasaki A, Foxman EF, Molony RD. Early local immune defences in the respiratory tract. Nature Reviews Immunology. 2017; 17: 7–20.

[56] Wong JJM, Leong JY, Lee JH, Albani S, Yeo JG. Insights into the immuno-pathogenesis of acute respiratory distress syndrome. Annals of Translational Medicine. 2019; 7: 504–504.

[57] Grommes J, Soehnlein O. Contribution of Neutrophils to Acute Lung Injury. Molecular Medicine. 2011; 17: 293–307.

[58] Johnston LK, Rims CR, Gill SE, McGuire JK, Manicone AM. Pulmonary macrophage subpopulations in the induction and resolution of acute lung injury. American Journal of Respiratory Cell and Molecular Biology. 2012; 47: 417–426.

[59] Rosseau S, Hammerl P, Maus U, Walmrath H, Schütte H, Grimminger F, et al. Phenotypic characterization of alveolar monocyte recruitment in acute respiratory distress syndrome. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2000; 279: L25–L35.

[60] Bittencourt-Mernak MI, Pinheiro NM, Santana FPR, Guerreiro MP, Saraiva-Romanholo BM, Grecco SS, et al. Prophylactic and therapeutic treatment with the flavonone sakuranetin ameliorates LPS-induced acute lung injury. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2017; 312: L217–L230.

[61] Pinheiro NM, Santana FPR, Almeida RR, Guerreiro M, Martins MA, Caperuto LC, et al. Acute lung injury is reduced by the a7nAChR agonist PNU-282987 through changes in the macrophage profile. FASEB Journal. 2017; 31: 320–332.

[62] Wang Y, Xu Y, Zhang P, Ruan W, Zhang L, Yuan S, et al. Smiglaside a ameliorates LPS-induced acute lung injury by modulating macrophage polarization via AMPK-PPARγ pathway. Biochemical Pharmacology. 2018; 156: 385–395.

[63] Zhuo Y, Li D, Cui L, Li C, Zhang S, Zhang Q, et al. Treatment with 3,4-dihydroxyphenylethyl alcohol glycoside ameliorates sepsis-induced ALI in mice by reducing inflammation and regulating M1 polarization. Biomedicine & Pharmacotherapy. 2019; 116: 109012.

[64] Mishra P, Pandey N, Pandey R, Tripathi YB. Role of macrophage po-larization in acute respiratory distress syndrome. Journal of Respiration. 2021; 1: 260–272.

[65] Wang F, Fu X, Wu X, Zhang J, Zhu J, Zou Y, et al. Bone marrow derived M 2 macrophages protected against lipopolysaccharide-induced acute lung injury through inhibiting oxidative stress and inflammation by modulating neutrophils and T lymphocytes responses. International Immunopharmacology. 2018; 61: 162–168.

[66] Chen X, Tang J, Shuai W, Meng J, Feng J, Han Z. Macrophage polarization and its role in the pathogenesis of acute lung injury/acute respiratory distress syndrome. Inflammation Research. 2020; 69: 883–895.

[67] Yang CY, Chen CS, Yiang GT, Cheng YL, Yong SB, Wu MY, et al. New insights into the immune molecular regulation of the pathogenesis of acute respiratory distress syndrome. International Journal of Molecular Sciences. 2018; 19: 588.

[68] Genschmer KR, Russell DW, Lal C, Szul T, Bratcher PE, Noerager BD, et al. Activated PMN exosomes: pathogenic entities causing matrix destruction and disease in the lung. Cell. 2019; 176: 113–126.e15.

[69] Vorobjeva NV, Chernyak BV. NETosis: molecular mechanisms, role in physiology and pathology. Biochemistry. 2020; 85: 1178–1190.

[70] Meegan JE, Yang X, Coleman DC, Jannaway M, Yuan SY. Neutrophil-mediated vascular barrier injury: role of neutrophil extracellular traps. Microcirculation. 2017; 24: e12352.

[71] Ma Y, Yang X, Chatterjee V, Meegan JE, Beard RS, Yuan SY. Role of neutrophil extracellular traps and vesicles in regulating vascular endothelial permeability. Frontiers in Immunology. 2019; 10: 1037.

[72] Thålin C, Hisada Y, Lundström S, Mackman N, Wallén H. Neutrophil extracellular traps. Arteriosclerosis, Thrombosis, and Vascular Biology. 2019; 39: 1724–1738.

[73] Zhang H, Zhou Y, Qu M, Yu Y, Chen Z, Zhu S, et al. Tissue factor-enriched neutrophil extracellular traps promote immunothrombosis and disease progression in sepsis-induced lung injury. Frontiers in Cellular and Infection Microbiology. 2021; 11: 677902.

