1
|
Zivancevic-Simonovic S, Minic R, Cupurdija V, Stanojevic-Pirkovic M, Milosevic-Djordjevic O, Jakovljevic V, Mihaljevic O. Transforming growth factor beta 1 (TGF-β1) in COVID-19 patients: relation to platelets and association with the disease outcome. Mol Cell Biochem 2023; 478:2461-2471. [PMID: 36869188 PMCID: PMC9984293 DOI: 10.1007/s11010-023-04674-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 02/07/2023] [Indexed: 03/05/2023]
Abstract
Transforming growth factor beta (TGF-β) is a ubiquitously distributed cytokine known to contribute to the pathogenesis of numerous pathological processes. The aim of this study was to measure serum concentrations of TGF-β1 in severely ill COVID-19 patients and to analyze its relationship with selected hematological and biochemical parameters and with the disease outcome. The study population included 53 COVID-19 patients with severe clinical expression of the disease and 15 control subjects. TGF-β1 was determined in serum samples and supernatants from PHA-stimulated whole blood cultures using ELISA assay. Biochemical and hematological parameters were analyzed using standard accepted methods. Our results showed that serum levels of TGF-β1 in COVID-19 patients and controls correlate with the platelet counts. Also, positive correlations of TGF-β1 with white blood cell and lymphocyte counts, platelet-to-lymphocyte (PLR) ratio, and fibrinogen level were shown, while negative correlations of this cytokine with platelet distribution width (PDW), D-dimer and activated partial thromboplastin time (a-PTT) values in COVID-19 patients were observed. The lower serum values of TGF-β1 were associated with the unfavorable outcome of COVID-19. In conclusion, TGF-β1 levels were strongly associated with platelet counts and unfavorable disease outcome of severely ill COVID-19 patients.
Collapse
Affiliation(s)
| | - Rajna Minic
- Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Vojislav Cupurdija
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marijana Stanojevic-Pirkovic
- University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | | | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Olgica Mihaljevic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
2
|
Role of pirfenidone in TGF-β pathways and other inflammatory pathways in acute respiratory syndrome coronavirus 2 (SARS-Cov-2) infection: a theoretical perspective. Pharmacol Rep 2021; 73:712-727. [PMID: 33880743 PMCID: PMC8057922 DOI: 10.1007/s43440-021-00255-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/14/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes pulmonary injury or multiple-organ injury by various pathological pathways. Transforming growth factor-beta (TGF-β) is a key factor that is released during SARS-CoV-2 infection. TGF-β, by internalization of the epithelial sodium channel (ENaC), suppresses the anti-oxidant system, downregulates the cystic fibrosis transmembrane conductance regulator (CFTR), and activates the plasminogen activator inhibitor 1 (PAI-1) and nuclear factor-kappa-light-chain-enhancer of activated B cells (NF-kB). These changes cause inflammation and lung injury along with coagulopathy. Moreover, reactive oxygen species play a significant role in lung injury, which levels up during SARS-CoV-2 infection. Drug Suggestion Pirfenidone is an anti-fibrotic drug with an anti-oxidant activity that can prevent lung injury during SARS-CoV-2 infection by blocking the maturation process of transforming growth factor-beta (TGF-β) and enhancing the protective role of peroxisome proliferator-activated receptors (PPARs). Pirfenidone is a safe drug for patients with hypertension or diabetes and its side effect tolerated well. Conclusion The drug as a theoretical perspective may be an effective and safe choice for suppressing the inflammatory response during COVID-19. The recommendation would be a combination of pirfenidone and N-acetylcysteine to achieve maximum benefit during SARS-CoV-2 treatment.
