1
|
Zhang FL, Chen YL, Luo ZY, Song ZB, Chen Z, Zhang JX, Zheng ZZ, Tan XM. Huashi baidu granule alleviates inflammation and lung edema by suppressing the NLRP3/caspase-1/GSDMD-N pathway and promoting fluid clearance in a porcine reproductive and respiratory syndrome (PRRS) model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119207. [PMID: 39653102 DOI: 10.1016/j.jep.2024.119207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huashi Baidu Granule (HSBDG), a traditional Chinese medicine (TCM), is used for treating coronavirus disease 2019 (COVID-19). Porcine reproductive and respiratory syndrome (PRRS) is considered the "COVID-19" for swine. According to the TCM theory, "dampness" is the main pathogenic factor in COVID-19 and PRRS, and "Huashi" means that this formula is good at removing "dampness". Studies have demonstrated that HSBDG's effect in COVID-19; but the mechanism of removing "dampness" remains elusive. AIM OF THE STUDY We aimed to assess the effect of HSBDG on PRRS, and elucidate its potential mechanism in removing "dampness". MATERIALS AND METHODS We established a PRRS-virus (PRRSV)-infected Marc-145 cells model, and performed qRT-PCR, Western blot analysis, and indirect immunofluorescence assay to examine the anti-PRRSV effects of HSBDG in vitro. PRRSV-infected pig model was established and used to investigate HSBDG's effect in PRRS and explore underlying mechanisms in removing "dampness" using ELISA and immunohistochemistry assay methods. RESULTS HSBDG exhibited anti-PRRSV activity and suppressed the viral replication and release phases. HSBDG treatment alleviated PRRS, lowered rectal temperature, reduced histopathological changes and viral load in lung tissues, and ameliorated organ lesions. Moreover, IL-1β, IL-6, IL-8, and TNF-α expressions were decreased in lung tissues. Mechanistically, HSBDG inhibited the NLRP3/Caspase-1/GSDMD-N pathway to reduce the inflammatory response and upregulated AQP1, AQP5, α-ENaC, and Na-K-ATPase expressions to promote lung fluid clearance. CONCLUSION HSBDG exerted anti-PRRSV effects and could attenuate PRRS. HSBDG potentially removes "dampness" by attenuating inflammation by suppressing the NLRP3/Caspase-1/GSDMD-N pathway and inhibiting pulmonary edema by upregulating the expression of AQP1, AQP5, α-ENaC, and Na-K-ATPase.
Collapse
Affiliation(s)
- Feng-Lin Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Yi-Lin Chen
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Zhen-Ye Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Ze-Bu Song
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Zhe Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Jia-Xuan Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Ze-Zhong Zheng
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Xiao-Mei Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| |
Collapse
|
2
|
Meegan JE, Rizzo AN, Schmidt EP, Bastarache JA. Cellular Mechanisms of Lung Injury: Current Perspectives. Clin Chest Med 2024; 45:821-833. [PMID: 39443000 PMCID: PMC11499619 DOI: 10.1016/j.ccm.2024.08.004] [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] [Indexed: 10/25/2024]
Abstract
The alveolar-capillary barrier includes microvascular endothelial and alveolar epithelial cells and their matrices, and its disruption is a critical driver of lung injury during development of acute respiratory distress syndrome. In this review, we provide an overview of the structure and function of the alveolar-capillary barrier during health and highlight several important signaling mechanisms that underlie endothelial and epithelial injury during critical illness, emphasizing areas with potential for development of therapeutic strategies targeting alveolar-capillary leak. We also emphasize the importance of biomarker and preclinical studies in developing novel therapies and highlight important areas warranting future investigation.
Collapse
Affiliation(s)
- Jamie E Meegan
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alicia N Rizzo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Bulfinch 148, Boston, MA 02114, USA
| | - Eric P Schmidt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Bulfinch 148, Boston, MA 02114, USA
| | - Julie A Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|
3
|
Whiteley J, Shan H, Yeung JC. Noninflammatory Causes of Pulmonary Edema During Ex Vivo Lung Perfusion. ANNALS OF THORACIC SURGERY SHORT REPORTS 2024; 2:848-850. [PMID: 39790629 PMCID: PMC11708558 DOI: 10.1016/j.atssr.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 01/12/2025]
Abstract
Ex vivo lung perfusion (EVLP) is used to evaluate donor lungs prior to lung transplantation. Development of pulmonary edema during EVLP is generally thought to represent inflammatory breakdown of the air-fluid barrier and these lungs are declined for transplant. We present the case of a donor lung that underwent stapled wedge resection during cold storage for air leak and the subsequent development of profound (∼650 mL) pulmonary edema around the staple line during EVLP. Nevertheless, the edema cleared shortly after implantation. This report illustrates the potential for significant alveolar fluid clearance and sealing of vascular injury after implantation when edema is not caused by inflammatory injury.
Collapse
Affiliation(s)
- Jennifer Whiteley
- Toronto Lung Transplant Program, Toronto General Hospital, Toronto, Canada
| | - Hongchao Shan
- Toronto Lung Transplant Program, Toronto General Hospital, Toronto, Canada
| | - Jonathan C. Yeung
- Toronto Lung Transplant Program, Toronto General Hospital, Toronto, Canada
| |
Collapse
|
4
|
Rocha NN, Silva PL, Battaglini D, Rocco PRM. Heart-lung crosstalk in acute respiratory distress syndrome. Front Physiol 2024; 15:1478514. [PMID: 39493867 PMCID: PMC11527665 DOI: 10.3389/fphys.2024.1478514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024] Open
Abstract
Acute Respiratory Distress Syndrome (ARDS) is initiated by a primary insult that triggers a cascade of pathological events, including damage to lung epithelial and endothelial cells, extracellular matrix disruption, activation of immune cells, and the release of pro-inflammatory mediators. These events lead to increased alveolar-capillary barrier permeability, resulting in interstitial/alveolar edema, collapse, and subsequent hypoxia and hypercapnia. ARDS not only affects the lungs but also significantly impacts the cardiovascular system. We conducted a comprehensive literature review on heart-lung crosstalk in ARDS, focusing on the pathophysiology, effects of mechanical ventilation, hypoxemia, and hypercapnia on cardiac function, as well as ARDS secondary to cardiac arrest and cardiac surgery. Mechanical ventilation, essential for ARDS management, can increase intrathoracic pressure, decrease venous return and right ventricle preload. Moreover, acidemia and elevations in transpulmonary pressures with mechanical ventilation both increase pulmonary vascular resistance and right ventricle afterload. Cardiac dysfunction can exacerbate pulmonary edema and impair gas exchange, creating a vicious cycle, which hinders both heart and lung therapy. In conclusion, understanding the heart-lung crosstalk in ARDS is important to optimize therapeutic strategies. Future research should focus on elucidating the precise mechanisms underlying this interplay and developing targeted interventions that address both organs simultaneously.
Collapse
Affiliation(s)
- Nazareth N. Rocha
- Biomedical Institute, Department of Physiology and Pharmacology, Fluminense Federal University, Niteroi, Brazil
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L. Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise Battaglini
- Anesthesia and Intensive Care, IRCCS Ospedale Policlinico, Genova, Italy
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Genova, Italy
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Li P, Liu H, Li F, Sui M, Liu K, Fu H. Genetic effect of thyroid function-related diseases on acute respiratory distress syndrome: a Mendelian randomization study. Aging (Albany NY) 2024; 16:12209-12224. [PMID: 39225634 PMCID: PMC11424573 DOI: 10.18632/aging.205996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/30/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Previous studies have shown an association between acute respiratory distress syndrome (ARDS) and thyroid function. However, their causal relationship remains unspecified. Therefore, this study aims to explore the causal relationship between ARDS and thyroid function-related diseases with Mendelian Randomization (MR) analysis. METHODS ARDS dataset finn-b-J10_ARDS, finn-b-E4_THYROID dataset of disorders of the thyroid gland (DTG) and finn-b-E4_HYTHYNAS of hypothyroidism were acquired from public database. In univariate MR (UVMR), causal effects between DTG, hypothyroidism and ARDS were investigated using 5 types of algorithms, and reliability was validated by sensitivity analysis. Moreover, multivariate MR (MVMR), enrichment and interaction network analyses of genes corresponding to SNPs of DTG and hypothyroidism were carried out. Significant level was chosen as p<0.05. RESULTS UVMR identified DTG and hypothyroidism (P < 0.05, OR > 1) as risk factors, and were causally related to ARDS. Reliability of UVMR results was confirmed through sensitivity analysis, and results were stable and reliable. However, DTG and hypothyroidism had no effect on ARDS in MVMR, possibly because these factors had independent effects on ARDS. Ultimately, 96 and 113 genes corresponding to SNPs of DTG and hypothyroidism were found closely related to immune-related pathways. CONCLUSIONS UVMR and MVMR analysis revealed a causal connection between DTG and hypothyroidism as risk factors with ARDS, providing robust evidence for investigation into relationship of hypothyroidism on ARDS and between DTG and ARDS.