[74] Lefrançais E, Mallavia B, Zhuo H, Calfee CS, Looney MR. Maladaptive role of neutrophil extracellular traps in pathogen-induced lung injury. JCI Insight. 2018; 3: e98178.

[75] Delano MJ, Ward PA. Sepsis-induced immune dysfunction: can immune therapies reduce mortality? Journal of Clinical Investigation. 2016; 126: 23–31.

[76] Li JT, Melton AC, Su G, Hamm DE, LaFemina M, Howard J, et al. Unexpected role for adaptive αβTh17 cells in acute respiratory distress syndrome. The Journal of Immunology. 2015; 195: 87–95.

[77] Yi X, Jian W, Di W, Jun Z, Mei Y. Prognostic values of Th17 cells level in bronchoalveolar lavage fluid in children of sepsis with acute lung injury. Chinese Journal of Contemporary Pediatrics. 2015; 17: 942–945.

[78] Holloway TL, Rani M, Cap AP, Stewart RM, Schwacha MG. The associ-ation between the Th-17 immune response and pulmonary complications in a trauma ICU population. Cytokine. 2015; 76: 328–333.

[79] Liston A, Gray DHD. Homeostatic control of regulatory T cell diversity. Nature Reviews Immunology. 2014; 14: 154–165.

[80] Lin S, Wu H, Wang C, Xiao Z, Xu F. Regulatory T cells and acute lung injury: cytokines, uncontrolled inflammation, and therapeutic implications. Frontiers in Immunology. 2018; 9: 1545.

[81] Yu Z, Ji M, Yan J, Cai Y, Liu J, Yang H, et al. The ratio of Th17/Treg cells as a risk indicator in early acute respiratory distress syndrome. Critical Care. 2015; 19: 82.

[82] Li Q, Hu X, Sun R, Tu Y, Gong F, Ni Y. Resolution acute respiratory distress syndrome through reversing the imbalance of Treg/Th17 by targeting the cAMP signaling pathway. Molecular Medicine Reports. 2016; 14: 343–348.

[83] Strukova S. Blood coagulation-dependent inflammation. Coagulation-dependent inflammation and inflammation-dependent thrombosis. Fron-tiers in Bioscience. 2006; 11: 59.

[84] Frantzeskaki F, Armaganidis A, Orfanos SE. Immunothrombosis in acute respiratory distress syndrome: cross talks between inflammation and coagulation. Respiration. 2017; 93: 212–225.

[85] Glas GJ, Van Der Sluijs KF, Schultz MJ, Hofstra J-H, Van Der Poll T, Levi M. Bronchoalveolar hemostasis in lung injury and acute respiratory distress syndrome. Journal of Thrombosis and Haemostasis. 2013; 11: 17–25.

[86] Taylor, Jr. FB. Response of anticoagulant pathways in disseminated intravascular coagulation. Seminars in Thrombosis and Hemostasis. 2001; 27: 619–632.

[87] Camprubí-Rimblas M, Tantinyà N, Bringué J, Guillamat-Prats R, Artigas A. Anticoagulant therapy in acute respiratory distress syndrome. Annals of Translational Medicine. 2018; 6: 36.

[88] Bernard GR, Vincent J, Laterre P, LaRosa SP, Dhainaut J, Lopez-Rodriguez A, et al. Efficacy and safety of recombinant human activated protein c for severe sepsis. New England Journal of Medicine. 2001; 344: 699–709.

[89] Ozolina A, Sarkele M, Sabelnikovs O, Skesters A, Jaunalksne I, Serova J, et al. Activation of coagulation and fibrinolysis in acute respiratory distress syndrome: a prospective pilot study. Frontiers in Medicine. 2016; 3: 64.

[90] Jiang J, Chou H, Wang L, Lang Y, Chen C. Effects of activated protein C on ventilator-induced lung injury in rats. Respiration. 2010; 80: 246–253.

[91] Ware LB, Fang X, Matthay MA. Protein C and thrombomodulin in human acute lung injury. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2003; 285: L514–L521.

[92] Asokananthan N, Graham PT, Fink J, Knight DA, Bakker AJ, McWilliam AS, et al. Activation of protease-activated receptor (PAR)-1, PAR-2, and PAR-4 stimulates IL-6, IL-8, and prostaglandin E 2 release from human respiratory epithelial cells. The Journal of Immunology. 2002; 168: 3577–3585.

[93] Camprubí-Rimblas M, Tantinyà N, Guillamat-Prats R, Bringué J, Puig F, Gómez MN, et al. Effects of nebulized antithrombin and heparin on inflammatory and coagulation alterations in an acute lung injury model in rats. Journal of Thrombosis and Haemostasis. 2020; 18: 571–583.