Collapse
|
3
|
Uckun FM, Saund S, Windlass H, Trieu V. Repurposing Anti-Malaria Phytomedicine Artemisinin as a COVID-19 Drug. Front Pharmacol 2021; 12:649532. [PMID: 33815126 PMCID: PMC8017220 DOI: 10.3389/fphar.2021.649532] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Artemisinin is an anti-inflammatory phytomedicine with broad-spectrum antiviral activity. Artemisinin and its antimalarial properties were discovered by the Chinese scientist Tu Youyu, who became one of the laureates of the 2015 Nobel Prize in Physiology or Medicine for this breakthrough in tropical medicine. It is a commonly used anti-malaria drug. Artemisinin has recently been repurposed as a potential COVID-19 drug. Its documented anti-SARS-CoV-2 activity has been attributed to its ability to inhibit spike-protein mediated and TGF-β-dependent early steps in the infection process as well as its ability to disrupt the post-entry intracellular events of the SARS-CoV-2 infection cycle required for viral replication. In addition, Artemisinin has anti-inflammatory activity and reduces the systemic levels of inflammatory cytokines that contribute to cytokine storm and inflammatory organ injury in high-risk COVID-19 patients. We postulate that Artemisinin may prevent the worsening of the health condition of patients with mild-moderate COVID-19 when administered early in the course of their disease.
Collapse
Affiliation(s)
| | - Saran Saund
- Oncotelic Inc., Agoura Hills, CA, United States
| | | | - Vuong Trieu
- Oncotelic Inc., Agoura Hills, CA, United States
| |
Collapse
|
4
|
O'Sullivan MJ, Mitchel JA, Mwase C, McGill M, Kanki P, Park JA. In well-differentiated primary human bronchial epithelial cells, TGF- β1 and TGF- β2 induce expression of furin. Am J Physiol Lung Cell Mol Physiol 2021; 320:L246-L253. [PMID: 33174447 PMCID: PMC7900917 DOI: 10.1152/ajplung.00423.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/04/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 pandemic is an ongoing threat to public health. Since the identification of COVID-19, the disease caused by SARS-CoV-2, no drugs have been developed to specifically target SARS-CoV-2. To develop effective and safe treatment options, a better understanding of cellular mechanisms underlying SARS-CoV-2 infection is required. To fill this knowledge gap, researchers require reliable experimental systems that express the host factor proteins necessary for the cellular entry of SARS-CoV-2. These proteins include the viral receptor, angiotensin-converting enzyme 2 (ACE2), and the proteases, transmembrane serine protease 2 (TMPRSS2) and furin. A number of studies have reported cell-type-specific expression of the genes encoding these molecules. However, less is known about the protein expression of these molecules. We assessed the suitability of primary human bronchial epithelial (HBE) cells maintained in an air-liquid interface (ALI) as an experimental system for studying SARS-CoV-2 infection in vitro. During cellular differentiation, we measured the expression of ACE2, TMPRSS2, and furin over progressive ALI days by reverse transcription-quantitative polymerase chain reaction (RT-qPCR), Western blot, and immunofluorescence staining. We also explored the effect of the fibrotic cytokine TGF-β on the expression of these proteins in well-differentiated HBE cells. Like ACE2, TMPRSS2 and furin proteins are localized in differentiated ciliated cells, as confirmed by immunofluorescence staining. These data suggest that well-differentiated HBE cells maintained in ALI are a reliable in vitro system for investigating cellular mechanisms of SARS-CoV-2 infection. We further identified that the profibrotic mediators, TGF-β1 and TGF-β2, increase the expression of furin, which is a protease required for the cellular entry of SARS-CoV-2.