Collapse
Affiliation(s)
- Peilong Li
- Children’s Hospital Affiliated to Kunming Medical University, Kunming Medical University and Kunming Children’s Hospital, Kunming, P.R. China
- Department of Pulmonary and Critical Care Medicine, Kunming Children’s Hospital and Yunnan Key Laboratory of Children’s Major Disease Research, Kunming, P.R. China
| | - Haifeng Liu
- Children’s Hospital Affiliated to Kunming Medical University, Kunming Medical University and Kunming Children’s Hospital, Kunming, P.R. China
- Department of Pulmonary and Critical Care Medicine, Kunming Children’s Hospital and Yunnan Key Laboratory of Children’s Major Disease Research, Kunming, P.R. China
| | - Feng Li
- Children’s Hospital Affiliated to Kunming Medical University, Kunming Medical University and Kunming Children’s Hospital, Kunming, P.R. China
| | - Mingze Sui
- Children’s Hospital Affiliated to Kunming Medical University, Kunming Medical University and Kunming Children’s Hospital, Kunming, P.R. China
- Department of Pulmonary and Critical Care Medicine, Kunming Children’s Hospital and Yunnan Key Laboratory of Children’s Major Disease Research, Kunming, P.R. China
| | - Kai Liu
- Children’s Hospital Affiliated to Kunming Medical University, Kunming Medical University and Kunming Children’s Hospital, Kunming, P.R. China
| | - Hongmin Fu
- Children’s Hospital Affiliated to Kunming Medical University, Kunming Medical University and Kunming Children’s Hospital, Kunming, P.R. China
| |
Collapse
|
6
|
Taenaka H, Wick KD, Sarma A, Matsumoto S, Ghale R, Fang X, Maishan M, Gotts JE, Langelier CR, Calfee CS, Matthay MA. Biological effects of corticosteroids on pneumococcal pneumonia in Mice-translational significance. Crit Care 2024; 28:185. [PMID: 38807178 PMCID: PMC11134653 DOI: 10.1186/s13054-024-04956-6] [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: 02/17/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Streptococcus pneumoniae is the most common bacterial cause of community acquired pneumonia and the acute respiratory distress syndrome (ARDS). Some clinical trials have demonstrated a beneficial effect of corticosteroid therapy in community acquired pneumonia, COVID-19, and ARDS, but the mechanisms of this benefit remain unclear. The primary objective of this study was to investigate the effects of corticosteroids on the pulmonary biology of pneumococcal pneumonia in a mouse model. A secondary objective was to identify shared transcriptomic features of pneumococcal pneumonia and steroid treatment in the mouse model and clinical samples. METHODS We carried out comprehensive physiologic, biochemical, and histological analyses in mice to identify the mechanisms of lung injury in Streptococcus pneumoniae with and without adjunctive steroid therapy. We also studied lower respiratory tract gene expression from a cohort of 15 mechanically ventilated patients (10 with Streptococcus pneumoniae and 5 controls) to compare with the transcriptional studies in the mice. RESULTS In mice with pneumonia, dexamethasone in combination with ceftriaxone reduced (1) pulmonary edema formation, (2) alveolar protein permeability, (3) proinflammatory cytokine release, (4) histopathologic lung injury score, and (5) hypoxemia but did not increase bacterial burden. Transcriptomic analyses identified effects of steroid therapy in mice that were also observed in the clinical samples. CONCLUSIONS In combination with appropriate antibiotic therapy in mice, treatment of pneumococcal pneumonia with steroid therapy reduced hypoxemia, pulmonary edema, lung permeability, and histologic criteria of lung injury, and also altered inflammatory responses at the protein and gene expression level. The transcriptional studies in patients suggest that the mouse model replicates some of the features of pneumonia in patients with Streptococcus pneumoniae and steroid treatment. Overall, these studies provide evidence for the mechanisms that may explain the beneficial effects of glucocorticoid therapy in patients with community acquired pneumonia from Streptococcus Pneumoniae.
Collapse
Affiliation(s)
- Hiroki Taenaka
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA.
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Japan.
| | - Katherine D Wick
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Aartik Sarma
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
| | - Shotaro Matsumoto
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
- Department of Intensive Care Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rajani Ghale
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
| | - Xiaohui Fang
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Mazharul Maishan
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Jeffrey E Gotts
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Charles R Langelier
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Chan Zuckerberg Biohub, San Francisco, USA
| | - Carolyn S Calfee
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Michael A Matthay
- Department of Medicine, University of California, 513 Parnassus Avenue, HSE RM-760, San Francisco, CA, 94143, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
7
|
Taenaka H, Wick KD, Sarma A, Matsumoto S, Ghale R, Fang X, Maishan M, Gotts JE, Langelier CR, Calfee CS, Matthay MA. Biological Effects of Corticosteroids on Pneumococcal Pneumonia in Mice and Humans. RESEARCH SQUARE 2024:rs.3.rs-3962861. [PMID: 38464245 PMCID: PMC10925444 DOI: 10.21203/rs.3.rs-3962861/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Streptococcus pneumoniae is the most common bacterial cause of community acquired pneumonia and the acute respiratory distress syndrome (ARDS). Some clinical trials have demonstrated a beneficial effect of corticosteroid therapy in community acquired pneumonia, COVID-19, and ARDS, but the mechanisms of this benefit remain unclear. The objective of this study was to investigate the effects of corticosteroids on the pulmonary biology of pneumococcal pneumonia in an observational cohort of mechanically ventilated patients and in a mouse model of bacterial pneumonia with Streptococcus pneumoniae. Methods We studied gene expression with lower respiratory tract transcriptomes from a cohort of mechanically ventilated patients and in mice. We also carried out comprehensive physiologic, biochemical, and histological analyses in mice to identify the mechanisms of lung injury in Streptococcus pneumoniae with and without adjunctive steroid therapy. Results Transcriptomic analysis identified pleiotropic effects of steroid therapy on the lower respiratory tract in critically ill patients with pneumococcal pneumonia, findings that were reproducible in mice. In mice with pneumonia, dexamethasone in combination with ceftriaxone reduced (1) pulmonary edema formation, (2) alveolar protein permeability, (3) proinflammatory cytokine release, (4) histopathologic lung injury score, and (5) hypoxemia but did not increase bacterial burden. Conclusions The gene expression studies in patients and in the mice support the clinical relevance of the mouse studies, which replicate several features of pneumococcal pneumonia and steroid therapy in humans. In combination with appropriate antibiotic therapy in mice, treatment of pneumococcal pneumonia with steroid therapy reduced hypoxemia, pulmonary edema, lung permeability, and histologic criteria of lung injury, and also altered inflammatory responses at the protein and gene expression level. The results from these studies provide evidence for the mechanisms that may explain the beneficial effects of glucocorticoid therapy in patients with community acquired pneumonia from Streptococcus Pneumoniae.
Collapse
|
8
|
Silva AR, de Souza e Souza KFC, Souza TBD, Younes-Ibrahim M, Burth P, de Castro Faria Neto HC, Gonçalves-de-Albuquerque CF. The Na/K-ATPase role as a signal transducer in lung inflammation. Front Immunol 2024; 14:1287512. [PMID: 38299144 PMCID: PMC10827986 DOI: 10.3389/fimmu.2023.1287512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/26/2023] [Indexed: 02/02/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is marked by damage to the capillary endothelium and alveolar epithelium following edema formation and cell infiltration. Currently, there are no effective treatments for severe ARDS. Pathologies such as sepsis, pneumonia, fat embolism, and severe trauma may cause ARDS with respiratory failure. The primary mechanism of edema clearance is the epithelial cells' Na/K-ATPase (NKA) activity. NKA is an enzyme that maintains the electrochemical gradient and cell homeostasis by transporting Na+ and K+ ions across the cell membrane. Direct injury on alveolar cells or changes in ion transport caused by infections decreases the NKA activity, loosening tight junctions in epithelial cells and causing edema formation. In addition, NKA acts as a receptor triggering signal transduction in response to the binding of cardiac glycosides. The ouabain (a cardiac glycoside) and oleic acid induce lung injury by targeting NKA. Besides enzymatic inhibition, the NKA triggers intracellular signal transduction, fostering proinflammatory cytokines production and contributing to lung injury. Herein, we reviewed and discussed the crucial role of NKA in edema clearance, lung injury, and intracellular signaling pathway activation leading to lung inflammation, thus putting the NKA as a protagonist in lung injury pathology.
Collapse
Affiliation(s)
- Adriana Ribeiro Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Thamires Bandeira De Souza
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | - Mauricio Younes-Ibrahim
- Departamento de Medicina Interna, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Burth
- Departamento de Biologia Celular e Molecular, Instituto de Biologia, Universidade Federal Fluminense, Niterói, Brazil
| | | | - Cassiano Felippe Gonçalves-de-Albuquerque
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
- Laboratório de Imunofarmacologia, Departamento de Ciências Fisiológicas, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Gałgańska H, Jarmuszkiewicz W, Gałgański Ł. Carbon dioxide and MAPK signalling: towards therapy for inflammation. Cell Commun Signal 2023; 21:280. [PMID: 37817178 PMCID: PMC10566067 DOI: 10.1186/s12964-023-01306-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/05/2023] [Indexed: 10/12/2023] Open
Abstract
Inflammation, although necessary to fight infections, becomes a threat when it exceeds the capability of the immune system to control it. In addition, inflammation is a cause and/or symptom of many different disorders, including metabolic, neurodegenerative, autoimmune and cardiovascular diseases. Comorbidities and advanced age are typical predictors of more severe cases of seasonal viral infection, with COVID-19 a clear example. The primary importance of mitogen-activated protein kinases (MAPKs) in the course of COVID-19 is evident in the mechanisms by which cells are infected with SARS-CoV-2; the cytokine storm that profoundly worsens a patient's condition; the pathogenesis of diseases, such as diabetes, obesity, and hypertension, that contribute to a worsened prognosis; and post-COVID-19 complications, such as brain fog and thrombosis. An increasing number of reports have revealed that MAPKs are regulated by carbon dioxide (CO2); hence, we reviewed the literature to identify associations between CO2 and MAPKs and possible therapeutic benefits resulting from the elevation of CO2 levels. CO2 regulates key processes leading to and resulting from inflammation, and the therapeutic effects of CO2 (or bicarbonate, HCO3-) have been documented in all of the abovementioned comorbidities and complications of COVID-19 in which MAPKs play roles. The overlapping MAPK and CO2 signalling pathways in the contexts of allergy, apoptosis and cell survival, pulmonary oedema (alveolar fluid resorption), and mechanical ventilation-induced responses in lungs and related to mitochondria are also discussed. Video Abstract.