[94] Sabharwal AK, Bajaj SP, Ameri A, Tricomi SM, Hyers TM, Dahms TE, et al. Tissue factor pathway inhibitor and von Willebrand factor antigen levels in adult respiratory distress syndrome and in a primate model of sepsis. American Journal of Respiratory and Critical Care Medicine. 1995; 151: 758–767.

[95] Gando S, Otomo Y. Local hemostasis, immunothrombosis, and systemic disseminated intravascular coagulation in trauma and traumatic shock. Critical Care. 2015; 19: 72.

[96] Haitsma JJ, Schultz MJ, Hofstra JJH, Kuiper JW, Juco J, Vaschetto R, et al. Ventilator-induced coagulopathy in experimental Streptococcus pneumoniae pneumonia. European Respiratory Journal. 2008; 32: 1599–1606.

[97] Hrenak J, Simko F. Renin-angiotensin system: an important player in the pathogenesis of acute respiratory distress syndrome. Int International Journal of Molecular Sciences. 2020; 21: 8038.

[98] Carvalho AC, Quinn DA, Demarinis SM, Beitz JG, Zapol WM. Platelet function in acute respiratory failure. American Journal of Hematology. 1987; 25: 377–388.

[99] Idell S, Maunder R, Fein AM, Switalska HI, Tuszynski GF, McLarty J, et al. Platelet-specific α-granule proteins and thrombospondin in bronchoalveolar lavage in the adult respiratory distress syndrome. Chest. 1989; 96: 1125–1132.

[100] Wijten P, van Holten T, Woo LL, Bleijerveld OB, Roest M, Heck AJR, et al. High precision platelet releasate definition by quantitative reversed protein profiling—brief report. Arteriosclerosis, Thrombosis, and Vascular Biology. 2013; 33: 1635–1638.

[101] Puneet P, Moochhala S, Bhatia M. Chemokines in acute respiratory distress syndrome. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2005; 288: L3–L15.

[102] Golebiewska EM, Poole AW. Platelet secretion: from haemostasis to wound healing and beyond. Blood Reviews. 2015; 29: 153–162.

[103] Yadav H, Kor DJ. Platelets in the pathogenesis of acute respiratory distress syndrome. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2015; 309: L915–L923.

[104] de Bont CM, Boelens WC, Pruijn GJM. NETosis, complement, and co-agulation: a triangular relationship. Cellular & Molecular Immunology. 2019; 16: 19–27.

[105] Kaplan MJ, Radic M. Neutrophil extracellular traps: double-edged swords of innate immunity. The Journal of Immunology. 2012; 189: 2689–2695.

[106] Song C, Li H, Li Y, Dai M, Zhang L, Liu S, et al. NETs promote ALI/ARDS inflammation by regulating alveolar macrophage polarization. Experimental Cell Research. 2019; 382: 111486.

[107] Zarbock A, Singbartl K, Ley K. Complete reversal of acid-induced acute lung injury by blocking of platelet-neutrophil aggregation. Journal of Clinical Investigation. 2006; 116: 3211–3219.

[108] Camerer E, Huang W, Coughlin SR. Tissue factor- and factor X-dependent activation of protease-activated receptor 2 by factor VIIa. Proceedings of the National Academy of Sciences. 2000; 97: 5255–5260.

[109] Ruf W, Dorfleutner A, Riewald M. Specificity of coagulation factor signaling. Journal of Thrombosis and Haemostasis. 2003; 1: 1495–1503.

[110] Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nature Reviews Immunology. 2013; 13: 34–45.

[111] Choi G, Schultz MJ, Levi M, van der Poll T. The relationship between inflammation and the coagulation system. Swiss Medical Weekly. 2006; 136: 139–144.

[112] Bester J, Pretorius E. Effects of IL-1β, IL-6 and IL-8 on erythrocytes, platelets and clot viscoelasticity. Scientific Reports. 2016; 6: 32188.

[113] Livingstone SA, Wildi KS, Dalton HJ, Usman A, Ki KK, Passmore MR, et al. Coagulation dysfunction in acute respiratory distress syndrome and its potential impact in inflammatory subphenotypes. Frontiers in Medicine. 2021; 8: 723217.

[114] Gupta N, Zhao YY, Evans CE. The stimulation of thrombosis by hypoxia. Thrombosis Research. 2019; 181: 77–83.

[115] Chan CK, Vanhoutte PM. Hypoxia, vascular smooth muscles and endothelium. Acta Pharmaceutica Sinica B. 2013; 3: 1–7.

[116] Palazon A, Goldrath A, Nizet V, Johnson R. HIF transcription factors, inflammation, and immunity. Immunity. 2014; 41: 518–528.

[117] Raposo G, Stoorvogel W. Extracellular vesicles: Exosomes, microvesi-cles, and friends. Journal of Cell Biology. 2013; 200: 373–383.

