1
|
Rathod SG, Latif R. Pediatric Ischemic Stroke Following Scorpion Sting: Correspondence. Indian J Pediatr 2024; 91:1219. [PMID: 39174747 DOI: 10.1007/s12098-024-05250-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024]
Affiliation(s)
- Santosh Govind Rathod
- Department of Clinical Hematology, Dr D Y Patil Medical College, Hospital and Research, Dr D Y Patil Vidyapeeth, Deemed to be University, Pune, Maharastra, India.
| | - Roman Latif
- Department of Pathology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
2
|
Silva-Lance F, Montejano-Montelongo I, Bautista E, Nielsen LK, Johansson PI, Marin de Mas I. Integrating Genome-Scale Metabolic Models with Patient Plasma Metabolome to Study Endothelial Metabolism In Situ. Int J Mol Sci 2024; 25:5406. [PMID: 38791446 PMCID: PMC11121795 DOI: 10.3390/ijms25105406] [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: 03/01/2024] [Revised: 04/30/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Patient blood samples are invaluable in clinical omics databases, yet current methodologies often fail to fully uncover the molecular mechanisms driving patient pathology. While genome-scale metabolic models (GEMs) show promise in systems medicine by integrating various omics data, having only exometabolomic data remains a limiting factor. To address this gap, we introduce a comprehensive pipeline integrating GEMs with patient plasma metabolome. This pipeline constructs case-specific GEMs using literature-based and patient-specific metabolomic data. Novel computational methods, including adaptive sampling and an in-house developed algorithm for the rational exploration of the sampled space of solutions, enhance integration accuracy while improving computational performance. Model characterization involves task analysis in combination with clustering methods to identify critical cellular functions. The new pipeline was applied to a cohort of trauma patients to investigate shock-induced endotheliopathy using patient plasma metabolome data. By analyzing endothelial cell metabolism comprehensively, the pipeline identified critical therapeutic targets and biomarkers that can potentially contribute to the development of therapeutic strategies. Our study demonstrates the efficacy of integrating patient plasma metabolome data into computational models to analyze endothelial cell metabolism in disease contexts. This approach offers a deeper understanding of metabolic dysregulations and provides insights into diseases with metabolic components and potential treatments.
Collapse
Affiliation(s)
- Fernando Silva-Lance
- Novo Nordisk Foundation Center for Biosustainability, Danish Technical University, 2800 Lyngby, Denmark
| | | | - Eric Bautista
- Novo Nordisk Foundation Center for Biosustainability, Danish Technical University, 2800 Lyngby, Denmark
| | - Lars K. Nielsen
- Novo Nordisk Foundation Center for Biosustainability, Danish Technical University, 2800 Lyngby, Denmark
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Pär I. Johansson
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Igor Marin de Mas
- Novo Nordisk Foundation Center for Biosustainability, Danish Technical University, 2800 Lyngby, Denmark
- CAG Center for Endotheliomics, Copenhagen University Hospital, Rigshospitalet, 2100 Copenhagen, Denmark
| |
Collapse
|
3
|
Gouchoe DA, Whitson BA, Zhu H. The Next Frontier in Lung Transplantation: Protecting the Endothelium and Repairing Organs for Transplant Utilizing MG53. CLINICAL AND TRANSLATIONAL DISCOVERY 2023; 3:e255. [PMID: 38774634 PMCID: PMC11104552 DOI: 10.1002/ctd2.255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 11/05/2023] [Indexed: 05/24/2024]
Affiliation(s)
- Doug A. Gouchoe
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, The United States
| | - Bryan A. Whitson
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, The United States
| | - Hua Zhu
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, The United States
| |
Collapse
|
4
|
Nassef NAA, Abd-El Hamid MS, Abusikkien SA, Ahmed AI. Quercetin ameliorates acute lung injury in a rat model of hepatopulmonary syndrome. BMC Complement Med Ther 2022; 22:320. [PMID: 36463144 PMCID: PMC9719635 DOI: 10.1186/s12906-022-03785-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/09/2022] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND Common bile duct ligation (BDL) is a rat experimental model to induce biliary cirrhosis. Lung fibrosis and pulmonary vascular angiogenesis and congestion are the most common complications of biliary cirrhosis that is known as hepatopulmonary syndrome. The aim of the present work is to investigate the acute lung injury in a BDL model and to investigate the possible protective effect of quercetin on this injury. METHODS Twenty-four adult male albino rats of the Wister strain (weighing 150-250 g). Animals were divided into 3 groups, with 8 rats each: Group I: Sham-operated group (control). Group II: Bile duct ligation group (BDL) sacrificed after 28 days from the surgery. Group III: Quercetin-treated bile duct ligation group (Q-BDL) was given orally by gastric gavage in a dose of 50 mg/kg/day, starting from the 4th day of the operation until the 28th day. At the end of the experiment, at day 28, all rats were sacrificed. Lung specimens were processed to measure Endothelin B receptor gene expression by PCR, lung surfactant by ELISA, "eNO" s by immunohistochemistry. Histological assessment was done using; H&E, Masson's trichrome, PAS, toluidine blue-stained semi-thin sections, transmission electron microscope. Histomorphometric and statistical studies were done. RESULTS BDL group showed significant increase in lung index together with mononuclear cellular infiltration denoting lung inflammatory state. Also, the significant increase in pulmonary endothelial nitric oxide synthase ("eNO" s) area percent and endothelin B receptor (ETB) gene expression indicates enhanced angiogenesis. Pulmonary surfactant concentration was significantly decreased together with thickening of interalveolar septa denoting lung injury and fibrosis. Quercetin led to significant decrease in lung index, pulmonary "eNO" s area percent, ETB gene expression and significant increase in pulmonary surfactant concentration. Quercetin treatment improved histological changes and morphometric measurements, limited mononuclear cellular infiltration and decreased perivascular and perialveolar collagen deposition. CONCLUSION Quercetin ameliorates the hepatopulmonary syndrome-induced lung injury through its anti-inflammatory, antioxidative and antifibrotic effects.
Collapse
Affiliation(s)
- Noha Abdel-Aziz Nassef
- grid.7269.a0000 0004 0621 1570Assistant Professor of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Manal S. Abd-El Hamid
- grid.7269.a0000 0004 0621 1570Assistant Professor of Physiology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Samy A. Abusikkien
- grid.7269.a0000 0004 0621 1570Lecturer of Anatomy, Rabigh Faculty of Medicine, King Abdulaziz University, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Asmaa Ibrahim Ahmed
- grid.7269.a0000 0004 0621 1570Assistant Professor of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
5
|
Belhaj A, Dewachter L, Hupkens E, Remmelink M, Galanti L, Rorive S, Melot C, Naeije R, Rondelet B. Tacrolimus Prevents Mechanical and Humoral Alterations in Brain Death-Induced Lung Injury in Pigs. Am J Respir Crit Care Med 2022; 206:584-595. [PMID: 35549669 DOI: 10.1164/rccm.202201-0033oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Donor brain death-induced lung injury may compromise graft function after transplantation. Establishing strategies to attenuate lung damage remains a challenge because the underlying mechanisms remain uncertain. OBJECTIVES The effects of tacrolimus pretreatment were evaluated in an experimental model of brain death-induced lung injury. METHODS Brain death was induced by slow intracranial infusion of blood in anesthetized pigs after randomization to tacrolimus (orally administered at 0.25 mg. kg-1 BID the day before the experiment and intravenously at 0.05 mg. kg-1 one hour before the experiment; n=8) or placebo (n=9) pretreatment. Hemodynamic measurements were performed 1, 3, 5 and 7 hours after brain death. After euthanasia of the animals, lung tissue was sampled for pathobiological and histological analysis, including lung injury scoring (LIS). MEASUREMENTS AND MAIN RESULTS Tacrolimus pretreatment prevented increases in pulmonary artery pressure, pulmonary vascular resistance and pulmonary capillary pressure and decreases in systemic artery pressure and thermodilution cardiac output associated with brain death. After brain death, the ratio of the partial arterial O2 pressure to the inspired O2 fraction (PaO2/FiO2) decreased, which was prevented by tacrolimus. Tacrolimus pretreatment prevented increases in the interleukin (IL)-6-to-IL-10 ratio, vascular cell adhesion molecule-1, circulating levels of IL-1β, IL-6-to-IL-10 ratio and glycocalyx-derived molecules. Tacrolimus partially decreased apoptosis [Bax-to-Bcl2 ratio (p=0.07) and the number of apoptotic cells in the lungs (p<0.05)] but failed to improve LIS. CONCLUSIONS Immunomodulation through tacrolimus pretreatment prevented pulmonary capillary hypertension as well as the activation of inflammatory and apoptotic processes in the lungs after brain death; however, LIS did not improve.