Collapse
Affiliation(s)
- Hanna Gałgańska
- Faculty of Biology, Molecular Biology Techniques Laboratory, Adam Mickiewicz University in Poznan, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Wieslawa Jarmuszkiewicz
- Faculty of Biology, Department of Bioenergetics, Adam Mickiewicz University in Poznan, Institute of Molecular Biology and Biotechnology, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland
| | - Łukasz Gałgański
- Faculty of Biology, Department of Bioenergetics, Adam Mickiewicz University in Poznan, Institute of Molecular Biology and Biotechnology, Uniwersytetu Poznanskiego 6, 61-614, Poznan, Poland.
| |
Collapse
|
10
|
Liu J, Schiralli-Lester GM, Norman R, Dean DA. Upregulation of alveolar fluid clearance is not sufficient for Na +,K +-ATPase β subunit-mediated gene therapy of LPS-induced acute lung injury in mice. Sci Rep 2023; 13:6792. [PMID: 37100889 PMCID: PMC10130817 DOI: 10.1038/s41598-023-33985-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 04/21/2023] [Indexed: 04/28/2023] Open
Abstract
Acute Lung Injury/Acute Respiratory Distress Syndrome (ALI/ARDS) is characterized by diffuse alveolar damage and significant edema accumulation, which is associated with impaired alveolar fluid clearance (AFC) and alveolar-capillary barrier disruption, leading to acute respiratory failure. Our previous data showed that electroporation-mediated gene delivery of the Na+, K+-ATPase β1 subunit not only increased AFC, but also restored alveolar barrier function through upregulation of tight junction proteins, leading to treatment of LPS-induced ALI in mice. More importantly, our recent publication showed that gene delivery of MRCKα, the downstream effector of β1 subunit-mediated signaling towards upregulation of adhesive junctions and epithelial and endothelial barrier integrity, also provided therapeutic potential for ARDS treatment in vivo but without necessarily accelerating AFC, indicating that for ARDS treatment, improving alveolar capillary barrier function may be of more benefit than improving fluid clearance. In the present study, we investigated the therapeutical potential of β2 and β3 subunits, the other two β isoforms of Na+, K+-ATPase, for LPS-induced ALI. We found that gene transfer of either the β1, β2, or β3 subunits significantly increased AFC compared to the basal level in naïve animals and each gave similar increased AFC to each other. However, unlike that of the β1 subunit, gene transfer of the β2 or β3 subunit into pre-injured animal lungs failed to show the beneficial effects of attenuated histological damage, neutrophil infiltration, overall lung edema, or increased lung permeability, indicating that β2 or β3 gene delivery could not treat LPS induced lung injury. Further, while β1 gene transfer increased levels of key tight junction proteins in the lungs of injured mice, that of either the β2 or β3 subunit had no effect on levels of tight junction proteins. Taken together, this strongly suggests that restoration of alveolar-capillary barrier function alone may be of equal or even more benefit than improving AFC for ALI/ARDS treatment.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Gillian M Schiralli-Lester
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - Rosemary Norman
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA
| | - David A Dean
- Department of Pediatrics, University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA.
- Department of Pharmacology and Physiology, University of Rochester, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
11
|
Duan Q, Zhang Y, Yang D. Perioperative fluid management for lung transplantation is challenging. Heliyon 2023; 9:e14704. [PMID: 37035359 PMCID: PMC10073756 DOI: 10.1016/j.heliyon.2023.e14704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/24/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
Lung transplantation is the definitive end-stage treatment for many lung diseases, and postoperative pulmonary oedema severely affects survival after lung transplantation. Optimizing perioperative fluid management can reduce the incidence of postoperative pulmonary oedema and improve the prognosis of lung transplant patients by removing the influence of patient, donor's lung and ECMO factors. Therefore, this article reviews seven aspects of lung transplant patients' pathophysiological characteristics, physiological characteristics of fluids, the influence of the donor lung on pulmonary oedema as well as current fluid rehydration concepts, advantages or disadvantages of intraoperative monitoring tools or types of fluids on postoperative pulmonary oedema, while showing the existing challenges in section 7. The aim is to show the specificity of perioperative fluid management in lung transplant patients and to provide new ideas for individualised fluid management in lung transplantation.
Collapse
Affiliation(s)
- Qirui Duan
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | - Yajun Zhang
- China-Japan Friendship Hospital, Beijing, 100020, China
- Corresponding author.
| | - Dong Yang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
- Corresponding author.,
| |
Collapse
|
12
|
Taenaka H, Matthay MA. Mechanisms of impaired alveolar fluid clearance. Anat Rec (Hoboken) 2023:10.1002/ar.25166. [PMID: 36688689 PMCID: PMC10564110 DOI: 10.1002/ar.25166] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/09/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023]
Abstract
Impaired alveolar fluid clearance (AFC) is an important cause of alveolar edema fluid accumulation in patients with acute respiratory distress syndrome (ARDS). Alveolar edema leads to insufficient gas exchange and worse clinical outcomes. Thus, it is important to understand the pathophysiology of impaired AFC in order to develop new therapies for ARDS. Over the last few decades, multiple experimental studies have been done to understand the molecular, cellular, and physiological mechanisms that regulate AFC in the normal and the injured lung. This review provides a review of AFC in the normal lung, focuses on the mechanisms of impaired AFC, and then outlines the regulation of AFC. Finally, we summarize ongoing challenges and possible future research that may offer promising therapies for ARDS.
Collapse
Affiliation(s)
- Hiroki Taenaka
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, USA
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Michael A. Matthay
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, USA
| |
Collapse
|
13
|
Zong HF, Guo G, Liu J, Yang CZ, Bao LL. Influence of Alveolar Fluid on Aquaporins and Na+/K+-ATPase and Its Possible Theoretical or Clinical Significance. Am J Perinatol 2022; 29:1586-1595. [PMID: 33611784 DOI: 10.1055/s-0041-1724001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Pulmonary edema is the most common pathophysiological change in pulmonary disease. Aquaporins (AQPs) and Na+/K+-ATPase play pivotal roles in alveolar fluid clearance. This study aimed to explore the influence of increased alveolar fluid on the absorption of lung fluid. STUDY DESIGN Eighty New Zealand rabbits were randomly divided into eight groups (n = 10 in each group), and models of different alveolar fluid contents were established by the infusion of different volumes of normal saline (NS) via the endotracheal tube. Five animals in each group were sacrificed immediately after infusion to determine the wet/dry ratio, while the remaining animals in each group were killed 4 hours later to determine the wet/dry ratio at 4 hours. Additionally, lung specimens were collected from each group, and quantitative real-time PCR (qRT-PCR), western blot, and immunohistochemical (IHC) analyses of AQPs and Na+/K+-ATPase were performed. RESULTS The qRT-PCR analysis and western blot studies showed markedly decreased mRNA and protein levels of AQP1 and Na+/K+-ATPase when the alveolar fluid volume was ≥6 mL/kg, and the mRNA level of AQP5 was significantly reduced when the alveolar fluid volume was ≥4 mL/kg. In addition, IHC analysis showed the same results. At 4 hours, the lung wet/dry ratio was significantly increased when the alveolar fluid volume was ≥6 mL/kg; however, compared with 0 hours after NS infusion, there was still a significant absorption of alveolar fluid for a period of 4 hours. CONCLUSION The results of this study suggest that increased alveolar fluid may induce the downregulation of the mRNA and protein expression of AQPs and Na+/K+-ATPase, which appear to affect alveolar fluid clearance in rabbit lungs. Early intervention is required to avoid excessive alveolar fluid accumulation. KEY POINTS · The expression levels of AQPs and Na+/K+--ATPase were significantly decreased as alveolar fluid increased.. · At 4 hours, wet/dry ratio was significantly increased when infusion volume was ≥ 6 mL/kg.. · Early intervention is required to avoid excessive alveolar fluid accumulation..
Collapse
Affiliation(s)
- Hai-Feng Zong
- Neonatal Intensive Care Unit, Southern Medical University, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
- Department of Neonatology and NICU, Beijing Chaoyang District Maternal and Child Healthcare Hospital, Beijing, China
| | - Guo Guo
- Department of Neonatology and NICU, Beijing Chaoyang District Maternal and Child Healthcare Hospital, Beijing, China
- Department of Pediatrics, Medical School of Chinese PLA, Beijing, China
- Department of Neonatology, The Fifth Medical Center of the PLA General Hospital, Beijing, China
| | - Jing Liu
- Department of Neonatology and NICU, Beijing Chaoyang District Maternal and Child Healthcare Hospital, Beijing, China
| | - Chuan-Zhong Yang
- Neonatal Intensive Care Unit, Southern Medical University, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Lin-Lin Bao
- Department of Dermatology, Shenzhen People's Hospital, Shenzhen, China
| |
Collapse
|
14
|
Marhuenda E, Villarino A, Narciso M, Elowsson L, Almendros I, Westergren-Thorsson G, Farré R, Gavara N, Otero J. Development of a physiomimetic model of acute respiratory distress syndrome by using ECM hydrogels and organ-on-a-chip devices. Front Pharmacol 2022; 13:945134. [PMID: 36188621 PMCID: PMC9517737 DOI: 10.3389/fphar.2022.945134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Acute Respiratory Distress Syndrome is one of the more common fatal complications in COVID-19, characterized by a highly aberrant inflammatory response. Pre-clinical models to study the effect of cell therapy and anti-inflammatory treatments have not comprehensively reproduced the disease due to its high complexity. This work presents a novel physiomimetic in vitro model for Acute Respiratory Distress Syndrome using lung extracellular matrix-derived hydrogels and organ-on-a-chip devices. Monolayres of primary alveolar epithelial cells were cultured on top of decellullarized lung hydrogels containing primary lung mesenchymal stromal cells. Then, cyclic stretch was applied to mimic breathing, and an inflammatory response was induced by using a bacteriotoxin hit. Having simulated the inflamed breathing lung environment, we assessed the effect of an anti-inflammatory drug (i.e., dexamethasone) by studying the secretion of the most relevant inflammatory cytokines. To better identify key players in our model, the impact of the individual factors (cyclic stretch, decellularized lung hydrogel scaffold, and the presence of mesenchymal stromal cells) was studied separately. Results showed that developed model presented a more reduced inflammatory response than traditional models, which is in line with what is expected from the response commonly observed in patients. Further, from the individual analysis of the different stimuli, it was observed that the use of extracellular matrix hydrogels obtained from decellularized lungs had the most significant impact on the change of the inflammatory response. The developed model then opens the door for further in vitro studies with a better-adjusted response to the inflammatory hit and more robust results in the test of different drugs or cell therapy.