[118] Esquivel-Ruiz S, González-Rodríguez P, Lorente JA, Pérez-Vizcaíno F, Herrero R, Moreno L. Extracellular Vesicles and alveolar epithelial-capillary barrier disruption in acute respiratory distress syndrome: patho-physiological role and therapeutic potential. Frontiers in Physiology. 2021; 12: 752287.

[119] Wang L, Luo H, Chen X, Jiang Y, Huang Q. Functional characterization of S100A8 and S100A9 in altering monolayer permeability of human umbilical endothelial cells. PLoS ONE. 2014; 9: e90472.

[120] Mitra S, Wewers MD, Sarkar A. Mononuclear phagocyte-derived microparticulate caspase-1 induces pulmonary vascular endothelial cell injury. PLoS ONE. 2015; 10: e0145607.

[121] Neudecker V, Brodsky KS, Clambey ET, Schmidt EP, Packard TA, Davenport B, et al. Neutrophil transfer of miR-223 to lung epithelial cells dampens acute lung injury in mice. Science Translational Medicine. 2017; 9: eaah5360.

[122] Combes V, Simon A, Grau G, Arnoux D, Camoin L, Sabatier F, et al. In vitro generation of endothelial microparticles and possible prothrombotic activity in patients with lupus anticoagulant. Journal of Clinical Investigation. 1999; 104: 93–102.

[123] Kulshreshtha A, Ahmad T, Agrawal A, Ghosh B. Proinflammatory role of epithelial cell–derived exosomes in allergic airway inflammation. Journal of Allergy and Clinical Immunology. 2013; 131: 1194–1203.e14.

[124] Iba T, Ogura H. Role of extracellular vesicles in the development of sepsis-induced coagulopathy. Journal of Intensive Care. 2018; 6: 68.

[125] Lu P, Takai K, Weaver VM, Werb Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harbor Perspectives in Biology. 2011; 3: a005058–a005058.

[126] Moon H, Kim S, Gao J, Quan T, Qin Z, Osorio JC, et al. CCN1 secretion and cleavage regulate the lung epithelial cell functions after cigarette smoke. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2014; 307: L326–L337.

[127] Moon H, Cao Y, Yang J, Lee JH, Choi HS, Jin Y. Lung epithelial cell-derived extracellular vesicles activate macrophage-mediated inflamma-tory responses via ROCK1 pathway. Cell Death & Disease. 2015; 6: e2016–e2016.

[128] Lee H, Zhang D, Wu J, Otterbein LE, Jin Y. Lung epithelial cell-derived microvesicles regulate macrophage migration via MicroRNA-17/221-induced integrin β 1 recycling. Journal of Immunology. 2017; 199: 1453–1464.

[129] Ismail N, Wang Y, Dakhlallah D, Moldovan L, Agarwal K, Batte K, et al. Macrophage microvesicles induce macrophage differentiation and miR-223 transfer. Blood. 2013; 121: 984–995.

[130] Tamari T, Brenner B, Aharon A. Monocyte-derived microparticles and exosomes induce procoagulant and apoptotic effects on endothelial cells. Thrombosis and Haemostasis. 2008; 100: 878–885.

[131] Li L, Cheng Y, Emrich S, Schorey J. Activation of endothelial cells by extracellular vesicles derived from Mycobacterium tuberculosis infected macrophages or mice. PLoS ONE. 2018; 13: e0198337.

[132] Wang J, Williams JC, Davis BK, Jacobson K, Doerschuk CM, Ting JP-, et al. Monocytic microparticles activate endothelial cells in an IL-1β–dependent manner. Blood. 2011; 118: 2366–2374.

[133] Buesing KL, Densmore JC, Kaul S, Pritchard KA, Jarzembowski JA, Gourlay DM, et al. Endothelial microparticles induce inflammation in acute lung injury. Journal of Surgical Research. 2011; 166: 32–39.

[134] Sun X, Singleton PA, Letsiou E, Zhao J, Belvitch P, Sammani S, et al. Sphingosine-1–phosphate receptor-3 is a novel biomarker in acute lung injury. American Journal of Respiratory Cell and Molecular Biology. 2012; 47: 628–636.

[135] Li H, Meng X, Liang X, Gao Y, Cai S. Administration of microparticles from blood of the lipopolysaccharide-treated rats serves to induce pathologic changes of acute respiratory distress syndrome. Experimental Biology and Medicine. 2015; 240: 1735–1741.

[136] Chatterjee V, Yang X, Ma Y, Wu MH, Yuan SY. Extracellular vesicles: new players in regulating vascular barrier function. American Journal of Physiology-Heart and Circulatory Physiology. 2020; 319: H1181–H1196.