Collapse
Affiliation(s)
- Asmae Belhaj
- CHU UCL Namur, 82470, cardiovascular, thoracic surgery and lung transplantation, Yvoir, Belgium.,Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium;
| | - Laurence Dewachter
- Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium
| | - Emeline Hupkens
- Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium
| | - Myriam Remmelink
- Université Libre de Bruxelles, 26659, Department of Pathology, Hôpital Erasme, Brussels, Belgium
| | - Laurence Galanti
- CHU UCL Namur, 82470, Department of Clinical Biology, Yvoir, Belgium
| | - Sandrine Rorive
- Université Libre de Bruxelles, 26659, Department of Pathology, Hôpital Erasme, Brussels, Belgium
| | - Christian Melot
- Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium
| | - Robert Naeije
- Department of Pathophysiology, Free University of Brussels, Brussels, Belgium
| | - Benoît Rondelet
- CHU UCL Namur, 82470, cardiovascular, thoracic surgery and lung transplantation, Yvoir, Belgium.,Université Libre de Bruxelles, 26659, Laboratory of Physiology and Pharmacology, Faculty of Medicine, Bruxelles, Belgium
| |
Collapse
|
6
|
Tsai HY, Hsu YJ, Lu CY, Tsai MC, Hung WC, Chen PC, Wang JC, Hsu LA, Yeh YH, Chu P, Tsai SH. Pharmacological Activation Of Aldehyde Dehydrogenase 2 Protects Against Heatstroke-Induced Acute Lung Injury by Modulating Oxidative Stress and Endothelial Dysfunction. Front Immunol 2021; 12:740562. [PMID: 34764958 PMCID: PMC8576434 DOI: 10.3389/fimmu.2021.740562] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
Heatstroke (HS) can cause acute lung injury (ALI). Heat stress induces inflammation and apoptosis via reactive oxygen species (ROS) and endogenous reactive aldehydes. Endothelial dysfunction also plays a crucial role in HS-induced ALI. Aldehyde dehydrogenase 2 (ALDH2) is a mitochondrial enzyme that detoxifies aldehydes such as 4-hydroxy-2-nonenal (4-HNE) protein adducts. A single point mutation in ALDH2 at E487K (ALDH2*2) intrinsically lowers the activity of ALDH2. Alda-1, an ALDH2 activator, attenuates the formation of 4-HNE protein adducts and ROS in several disease models. We hypothesized that ALDH2 can protect against heat stress-induced vascular inflammation and the accumulation of ROS and toxic aldehydes. Homozygous ALDH2*2 knock-in (KI) mice on a C57BL/6J background and C57BL/6J mice were used for the animal experiments. Human umbilical vein endothelial cells (HUVECs) were used for the in vitro experiment. The mice were directly subjected to whole-body heating (WBH, 42°C) for 1 h at 80% relative humidity. Alda-1 (16 mg/kg) was administered intraperitoneally prior to WBH. The severity of ALI was assessed by analyzing the protein levels and cell counts in the bronchoalveolar lavage fluid, the wet/dry ratio and histology. ALDH2*2 KI mice were susceptible to HS-induced ALI in vivo. Silencing ALDH2 induced 4-HNE and ROS accumulation in HUVECs subjected to heat stress. Alda-1 attenuated the heat stress-induced activation of inflammatory pathways, senescence and apoptosis in HUVECs. The lung homogenates of mice pretreated with Alda-1 exhibited significantly elevated ALDH2 activity and decreased ROS accumulation after WBH. Alda-1 significantly decreased the WBH-induced accumulation of 4-HNE and p65 and p38 activation. Here, we demonstrated the crucial roles of ALDH2 in protecting against heat stress-induced ROS production and vascular inflammation and preserving the viability of ECs. The activation of ALDH2 by Alda-1 attenuates WBH-induced ALI in vivo.
Collapse
Affiliation(s)
- Hsiao-Ya Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Center for the Prevention and Treatment of Heat Stroke, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Cheng-Yo Lu
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Wan-Chu Hung
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Po-Chuan Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Jen-Chun Wang
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Lung-An Hsu
- Cardiovascular Department, Chang-Gung Memorial Hospital and School of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang-Gung Memorial Hospital and School of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Pauling Chu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Center for the Prevention and Treatment of Heat Stroke, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
7
|
Bai H, Zhou R, Barravecchia M, Norman R, Friedman A, Yu D, Lin X, Young JL, Dean DA. The Na+, K+-ATPase β1 subunit regulates epithelial tight junctions via MRCKα. JCI Insight 2021; 6:134881. [PMID: 33507884 PMCID: PMC7934944 DOI: 10.1172/jci.insight.134881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
An intact lung epithelial barrier is essential for lung homeostasis. The Na+, K+-ATPase (NKA), primarily serving as an ion transporter, also regulates epithelial barrier function via modulation of tight junctions. However, the underlying mechanism is not well understood. Here, we show that overexpression of the NKA β1 subunit upregulates the expression of tight junction proteins, leading to increased alveolar epithelial barrier function by an ion transport–independent mechanism. Using IP and mass spectrometry, we identified a number of unknown protein interactions of the β1 subunit, including a top candidate, myotonic dystrophy kinase–related cdc42-binding kinase α (MRCKα), which is a protein kinase known to regulate peripheral actin formation. Using a doxycycline-inducible gene expression system, we demonstrated that MRCKα and its downstream activation of myosin light chain is required for the regulation of alveolar barrier function by the NKA β1 subunit. Importantly, MRCKα is expressed in both human airways and alveoli and has reduced expression in patients with acute respiratory distress syndrome (ARDS), a lung illness that can be caused by multiple direct and indirect insults, including the infection of influenza virus and SARS-CoV-2. Our results have elucidated a potentially novel mechanism by which NKA regulates epithelial tight junctions and have identified potential drug targets for treating ARDS and other pulmonary diseases that are caused by barrier dysfunction.