Collapse
Affiliation(s)
- Esther Marhuenda
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Alvaro Villarino
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
| | - Maria Narciso
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- The Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Linda Elowsson
- Lung Biology, Biomedical Center, Department of Medical Science,Lund University, Lund, Sweden
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | | | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Núria Gavara
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- The Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jorge Otero
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, University de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- The Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
- *Correspondence: Jorge Otero,
| |
Collapse
|
15
|
Velankar KY, Mou M, Hartmeier PR, Clegg B, Gawalt ES, Jiang M, Meng WS. Recrystallization of Adenosine for Localized Drug Delivery. Mol Pharm 2022; 19:3394-3404. [PMID: 36001090 DOI: 10.1021/acs.molpharmaceut.2c00527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Adenosine (ADO) is an endogenous metabolite with immense potential to be repurposed as an immunomodulatory therapeutic, as preclinical studies have demonstrated in models of epilepsy, acute respiratory distress syndrome, and traumatic brain injury, among others. The currently licensed products Adenocard and Adenoscan are formulated at 3 mg/mL of ADO for rapid bolus intravenous injection, but the systemic administration of the saline formulations for anti-inflammatory purposes is limited by the nucleoside's profound hemodynamic effects. Moreover, concentrations that can be attained in the airway or the brain through direct instillation or injection are limited by the volumes that can be accommodated in the anatomical space (<5 mL in humans) and the rapid elimination by enzymatic and transport mechanisms in the interstitium (half-life <5 s). As such, highly concentrated formulations of ADO are needed to attain pharmacologically relevant concentrations at sites of tissue injury. Herein, we report a previously uncharacterized crystalline form of ADO (rcADO) in which 6.7 mg/mL of the nucleoside is suspended in water. Importantly, the crystallinity is not diminished in a protein-rich environment, as evidenced by resuspending the crystals in albumin (15% w/v). To the best of our knowledge, this is the first report of crystalline ADO generated using a facile and organic solvent-free method aimed at localized drug delivery. The crystalline suspension may be suitable for developing ADO into injectable formulations for attaining high concentrations of the endogenous nucleoside in inflammatory locales.
Collapse
Affiliation(s)
- Ketki Y Velankar
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Mingyao Mou
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Paul R Hartmeier
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Benjamin Clegg
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, Pennsylvania 15282, United States
| | - Ellen S Gawalt
- Department of Chemistry and Biochemistry, Duquesne University, Pittsburgh, Pennsylvania 15282, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| | - Mo Jiang
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23284, United States.,Center for Pharmaceutical Engineering and Sciences, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Wilson S Meng
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, Pennsylvania 15282, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, United States
| |
Collapse
|
16
|
Griesser E, Schönberger T, Stierstorfer B, Wyatt H, Rist W, Lamla T, Thomas MJ, Lamb D, Geillinger-Kästle KE. Characterization of a flexible AAV-DTR/DT mouse model of acute epithelial lung injury. Am J Physiol Lung Cell Mol Physiol 2022; 323:L206-L218. [PMID: 35762632 DOI: 10.1152/ajplung.00364.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Animal models are important to mimic certain pathways or biological aspects of human pathologies including acute and chronic pulmonary diseases. We developed a novel and flexible mouse model of acute epithelial lung injury based on adeno-associated virus (AAV) variant 6.2 mediated expression of the human diphtheria toxin receptor (DTR). Following intratracheal administration of diphtheria toxin (DT), a cell-specific death of bronchial and alveolar epithelial cells can be observed. In contrast to other lung injury models, the here described mouse model provides the possibility of targeted injury using specific tropisms of AAV vectors or cell type specific promotors to drive the human DTR expression. Also, generation of cell specific mouse lines is not required. Detailed characterization of the AAV-DTR/DT mouse model including titration of viral genome (vg) load and administered DT amount revealed increasing cell numbers in bronchoalveolar lavage (BAL; macrophages, neutrophils, and unspecified cells) and elevation of degenerated cells and infiltrated leukocytes in lung tissue, dependent of vg load and DT dose. Cytokine levels in BAL fluid showed different patterns with higher vg load, e.g. IFNγ, TNFα, and IP10 increasing and IL-5 and IL-6 decreasing, while lung function was not affected. Additionally, laser-capture microdissection (LCM)-based proteomics of bronchial epithelium and alveolar tissue revealed upregulated immune and inflammatory response in all regions and extracellular matrix deposition in infiltrated alveoli. Overall, our novel AAV-DTR/DT model allows investigation of repair mechanisms following epithelial injury and resembles specific mechanistic aspects of acute and chronic pulmonary diseases.
Collapse
Affiliation(s)
- Eva Griesser
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Tanja Schönberger
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Birgit Stierstorfer
- Non-clinical Drug Safety, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Hannah Wyatt
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Wolfgang Rist
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Thorsten Lamla
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Matthew James Thomas
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany.,University of Bath, Bath, United Kingdom
| | - David Lamb
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany
| | - Kerstin E Geillinger-Kästle
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
17
|
Chen B, Gong S, Li M, Liu Y, Nie J, Zheng J, Zheng X, Li J, Gan Y, Su Z, Chen J, Li Y, Xie Q, Yan F. Protective effect of oxyberberine against acute lung injury in mice via inhibiting RhoA/ROCK signaling pathway. Biomed Pharmacother 2022; 153:113307. [PMID: 35753262 DOI: 10.1016/j.biopha.2022.113307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Acute lung injury (ALI), hallmarked with alveolar epithelial barrier impairment and pulmonary edema induced by acute inflammation, presents a severe health burden to the public, due to the limited available interventions. Oxyberberine (OBB), having improved anti-inflammatory activity and safety, is a representative component with various activities derived from berberine, whereas its role against ALI with alveolar epithelial barrier injury remains uncertain. To investigate the influence and underlying mechanisms of OBB on ALI, we induced acute inflammation in mice and A549 cells by using lipopolysaccharide (LPS). Changes in alveolar permeability were assessed by analyzing lung histopathology, measuring the dry/wet weight ratio of the lungs, and altering proinflammatory cytokines and neutrophils levels in the bronchoalveolar lavage fluid (BALF). Parameters of pulmonary permeability were assessed through ELISA, western blotting, quantitative real-time PCR, and immunofluorescence analysis. U46619, the agonist of RhoA/ROCK, was employed to further investigate the mechanism of OBB on ALI. Unexpectedly, we found OBB mitigated lung impairment, pulmonary edema, inflammatory reactions in BALF and lung tissue, reduction in ZO-1, and addition of connexin-43. Besides, OBB markedly reduced the expression of RhoA in association with its downstream factors, which are linked to the intercellular junctions and permeability both in vivo and in vitro. Nevertheless, U46619 abolished the benefits obtained from OBB in A549 cells. In conclusion, these outcomes indicated that OBB exerted RhoA/ROCK inhibitor-like effect to moderate alveolar epithelial barrier impairment and permeability, ultimately preventing ALI progression.
Collapse
Affiliation(s)
- Baoyi Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Shiting Gong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Minhua Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yanlu Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Juan Nie
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Medical School, Hubei Minzu University, Hubei 445000, China
| | - Jingna Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiaohong Zheng
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jincan Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yuxuan Gan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; South China Hospital of Shenzhen University, Shenzhen 518116, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qingfeng Xie
- Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, China; The Second Clinical College Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Fang Yan
- Li Ke and Qi Yu-ru Academic Experience Inheritance Studio, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou 510006, China; The Second Clinical College Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
18
|
Hua Y, Han A, Yu T, Hou Y, Ding Y, Nie H. Small Extracellular Vesicles Containing miR-34c Derived from Bone Marrow Mesenchymal Stem Cells Regulates Epithelial Sodium Channel via Targeting MARCKS. Int J Mol Sci 2022; 23:ijms23095196. [PMID: 35563590 PMCID: PMC9101277 DOI: 10.3390/ijms23095196] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/18/2022] Open
Abstract
Epithelial sodium channel (ENaC) is a pivotal regulator of alveolar fluid clearance in the airway epithelium and plays a key role in the treatment of acute lung injury (ALI), which is mainly composed of the three homologous subunits (α, β and γ). The mechanisms of microRNAs in small extracellular vesicles (sEVs) derived from mesenchymal stem cell (MSC-sEVs) on the regulation of lung ion transport are seldom reported. In this study, we aimed at investigating whether miR-34c had an effect on ENaC dysfunction induced by lipopolysaccharide and explored the underlying mechanism in this process. Primarily, the effect of miR-34c on lung edema and histopathology changes in an ALI mouse model was investigated. Then the uptake of PKH26-labeled sEVs was observed in recipient cells, and we observed that the overexpression of miR-34c in MSC-sEVs could upregulate the LPS-inhibited γ-ENaC expression. The dual luciferase reporter gene assay demonstrated that myristoylated alanine-rich C kinase substrate (MARCKS) was one of target genes of miR-34c, the protein expression of which was negatively correlated with miR-34c. Subsequently, either upregulating miR-34c or knocking down MARCKS could increase the protein expression of phospho-phosphatidylinositol 3-kinase (p-PI3K) and phospho-protein kinase B (p-AKT), implying a downstream regulation pathway was involved. All of the above suggest that miR-34c in MSC-sEVs can attenuate edematous lung injury via enhancing γ-ENaC expression, at least partially, through targeting MARCKS and activating the PI3K/AKT signaling pathway subsequently.