[137] Chatterjee V, Yang X, Ma Y, Cha B, Meegan JE, Wu M, et al. En-dothelial microvesicles carrying Src-rich cargo impair adherens junction integrity and cytoskeleton homeostasis. Cardiovascular Research. 2020; 116: 1525–1538.

[138] Hosseinkhani B, Kuypers S, van den Akker NMS, Molin DGM, Michiels L. Extracellular vesicles work as a functional inflammatory mediator between vascular endothelial cells and immune cells. Frontiers in Immunology. 2018; 9: 1789.

[139] Matsumoto H, Yamakawa K, Ogura H, Koh T, Matsumoto N, Shimazu

T. Enhanced expression of cell-specific surface antigens on endothelial microparticles in sepsis-induced disseminated intravascular coagulation. Shock. 2015; 43: 443–449.

[140] Rondina MT, Tatsumi K, Bastarache JA, Mackman N. Microvesicle tissue factor activity and interleukin-8 levels are associated with mortality in patients with influenza a/H1N1 infection. Critical Care Medicine. 2016; 44: e574–e578.

[141] Rosell A, Havervall S, von Meijenfeldt F, Hisada Y, Aguilera K, Grover SP, et al. Patients with COVID-19 have elevated levels of circulating extracellular vesicle tissue factor activity that is associated with severity and mortality—brief report. Arteriosclerosis, Thrombosis, and Vascular Biology. 2021; 41: 878–882.

[142] Hong CW. Extracellular vesicles of neutrophils. Immune Network. 2018; 18: e43.

[143] Kolonics F, Szeifert V, Timár CI, Ligeti E, Lőrincz ÁM. The functional heterogeneity of neutrophil-derived extracellular vesicles reflects the status of the parent cell. Cells. 2020; 9: 2718.

[144] Youn Y, Shrestha S, Lee Y, Kim J, Lee JH, Hur K, et al. Neutrophil-derived trail is a proinflammatory subtype of neutrophil-derived extra-cellular vesicles. Theranostics. 2021; 11: 2770–2787.

[145] Mesri M, Altieri DC. Endothelial cell activation by leukocyte micropar-ticles. Journal of Immunology. 1998; 161: 4382–4387.

[146] Mesri M, Altieri DC. Leukocyte microparticles stimulate endothelial cell cytokine release and tissue factor induction in a jnk1 signaling pathway. Journal of Biological Chemistry. 1999; 274: 23111–23118.

[147] Dalli J, Montero-Melendez T, Norling LV, Yin X, Hinds C, Haskard D, et al. Heterogeneity in neutrophil microparticles reveals distinct proteome and functional properties. Molecular & Cellular Proteomics. 2013; 12: 2205–2219.

[148] Rossaint J, Kühne K, Skupski J, Van Aken H, Looney MR, Hidalgo A, et al. Directed transport of neutrophil-derived extracellular vesicles enables platelet-mediated innate immune response. Nature Communications. 2016; 7: 13464.

[149] Barry OP, Pratico D, Lawson JA, FitzGerald GA. Transcellular activation of platelets and endothelial cells by bioactive lipids in platelet microparticles. Journal of Clinical Investigation. 1997; 99: 2118–2127.

[150] Nomura S, Tandon NN, Nakamura T, Cone J, Fukuhara S, Kambayashi J. High-shear-stress-induced activation of platelets and microparticles enhances expression of cell adhesion molecules in THP-1 and endothelial cells. Atherosclerosis. 2001; 158: 277–287.

[151] Chiva-Blanch G, Padró T, Alonso R, Crespo J, Perez de Isla L, Mata P, et al. Liquid biopsy of extracellular microvesicles maps coronary calcification and atherosclerotic plaque in asymptomatic patients with fa-milial hypercholesterolemia. Arteriosclerosis, Thrombosis, and Vascular Biology. 2019; 39: 945–955.

[152] Oggero S, Austin-Williams S, Norling LV. The contrasting role of extracellular vesicles in vascular inflammation and tissue repair. Frontiers in Pharmacology. 2019; 10: 1479.

[153] de Freitas RCC, Hirata RDC, Hirata MH, Aikawa E. Circulating extracellular vesicles as biomarkers and drug delivery vehicles in cardiovascular diseases. Biomolecules. 2021; 11: 388.

[154] Hottz ED, Lopes JF, Freitas C, Valls-De-Souza R, Oliveira MF, Bozza MT, et al. Platelets mediate increased endothelium permeability in dengue through NLRP3-inflammasome activation. Blood. 2013; 122: 3405–3414.

[155] Bao H, Yao QP, Huang K, Chen XH, Han Y, Jiang ZL, et al. Platelet-derived miR-142-3p induces apoptosis of endothelial cells in hypertension. Cellular and Molecular Biology. 2017; 63: 3–9.