Collapse
Affiliation(s)
- Haiqing Bai
- Department of Pediatrics and.,Department of Pathology, School of Medicine and Dentistry, University of Rochester, Rochester, New York, USA
| | | | | | | | - Alan Friedman
- Department of Pediatrics and.,Department of Materials Design and Innovation, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, New York, USA
| | | | - Xin Lin
- Department of Pediatrics and
| | | | | |
Collapse
|
8
|
Walweel K, Skeggs K, Boon AC, See Hoe LE, Bouquet M, Obonyo NG, Pedersen SE, Diab SD, Passmore MR, Hyslop K, Wood ES, Reid J, Colombo SM, Bartnikowski NJ, Wells MA, Black D, Pimenta LP, Stevenson AK, Bisht K, Marshall L, Prabhu DA, James L, Platts DG, Macdonald PS, McGiffin DC, Suen JY, Fraser JF. Endothelin receptor antagonist improves donor lung function in an ex vivo perfusion system. J Biomed Sci 2020; 27:96. [PMID: 33008372 PMCID: PMC7532654 DOI: 10.1186/s12929-020-00690-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/24/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND A lung transplant is the last resort treatment for many patients with advanced lung disease. The majority of donated lungs come from donors following brain death (BD). The endothelin axis is upregulated in the blood and lung of the donor after BD resulting in systemic inflammation, lung damage and poor lung graft outcomes in the recipient. Tezosentan (endothelin receptor blocker) improves the pulmonary haemodynamic profile; however, it induces adverse effects on other organs at high doses. Application of ex vivo lung perfusion (EVLP) allows the development of organ-specific hormone resuscitation, to maximise and optimise the donor pool. Therefore, we investigate whether the combination of EVLP and tezosentan administration could improve the quality of donor lungs in a clinically relevant 6-h ovine model of brain stem death (BSD). METHODS After 6 h of BSD, lungs obtained from 12 sheep were divided into two groups, control and tezosentan-treated group, and cannulated for EVLP. The lungs were monitored for 6 h and lung perfusate and tissue samples were processed and analysed. Blood gas variables were measured in perfusate samples as well as total proteins and pro-inflammatory biomarkers, IL-6 and IL-8. Lung tissues were collected at the end of EVLP experiments for histology analysis and wet-dry weight ratio (a measure of oedema). RESULTS Our results showed a significant improvement in gas exchange [elevated partial pressure of oxygen (P = 0.02) and reduced partial pressure of carbon dioxide (P = 0.03)] in tezosentan-treated lungs compared to controls. However, the lungs hematoxylin-eosin staining histology results showed minimum lung injuries and there was no difference between both control and tezosentan-treated lungs. Similarly, IL-6 and IL-8 levels in lung perfusate showed no difference between control and tezosentan-treated lungs throughout the EVLP. Histological and tissue analysis showed a non-significant reduction in wet/dry weight ratio in tezosentan-treated lung tissues (P = 0.09) when compared to control. CONCLUSIONS These data indicate that administration of tezosentan could improve pulmonary gas exchange during EVLP.
Collapse
Affiliation(s)
- K Walweel
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| | - K Skeggs
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.,Princess Alexandra Hospital, Woolloongabba, Brisbane, QLD, 4102, Australia
| | - A C Boon
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L E See Hoe
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - M Bouquet
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - N G Obonyo
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.,Initiative to Develop African Research Leaders, KEMRI-Wellcome, Trust Research Programme, Kilifi, Kenya
| | - S E Pedersen
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - S D Diab
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - M R Passmore
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - K Hyslop
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - E S Wood
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - J Reid
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - S M Colombo
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.,University of Milan, Milan, Italy
| | | | - M A Wells
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.,School of Medical Science, Griffith University, Brisbane, Australia
| | - D Black
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L P Pimenta
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - A K Stevenson
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - K Bisht
- Mater Research Institute-The University of Queensland, Woolloongabba, QLD, Australia
| | - L Marshall
- The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - D A Prabhu
- The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - L James
- Princess Alexandra Hospital, Woolloongabba, Brisbane, QLD, 4102, Australia
| | - D G Platts
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia
| | - P S Macdonald
- Cardiac Mechanics Research Laboratory, St. Vincent's Hospital and the Victor Chang Cardiac Research Institute, Victoria Street, Darlinghurst, Sydney, NSW, 2061, Australia
| | - D C McGiffin
- Cardiothoracic Surgery and Transplantation, The Alfred Hospital, Melbourne, Australia
| | - J Y Suen
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| | - J F Fraser
- Critical Care Research Group, Level 3, Clinical Sciences Building, The Prince Charles Hospital, Rode Road, Brisbane, Australia.
| |
Collapse
|
9
|
Rathod SG. Uncommon presentation of scorpion sting at teaching hospital. J Family Med Prim Care 2020; 9:4480. [PMID: 33110893 PMCID: PMC7586568 DOI: 10.4103/jfmpc.jfmpc_1149_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 11/25/2022] Open
Affiliation(s)
- Santosh Govind Rathod
- Department of Pathology, Subdistrict Hospital Mangaon, Dist Raigad, Maharashtra, India
| |
Collapse
|
10
|
Abstract
BACKGROUND The aim of the study was to provide a theoretical basis for the early diagnosis and prediction of acute altitude sickness, to provide a better entry mode for healthy people from plain areas to plateau areas, and to preliminarily clarify the possible mechanism of this approach. METHODS We measured endothelin-1 (ET-1), asymmetric dimethylarginine (ADMA), vascular endothelial growth factor (VEGF), nitric oxide (NO), and hypoxia-inducible factor 1 (HIF-1) levels in each sample and determined flow-mediated dilation (FMD) values using a portable OMRON color Doppler with a 7.0- to 12.0-MHz linear array probe. We used the Lewis Lake score to diagnose acute mountain sickness (AMS) and to stratify the disease severity. RESULTS We found no cases of AMS at any of the studied elevation gradients. We found significant differences in FMD values between individuals when at 400 m above sea level and when at 2200, 3200, and 4200 m above sea level (P < .05) but found no significant differences among those at 2200, 3200, and 4200 m. Our variance analysis showed that serum ET-1, VEGF, ADMA, NO, and HIF-1 levels in individuals at ≥3000 m and those at subplateau and plain areas (<3000 m) significantly differed (P < .05). The level of these factors also significantly differed between individuals at elevation gradients of plateau areas (3260 m vs 4270 m) (P < .05). We found no significant differences in serum ET-1, VEGF, and ADMA levels between individuals at the plateau (2260 m) and plain (400 m) areas (P > .05). NO and HIF-1 levels were significantly different in serum samples from individuals between the plateau (2260 m) and plain (400 m) areas (P < .05). However, with increasing altitude, the NO level gradually increased, whereas ET-1, ADMA, VEGF, and HIF-1 levels showed a decreasing trend. With the increase of altitude, there is no correlation between the trend of FMD and hematologic-related factors such as VEGF, NO, and HIF-1. CONCLUSION A healthy young male population ascending to a high-altitude area experiences a low incidence of AMS. Entering an acute plateau exposure environment from different altitude gradients may weaken the effect of acute highland exposure on vascular endothelial dysfunction in healthy individuals. Changes in serum ET-1, VEGF, ADMA, NO, and HIF-1 levels in healthy young men may be related to the body's self-regulation and protect healthy individuals from AMS. A short stay in a subplateau region may initiate an oxygen-free preconditioning process in healthy individuals, thereby protecting them from AMS. Noninvasive brachial artery endothelial function test instead of the detection of invasive hematologic-related factors for early diagnosis and prediction of the occurrence and severity of acute high-altitude disease is still lack of sufficient theoretical basis.
Collapse
Affiliation(s)
- Ning Fan
- Graduate School of Qinghai University
| | - Cun Liu
- Qinghai Cardiovascular Hospital
| | - Ming Ren
- The Affiliated Hospital of Qing Hai University, Xi Ning, Qing Hai, China
| |
Collapse
|
11
|
Suleiman S, Klassen S, Katz I, Balakirski G, Krabbe J, von Stillfried S, Kintsler S, Braunschweig T, Babendreyer A, Spillner J, Kalverkamp S, Schröder T, Moeller M, Coburn M, Uhlig S, Martin C, Rieg AD. Argon reduces the pulmonary vascular tone in rats and humans by GABA-receptor activation. Sci Rep 2019; 9:1902. [PMID: 30760775 PMCID: PMC6374423 DOI: 10.1038/s41598-018-38267-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Abstract
Argon exerts neuroprotection. Thus, it might improve patients' neurological outcome after cerebral disorders or cardiopulmonary resuscitation. However, limited data are available concerning its effect on pulmonary vessel and airways. We used rat isolated perfused lungs (IPL) and precision-cut lung slices (PCLS) of rats and humans to assess this topic. IPL: Airway and perfusion parameters, oedema formation and the pulmonary capillary pressure (Pcap) were measured and the precapillary and postcapillary resistance (Rpost) was calculated. In IPLs and PCLS, the pulmonary vessel tone was enhanced with ET-1 or remained unchanged. IPLs were ventilated and PCLS were gassed with argon-mixture or room-air. IPL: Argon reduced the ET-1-induced increase of Pcap, Rpost and oedema formation (p < 0.05). PCLS (rat): Argon relaxed naïve pulmonary arteries (PAs) (p < 0.05). PCLS (rat/human): Argon attenuated the ET-1-induced contraction in PAs (p < 0.05). Inhibition of GABAB-receptors abolished argon-induced relaxation (p < 0.05) in naïve or ET-1-pre-contracted PAs; whereas inhibition of GABAA-receptors only affected ET-1-pre-contracted PAs (p < 0.01). GABAA/B-receptor agonists attenuated ET-1-induced contraction in PAs and baclofen (GABAB-agonist) even in pulmonary veins (p < 0.001). PLCS (rat): Argon did not affect the airways. Finally, argon decreases the pulmonary vessel tone by activation of GABA-receptors. Hence, argon might be applicable in patients with pulmonary hypertension and right ventricular failure.