Collapse
Affiliation(s)
- Michael J O'Sullivan
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jennifer A Mitchel
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Chimwemwe Mwase
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Maureen McGill
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Phyllis Kanki
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Jin-Ah Park
- Program in Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
5
|
Uckun FM, Carlson J, Orhan C, Powell J, Pizzimenti NM, van Wyk H, Ozercan IH, Volk M, Sahin K. Rejuveinix Shows a Favorable Clinical Safety Profile in Human Subjects and Exhibits Potent Preclinical Protective Activity in the Lipopolysaccharide-Galactosamine Mouse Model of Acute Respiratory Distress Syndrome and Multi-Organ Failure. Front Pharmacol 2020; 11:594321. [PMID: 33244300 PMCID: PMC7683794 DOI: 10.3389/fphar.2020.594321] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 09/25/2020] [Indexed: 12/22/2022] Open
Abstract
Background: New treatment platforms that can prevent acute respiratory distress syndrome (ARDS) or reduce its mortality rate in high-risk coronavirus disease 2019 (COVID-19) patients, such as those with an underlying cancer, are urgently needed. Rejuveinix (RJX) is an intravenous formulation of anti-oxidants and anti-inflammatory agents. Its active ingredients include ascorbic acid, cyanocobalamin, thiamine hydrochloride, riboflavin 5' phosphate, niacinamide, pyridoxine hydrochloride, and calcium D-pantothenate. RJX is being developed as an anti-inflammatory and anti-oxidant treatment platform for patients with sepsis, including COVID-19 patients with viral sepsis and ARDS. Here, we report its clinical safety profile in a phase 1 clinical study (ClinicalTrials.gov Identifier: NCT03680105) and its potent protective activity in the lipopolysaccharide galactosamine (LPS-GalN) mouse model of ARDS. Methods: A phase 1, double-blind, placebo-controlled, randomized, two-part, ascending dose-escalation study was performed in participating 76 healthy volunteer human subjects in compliance with the ICH (E6) good clinical practice guidelines to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of RJX (Protocol No. RPI003; ClinicalTrials.gov Identifier: NCT03680105). The ability of RJX to prevent fatal shock, ARDS, and multi-organ failure was examined in the well-established LPS-GalN mouse model of sepsis and ARDS. Standard methods were employed for the statistical analysis of data in both studies. Findings: In the phase 1 clinical study, no participant developed serious adverse events (SAEs) or Grade 3-Grade 4 adverse events (AEs) or prematurely discontinued participation in the study. In the non-clinical study, RJX exhibited potent and dose-dependent protective activity, decreased the inflammatory cytokine responses (interleukin-6, tumor necrosis factor alpha, transforming growth factor beta), and improved survival in the LPS-GalN mouse model of sepsis and ARDS. Histopathological examinations showed that RJX attenuated the LPS-GalN induced acute lung injury (ALI) and pulmonary edema as well as liver damage. Conclusion: RJX showed a very favorable safety profile and tolerability in human subjects. It shows potential to favorably affect the clinical course of high-risk COVID-19 by preventing ARDS and its complications.
Collapse
Affiliation(s)
- Fatih M. Uckun
- Drug Discovery Program, Reven Pharmaceuticals, Golden, CO, United States
- Department of Developmental Therapeutics, Immunology, and Integrative Medicine, Ares Pharmaceuticals, St. Paul, MN, United States
| | - James Carlson
- Drug Discovery Program, Reven Pharmaceuticals, Golden, CO, United States
| | - Cemal Orhan
- Department of Animal Nutrition, Faculty of Veterinary, Firat University, Elazig, Turkey
| | - Joy Powell
- Drug Discovery Program, Reven Pharmaceuticals, Golden, CO, United States
| | | | - Hendrik van Wyk
- Drug Discovery Program, Reven Pharmaceuticals, Golden, CO, United States
| | - Ibrahim H. Ozercan
- Department of Pathology Faculty of Medicine, Firat University, Elazig, Turkey
| | - Michael Volk
- Drug Discovery Program, Reven Pharmaceuticals, Golden, CO, United States
| | - Kazim Sahin
- Department of Animal Nutrition, Faculty of Veterinary, Firat University, Elazig, Turkey
| |
Collapse
|
6
|