Collapse
|
19
|
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV) and the closely related SARS-CoV-2 are emergent highly pathogenic human respiratory viruses causing acute lethal disease associated with lung damage and dysregulated inflammatory responses. SARS-CoV envelope protein (E) is a virulence factor involved in the activation of various inflammatory pathways. Here, we study the contribution of host miRNAs to the virulence mediated by E protein. Small RNAseq analysis of infected mouse lungs identified miRNA-223 as a potential regulator of pulmonary inflammation, since it was significantly increased in SARS-CoV-WT virulent infection compared to the attenuated SARS-CoV-ΔE infection. In vivo inhibition of miRNA-223-3p increased mRNA levels of pro-inflammatory cytokines and NLRP3 inflammasome, suggesting that during lung infection, miRNA-223 might contribute to restrict an excessive inflammatory response. Interestingly, miRNA-223-3p inhibition also increased the levels of the CFTR transporter, which is involved in edema resolution and was significantly downregulated in the lungs of mice infected with the virulent SARS-CoV-WT virus. At the histopathological level, a decrease in the pulmonary edema was observed when miR-223-3p was inhibited, suggesting that miRNA-223-3p was involved in the regulation of the SARS-CoV-induced inflammatory pathology. These results indicate that miRNA-223 participates in the regulation of E protein-mediated inflammatory response during SARS-CoV infection by targeting different host mRNAs involved in the pulmonary inflammation, and identify miRNA-223 as a potential therapeutic target in SARS-CoV infection.
Collapse
|
20
|
Van Slambrouck J, Van Raemdonck D, Vos R, Vanluyten C, Vanstapel A, Prisciandaro E, Willems L, Orlitová M, Kaes J, Jin X, Jansen Y, Verleden GM, Neyrinck AP, Vanaudenaerde BM, Ceulemans LJ. A Focused Review on Primary Graft Dysfunction after Clinical Lung Transplantation: A Multilevel Syndrome. Cells 2022; 11:cells11040745. [PMID: 35203392 PMCID: PMC8870290 DOI: 10.3390/cells11040745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 02/01/2023] Open
Abstract
Primary graft dysfunction (PGD) is the clinical syndrome of acute lung injury after lung transplantation (LTx). However, PGD is an umbrella term that encompasses the ongoing pathophysiological and -biological mechanisms occurring in the lung grafts. Therefore, we aim to provide a focused review on the clinical, physiological, radiological, histological and cellular level of PGD. PGD is graded based on hypoxemia and chest X-ray (CXR) infiltrates. High-grade PGD is associated with inferior outcome after LTx. Lung edema is the main characteristic of PGD and alters pulmonary compliance, gas exchange and circulation. A conventional CXR provides a rough estimate of lung edema, while a chest computed tomography (CT) results in a more in-depth analysis. Macroscopically, interstitial and alveolar edema can be distinguished below the visceral lung surface. On the histological level, PGD correlates to a pattern of diffuse alveolar damage (DAD). At the cellular level, ischemia-reperfusion injury (IRI) is the main trigger for the disruption of the endothelial-epithelial alveolar barrier and inflammatory cascade. The multilevel approach integrating all PGD-related aspects results in a better understanding of acute lung failure after LTx, providing novel insights for future therapies.
Collapse
Affiliation(s)
- Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Dirk Van Raemdonck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Robin Vos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Cedric Vanluyten
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Elena Prisciandaro
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Pulmonary Circulation Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium;
| | - Michaela Orlitová
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.O.); (A.P.N.)
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Yanina Jansen
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Geert M. Verleden
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Respiratory Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Arne P. Neyrinck
- Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; (M.O.); (A.P.N.)
- Department of Anesthesiology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Bart M. Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
| | - Laurens J. Ceulemans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Lung Transplant Unit, Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; (J.V.S.); (D.V.R.); (R.V.); (C.V.); (A.V.); (E.P.); (J.K.); (X.J.); (Y.J.); (G.M.V.); (B.M.V.)
- Department of Thoracic Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
- Correspondence:
| |
Collapse
|
21
|
Li J, Peng Q, Yang R, Li K, Zhu P, Zhu Y, Zhou P, Szabó G, Zheng S. Application of Mesenchymal Stem Cells During Machine Perfusion: An Emerging Novel Strategy for Organ Preservation. Front Immunol 2022; 12:713920. [PMID: 35024039 PMCID: PMC8744145 DOI: 10.3389/fimmu.2021.713920] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 12/03/2021] [Indexed: 12/24/2022] Open
Abstract
Although solid organ transplantation remains the definitive management for patients with end-stage organ failure, this ultimate treatment has been limited by the number of acceptable donor organs. Therefore, efforts have been made to expand the donor pool by utilizing marginal organs from donation after circulatory death or extended criteria donors. However, marginal organs are susceptible to ischemia-reperfusion injury (IRI) and entail higher requirements for organ preservation. Recently, machine perfusion has emerged as a novel preservation strategy for marginal grafts. This technique continually perfuses the organs to mimic the physiologic condition, allows the evaluation of pretransplant graft function, and more excitingly facilitates organ reconditioning during perfusion with pharmacological, gene, and stem cell therapy. As mesenchymal stem cells (MSCs) have anti-oxidative, immunomodulatory, and regenerative properties, mounting studies have demonstrated the therapeutic effects of MSCs on organ IRI and solid organ transplantation. Therefore, MSCs are promising candidates for organ reconditioning during machine perfusion. This review provides an overview of the application of MSCs combined with machine perfusion for lung, kidney, liver, and heart preservation and reconditioning. Promising preclinical results highlight the potential clinical translation of this innovative strategy to improve the quality of marginal grafts.
Collapse
Affiliation(s)
- Jiale Li
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qinbao Peng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ronghua Yang
- Department of Burn Surgery and Skin Regeneration, The First People's Hospital of Foshan, Foshan, China
| | - Kunsheng Li
- Department of Cardiothoracic Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Peng Zhu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yufeng Zhu
- Laboratory Animal Research Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Pengyu Zhou
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Gábor Szabó
- Department of Cardiac Surgery, Heidelberg University Hospital, Heidelberg, Germany.,Department of Cardiac Surgery, University Hospital Halle (Saale), Halle, Germany
| | - Shaoyi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Liu J, Dean DA. Gene Therapy for Acute Respiratory Distress Syndrome. Front Physiol 2022; 12:786255. [PMID: 35111077 PMCID: PMC8801611 DOI: 10.3389/fphys.2021.786255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating clinical syndrome that leads to acute respiratory failure and accounts for over 70,000 deaths per year in the United States alone, even prior to the COVID-19 pandemic. While its molecular details have been teased apart and its pathophysiology largely established over the past 30 years, relatively few pharmacological advances in treatment have been made based on this knowledge. Indeed, mortality remains very close to what it was 30 years ago. As an alternative to traditional pharmacological approaches, gene therapy offers a highly controlled and targeted strategy to treat the disease at the molecular level. Although there is no single gene or combination of genes responsible for ARDS, there are a number of genes that can be targeted for upregulation or downregulation that could alleviate many of the symptoms and address the underlying mechanisms of this syndrome. This review will focus on the pathophysiology of ARDS and how gene therapy has been used for prevention and treatment. Strategies for gene delivery to the lung, such as barriers encountered during gene transfer, specific classes of genes that have been targeted, and the outcomes of these approaches on ARDS pathogenesis and resolution will be discussed.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| | - David A. Dean
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
23
|
You T, Zhou YR, Liu XC, Li LQ. Risk Factors and Clinical Characteristics of Neonatal Acute Respiratory Distress Syndrome Caused by Early Onset Sepsis. Front Pediatr 2022; 10:847827. [PMID: 35419326 PMCID: PMC8995893 DOI: 10.3389/fped.2022.847827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE To identify risk factors associated with the development of acute respiratory distress syndrome (ARDS) in infants with early onset sepsis (EOS) and to describe the clinical features. METHODS A retrospective study was conducted at the Children's Hospital of Chongqing Medical University between January 2000 and October 2020. The infants were divided into ARDS and non-ARDS groups. Clinical characteristics and risk factors were compared between the two groups. RESULTS Two hundred fifty infants (58 with ARDS) were included. Smaller gestational age, lower birth weight (LBW), lower serum albumin level, a higher rate of preterm birth, premature rupture of membranes, antenatal steroid exposure, and lower Apgar score were associated with an increased development of ARDS by univariate analysis (P < 0.05). LBW (β = -0.001, P = 0.000, OR: 0.999, 95% CI: 0.998-0.999) and low serum albumin levels (β = -0.063, P = 0.022, OR: 0.939, 95% CI: 0.889-0.991) were identified as independent risk factors for the development of ARDS by logistic regression analysis. A higher frequency of complications, including persistent pulmonary hypertension, intraventricular hemorrhage, pulmonary hemorrhage, septic shock, and bronchopulmonary dysplasia, was found in the ARDS group (P < 0.05). The rate of mortality was higher for those in the ARDS group than for those in the non-ARDS group (46.6% vs. 15.6%, χ2 = 24.205, P = 0.000). CONCLUSION Acute respiratory distress syndrome (ARDS) in EOS could lead to a higher frequency of complications and mortality. The risk factors for the development of ARDS were LBW and low serum albumin levels.
Collapse
Affiliation(s)
- Ting You
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yan-Rong Zhou
- Jiulongpo People's Hospital of Chongqing, Chongqing, China
| | - Xiao-Chen Liu
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lu-Quan Li
- Department of Neonatology, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Children's Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
24
|
Ebrahimi M, Rad MTS, Zebardast A, Ayyasi M, Goodarzi G, Tehrani SS. The critical role of mesenchymal stromal/stem cell therapy in COVID-19 patients: An updated review. Cell Biochem Funct 2021; 39:945-954. [PMID: 34545605 PMCID: PMC8652792 DOI: 10.1002/cbf.3670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/02/2021] [Accepted: 09/04/2021] [Indexed: 12/20/2022]
Abstract
New coronavirus disease 2019 (COVID-19), as a pandemic disaster, has drawn the attention of researchers in various fields to discover suitable therapeutic approaches for the management of COVID-19 patients. Currently, there are many worries about the rapid spread of COVID-19; there is no approved treatment for this infectious disease, despite many efforts to develop therapeutic procedures for COVID-19. Emerging evidence shows that mesenchymal stromal/stem cell (MSC) therapy can be a suitable option for the management of COVID-19. These cells have many biological features (including the potential of differentiation, high safety and effectiveness, secretion of trophic factors and immunoregulatory features) that make them suitable for the treatment of various diseases. However, some studies have questioned the positive role of MSC therapy in the treatment of COVID-19. Accordingly, in this paper, we will focus on the therapeutic impacts of MSCs and their critical role in cytokine storm of COVID-19 patients.