Collapse
Affiliation(s)
- Said Suleiman
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Sergej Klassen
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Ira Katz
- Medical Research & Development, Air Liquide Santé Internationale, Centre de Recherche Paris-Saclay, 78354, Jouy-en-Josas, France
| | - Galina Balakirski
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Julia Krabbe
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | | | - Svetlana Kintsler
- Institute of Pathology, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Till Braunschweig
- Institute of Pathology, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Jan Spillner
- Department of Cardiac and Thoracic Surgery, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Sebastian Kalverkamp
- Department of Cardiac and Thoracic Surgery, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Thomas Schröder
- Department of Surgery, Luisenhospital Aachen, 52064, Aachen, Germany
| | - Manfred Moeller
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Mark Coburn
- Department of Anaesthesiology, Medical Faculty RWTH Aachen, 52074, Aachen, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology, Medical Faculty RWTH-Aachen, 52074, Aachen, Germany
| | - Annette D Rieg
- Department of Anaesthesiology, Medical Faculty RWTH Aachen, 52074, Aachen, Germany.
| |
Collapse
|
12
|
Gal Y, Mazor O, Falach R, Sapoznikov A, Kronman C, Sabo T. Treatments for Pulmonary Ricin Intoxication: Current Aspects and Future Prospects. Toxins (Basel) 2017; 9:E311. [PMID: 28972558 PMCID: PMC5666358 DOI: 10.3390/toxins9100311] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Ricin, a plant-derived toxin originating from the seeds of Ricinus communis (castor beans), is one of the most lethal toxins known, particularly if inhaled. Ricin is considered a potential biological threat agent due to its high availability and ease of production. The clinical manifestation of pulmonary ricin intoxication in animal models is closely related to acute respiratory distress syndrome (ARDS), which involves pulmonary proinflammatory cytokine upregulation, massive neutrophil infiltration and severe edema. Currently, the only post-exposure measure that is effective against pulmonary ricinosis at clinically relevant time-points following intoxication in pre-clinical studies is passive immunization with anti-ricin neutralizing antibodies. The efficacy of this antitoxin treatment depends on antibody affinity and the time of treatment initiation within a limited therapeutic time window. Small-molecule compounds that interfere directly with the toxin or inhibit its intracellular trafficking may also be beneficial against ricinosis. Another approach relies on the co-administration of antitoxin antibodies with immunomodulatory drugs, thereby neutralizing the toxin while attenuating lung injury. Immunomodulators and other pharmacological-based treatment options should be tailored according to the particular pathogenesis pathways of pulmonary ricinosis. This review focuses on the current treatment options for pulmonary ricin intoxication using anti-ricin antibodies, disease-modifying countermeasures, anti-ricin small molecules and their various combinations.
Collapse
Affiliation(s)
- Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Ohad Mazor
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona 76100, Israel.
| |
Collapse
|
13
|
Azzam ZS, Kinaneh S, Bahouth F, Ismael-Badarneh R, Khoury E, Abassi Z. Involvement of Cytokines in the Pathogenesis of Salt and Water Imbalance in Congestive Heart Failure. Front Immunol 2017; 8:716. [PMID: 28674538 PMCID: PMC5474564 DOI: 10.3389/fimmu.2017.00716] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/02/2017] [Indexed: 12/28/2022] Open
Abstract
Congestive heart failure (CHF) has become a major medical problem in the western world with high morbidity and mortality rates. CHF adversely affects several systems, mainly the kidneys and the lungs. While the involvement of the renin-angiotensin-aldosterone system and the sympathetic nervous system in the progression of cardiovascular, pulmonary, and renal dysfunction in experimental and clinical CHF is well established, the importance of pro-inflammatory mediators in the pathogenesis of this clinical setting is still evolving. In this context, CHF is associated with overexpression of pro-inflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-1, and IL-6, which are activated in response to environmental injury. This family of cytokines has been implicated in the deterioration of CHF, where it plays an important role in initiating and integrating homeostatic responses both at the myocardium and circulatory levels. We and others showed that angiotensin II decreased the ability of the lungs to clear edema and enhanced the fibrosis process via phosphorylation of the mitogen-activated protein kinases p38 and p42/44, which are generally involved in cellular responses to pro-inflammatory cytokines. Literature data also indicate the involvement of these effectors in modulating ion channel activity. It has been reported that in heart failure due to mitral stenosis; there were varying degrees of vascular and other associated parenchymal changes such as edema and fibrosis. In this review, we will discuss the effects of cytokines and other inflammatory mediators on the kidneys and the lungs in heart failure; especially their role in renal and alveolar ion channels activity and fluid balance.
Collapse
Affiliation(s)
- Zaher S. Azzam
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
- Internal Medicine “B”, Rambam Health Care Campus, Haifa, Israel
| | - Safa Kinaneh
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| | - Fadel Bahouth
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| | - Reem Ismael-Badarneh
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| | - Emad Khoury
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
14
|
Katalan S, Falach R, Rosner A, Goldvaser M, Brosh-Nissimov T, Dvir A, Mizrachi A, Goren O, Cohen B, Gal Y, Sapoznikov A, Ehrlich S, Sabo T, Kronman C. A novel swine model of ricin-induced acute respiratory distress syndrome. Dis Model Mech 2017; 10:173-183. [PMID: 28067630 PMCID: PMC5312011 DOI: 10.1242/dmm.027847] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/13/2016] [Indexed: 12/22/2022] Open
Abstract
Pulmonary exposure to the plant toxin ricin leads to respiratory insufficiency and death. To date, in-depth study of acute respiratory distress syndrome (ARDS) following pulmonary exposure to toxins is hampered by the lack of an appropriate animal model. To this end, we established the pig as a large animal model for the comprehensive study of the multifarious clinical manifestations of pulmonary ricinosis. Here, we report for the first time, the monitoring of barometric whole body plethysmography for pulmonary function tests in non-anesthetized ricin-treated pigs. Up to 30 h post-exposure, as a result of progressing hypoxemia and to prevent carbon dioxide retention, animals exhibited a compensatory response of elevation in minute volume, attributed mainly to a large elevation in respiratory rate with minimal response in tidal volume. This response was followed by decompensation, manifested by a decrease in minute volume and severe hypoxemia, refractory to oxygen treatment. Radiological evaluation revealed evidence of early diffuse bilateral pulmonary infiltrates while hemodynamic parameters remained unchanged, excluding cardiac failure as an explanation for respiratory insufficiency. Ricin-intoxicated pigs suffered from increased lung permeability accompanied by cytokine storming. Histological studies revealed lung tissue insults that accumulated over time and led to diffuse alveolar damage. Charting the decline in PaO2/FiO2 ratio in a mechanically ventilated pig confirmed that ricin-induced respiratory damage complies with the accepted diagnostic criteria for ARDS. The establishment of this animal model of pulmonary ricinosis should help in the pursuit of efficient medical countermeasures specifically tailored to deal with the respiratory deficiencies stemming from ricin-induced ARDS.