Wu D, Fu X, Zhang Y, Li Q, Ye L, Han S, Zhang M. The protective effects of C16 peptide and angiopoietin-1 compound in lipopolysaccharide-induced acute respiratory distress syndrome. Exp Biol Med (Maywood) 2020; 245:1683-1696. [PMID: 32915636 DOI: 10.1177/1535370220953791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
C16 peptide and angiopoietin-1 (Ang-1) have been found to have anti-inflammatory activity in various inflammation-related diseases. However, their combined role in acute respiratory distress syndrome (ARDS) has not been investigated yet. The objective of this study was to investigate the effects of C16 peptide and Ang-1 in combination with lipopolysaccharide (LPS)-induced inflammatory insult in vitro and in vivo. Human pulmonary microvascular endothelial cells and human pulmonary alveolar epithelial cells were used as cell culture systems, and an ARDS rodent model was used for in vivo studies. Our results demonstrated that C16 and Ang-1 in combination significantly suppressed inflammatory cell transmigration by 33% in comparison with the vehicle alone, and decreased the lung tissue wet-to-dry lung weight ratio to a maximum of 1.53, compared to 3.55 in the vehicle group in ARDS rats. Moreover, C + A treatment reduced the histology injury score to 60% of the vehicle control, enhanced arterial oxygen saturation (SO2), decreased arterial carbon dioxide partial pressure (PCO2), and increased oxygen partial pressure (PO2) in ARDS rats, while also improving the survival rate from 47% (7/15) to 80% (12/15) and diminishing fibrosis, necrosis, and apoptosis in lung tissue. Furthermore, when C + A therapy was administered 4 h following LPS injection, the treatment showed significant alleviating effects on pulmonary inflammatory cell infiltration 24 h postinsult. In conclusion, our in vitro and in vivo studies show that C16 and Ang-1 exert protective effects against LPS-induced inflammatory insult. C16 and Ang-1 hold promise as a novel agent against LPS-induced ARDS. Further studies are needed to determine the potential for C16 and Ang-1 in combination in treating inflammatory lung diseases.
Collapse
Affiliation(s)
- Dingqian Wu
- The Emergency Medicine Department; the Second Affiliated Hospital Zhejiang University School of Medicine (SAHZU), Hangzhou 310058, China
| | - Xiaoxiao Fu
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, Hangzhou 310058, China
| | - Yuanyuan Zhang
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, Hangzhou 310058, China
| | - Qiang Li
- The Emergency Medicine Department; the Second Affiliated Hospital Zhejiang University School of Medicine (SAHZU), Hangzhou 310058, China
| | - Ligang Ye
- The Emergency Medicine Department; the Second Affiliated Hospital Zhejiang University School of Medicine (SAHZU), Hangzhou 310058, China
| | - Shu Han
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, Hangzhou 310058, China
| | - Mao Zhang
- The Emergency Medicine Department; the Second Affiliated Hospital Zhejiang University School of Medicine (SAHZU), Hangzhou 310058, China
| |
Collapse
|
7
|
A Staphylococcus pro-apoptotic peptide induces acute exacerbation of pulmonary fibrosis. Nat Commun 2020; 11:1539. [PMID: 32210242 PMCID: PMC7093394 DOI: 10.1038/s41467-020-15344-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 03/03/2020] [Indexed: 11/08/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal disease of unknown etiology; however, apoptosis of lung alveolar epithelial cells plays a role in disease progression. This intractable disease is associated with increased abundance of Staphylococcus and Streptococcus in the lungs, yet their roles in disease pathogenesis remain elusive. Here, we report that Staphylococcus nepalensis releases corisin, a peptide conserved in diverse staphylococci, to induce apoptosis of lung epithelial cells. The disease in mice exhibits acute exacerbation after intrapulmonary instillation of corisin or after lung infection with corisin-harboring S. nepalensis compared to untreated mice or mice infected with bacteria lacking corisin. Correspondingly, the lung corisin levels are significantly increased in human IPF patients with acute exacerbation compared to patients without disease exacerbation. Our results suggest that bacteria shedding corisin are involved in acute exacerbation of IPF, yielding insights to the molecular basis for the elevation of staphylococci in pulmonary fibrosis.