Collapse
Affiliation(s)
- Mohsen Ebrahimi
- Neonatal and Child Health Research CenterGolestan University of Medical SciencesGorganIran
| | - Mohammad Taha Saadati Rad
- Psychiatric and Behavioral Sciences Research Center, Addiction Research InstituteMazandaran University of Medical SciencesSariIran
| | - Arghavan Zebardast
- Department of Virology, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Mitra Ayyasi
- Critical Care NursingIslamic Azad University, Sari BranchSariIran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, School of MedicineTehran University of Medical SciencesTehranIran
- Scientific Research CenterTehran University of Medical SciencesTehranIran
| | - Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of MedicineTehran University of Medical SciencesTehranIran
- Scientific Research CenterTehran University of Medical SciencesTehranIran
| |
Collapse
|
25
|
Sriram K, Insel MB, Insel PA. Inhaled β2 Adrenergic Agonists and Other cAMP-Elevating Agents: Therapeutics for Alveolar Injury and Acute Respiratory Disease Syndrome? Pharmacol Rev 2021; 73:488-526. [PMID: 34795026 DOI: 10.1124/pharmrev.121.000356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022] Open
Abstract
Inhaled long-acting β-adrenergic agonists (LABAs) and short-acting β-adrenergic agonists are approved for the treatment of obstructive lung disease via actions mediated by β2 adrenergic receptors (β2-ARs) that increase cellular cAMP synthesis. This review discusses the potential of β2-AR agonists, in particular LABAs, for the treatment of acute respiratory distress syndrome (ARDS). We emphasize ARDS induced by pneumonia and focus on the pathobiology of ARDS and actions of LABAs and cAMP on pulmonary and immune cell types. β2-AR agonists/cAMP have beneficial actions that include protection of epithelial and endothelial cells from injury, restoration of alveolar fluid clearance, and reduction of fibrotic remodeling. β2-AR agonists/cAMP also exert anti-inflammatory effects on the immune system by actions on several types of immune cells. Early administration is likely critical for optimizing efficacy of LABAs or other cAMP-elevating agents, such as agonists of other Gs-coupled G protein-coupled receptors or cyclic nucleotide phosphodiesterase inhibitors. Clinical studies that target lung injury early, prior to development of ARDS, are thus needed to further assess the use of inhaled LABAs, perhaps combined with inhaled corticosteroids and/or long-acting muscarinic cholinergic antagonists. Such agents may provide a multipronged, repurposing, and efficacious therapeutic approach while minimizing systemic toxicity. SIGNIFICANCE STATEMENT: Acute respiratory distress syndrome (ARDS) after pulmonary alveolar injury (e.g., certain viral infections) is associated with ∼40% mortality and in need of new therapeutic approaches. This review summarizes the pathobiology of ARDS, focusing on contributions of pulmonary and immune cell types and potentially beneficial actions of β2 adrenergic receptors and cAMP. Early administration of inhaled β2 adrenergic agonists and perhaps other cAMP-elevating agents after alveolar injury may be a prophylactic approach to prevent development of ARDS.
Collapse
Affiliation(s)
- Krishna Sriram
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| | - Michael B Insel
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| | - Paul A Insel
- Departments of Pharmacology (K.S., P.A.I.) and Medicine (P.A.I.), University of California San Diego, La Jolla, California; Department of Medicine (M.B.I.) University of Arizona, Tucson, Arizona
| |
Collapse
|
26
|
Loke XY, Imran SAM, Tye GJ, Wan Kamarul Zaman WS, Nordin F. Immunomodulation and Regenerative Capacity of MSCs for Long-COVID. Int J Mol Sci 2021; 22:ijms222212421. [PMID: 34830303 PMCID: PMC8625432 DOI: 10.3390/ijms222212421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/26/2022] Open
Abstract
The rapid mutation of the SARS-CoV-2 virus is now a major concern with no effective drugs and treatments. The severity of the disease is linked to the induction of a cytokine storm that promotes extensive inflammation in the lung, leading to many acute lung injuries, pulmonary edema, and eventually death. Mesenchymal stem cells (MSCs) might prove to be a treatment option as they have immunomodulation and regenerative properties. Clinical trials utilizing MSCs in treating acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) have provided a basis in treating post-COVID-19 patients. In this review, we discussed the effects of MSCs as an immunomodulator to reduce the severity and death in patients with COVID-19, including the usage of MSCs as an alternative regenerative therapy in post-COVID-19 patients. This review also includes the current clinical trials in utilizing MSCs and their potential future utilization for long-COVID treatments.
Collapse
Affiliation(s)
- Xin Ya Loke
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (X.Y.L.); (S.A.M.I.)
| | - Siti A. M. Imran
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (X.Y.L.); (S.A.M.I.)
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor 11800, Malaysia;
| | - Wan Safwani Wan Kamarul Zaman
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (X.Y.L.); (S.A.M.I.)
- Correspondence: ; Tel.: +60-38921-5555
| |
Collapse
|
27
|
Licker M, Hagerman A, Bedat B, Ellenberger C, Triponez F, Schorer R, Karenovics W. Restricted, optimized or liberal fluid strategy in thoracic surgery: A narrative review. Saudi J Anaesth 2021; 15:324-334. [PMID: 34764839 PMCID: PMC8579501 DOI: 10.4103/sja.sja_1155_20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/05/2020] [Accepted: 12/06/2020] [Indexed: 11/16/2022] Open
Abstract
Perioperative fluid balance has a major impact on clinical and functional outcome, regardless of the type of interventions. In thoracic surgery, patients are more vulnerable to intravenous fluid overload and to develop acute respiratory distress syndrome and other complications. New insight has been gained on the mechanisms causing pulmonary complications and the role of the endothelial glycocalix layer to control fluid transfer from the intravascular to the interstitial spaces and to promote tissue blood flow. With the implementation of standardized processes of care, the preoperative fasting period has become shorter, surgical approaches are less invasive and patients are allowed to resume oral intake shortly after surgery. Intraoperatively, body fluid homeostasis and adequate tissue oxygen delivery can be achieved using a normovolemic therapy targeting a “near-zero fluid balance” or a goal-directed hemodynamic therapy to maximize stroke volume and oxygen delivery according to the Franck–Starling relationship. In both fluid strategies, the use of cardiovascular drugs is advocated to counteract the anesthetic-induced vasorelaxation and maintain arterial pressure whereas fluid intake is limited to avoid cumulative fluid balance exceeding 1 liter and body weight gain (~1-1.5 kg). Modern hemodynamic monitors provide valuable physiological parameters to assess patient volume responsiveness and circulatory flow while guiding fluid administration and cardiovascular drug therapy. Given the lack of randomized clinical trials, controversial debate still surrounds the issues of the optimal fluid strategy and the type of fluids (crystalloids versus colloids). To avoid the risk of lung hydrostatic or inflammatory edema and to enhance the postoperative recovery process, fluid administration should be prescribed as any drug, adapted to the patient's requirement and the context of thoracic intervention.
Collapse
Affiliation(s)
- Marc Licker
- Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospital of Geneva, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Andres Hagerman
- Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Benoit Bedat
- Division of Thoracic and Endocrine Surgery, University Hospital of Geneva, Geneva, Switzerland
| | - Christoph Ellenberger
- Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospital of Geneva, Geneva, Switzerland.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Frederic Triponez
- Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Thoracic and Endocrine Surgery, University Hospital of Geneva, Geneva, Switzerland
| | - Raoul Schorer
- Department of Anesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospital of Geneva, Geneva, Switzerland
| | - Wolfram Karenovics
- Division of Thoracic and Endocrine Surgery, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
28
|
Liu J, Dean DA. Gene transfer of MRCKα rescues lipopolysaccharide-induced acute lung injury by restoring alveolar capillary barrier function. Sci Rep 2021; 11:20862. [PMID: 34675326 PMCID: PMC8531330 DOI: 10.1038/s41598-021-99897-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/27/2021] [Indexed: 01/08/2023] Open
Abstract
Acute Lung Injury/Acute Respiratory Distress Syndrome (ALI/ARDS) is characterized by alveolar edema accumulation with reduced alveolar fluid clearance (AFC), alveolar-capillary barrier disruption, and substantial inflammation, all leading to acute respiratory failure. Enhancing AFC has long been considered one of the primary therapeutic goals in gene therapy treatments for ARDS. We previously showed that electroporation-mediated gene delivery of the Na+, K+-ATPase β1 subunit not only increased AFC, but also restored alveolar barrier function through upregulation of tight junction proteins, leading to treatment of LPS-induced ALI in mice. We identified MRCKα as an interaction partner of β1 which mediates this upregulation in cultured alveolar epithelial cells. In this study, we investigate whether electroporation-mediated gene transfer of MRCKα to the lungs can attenuate LPS-induced acute lung injury in vivo. Compared to mice that received a non-expressing plasmid, those receiving the MRCKα plasmid showed attenuated LPS-increased pulmonary edema and lung leakage, restored tight junction protein expression, and improved overall outcomes. Interestingly, gene transfer of MRCKα did not alter AFC rates. Studies using both cultured microvascular endothelial cells and mice suggest that β1 and MRCKα upregulate junctional complexes in both alveolar epithelial and capillary endothelial cells, and that one or both barriers may be positively affected by our approach. Our data support a model of treatment for ALI/ARDS in which improvement of alveolar-capillary barrier function alone may be of more benefit than improvement of alveolar fluid clearance.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA.,Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - David A Dean
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, 601 Elmwood Avenue, Box 850, Rochester, NY, 14642, USA. .,Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA.