Collapse
Affiliation(s)
- Shahaf Katalan
- Department of Pharmacology, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Reut Falach
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Amir Rosner
- Veterinary Center for Preclinical Research, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Michael Goldvaser
- Department of Organic Chemistry, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Tal Brosh-Nissimov
- Infectious Disease Unit, Sheba Medical Center, 5262160 Tel-Hashomer, Israel
| | - Ayana Dvir
- General Intensive Care Unit, Asaf Harofeh Medical Center, 70300 Zerifin, Israel
| | - Avi Mizrachi
- General Intensive Care Unit, Kaplan Medical Center, 7661041 Rehovot, Israel
| | - Orr Goren
- Anesthesia, Pain and Intensive Care Division, Tel-Aviv Medical Center, Tel-Aviv University, 6093000 Tel-Aviv, Israel
| | - Barak Cohen
- Anesthesia, Pain and Intensive Care Division, Tel-Aviv Medical Center, Tel-Aviv University, 6093000 Tel-Aviv, Israel
| | - Yoav Gal
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Anita Sapoznikov
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Sharon Ehrlich
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Tamar Sabo
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| | - Chanoch Kronman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, 7410001 Ness-Ziona, Israel
| |
Collapse
|
15
|
Jiang Y, Zeng Y, Huang X, Qin Y, Luo W, Xiang S, Sooranna SR, Pinhu L. Nur77 attenuates endothelin-1 expression via downregulation of NF-κB and p38 MAPK in A549 cells and in an ARDS rat model. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1023-L1035. [PMID: 27765761 PMCID: PMC5206403 DOI: 10.1152/ajplung.00043.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 10/03/2016] [Indexed: 02/07/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by inflammatory injury to the alveolar and capillary barriers that results in impaired gas exchange and severe acute respiratory failure. Nuclear orphan receptor Nur77 has emerged as a regulator of gene expression in inflammation, and its role in the pathogenesis of ARDS is not clear. The objective of this study is to investigate the potential role of Nur77 and its underlying mechanism in the regulation of endothelin-1 (ET-1) expression in lipopolysaccharide (LPS)-induced A549 cells and an ARDS rat model. We demonstrate that LPS induced Nur77 expression and nuclear export in A549 cells. Overexpression of Nur77 markedly decreased basal and LPS-induced ET-1 expression in A549 cells, whereas knockdown of Nur77 increased the ET-1 expression. LPS-induced phosphorylation and nuclear translocation of NF-κB and p38 MAPK were blocked by Nur77 overexpression and augmented by Nur77 knockdown in A549 cells. In vivo, LPS induced Nur77 expression in lung in ARDS rats. Pharmacological activation of Nur77 by cytosporone B (CsnB) inhibited ET-1 expression in ARDS rats, decreased LPS-induced phosphorylation of NF-κB and p38 MAPK, and relieved lung, liver, and kidney injury. Pharmacological deactivation of Nur77 by 1,1-bis-(3'-indolyl)-1-(p-hydroxyphenyl)methane (DIM-C-pPhOH, C-DIM8) had no effect on ET-1 expression and lung injury. These results indicated that Nur77 decreases ET-1 expression by suppressing NF-κB and p38 MAPK in LPS-stimulated A549 cells in vitro, and, in an LPS-induced ARDS rat model, CsnB reduced ET-1 expression and lung injury in ARDS rats.
Collapse
MESH Headings
- A549 Cells
- Active Transport, Cell Nucleus/drug effects
- Animals
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Disease Models, Animal
- Down-Regulation/drug effects
- Endothelin-1/metabolism
- Kidney/drug effects
- Kidney/pathology
- Lipopolysaccharides/pharmacology
- Liver/drug effects
- Liver/pathology
- Lung/drug effects
- Lung/metabolism
- Male
- NF-kappa B/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Phenylacetates/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Respiratory Distress Syndrome/enzymology
- Respiratory Distress Syndrome/genetics
- Respiratory Distress Syndrome/pathology
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Yujie Jiang
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong Province, China
- Department of Respiratory Medicine
| | - Yi Zeng
- Department of Central Laboratory, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Xia Huang
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong Province, China
- Department of Respiratory Medicine
| | - Yueqiu Qin
- Department of Digestive, Youjiang Medical University for Nationalities, Baise, Guangxi, China; Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | | | - Shulin Xiang
- Department of Intensive Care Unit, the People's Hospital of Guangxi, Nanning, Guangxi, China
| | - Suren R Sooranna
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdon; and
| | - Liao Pinhu
- Department of Intensive Care Medicine, Youjiang Medical University for Nationalities, Baise, Guangxi, China
| |
Collapse
|
16
|
Kurt A, Tumkaya L, Turut H, Cure MC, Cure E, Kalkan Y, Sehitoglu I, Acipayam A. Protective Effects of Infliximab on Lung Injury Induced by Methotrexate. Arch Bronconeumol 2015. [PMID: 26071367 DOI: 10.1016/j.arbr.2015.05.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Methotrexate (MTX) is used to treat cancers, several forms of arthritis and other rheumatic conditions, although MTX may cause pulmonary toxicity related to the production of free oxygen radicals, various cytokines. Infliximab (IB) with its potent effect on tumor necrosis factor-alpha (TNF-α) inhibition also inhibits the release of endothelin-1 (ET-1). We aimed to investigate whether IB reduces pulmonary damage induced by an overdose of MTX. METHOD The rats were divided into 3 groups of 8 animals. The control group was given only saline. One dose of 20mg/kg MTX intraperitoneal was administered in the MTX group. IB 7 mg/kg was given to the MTX+IB (MI) group. Three days after IB was administered, 20mg/kg MTX was given. Five days after MTX was administered, all rats were sacrificed. RESULTS The TNF-α, ET-1, malondialdehyde (MDA), myeloperoxidase (MPO) and caspase-3 levels in MTX group were significantly higher than in control groups of TNF-α (P=.001), ET-1 (P=.001), MDA (P=.001), MPO (P=.001) and caspase-3 levels (P=.001) and MI groups of TNF-α (P=.009), ET-1 (P=.001), MDA (P=.047), MPO (P=.007) and caspase-3 levels (P=.003). The MI group had less histopathological damage in lung tissue than the MTX group. CONCLUSION Overdose of MTX leads to cytokine release and the formation of reactive oxygen species in addition to increased ET-1 secretion release that causes lung damage. IB, as a potent proinflammatory agent, TNF-α blocker, can decrease ET-1 release and oxidative stress, it may show significant protective effects in lung tissue against damage caused by MTX overdose.
Collapse
Affiliation(s)
- Aysel Kurt
- Department of Thoracic Surgery, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía.
| | - Levent Tumkaya
- Department of Histology and Embryology, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Hasan Turut
- Department of Thoracic Surgery, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Medine Cumhur Cure
- Department of Biochemistry, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Erkan Cure
- Department of Internal Medicine, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Yildiray Kalkan
- Department of Histology and Embryology, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Ibrahim Sehitoglu
- Department of Pathology, School of Medicine, RecepTayyip Erdogan University, Rize, Turquía
| | - Ahmet Acipayam
- Department of Thoracic Surgery, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| |
Collapse
|
17
|
Kurt A, Tumkaya L, Turut H, Cure MC, Cure E, Kalkan Y, Sehitoglu I, Acipayam A. Protective Effects of Infliximab on Lung Injury Induced by Methotrexate. Arch Bronconeumol 2015; 51:551-7. [PMID: 26071367 DOI: 10.1016/j.arbres.2015.03.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/26/2015] [Accepted: 03/24/2015] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Methotrexate (MTX) is used to treat cancers, several forms of arthritis and other rheumatic conditions, although MTX may cause pulmonary toxicity related to the production of free oxygen radicals, various cytokines. Infliximab (IB) with its potent effect on tumor necrosis factor-alpha (TNF-α) inhibition also inhibits the release of endothelin-1 (ET-1). We aimed to investigate whether IB reduces pulmonary damage induced by an overdose of MTX. METHOD The rats were divided into 3 groups of 8 animals. The control group was given only saline. One dose of 20mg/kg MTX intraperitoneal was administered in the MTX group. IB 7 mg/kg was given to the MTX+IB (MI) group. Three days after IB was administered, 20mg/kg MTX was given. Five days after MTX was administered, all rats were sacrificed. RESULTS The TNF-α, ET-1, malondialdehyde (MDA), myeloperoxidase (MPO) and caspase-3 levels in MTX group were significantly higher than in control groups of TNF-α (P=.001), ET-1 (P=.001), MDA (P=.001), MPO (P=.001) and caspase-3 levels (P=.001) and MI groups of TNF-α (P=.009), ET-1 (P=.001), MDA (P=.047), MPO (P=.007) and caspase-3 levels (P=.003). The MI group had less histopathological damage in lung tissue than the MTX group. CONCLUSION Overdose of MTX leads to cytokine release and the formation of reactive oxygen species in addition to increased ET-1 secretion release that causes lung damage. IB, as a potent proinflammatory agent, TNF-α blocker, can decrease ET-1 release and oxidative stress, it may show significant protective effects in lung tissue against damage caused by MTX overdose.