Collapse
|
8
|
Koutsogiannaki S, Shimaoka M, Yuki K. The Use of Volatile Anesthetics as Sedatives for Acute Respiratory Distress Syndrome. ACTA ACUST UNITED AC 2019; 6:27-38. [PMID: 30923729 PMCID: PMC6433148 DOI: 10.31480/2330-4871/084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) remains to pose a high morbidity and mortality without any targeted therapies. Sedation, usually given intravenously, is an important part of clinical practice in intensive care unit (ICU), and the effect of sedatives on patients’ outcomes has been studied intensively. Although volatile anesthetics are not routine sedatives in ICU, preclinical and clinical studies suggested their potential benefit in pulmonary pathophysiology. This review will summarize the current knowledge of ARDS and the role of volatile anesthetic sedation in this setting from both clinical and mechanistic standpoints. In addition, we will review the infrastructure to use volatile anesthetics.
Collapse
Affiliation(s)
- Sophia Koutsogiannaki
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts, USA.,Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsushi, Mie, Japan
| | - Koichi Yuki
- Department of Anaesthesia, Harvard Medical School, Boston, Massachusetts, USA.,Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
9
|
Lin S, Wu H, Wang C, Xiao Z, Xu F. Regulatory T Cells and Acute Lung Injury: Cytokines, Uncontrolled Inflammation, and Therapeutic Implications. Front Immunol 2018; 9:1545. [PMID: 30038616 PMCID: PMC6046379 DOI: 10.3389/fimmu.2018.01545] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 12/24/2022] Open
Abstract
Acute respiratory distress syndrome/acute lung injury (ALI) was described in 1967. The uncontrolled inflammation is a central issue of the syndrome. The regulatory T cells (Tregs), formerly known as suppressor T cells, are a subpopulation of T cells. Tregs indirectly limits immune inflammation-inflicted tissue damage by employing multiple mechanisms and creating the appropriate immune environment for successful tissue repair. And it plays a central role in the resolution of ALI. Accordingly, for this review, we will focus on Treg populations which are critical for inflammatory immunity of ALI, and the effect of interaction between Treg subsets and cytokines on ALI. And then explore the possibility of cytokines as beneficial factors in inflammation resolution of ALI.
Collapse
Affiliation(s)
- Shihui Lin
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Wu
- Center for Cognitive and Neurobiological Imaging, Stanford University, Stanford, CA, United States
| | - Chuanjiang Wang
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhibo Xiao
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Emergency and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Gao X, Qian P, Cen D, Hong W, Peng Q, Xue M. Synthesis of phosphatidylcholine in rats with oleic acid-induced pulmonary edema and effect of exogenous pulmonary surfactant on its De Novo synthesis. PLoS One 2018; 13:e0193719. [PMID: 29554114 PMCID: PMC5858825 DOI: 10.1371/journal.pone.0193719] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 02/14/2018] [Indexed: 11/26/2022] Open
Abstract
In mammals, oleic acid (OA) induces pulmonary edema (PE), which can initiate acute lung injury (ALI) and lead to acute respiratory distress syndrome (ARDS). Pulmonary surfactant (PS) plays a key role in a broad range of treatments for ARDS. The aim of the present investigation was to assess changes in the synthesis of phosphatidylcholine (PC) from choline and determine the effect of exogenous PS on its de novo synthesis in rats with OA-induced PE. Experimental rats were randomized into three groups, including a control group, OA-induced PE group, and OA-induced group treated with exogenous PS (OA-PS). Twenty-four rats were sacrificed 4 h after induction of the OA model, and tissue was examined by light and electron microscopy to assess the severity of ALI using an established scoring system at the end of the experiment. After 15 μCi 3H-choline chloride was injected intravenously, eight rats in each