[156] Nieuwland R, Berckmans RJ, McGregor S, Böing AN, Th. M. Romijn FPH, Westendorp RGJ, et al. Cellular origin and procoagulant properties of microparticles in meningococcal sepsis. Blood. 2000; 95: 930–935.

[157] Tripisciano C, Weiss R, Eichhorn T, Spittler A, Heuser T, Fischer MB, et al. Different potential of extracellular vesicles to support thrombin generation: contributions of phosphatidylserine, tissue factor, and cellular origin. Scientific Reports. 2017; 7: 6522.

[158] Puhm F, Boilard E, MacHlus KR. Platelet extracellular vesicles; beyond the blood. Arteriosclerosis, Thrombosis, and Vascular Biology. 2021; 41: 87–96.

[159] Darmon M, Clec’h C, Adrie C, Argaud L, Allaouchiche B, Azoulay E, et al. Acute respiratory distress syndrome and risk of aki among critically ill patients. Clinical Journal of the American Society of Nephrology. 2014; 9: 1347–1353.

[160] Del Sorbo L, Slutsky AS. Acute respiratory distress syndrome and multiple organ failure. Current Opinion in Critical Care. 2011; 17: 1–6.

[161] Matuschak GM. Liver-lung interactions in critical illness. Update in Intensive Care and Emergency Medicine. 1995; 256: 73–93.

[162] Kallet RH, Lipnick MS, Zhuo H, Pangilinan LP, Gomez A. Characteris-tics of nonpulmonary organ dysfunction at onset of ARDS based on the berlin definition. Respiratory Care. 2019; 64: 493–501.

[163] Trefts E, Gannon M, Wasserman DH. The liver. Current Biology. 2017; 27: R1147–R1151.

[164] Yang P, Formanek P, Scaglione S, Afshar M. Risk factors and outcomes of acute respiratory distress syndrome in critically ill patients with cirrhosis. Hepatology Research. 2019; 49: 335–343.

[165] Dong V, Sun K, Gottfried M, Cardoso FS, McPhail MJ, Stravitz RT, et al. Significant lung injury and its prognostic significance in acute liver failure: a cohort analysis. Liver International. 2020; 40: 654–663.

[166] Dizier S, Forel JM, Ayzac L, Richard JC, Hraiech S, Lehingue S, et al. Early hepatic dysfunction is associated with a worse outcome in patients presenting with acute respiratory distress syndrome: a post-hoc analysis of the ACURASYS and PROSEVA studies. PLoS ONE. 2015; 10: e0144278.

[167] Herrero R, Sánchez G, Asensio I, López E, Ferruelo A, Vaquero J, et al. Liver–lung interactions in acute respiratory distress syndrome. Intensive Care Medicine Experimental. 2020; 8: 48.

[168] Bilzer M, Roggel F, Gerbes AL. Role of Kupffer cells in host defense and liver disease. Liver International. 2006; 26: 1175–1186.

[169] Triantafyllou E, Woollard KJ, McPhail MJW, Antoniades CG, Possamai LA. The role of monocytes and macrophages in acute and acute-on-chronic liver failure. Frontiers in Immunology. 2018; 9: 2948.

[170] Wiedermann FJ, Mayr AJ, Kaneider NC, Fuchs D, Mutz NJ, Schobers-berger W. Alveolar granulocyte colony-stimulating factor and alpha-chemokines in relation to serum levels, pulmonary neutrophilia, and severity of lung injury in ARDS. Chest. 2004; 125: 212–219.

[171] Kobayashi K, Horikami D, Omori K, Nakamura T, Yamazaki A, Maeda S, et al. Thromboxane a2 exacerbates acute lung injury via promoting edema formation. Scientific Reports. 2016; 6: 32109.

[172] Panos RJ, Baker SK. Mediators, cytokines, and growth factors in liver-lung interactions. Clinics in Chest Medicine. 1996; 17: 151–169.

[173] Callery MP. Organ Interactions in Sepsis. Archives of Surgery. 1991; 126: 28.

[174] Matuschak GM. Lung-liver interactions in sepsis and multiple organ failure syndrome. Clinics in Chest Medicine. 1996; 17: 83–98.

[175] Salatti Ferrari R, da Rosa DP, Forgiarini LF, Bona S, Simões Dias A, Marroni NP. Oxidative stress and pulmonary changes in experimental liver cirrhosis. Oxidative Medicine and Cellular Longevity. 2012; 2012: 1–8.

[176] Ferrari RS, Tieppo M, Rosa DPD, Forgiarini JR LA, Dias AS, Marroni NP. Lung and liver changes due to the induction of cirrhosis in two experimental models. Archives of Gastroenterology. 2013; 50: 208–213.