Collapse
Affiliation(s)
- Aysel Kurt
- Department of Thoracic Surgery, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía.
| | - Levent Tumkaya
- Department of Histology and Embryology, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Hasan Turut
- Department of Thoracic Surgery, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Medine Cumhur Cure
- Department of Biochemistry, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Erkan Cure
- Department of Internal Medicine, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Yildiray Kalkan
- Department of Histology and Embryology, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| | - Ibrahim Sehitoglu
- Department of Pathology, School of Medicine, RecepTayyip Erdogan University, Rize, Turquía
| | - Ahmet Acipayam
- Department of Thoracic Surgery, School of Medicine, Recep Tayyip Erdogan University, Rize, Turquía
| |
Collapse
|
18
|
Abstract
Pulmonary edema clearance is necessary for patients with lung injury to recover and survive. The mechanisms regulating edema clearance from the lungs are distinct from the factors contributing edema formation during injury. Edema clearance is effected via vectorial transport of Na(+) out of the airspaces which generates an osmotic gradient causing water to follow the gradient out of the cells. This Na(+) transport across the alveolar epithelium is mostly effected via apical Na(+) and chloride channels and basolateral Na,K-ATPase. The Na,K-ATPase pumps Na(+) out of the cell and K(+) into the cell against their respective gradients in an ATP-consuming reaction. Two mechanisms contribute to the regulation of the Na,K-ATPase activity:recruitment of its subunits from intracellular compartments into the basolateral membrane, and transcriptional/translational regulation. Na,K-ATPase activity and edema clearance are increased by catecholamines, aldosterone, vasopressin, overexpression of the pump genes, and others. During lung injury, mechanisms regulating edema clearance are inhibited by yet unclear pathways. Better understanding of the mechanisms that regulate pulmonary edema clearance may lead to therapeutic interventions that counterbalance the inhibition of edema clearance during lung injury and improve the lungs' ability to clear fluid, which is crucial for patient survival.
Collapse
Affiliation(s)
- Zaher S. Azzam
- Internal Medicine “B”, Rambam Health Care Campus, Department of Physiology and Biophysics, The Rappaport Family Faculty of Medicine and Research Institute, Technion, Israel Institute of Technology, Haifa, Israel
| | - Jacob I. Sznajder
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
19
|
Machuca TN, Cypel M, Zhao Y, Grasemann H, Tavasoli F, Yeung JC, Bonato R, Chen M, Zamel R, Chun YM, Guan Z, de Perrot M, Waddell TK, Liu M, Keshavjee S. The role of the endothelin-1 pathway as a biomarker for donor lung assessment in clinical ex vivo lung perfusion. J Heart Lung Transplant 2015; 34:849-57. [DOI: 10.1016/j.healun.2015.01.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/27/2014] [Accepted: 01/13/2015] [Indexed: 11/15/2022] Open
|
20
|
Kholdani CA, Fares WH, Trow TK. Macitentan for the treatment of pulmonary arterial hypertension. Vasc Health Risk Manag 2014; 10:665-73. [PMID: 25473292 PMCID: PMC4251661 DOI: 10.2147/vhrm.s33904] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Macitentan is the most recently approved dual endothelin-receptor antagonist (ERA) for the treatment of symptomatic pulmonary arterial hypertension. Compared to other available ERAs, it demonstrates superior receptor-binding properties, with consequently improved tissue penetration, and a longer duration of action allowing for once-daily dosing. It has a favorable adverse-effect profile, with notably no demonstrable increase in the risk of hepatotoxicity or peripheral edema, but like other ERAs, it is potentially limited by significant anemia. Phase I data have demonstrated a favorable drug-drug interaction profile and no need for dose adjustment with hepatic and renal impairment. In the pivotal SERAPHIN study, treatment of symptomatic pulmonary arterial hypertension patients with macitentan led to statistically significant improvements in functional class, exercise tolerance, and hemodynamic parameters, in addition to a reduction in morbidity in an event-driven long-term trial.
Collapse
Affiliation(s)
- Cyrus A Kholdani
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Wassim H Fares
- Yale Pulmonary Vascular Disease Program, Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Terence K Trow
- Yale Pulmonary Vascular Disease Program, Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
21
|
|
22
|
Sevoflurane anesthesia deteriorates pulmonary surfactant promoting alveolar collapse in male Sprague–Dawley rats. Pulm Pharmacol Ther 2014; 28:122-9. [DOI: 10.1016/j.pupt.2013.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 12/06/2013] [Accepted: 12/24/2013] [Indexed: 01/01/2023]
|
23
|
DuBrock HM, Channick RN. Macitentan for the treatment of pulmonary arterial hypertension. Expert Rev Respir Med 2014; 8:393-9. [PMID: 24998329 DOI: 10.1586/17476348.2014.937708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Macitentan is a novel dual endothelin receptor antagonist recently approved for the treatment of symptomatic pulmonary arterial hypertension (PAH). Compared to other endothelin receptor antagonists, in vitro and in vivo studies have demonstrated that macitentan has improved tissue targeting, a longer duration of action and an improved safety profile. Macitentan is available as a once daily oral medication and has been well tolerated in clinical trials. The recently published Study with an Endothelin Receptor Antagonist in PAH to Improve cliNical Outcomes (SERAPHIN), which was the first event-driven trial ever done in PAH, also demonstrated the benefit of macitentan on reducing the likelihood of a composite endpoint of morbidity and mortality events without a significant difference in mortality.
Collapse
Affiliation(s)
- Hilary M DuBrock
- Massachusetts General Hospital, Division of Pulmonary and Critical Care, 55 Fruit St Boston, MA 02114, USA
| | | |
Collapse
|
24
|
Abstract
High-altitude pulmonary edema (HAPE), a not uncommon form of acute altitude illness, can occur within days of ascent above 2500 to 3000 m. Although life-threatening, it is avoidable by slow ascent to permit acclimatization or with drug prophylaxis. The critical pathophysiology is an excessive rise in pulmonary vascular resistance or hypoxic pulmonary vasoconstriction (HPV) leading to increased microvascular pressures. The resultant hydrostatic stress causes dynamic changes in the permeability of the alveolar capillary barrier and mechanical injurious damage leading to leakage of large proteins and erythrocytes into the alveolar space in the absence of inflammation. Bronchoalveolar lavage and hemodynamic pressure measurements in humans confirm that elevated capillary pressure induces a high-permeability noninflammatory lung edema. Reduced nitric oxide availability and increased endothelin in hypoxia are the major determinants of excessive HPV in HAPE-susceptible individuals. Other hypoxia-dependent differences in ventilatory control, sympathetic nervous system activation, endothelial function, and alveolar epithelial active fluid reabsorption likely contribute additionally to HAPE susceptibility. Recent studies strongly suggest nonuniform regional hypoxic arteriolar vasoconstriction as an explanation for how HPV occurring predominantly at the arteriolar level causes leakage. In areas of high blood flow due to lesser HPV, edema develops due to pressures that exceed the dynamic and structural capacity of the alveolar capillary barrier to maintain normal fluid balance. This article will review the pathophysiology of the vasculature, alveolar epithelium, innervation, immune response, and genetics of the lung at high altitude, as well as therapeutic and prophylactic strategies to reduce the morbidity and mortality of HAPE.