group were sacrificed at 4, 8, and 16 h. The radioactivity of 3H incorporated into total phospholipid (TPL) and desaturated phosphatidylcholine (DSPC) was measured in bronchoalveolar lavage fluid (BALF) and lung tissue (LT) using a liquid scintillation counter and was expressed as counts per minute (CPM). Results showed that TPL, DSPC, and the ratio of DSPC/total protein (TP) in lung tissue decreased 4 h after challenge with OA, but the levels recovered after 8 and 16 h. At 8 h after injection, 3H-TPL and 3H-DSPC radioactivity in the lungs reached its peak. Importantly, 3H-DSPC CPM were significantly lower in the PS treatment group (LT: Control: 62327 ± 9108; OA-PE: 97315 ± 10083; OA-PS: 45127 ± 10034, P < 0.05; BALF: Control: 7771 ± 1768; OA-PE: 8097 ± 1799; OA-PE: 3651 ± 1027, P < 0.05). Furthermore, DSPC secretory rate (SR) in the lungs was significantly lower in the PS treatment group at 4 h after injection (Control: 0.014 ± 0.003; OA-PE: 0.011 ± 0.004; OA-PS: 0.023 ± 0.006, P < 0.05). Therefore, we hypothesize that exogenous PS treatments may adversely affect endogenous de novo synthetic and secretory phospholipid pathways via feedback inhibition. This novel finding reveals the specific involvement of exogenous PS in endogenous synthetic and secretory phospholipid pathways during the treatment of ARDS. This information improves our understanding of how PS treatment is beneficial against ARDS and opens new opportunities for expanding its use.
Collapse
Affiliation(s)
- Xiwen Gao
- Department of Respiratory Diseases, Minhang Hospital, Fudan University, Minhang District, Shanghai, P.R. China
- * E-mail:
| | - Peiyu Qian
- Oncology Bioinformatic Research Center, Minhang Hospital, Fudan University, Minhang District, Shanghai, P.R. China
| | - Dong Cen
- Centre for Clinical Laboratory, Ningbo Yinzhou No 2 Hospital, Zhejiang
| | - Weijun Hong
- Department of Respiratory Diseases, Minhang Hospital, Fudan University, Minhang District, Shanghai, P.R. China
| | - Qing Peng
- Department of Respiratory Diseases, Minhang Hospital, Fudan University, Minhang District, Shanghai, P.R. China
| | - Min Xue
- Department of Respiratory Diseases, Minhang Hospital, Fudan University, Minhang District, Shanghai, P.R. China
| |
Collapse
|
11
|
Hamacher J, Hadizamani Y, Borgmann M, Mohaupt M, Männel DN, Moehrlen U, Lucas R, Stammberger U. Cytokine-Ion Channel Interactions in Pulmonary Inflammation. Front Immunol 2018; 8:1644. [PMID: 29354115 PMCID: PMC5758508 DOI: 10.3389/fimmu.2017.01644] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
The lungs conceptually represent a sponge that is interposed in series in the bodies’ systemic circulation to take up oxygen and eliminate carbon dioxide. As such, it matches the huge surface areas of the alveolar epithelium to the pulmonary blood capillaries. The lung’s constant exposure to the exterior necessitates a competent immune system, as evidenced by the association of clinical immunodeficiencies with pulmonary infections. From the in utero to the postnatal and adult situation, there is an inherent vital need to manage alveolar fluid reabsorption, be it postnatally, or in case of hydrostatic or permeability edema. Whereas a wealth of literature exists on the physiological basis of fluid and solute reabsorption by ion channels and water pores, only sparse knowledge is available so far on pathological situations, such as in microbial infection, acute lung injury or acute respiratory distress syndrome, and in the pulmonary reimplantation response in transplanted lungs. The aim of this review is to discuss alveolar liquid clearance in a selection of lung injury models, thereby especially focusing on cytokines and mediators that modulate ion channels. Inflammation is characterized by complex and probably time-dependent co-signaling, interactions between the involved cell types, as well as by cell demise and barrier dysfunction, which may not uniquely determine a clinical picture. This review, therefore, aims to give integrative thoughts and wants to foster the unraveling of unmet needs in future research.