| |
Collapse
|
29
|
Yang Q, Xu HR, Xiang SY, Zhang C, Ye Y, Shen CX, Mei HX, Zhang PH, Ma HY, Zheng SX, Smith FG, Jin SW, Wang Q. Resolvin Conjugates in Tissue Regeneration 1 Promote Alveolar Fluid Clearance by Activating Alveolar Epithelial Sodium Channels and Na, K-ATPase in Lipopolysaccharide-Induced Acute Lung Injury. J Pharmacol Exp Ther 2021; 379:156-165. [PMID: 34465632 DOI: 10.1124/jpet.121.000712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/18/2021] [Indexed: 01/09/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS), a common and fatal clinical condition, is characterized by the destruction of epithelium and augmented permeability of the alveolar-capillary barrier. Resolvin conjugates in tissue regeneration 1 (RCTR1) is an endogenous lipid mediator derived from docosahexaenoic acid , exerting proresolution effects in the process of inflammation. In our research, we evaluated the role of RCTR1 in alveolar fluid clearance (AFC) in lipopolysaccharide-induced ARDS/acute lung injury (ALI) rat model. Rats were injected with RCTR1 (5 μg/kg) via caudal veins 8 hours after lipopolysaccharide (LPS) (14 mg/kg) treatment, and then AFC was estimated after 1 hour of ventilation. Primary type II alveolar epithelial cells were incubated with LPS (1 ug/ml) with or without RCTR1 (10 nM) for 8 hours. Our results showed that RCTR1 significantly enhanced the survival rate, promoted the AFC, and alleviated LPS-induced ARDS/ALI in vivo. Furthermore, RCTR1 remarkably elevated the protein expression of sodium channels and Na, K-ATPase and the activity of Na, K-ATPase in vivo and in vitro. Additionally, RCTR1 also decreased neural precursor cell expressed developmentally downregulated 4-2 (Nedd4-2) level via upregulating Ser473-phosphorylated-Akt expression. Besides this, inhibitors of receptor for lipoxin A4 (ALX), cAMP, and phosphatidylinositol 3-kinase (PI3K) (BOC-2, KH-7, and LY294002) notably inhibited the effects of RCTR1 on AFC. In summary, RCTR1 enhances the protein levels of sodium channels and Na, K-ATPase and the Na, K-ATPase activity to improve AFC in ALI through ALX/cAMP/PI3K/Nedd4-2 pathway, suggesting that RCTR1 may become a therapeutic drug for ARDS/ALI. SIGNIFICANCE STATEMENT: RCTR1, an endogenous lipid mediator, enhanced the rate of AFC to accelerate the resolution of inflammation in the LPS-induced murine lung injury model. RCTR1 upregulates the expression of epithelial sodium channels (ENaCs) and Na, K-ATPase in vivo and in vitro to accelerate the AFC. The efficacy of RCTR1 on the ENaC and Na, K-ATPase level was in an ALX/cAMP/PI3K/Nedd4-2-dependent manner.
Collapse
Affiliation(s)
- Qian Yang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Hao-Ran Xu
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Shu-Yang Xiang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Chen Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Yang Ye
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Chen-Xi Shen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Hong-Xia Mei
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Pu-Hong Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Hong-Yu Ma
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Sheng-Xing Zheng
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Fang-Gao Smith
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| | - Qian Wang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University (Q.Y., H.X., S.X., Y.Y., C.S., H.M., P.Z., H.Ma, S.Z. F.S., S.J., Q.W.), and Wenzhou Medical University (C.Z.), Zhejiang, China; and Institute of Inflammation and Aging, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom (F.S.)
| |
Collapse
|
30
|
Miyamoto E, Takahagi A, Ohsumi A, Martinu T, Hwang D, Boonstra KM, Joe B, Umana JM, Bei KF, Vosoughi D, Liu M, Cypel M, Keshavjee S, Juvet SC. Ex vivo delivery of regulatory T cells for control of alloimmune priming in the donor lung. Eur Respir J 2021; 59:13993003.00798-2021. [PMID: 34475226 DOI: 10.1183/13993003.00798-2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/17/2021] [Indexed: 11/05/2022]
Abstract
Survival after lung transplantation (LTx) is hampered by uncontrolled inflammation and alloimmunity. Regulatory T cells (Tregs) are being studied as a cellular therapy in solid organ transplantation. Whether these systemically administered Tregs can function at the appropriate location and time is an important concern. We hypothesized that in vitro expanded, recipient-derived Tregs can be delivered to donor lungs prior to LTx via ex vivo lung perfusion (EVLP), maintaining their immunomodulatory ability.In a rat model, Wistar Kyoto (WKy) CD4+CD25high Tregs were expanded in vitro prior to EVLP. Expanded Tregs were administered to Fisher 344 (F344) donor lungs during EVLP; left lungs were transplanted into WKy recipients. Treg localisation and function post-transplant were assessed. In a proof-of-concept experiment, cryopreserved expanded human CD4+CD25+CD127low Tregs were thawed and injected into discarded human lungs during EVLP.Rat Tregs entered the lung parenchyma and retained suppressive function. Expanded Tregs had no adverse effect on donor lung physiology during EVLP; lung water as measured by wet-to-dry weight ratio was reduced by Treg therapy. The administered cells remained in the graft at 3 days post-transplant where they reduced activation of intragraft effector CD4+ T cells; these effects were diminished by day 7. Human Tregs entered the lung parenchyma during EVLP where they expressed key immunoregulatory molecules (CTLA4+, 4-1BB+, CD39+, and CD15s+).Pre-transplant Treg administration can inhibit alloimmunity within the lung allograft at early time points post- transplant. Our organ-directed approach has potential for clinical translation.
Collapse
Affiliation(s)
- Ei Miyamoto
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Akihiro Takahagi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Akihiro Ohsumi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Tereza Martinu
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David Hwang
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Kristen M Boonstra
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Betty Joe
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Juan Mauricio Umana
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ke F Bei
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Vosoughi
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Marcelo Cypel
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Stephen C Juvet
- Latner Thoracic Surgery Research Laboratories, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Wu SW, Peng CK, Wu SY, Wang Y, Yang SS, Tang SE, Huang KL. Obesity Attenuates Ventilator-Induced Lung Injury by Modulating the STAT3-SOCS3 Pathway. Front Immunol 2021; 12:720844. [PMID: 34489970 PMCID: PMC8417798 DOI: 10.3389/fimmu.2021.720844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background Ventilator-induced lung injury (VILI) is characterized by vascular barrier dysfunction and suppression of alveolar fluid clearance (AFC). Obesity itself leads to chronic inflammation, which may initiate an injurious cascade to the lungs and simultaneously induce a protective feedback. In this study, we investigated the protective mechanism of obesity on VILI in a mouse model. Methods The VILI model was set up via 6-h mechanical ventilation with a high tidal volume. Parameters including lung injury score, STAT3/NFκB pathway, and AFC were assessed. Mice with diet-induced obesity were obtained by allowing free access to a high-fat diet since the age of 3 weeks. After a 9-week diet intervention, these mice were sacrificed at the age of 12 weeks. The manipulation of SOCS3 protein was achieved by siRNA knockdown and pharmaceutical stimulation using hesperetin. WNK4 knockin and knockout obese mice were used to clarify the pathway of AFC modulation. Results Obesity itself attenuated VILI. Knockdown of SOCS3 in obese mice offset the protection against VILI afforded by obesity. Hesperetin stimulated SOCS3 upregulation in nonobese mice and provided protection against VILI. In obese mice, the WNK4 axis was upregulated at the baseline, but was significantly attenuated after VILI compared with nonobese mice. At the baseline, the manipulation of SOCS3 by siRNA and hesperetin also led to the corresponding alteration of WNK4, albeit to a lesser extent. After VILI, WNK4 expression correlated with STAT3/NFκB activation, regardless of SOCS3 status. Obese mice carrying WNK4 knockout had VILI with a severity similar to that of wild-type obese mice. The severity of VILI in WNK4-knockin obese mice was counteracted by obesity, similar to that of wild-type nonobese mice only. Conclusions Obesity protects lungs from VILI by upregulating SOCS3, thus suppressing the STAT3/NFκB inflammatory pathway and enhancing WNK4-related AFC. However, WNK4 activation is mainly from direct NFκB downstreaming, and less from SOCS3 upregulation. Moreover, JAK2-STAT3/NFκB signaling predominates the pathogenesis of VILI. Nevertheless, the interaction between SOCS3 and WNK4 in modulating VILI in obesity warrants further investigation.