Collapse
Affiliation(s)
- Erik R Swenson
- VA Puget Sound Health Care System, Department of Medicine, University of Washington, Seattle, Washington, USA.
| | | |
Collapse
|
25
|
Guetta J, Klorin G, Tal R, Berger G, Ismael-Badarneh R, Bishara B, Sabo E, Abassi Z, Azzam ZS. Vasopressin-2 Receptor Antagonist Attenuates the Ability of the Lungs to Clear Edema in an Experimental Model. Am J Respir Cell Mol Biol 2012; 47:583-8. [DOI: 10.1165/rcmb.2012-0117oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
26
|
de Bisschop C, Martinot JB, Leurquin-Sterk G, Faoro V, Guénard H, Naeije R. Improvement in lung diffusion by endothelin A receptor blockade at high altitude. J Appl Physiol (1985) 2011; 112:20-5. [PMID: 21979801 DOI: 10.1152/japplphysiol.00670.2011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lung diffusing capacity has been reported variably in high-altitude newcomers and may be in relation to different pulmonary vascular resistance (PVR). Twenty-two healthy volunteers were investigated at sea level and at 5,050 m before and after random double-blind intake of the endothelin A receptor blocker sitaxsentan (100 mg/day) vs. a placebo during 1 wk. PVR was estimated by Doppler echocardiography, and exercise capacity by maximal oxygen uptake (Vo(2 max)). The diffusing capacities for nitric oxide (DL(NO)) and carbon monoxide (DL(CO)) were measured using a single-breath method before and 30 min after maximal exercise. The membrane component of DL(CO) (Dm) and capillary volume (Vc) was calculated with corrections for hemoglobin, alveolar volume, and barometric pressure. Altitude exposure was associated with unchanged DL(CO), DL(NO), and Dm but a slight decrease in Vc. Exercise at altitude decreased DL(NO) and Dm. Sitaxsentan intake improved Vo(2 max) together with an increase in resting and postexercise DL(NO) and Dm. Sitaxsentan-induced decrease in PVR was inversely correlated to DL(NO). Both DL(CO) and DL(NO) were correlated to Vo(2 max) at sea level (r = 0.41-0.42, P < 0.1) and more so at altitude (r = 0.56-0.59, P < 0.05). Pharmacological pulmonary vasodilation improves the membrane component of lung diffusion in high-altitude newcomers, which may contribute to exercise capacity.
Collapse
Affiliation(s)
- Claire de Bisschop
- Laboratory of Physiologic Adaptations to Physical Activities, UPRES EA 3813, Poitiers University, Poitiers, France
| | | | | | | | | | | |
Collapse
|
27
|
Sidharta PN, van Giersbergen PLM, Halabi A, Dingemanse J. Macitentan: entry-into-humans study with a new endothelin receptor antagonist. Eur J Clin Pharmacol 2011; 67:977-84. [PMID: 21541781 PMCID: PMC3169777 DOI: 10.1007/s00228-011-1043-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Accepted: 03/23/2011] [Indexed: 01/23/2023]
Abstract
Purpose To study the pharmacokinetics, pharmacodynamics, and tolerability of rising single doses of macitentan, an endothelin receptor antagonist, in healthy male subjects. Methods This double-blind, placebo-controlled study was performed in seven groups of eight healthy male subjects. Doses of 0.2, 1, 5, 25, 100, 300 and 600 mg or placebo (two subjects per group) were administered. Plasma macitentan and endothelin-1 and serum total bile salt concentrations were measured and analysed non-compartmentally. Plasma and urine were analysed qualitatively for the presence of metabolites and one of these, ACT-132577, was also measured quantitatively in plasma. Standard tolerability measurements were performed throughout the study. Results Macitentan was slowly absorbed and, at a dose of 300 mg, the t1/2 (95% confidence interval, CI) was 17.5 h (14.1, 21.8). The dose-proportionality coefficient β for Cmax (95% CI) was 0.83 (0.79, 0.87) indicating less than dose-proportional pharmacokinetics of macitentan. In plasma, a pharmacologically active oxidative depropyl metabolite, ACT-132577, was found whereas in urine two minor metabolites were detected. The t1/2 of ACT-132577 (95% CI) was 65.6 h (53.1, 80.9). Macitentan dose-dependently increased endothelin-1 concentrations up to 2.2-fold (95% CI 1.4, 2.4) at a dose of 600 mg, but had no consistent effect on total bile salts. Macitentan was well tolerated up to and including a dose of 300 mg, the maximum tolerated dose. Headache, nausea and vomiting were dose-limiting adverse events. Conclusion The pharmacokinetic and tolerability profile of macitentan is consistent with a once-a-day dosing regimen and warrants further investigation in clinical studies.
Collapse
Affiliation(s)
- Patricia N Sidharta
- Department of Clinical Pharmacology, Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, 4123 Allschwil, Switzerland.
| | | | | | | |
Collapse
|
28
|
Feng S, Chen W, Cao D, Bian J, Gong FY, Cheng W, Cheng S, Xu Q, Hua ZC, Yin W. Involvement of Na(+), K (+)-ATPase and its inhibitors in HuR-mediated cytokine mRNA stabilization in lung epithelial cells. Cell Mol Life Sci 2011; 68:109-24. [PMID: 20614158 PMCID: PMC11115110 DOI: 10.1007/s00018-010-0444-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2010] [Revised: 06/15/2010] [Accepted: 06/22/2010] [Indexed: 02/02/2023]
Abstract
Increasing evidence demonstrates that Na(+), K(+)-ATPase plays an important role in pulmonary inflammation, but the mechanism remains largely unknown. In this study, we used cardiotonic steroids as Na(+), K(+)-ATPase inhibitors to explore the possible involvement of Na(+), K(+)-ATPase in pulmonary epithelial inflammation. The results demonstrated that mice after ouabain inhalation developed cyclooxygenase-2-dependent acute lung inflammation. The in vitro experiments further confirmed that Na(+), K(+)-ATPase inhibitors significantly stimulated cyclooxygenase-2 expression in lung epithelial cells of human or murine origin, the process of which was participated by multiple cis-elements and trans-acting factors. Most importantly, we first described here that Na(+), K(+)-ATPase inhibitors could evoke a significant Hu antigen R nuclear export in lung epithelial cells, which stabilized cyclooxygenase-2 mRNA by binding with a proximal AU-rich element within its 3'-untranslated region. In conclusion, HuR-mediated mRNA stabilization opens new avenues in understanding the importance of Na(+), K(+)-ATPase, as well as its inhibitors in inflammation.