Collapse
Affiliation(s)
- Jürg Hamacher
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Internal Medicine V - Pneumology, Allergology, Respiratory and Environmental Medicine, Faculty of Medicine, Saarland University, Saarbrücken, Germany.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Yalda Hadizamani
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Michèle Borgmann
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Markus Mohaupt
- Internal Medicine, Sonnenhofspital Bern, Bern, Switzerland
| | | | - Ueli Moehrlen
- Paediatric Visceral Surgery, Universitäts-Kinderspital Zürich, Zürich, Switzerland
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Vascular Biology Center, Medical College of Georgia, Augusta, GA, United States
| | - Uz Stammberger
- Lungen- und Atmungsstiftung Bern, Bern, Switzerland.,Novartis Institutes for Biomedical Research, Translational Clinical Oncology, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
12
|
Wynne BM, Zou L, Linck V, Hoover RS, Ma HP, Eaton DC. Regulation of Lung Epithelial Sodium Channels by Cytokines and Chemokines. Front Immunol 2017; 8:766. [PMID: 28791006 PMCID: PMC5524836 DOI: 10.3389/fimmu.2017.00766] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 06/16/2017] [Indexed: 12/20/2022] Open
Abstract
Acute lung injury leading to acute respiratory distress (ARDS) is a global health concern. ARDS patients have significant pulmonary inflammation leading to flooding of the pulmonary alveoli. This prevents normal gas exchange with consequent hypoxemia and causes mortality. A thin fluid layer in the alveoli is normal. The maintenance of this thin layer results from fluid movement out of the pulmonary capillaries into the alveolar interstitium driven by vascular hydrostatic pressure and then through alveolar tight junctions. This is then balanced by fluid reabsorption from the alveolar space mediated by transepithelial salt and water transport through alveolar cells. Reabsorption is a two-step process: first, sodium enters via sodium-permeable channels in the apical membranes of alveolar type 1 and 2 cells followed by active extrusion of sodium into the interstitium by the basolateral Na+, K+-ATPase. Anions follow the cationic charge gradient and water follows the salt-induced osmotic gradient. The proximate cause of alveolar flooding is the result of a failure to reabsorb sufficient salt and water or a failure of the tight junctions to prevent excessive movement of fluid from the interstitium to alveolar lumen. Cytokine- and chemokine-induced inflammation can have a particularly profound effect on lung sodium transport since they can alter both ion channel and barrier function. Cytokines and chemokines affect alveolar amiloride-sensitive epithelial sodium channels (ENaCs), which play a crucial role in sodium transport and fluid reabsorption in the lung. This review discusses the regulation of ENaC via local and systemic cytokines during inflammatory disease and the effect on lung fluid balance.
Collapse
Affiliation(s)
- Brandi M Wynne
- Department of Medicine, Nephrology, Emory University, Atlanta, GA, United States.,Department of Physiology, Emory University, Atlanta, GA, United States.,The Center for Cell and Molecular Signaling, Emory University, Atlanta, GA, United States
| | - Li Zou
- Department of Physiology, Emory University, Atlanta, GA, United States
| | - Valerie Linck
- Department of Physiology, Emory University, Atlanta, GA, United States
| | - Robert S Hoover
- Department of Medicine, Nephrology, Emory University, Atlanta, GA, United States.,Department of Physiology, Emory University, Atlanta, GA, United States.,Research Service, Atlanta Veteran's Administration Medical Center, Decatur, GA, United States
| | - He-Ping Ma
- Department of Physiology, Emory University, Atlanta, GA, United States.,The Center for Cell and Molecular Signaling, Emory University, Atlanta, GA, United States
| | - Douglas C Eaton
- Department of Physiology, Emory University, Atlanta, GA, United States.,The Center for Cell and Molecular Signaling, Emory University, Atlanta, GA, United States
| |
Collapse
|
13
|
Middleton EA, Weyrich AS, Zimmerman GA. Platelets in Pulmonary Immune Responses and Inflammatory Lung Diseases. Physiol Rev 2016; 96:1211-59. [PMID: 27489307 PMCID: PMC6345245 DOI: 10.1152/physrev.00038.2015] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Platelets are essential for physiological hemostasis and are central in pathological thrombosis. These are their traditional and best known activities in health and disease. In addition, however, platelets have specializations that broaden their functional repertoire considerably. These functional capabilities, some of which are recently discovered, include the ability to sense and respond to infectious and immune signals and to act as inflammatory effector cells. Human platelets and platelets from mice and other experimental animals can link the innate and adaptive limbs of the immune system and act across the immune continuum, often also linking immune and hemostatic functions. Traditional and newly recognized facets of the biology of platelets are relevant to defensive, physiological immune responses of the lungs and to inflammatory lung diseases. The emerging view of platelets as blood cells that are much more diverse and versatile than previously thought further predicts that additional features of the biology of platelets and of megakaryocytes, the precursors of platelets, will be discovered and that some of these will also influence pulmonary immune defenses and inflammatory injury.