[177] Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. New England Journal of Medicine. 1999; 340: 448–454.

[178] Quinton LJ, Jones MR, Robson BE, Mizgerd JP. Mechanisms of the hepatic acute-phase response during bacterial pneumonia. Infection and Immunity. 2009; 77: 2417–2426.

[179] Quinton LJ, Blahna MT, Jones MR, Allen E, Ferrari JD, Hilliard KL, et al. Hepatocyte-specific mutation of both NF-κB RelA and STAT3 abrogates the acute phase response in mice. Journal of Clinical Investigation. 2012; 122: 1758–1763.

[180] Hilliard KL, Allen E, Traber KE, Yamamoto K, Stauffer NM, Wasserman GA, et al. The lung-liver axis: a requirement for maximal innate immunity and hepatoprotection during pneumonia. American Journal of Respiratory Cell and Molecular Biology. 2015; 53: 378–390.

[181] Frank JA, Wray CM, McAuley DF, Schwendener R, Matthay MA. Alveolar macrophages contribute to alveolar barrier dysfunction in ventilator-induced lung injury. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2006; 291: L1191–L1198.

[182] Dani C, Martelli E, Tronchin M, Buonocore G, Longini M, Di Filippo A, et al. Bilirubin influence on oxidative lung damage and surfactant surface tension properties. Pediatric Pulmonology. 2004; 38: 179–185.

[183] Rodrigues CMP, Solá S, Brito MA, Brites D, Moura JJG. Bilirubin directly disrupts membrane lipid polarity and fluidity, protein order, and redox status in rat mitochondria. Journal of Hepatology. 2002; 36: 335–341.

[184] Fernandes A, Falcão AS, Silva RFM, Gordo AC, Gama MJ, Brito MA, et al. Inflammatory signalling pathways involved in astroglial activation by unconjugated bilirubin. Journal of Neurochemistry. 2006; 96: 1667–1679.

[185] Mohan A, Agarwal S, Clauss M, Britt NS, Dhillon NK. Extracellular vesicles: novel communicators in lung diseases. Respiratory Research. 2020; 21: 175.

[186] Chacón-Aponte AA, Durán-Vargas ÉA, Arévalo-Carrillo JA, Lozada-Martínez ID, Bolaño-Romero MP, Moscote-Salazar LR, et al. Brain-lung interaction: a vicious cycle in traumatic brain injury. Acute and Critical Care. 2022; 37: 35–44.

[187] Mikkelsen ME, Christie JD, Lanken PN, Biester RC, Thompson BT, Bellamy SL, et al. The adult respiratory distress syndrome cognitive outcomes study. American Journal of Respiratory and Critical Care Medicine. 2012; 185: 1307–1315.

[188] Bassi TG, Rohrs EC, Reynolds SC. Systematic review of cognitive impairment and brain insult after mechanical ventilation. Critical Care. 2021; 25: 99.

[189] Fries M, Bickenbach J, Henzler D, Beckers S, Dembinski R, Sellhaus B, et al. S-100 protein and neurohistopathologic changes in a porcine model of acute lung injury. Anesthesiology. 2005; 102: 761–767.

[190] Bernardo-Castro S, Sousa JA, Brás A, Cecília C, Rodrigues B, Almendra L, et al. Pathophysiology of blood-brain barrier permeability throughout the different stages of ischemic stroke and its implication on hemorrhagic transformation and recovery. Frontiers in Neurology. 2020; 11: 594672.

[191] Galea I. The blood–brain barrier in systemic infection and inflammation. Cellular & Molecular Immunology. 2021; 18: 2489–2501.

[192] Santos CCD, Shan Y, Akram A, Slutsky AS, Haitsma JJ. Neuroimmune regulation of ventilator-induced lung injury. American Journal of Respiratory and Critical Care Medicine. 2011; 183: 471–482.

[193] Ko GJ, Rabb H, Hassoun HT. Kidney-lung crosstalk in the critically ill patient. Blood Purification. 2009; 28: 75–83.

[194] Koyner JL, Murray PT. Mechanical ventilation and the kidney. Blood Purification. 2010; 29: 52–68.

[195] Murdaugh HV, Sieker HO, Manfredi F. Effect of altered intrathoracic pressure on renal hemodynamics, electrolyte excretion and water clearance. Journal of Clinical Investigation. 1959; 38: 834–842.

[196] Kuiper JW, Groeneveld ABJ, Slutsky AS, Plötz FB. Mechanical ventilation and acute renal failure. Critical Care Medicine. 2005; 33: 1408–1415.

[197] Moore ES, Galvez MB, Paton JB, Fisher DE, Behrman RE. Effects of positive pressure ventilation on intrarenal blood flow in infant primates. Pediatric Research. 1974; 8: 792–796.