Collapse
Affiliation(s)
- Su Feng
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| | - Wei Chen
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| | - Dan Cao
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| | - Jinjun Bian
- Department of Anesthesia and Intensive Care Unit, Changhai Hospital, Affiliated Hospital of the Second Military Medical University, Shanghai, 200433 China
| | - Fang-Yuan Gong
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| | - Wei Cheng
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| | - Shun Cheng
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| | - Qiang Xu
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| | - Zi-Chun Hua
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| | - Wu Yin
- The State Key Lab of Pharmaceutical Biotechnology, College of Life Sciences, Nanjing University, Nanjing, 210093 China
| |
Collapse
|
29
|
Yuan JXJ, Garcia JG, West JB, Hales CA, Rich S, Archer SL. High-Altitude Pulmonary Edema. TEXTBOOK OF PULMONARY VASCULAR DISEASE 2011. [PMCID: PMC7122766 DOI: 10.1007/978-0-387-87429-6_61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
High-altitude pulmonary edema (HAPE) is an uncommon form of pulmonary edema that occurs in healthy individuals within a few days of arrival at altitudes above 2,500–3,000 m. The crucial pathophysiology is an excessive hypoxia-mediated rise in pulmonary vascular resistance (PVR) or hypoxic pulmonary vasoconstriction (HPV) leading to increased microvascular hydrostatic pressures despite normal left atrial pressure. The resultant hydrostatic stress can cause both dynamic changes in the permeability of the alveolar capillary barrier and mechanical damage leading to leakage of large proteins and erythrocytes into the alveolar space in the absence of inflammation. Bronchoalveolar lavage (BAL) and pulmonary artery (PA) and microvascular pressure measurements in humans confirm that high capillary pressure induces a high-permeability non-inflammatory-type lung edema; a concept termed “capillary stress failure.” Measurements of endothelin and nitric oxide (NO) in exhaled air, NO metabolites in BAL fluid, and NO-dependent endothelial function in the systemic circulation all point to reduced NO availability and increased endothelin in hypoxia as a major cause of the excessive hypoxic PA pressure rise in HAPE-susceptible individuals. Other hypoxia-dependent differences in ventilatory control, sympathetic nervous system activation, endothelial function, and alveolar epithelial sodium and water reabsorption likely contribute additionally to the phenotype of HAPE susceptibility. Recent studies using magnetic resonance imaging in humans strongly suggest nonuniform regional hypoxic arteriolar vasoconstriction as an explanation for how HPV occurring predominantly at the arteriolar level can cause leakage. This compelling but not yet fully proven mechanism predicts that in areas of high blood flow due to lesser vasoconstriction edema will develop owing to pressures that exceed the structural and dynamic capacity of the alveolar capillary barrier to maintain normal alveolar fluid balance. Numerous strategies aimed at lowering HPV and possibly enhancing active alveolar fluid reabsorption are effective in preventing and treating HAPE. Much has been learned about HAPE in the past four decades such that what was once a mysterious alpine malady is now a well-characterized and preventable lung disease. This chapter will relate the history, pathophysiology, and treatment of HAPE, using it not only to illuminate the condition, but also for the broader lessons it offers in understanding pulmonary vascular regulation and lung fluid balance.
Collapse
Affiliation(s)
- Jason X. -J. Yuan
- Departments of Medicine, COMRB Rm. 3131 (MC 719), University of Illinois at Chicago, 909 South Wolcott Avenue, Chicago, 60612 Illinois USA
| | - Joe G.N. Garcia
- 310 Admin.Office Building (MC 672), University of Illinois at Chicago, 1737 W. Polk Street, Suite 310, Chicago, 60612 Illinois USA
| | - John B. West
- Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093-0623 California USA
| | - Charles A. Hales
- Dept. Pulmonary & Critical Care Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, 02114 Massachusetts USA
| | - Stuart Rich
- Department of Medicine, University of Chicago Medical Center, 5841 S. Maryland Ave., Chicago, 60637 Illinois USA
| | - Stephen L. Archer
- Department of Medicine, University of Chicago School of Medicine, 5841 S. Maryland Ave., Chicago, 60637 Illinois USA
| |
Collapse
|
30
|
Briva A, Lecuona E, Sznajder JI. [Permissive and non-permissive hypercapnia: mechanisms of action and consequences of high carbon dioxide levels]. Arch Bronconeumol 2010; 46:378-82. [PMID: 20303638 PMCID: PMC3858013 DOI: 10.1016/j.arbres.2010.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 01/11/2010] [Indexed: 01/11/2023]
Abstract
Acute lung injury is a disease with high incidence of mortality and its treatment is still controversial. Increasing the levels of CO2 beyond the physiological range has been proposed as a potential protective strategy for patients on mechanical ventilation, as it could moderate the inflammatory response. In this article we review the published evidence on the role of CO2 during acute lung injury. We conclude that although there are reports suggesting benefits from hypercapnia, more recent evidence suggests that hypercapnia could be deleterious, contributing to worsening of the lung injury.
Collapse
Affiliation(s)
- Arturo Briva
- Medicina Intensiva, Departamento de Fisiopatología, Hospital de Clínicas, Montevideo, Uruguay.
| | | | | |
Collapse
|
31
|
Matthay MA, Idell S. Update on acute lung injury and critical care medicine 2009. Am J Respir Crit Care Med 2010; 181:1027-32. [PMID: 20460547 PMCID: PMC3269230 DOI: 10.1164/rccm.201001-0074up] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 02/12/2010] [Indexed: 01/23/2023] Open
Affiliation(s)
- Michael A Matthay
- Department of Medicine, University of California-San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143-0624, USA.
| | | |
Collapse
|
32
|
Salama M, Andrukhova O, Hoda MA, Taghavi S, Jaksch P, Heinze G, Klepetko W, Aharinejad S. Concomitant endothelin-1 overexpression in lung transplant donors and recipients predicts primary graft dysfunction. Am J Transplant 2010; 10:628-36. [PMID: 20055806 DOI: 10.1111/j.1600-6143.2009.02957.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Primary graft dysfunction (PGD) causes significant morbidity following lung transplantation (LTX). Mortality is high in PGD and therapeutic strategies are limited. To investigate whether endothelin-1 (ET-1) that mediates increased vascular permeability and edema formation in lung grafts can predict PGD, ET-1 mRNA expression was examined in lung tissue biopsies of 105 donors and recipients obtained shortly before LTX. Serum ET-1 concentration was assessed by ELISA. PGD grade was diagnosed and scored by oxygenation and radiological characteristics according to ISHLT guidelines. PGD grade 3 developed in 11% of patients. ET-1 mRNA expression was significantly increased in both donor (p < 0.0001) and recipient (p = 0.01) developing PGD as compared to no PGD group. Pretransplant ET-1 serum concentrations were elevated in recipients with PGD as compared to no PGD group (p < 0.0001), although serum ET-1 was not different between donors whose grafts developed PGD grades 0-3. In regression analysis, concomitant elevated donor tissue ET-1 and recipient serum ET-1 predicted PGD grade 3. This study indicates that pretransplant ET-1 mRNA overexpression in donors associated with elevated pretransplant serum ET-1 in recipients contribute to PGD development and that their assessment might be beneficial to predict PGD and to identify recipients who could benefit from a targeted ET-1 blockade.
Collapse
Affiliation(s)
- M Salama
- Department of Cardiothoracic Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Comellas AP, Briva A. Role of endothelin-1 in acute lung injury. Transl Res 2009; 153:263-71. [PMID: 19446279 PMCID: PMC3046772 DOI: 10.1016/j.trsl.2009.02.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2009] [Revised: 02/19/2009] [Accepted: 02/20/2009] [Indexed: 01/11/2023]
Abstract
The alveolar-capillary membrane serves as a barrier that prevents the accumulation of fluid in the alveolar space and restricts the diffusion of large solutes while facilitating an efficient gas exchange. When this barrier becomes dysfunctional, patients develop acute lung injury (ALI), which is characterized by pulmonary edema and increased lung inflammation that leads to a life-threatening impairment of gas exchange. In addition to the increase of inflammatory cytokines, plasma levels of endothelin-1 (ET-1), which is a primarily endothelium-derived vasoconstrictor, are increased in patients with ALI. As patients recover, ET-1 levels decrease, which suggests that ET-1 may not only be a marker of endothelial dysfunction but may have a role in the pathogenesis of ALI. While pulmonary edema accumulates, alveolar fluid clearance (AFC) is of critical importance, as failure to return to normal clearance is associated with poor prognosis in patients with pulmonary edema. AFC involves active transport mechanisms where sodium (Na(+)) is actively transported from the alveolar airspaces, across the alveolar epithelium, and into the pulmonary circulation, which creates an osmotic gradient that is responsible for the clearance of lung edema. In this article, we review the relevance of ET-1 in the development of ALI, not only as a vasoconstrictor molecule but also by inhibiting AFC via the activation of endothelial ET-B receptors and generation. Furthermore, this review highlights the therapeutic role of drugs such as beta-adrenergic agonists and, in particular, of endothelin receptor antagonists in patients with ALI.
Collapse
Affiliation(s)
- Alejandro P Comellas
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa 52242, USA.
| | | |
Collapse
|
34
|
Severinghaus JW. SIGHTINGS. High Alt Med Biol 2009. [DOI: 10.1089/ham.2009.10103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|