Collapse
Affiliation(s)
- Elizabeth A Middleton
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and the Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Andrew S Weyrich
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and the Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Guy A Zimmerman
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, and the Program in Molecular Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
14
|
The Contribution of the Airway Epithelial Cell to Host Defense. Mediators Inflamm 2015; 2015:463016. [PMID: 26185361 PMCID: PMC4491388 DOI: 10.1155/2015/463016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 12/11/2014] [Indexed: 12/19/2022] Open
Abstract
In the context of cystic fibrosis, the epithelial cell has been characterized in terms of its ion transport capabilities. The ability of an epithelial cell to initiate CFTR-mediated chloride and bicarbonate transport has been recognized early as a means to regulate the thickness of the epithelial lining fluid and recently as a means to regulate the pH, thereby determining critically whether or not host defense proteins such as mucins are able to fold appropriately. This review describes how the epithelial cell senses the presence of pathogens and inflammatory conditions, which, in turn, facilitates the activation of CFTR and thus directly promotes pathogens clearance and innate immune defense on the surface of the epithelial cell. This paper summarizes functional data that describes the effect of cytokines, chemokines, infectious agents, and inflammatory conditions on the ion transport properties of the epithelial cell and relates these key properties to the molecular pathology of cystic fibrosis. Recent findings on the role of cystic fibrosis modifier genes that underscore the role of the epithelial ion transport in host defense and inflammation are discussed.
Collapse
|
15
|
Sharp C, Millar AB, Medford ARL. Advances in understanding of the pathogenesis of acute respiratory distress syndrome. Respiration 2015; 89:420-34. [PMID: 25925331 DOI: 10.1159/000381102] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 02/12/2015] [Indexed: 02/05/2023] Open
Abstract
The clinical syndrome of acute lung injury (ALI) occurs as a result of an initial acute systemic inflammatory response. This can be consequent to a plethora of insults, either direct to the lung or indirect. The insult results in increased epithelial permeability, leading to alveolar flooding with a protein-rich oedema fluid. The resulting loss of gas exchange leads to acute respiratory failure and typically catastrophic illness, termed acute respiratory distress syndrome (ARDS), requiring ventilatory and critical care support. There remains a significant disease burden, with some estimates showing 200,000 cases each year in the USA with a mortality approaching 50%. In addition, there is a significant burden of morbidity in survivors. There are currently no disease-modifying therapies available, and the most effective advances in caring for these patients have been in changes to ventilator strategy as a result of the ARDS network studies nearly 15 years ago. Here, we will give an overview of more recent advances in the understanding of the cellular biology of ALI and highlight areas that may prove fertile for future disease-modifying therapies.
Collapse
Affiliation(s)
- Charles Sharp
- Academic Respiratory Unit, University of Bristol, Southmead Hospital, Westbury-on-Trym, UK
| | | | | |
Collapse
|