[198] Priebe HJ, Heimann JC, Hedley-Whyte J. Mechanisms of renal dysfunction during positive end-expiratory pressure ventilation. Journal of Applied Physiology. 1981; 50: 643–649.

[199] Choi WI, Quinn DA, Park KM, Moufarrej RK, Jafari B, Syrkina O, et al. Systemic microvascular leak in an in vivo rat model of ventilator-induced lung injury. American Journal of Respiratory and Critical Care Medicine. 2003; 167: 1627–1632.

[200] Breslin JW, Pappas PJ, Cerveira JJ, Hobson RW, Durán WN. VEGF increases endothelial permeability by separate signaling pathways involving ERK-1/2 and nitric oxide. American Journal of Physiology-Heart and Circulatory Physiology. 2003; 284: H92–H100.

[201] Gurkan OU, O’Donnell C, Brower R, Ruckdeschel E, Becker PM. Differential effects of mechanical ventilatory strategy on lung injury and systemic organ inflammation in mice. American Journal of Physiology-Lung Cellular and Molecular Physiology. 2003; 285: L710–L718.

[202] Imai Y. Injurious mechanical ventilation and end-organ epithelial cell apoptosis and organ dysfunction in an experimental model of acute respiratory distress syndrome. JAMA. 2003; 289: 2104.

[203] Semama DS, Thonney M, Guignard JP. Does endothelin-1 mediate the hypoxemia-induced renal dysfunction in newborn rabbits? Biology of the Neonate. 1995; 67: 216–222.

[204] Ballèvre L, Thonney M, Guignard J-. Role of nitric oxide in the hypoxemia-induced renal dysfunction of the newborn rabbit. Pediatric Research. 1996; 39: 725–730.

[205] Hotter G, Palacios L, Sola A. Low O2 and high CO2 in LLC-PK1 cells culture mimics renal ischemia-induced apoptosis. Laboratory Investigation. 2004; 84: 213–220.

[206] Rabb H, Wang Z, Nemoto T, Hotchkiss J, Yokota N, Soleimani M. Acute renal failure leads to dysregulation of lung salt and water channels. Kidney International. 2003; 63: 600–606.

[207] Hoke TS, Douglas IS, Klein CL, He Z, Fang W, Thurman JM, et al. Acute renal failure after bilateral nephrectomy is associated with cytokine-mediated pulmonary injury. Journal of the American Society of Nephrology. 2007; 18: 155–164.

[208] Klein CL, Hoke TS, Fang WF, Altmann CJ, Douglas IS, Faubel S. Interleukin-6 mediates lung injury following ischemic acute kidney injury or bilateral nephrectomy. Kidney International. 2008; 74: 901–909.



Abstracted / indexed in

Science Citation Index Expanded (SciSearch) Created as SCI in 1964, Science Citation Index Expanded now indexes over 9,200 of the world’s most impactful journals across 178 scientific disciplines. More than 53 million records and 1.18 billion cited references date back from 1900 to present.

Journal Citation Reports/Science Edition Journal Citation Reports/Science Edition aims to evaluate a journal’s value from multiple perspectives including the journal impact factor, descriptive data about a journal’s open access content as well as contributing authors, and provide readers a transparent and publisher-neutral data & statistics information about the journal.

Chemical Abstracts Service Source Index The CAS Source Index (CASSI) Search Tool is an online resource that can quickly identify or confirm journal titles and abbreviations for publications indexed by CAS since 1907, including serial and non-serial scientific and technical publications.

Index Copernicus The Index Copernicus International (ICI) Journals database’s is an international indexation database of scientific journals. It covered international scientific journals which divided into general information, contents of individual issues, detailed bibliography (references) sections for every publication, as well as full texts of publications in the form of attached files (optional). For now, there are more than 58,000 scientific journals registered at ICI.

Geneva Foundation for Medical Education and Research The Geneva Foundation for Medical Education and Research (GFMER) is a non-profit organization established in 2002 and it works in close collaboration with the World Health Organization (WHO). The overall objectives of the Foundation are to promote and develop health education and research programs.

Scopus: CiteScore 1.0 (2022) Scopus is Elsevier's abstract and citation database launched in 2004. Scopus covers nearly 36,377 titles (22,794 active titles and 13,583 Inactive titles) from approximately 11,678 publishers, of which 34,346 are peer-reviewed journals in top-level subject fields: life sciences, social sciences, physical sciences and health sciences.

Embase Embase (often styled EMBASE for Excerpta Medica dataBASE), produced by Elsevier, is a biomedical and pharmacological database of published literature designed to support information managers and pharmacovigilance in complying with the regulatory requirements of a licensed drug.

Submission Turnaround Time

Conferences

Top