Collapse
Affiliation(s)
- Shih-Wei Wu
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Yu Wang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Sung-Sen Yang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Division of Nephrology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-En Tang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Tri-Service General Hospital, Taipei, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
32
|
Lin S, Chen Q, Zhang L, Ge S, Luo Y, He W, Xu C, Zeng M. Overexpression of HOXB4 Promotes Protection of Bone Marrow Mesenchymal Stem Cells Against Lipopolysaccharide-Induced Acute Lung Injury Partially Through the Activation of Wnt/β-Catenin Signaling. J Inflamm Res 2021; 14:3637-3649. [PMID: 34349541 PMCID: PMC8326777 DOI: 10.2147/jir.s319416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Pulmonary vascular endothelial cell (EC) injury is recognized as one of the pathological factors of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Bone marrow mesenchymal stem cell (BMSC)-based cytotherapy has attracted substantial attention over recent years as a promising therapeutic approach for ALI/ARDS; however, its use remains limited due to inconsistent efficacy. Currently, gene modification techniques are widely applied to MSCs. In the present study, we aimed to investigate the effect of BMSCs overexpressing Homeobox B4 (HOXB4) on lipopolysaccharide (LPS)-induced EC injury. Methods We used LPS to induce EC injury and established EC-BMSC coculture system using transwell chambers. The effect of BMSCs on ECs was explored by detecting EC proliferation, apoptosis, migration, tube formation, and permeability, and determining whether the Wnt/β-catenin pathway is involved in the regulatory mechanism using XAV-939, inhibitor of Wnt/ β-catenin. Results As compared to BMSCWT, BMSCHOXB4 coculture promoted EC proliferation, migration, and tube formation after LPS stimulation and attenuated LPS-induced EC apoptosis and vascular permeability. Mechanistically, BMSCHOXB4 coculture prevented LPS-induced EC injury by activating the Wnt/β-catenin pathway, which is partially reversible by XAV-939. When cocultured with BMSCHOXB4, pro-inflammatory factors were dramatically decreased and anti-inflammatory factors were greatly increased in the EC medium compared to those in the LPS group (P<0.05). Additionally, when compared to BMSCWT coculture, the BMSCHOXB4 coculture showed an enhanced modulation of IL-6, TNF-α, and IL-10, but there was no statistically significant effect on IL-1β and IL-4. Conclusion Coculturing of BMSCHOXB4 prevented LPS-induced EC injury by reversing the inactivation of the Wnt/β-catenin signaling pathway. An in vivo study remains warranted to ascertain whether engraftment of BMSCHOXB4 can be an attractive strategy for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Shan Lin
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Qingui Chen
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Lishan Zhang
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shanhui Ge
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yuling Luo
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Caixia Xu
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People's Republic of China.,Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
33
|
SOFU M, TOMRUK C, BAŞALOĞLU HK, ÇETİN UYANIKGİL EÖ, UYANIKGİL Y. Koronavirüslerin moleküler yapısı ve tedavide kök hücre kullanımı. EGE TIP DERGISI 2021. [DOI: 10.19161/etd.950623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
34
|
Kryvenko V, Vadász I. Molecular mechanisms of Na,K-ATPase dysregulation driving alveolar epithelial barrier failure in severe COVID-19. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1186-L1193. [PMID: 33689516 PMCID: PMC8238442 DOI: 10.1152/ajplung.00056.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
A significant number of patients with coronavirus disease 2019 (COVID-19) develop acute respiratory distress syndrome (ARDS) that is associated with a poor outcome. The molecular mechanisms driving failure of the alveolar barrier upon severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection remain incompletely understood. The Na,K-ATPase is an adhesion molecule and a plasma membrane transporter that is critically required for proper alveolar epithelial function by both promoting barrier integrity and resolution of excess alveolar fluid, thus enabling appropriate gas exchange. However, numerous SARS-CoV-2-mediated and COVID-19-related signals directly or indirectly impair the function of the Na,K-ATPase, thereby potentially contributing to disease progression. In this Perspective, we highlight some of the putative mechanisms of SARS-CoV-2-driven dysfunction of the Na,K-ATPase, focusing on expression, maturation, and trafficking of the transporter. A therapeutic mean to selectively inhibit the maladaptive signals that impair the Na,K-ATPase upon SARS-CoV-2 infection might be effective in reestablishing the alveolar epithelial barrier and promoting alveolar fluid clearance and thus advantageous in patients with COVID-19-associated ARDS.
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,The Cardio-Pulmonary Institute (CPI), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,The Cardio-Pulmonary Institute (CPI), Giessen, Germany
| |
Collapse
|
35
|
Majolo F, da Silva GL, Vieira L, Timmers LFSM, Laufer S, Goettert MI. Review of Trials Currently Testing Stem Cells for Treatment of Respiratory Diseases: Facts Known to Date and Possible Applications to COVID-19. Stem Cell Rev Rep 2021; 17:44-55. [PMID: 32827081 PMCID: PMC7442550 DOI: 10.1007/s12015-020-10033-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Therapeutic clinical and preclinical studies using cultured cells are on the rise, especially now that the World Health Organization (WHO) declared coronavirus disease 2019 (COVID-19) a "public health emergency of international concern", in January, 2020. Thus, this study aims to review the outcomes of ongoing clinical studies on stem cells in Severe Acute Respiratory Syndrome (SARS), Acute Respiratory Distress Syndrome (ARDS), and Middle East Respiratory Syndrome (MERS). The results will be associated with possible applications to COVID-19. Only three clinical trials related to stem cells are considered complete, whereby two are in Phase 1 and one is in Phase 2. Basically, the ongoing studies on coronavirus are using mesenchymal stem cells (MSCs) derived from bone marrow or the umbilical cord to demonstrate their feasibility, safety, and tolerability. The studies not related to coronavirus are all in ARDS conditions; four of them are in Phase 1 and three in Phase 2. With the COVID-19 boom, many clinical trials are being carried out using different sources with an emphasis on MSC-based therapy used to inhibit inflammation. One of the biggest challenges in the current treatment of COVID-19 is the cytokine storm, however MSCs can prevent or mitigate this cytokine storm through their immunomodulatory capacity. We look forward to the results of the ongoing clinical trials to find a treatment for the disease. Researchers around the world are joining forces to help fight COVID-19. Stem cells used in the current clinical studies are a new therapeutic promise for COVID-19 where pharmacological treatments seem insufficient.Graphical Abstract.
Collapse
Affiliation(s)
- Fernanda Majolo
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil
| | - Guilherme Liberato da Silva
- Medical Sciences Center, Universidade do Vale do Taquari - Univates, Lajeado, Rio Grande do Sul, 95914-014, Brazil
| | - Lucas Vieira
- Medical Sciences Center, Universidade do Vale do Taquari - Univates, Lajeado, Rio Grande do Sul, 95914-014, Brazil
| | - Luís Fernando Saraiva Macedo Timmers
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil
| | - Stefan Laufer
- Medicinal Chemistry, University of Tuebingen, D-72076, Tubingen, Germany
| | - Márcia Inês Goettert
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil.
| |
Collapse
|
36
|
Aubin Vega M, Chupin C, Massé C, Dagenais A, Berthiaume Y, Brochiero E. Impact of ENaC downregulation in transgenic mice on the outcomes of acute lung injury induced by bleomycin. Exp Physiol 2021; 106:1110-1119. [PMID: 33502034 DOI: 10.1113/ep089060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/22/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? How does the downregulation of ENaC, the major driving force for alveolar fluid clearance, impact acute lung injury outcomes induced by bleomycin, featuring alveolar damage, as observed during ARDS exudative phase? What is the main finding and its importance? ENaC downregulation in αENaC(-/-)Tg+ mice did not elicit a substantial worsening impact on the main bleomycin outcomes. In ARDS patients, both ENaC alteration and alveolar damage are observed. Thus, novel therapeutic avenues, favouring alveolar integrity restauration, in addition to lung oedema resolution capacity, mainly driven by ENaC, would be essential. ABSTRACT The exudative phase of acute respiratory distress syndrome (ARDS) is characterized by extended alveolar damage, resulting in accumulation of protein-rich inflammatory oedematous fluid in the alveolar space. Na+ reabsorption through ENaC channels is a major driving force for alveolar fluid clearance (AFC) in physiological and pathological conditions. It has previously been shown that partial αENaC impairment in transgenic (αENaC(-/-)Tg+) mice results in reduced AFC in basal conditions and increased wet/dry ratio after thiourea-induced lung oedema, a model in which the integrity of the alveolar epithelium is preserved. The goal of this study was to further investigate the impact of αENaC downregulation in αENaC(-/-)Tg+ mice using an experimental model of acute lung injury induced by bleomycin. A non-significant trend in enhanced weight loss and mortality rates was observed after the bleomycin challenge in αENaC(-/-)Tg+ compared to wild-type (WT) mice. Bronchoalveolar lavage analyses revealed increased TNFα levels and protein concentrations, as indexes of lung inflammation and alveolar damage, in αENaC(-/-)Tg+ mice, compared to WT, at day 3 post-bleomycin, although a statistical difference was no longer measured at day 7. Differential immune cell counts were similar in WT and αENaC(-/-)Tg+ mice challenged with bleomycin. Moreover, lung weight measurements indicated similar oedema levels in WT mice and in transgenic mice with impaired ENaC channels. Altogether, our data indicated that change in ENaC expression does not elicit a significant impact on lung oedema level/resolution in the bleomycin model, featuring alveolar damage.
Collapse
Affiliation(s)
- Mélissa Aubin Vega
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Cécile Chupin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Chantal Massé
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - André Dagenais
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Yves Berthiaume
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada.,Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec, Canada
| | - Emmanuelle Brochiero
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montréal, Québec, Canada.,Département de Médecine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
37
|
Ellison-Hughes GM, Colley L, O'Brien KA, Roberts KA, Agbaedeng TA, Ross MD. The Role of MSC Therapy in Attenuating the Damaging Effects of the Cytokine Storm Induced by COVID-19 on the Heart and Cardiovascular System. Front Cardiovasc Med 2020; 7:602183. [PMID: 33363221 PMCID: PMC7756089 DOI: 10.3389/fcvm.2020.602183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes coronavirus disease 2019 (COVID-19) has led to 47 m infected cases and 1. 2 m (2.6%) deaths. A hallmark of more severe cases of SARS-CoV-2 in patients with acute respiratory distress syndrome (ARDS) appears to be a virally-induced over-activation or unregulated response of the immune system, termed a "cytokine storm," featuring elevated levels of pro-inflammatory cytokines such as IL-2, IL-6, IL-7, IL-22, CXCL10, and TNFα. Whilst the lungs are the primary site of infection for SARS-CoV-2, in more severe cases its effects can be detected in multiple organ systems. Indeed, many COVID-19 positive patients develop cardiovascular complications, such as myocardial injury, myocarditis, cardiac arrhythmia, and thromboembolism, which are associated with higher mortality. Drug and cell therapies targeting immunosuppression have been suggested to help combat the cytokine storm. In particular, mesenchymal stromal cells (MSCs), owing to their powerful immunomodulatory ability, have shown promise in early clinical studies to avoid, prevent or attenuate the cytokine storm. In this review, we will discuss the mechanistic underpinnings of the cytokine storm on the cardiovascular system, and how MSCs potentially attenuate the damage caused by the cytokine storm induced by COVID-19. We will also address how MSC transplantation could alleviate the long-term complications seen in some COVID-19 patients, such as improving tissue repair and regeneration.
Collapse
Affiliation(s)
- Georgina M. Ellison-Hughes
- Faculty of Life Sciences & Medicine, Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, King's College London Guy's Campus, London, United Kingdom
| | - Liam Colley
- School of Sport, Health, and Exercise Sciences, Bangor University, Bangor, United Kingdom
| | - Katie A. O'Brien
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Kirsty A. Roberts
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Thomas A. Agbaedeng
- Faculty of Health & Medical Sciences, Centre for Heart Rhythm Disorders, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Mark D. Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| |
Collapse
|
38
|
Flory CM, Norris BJ, Larson NA, Coicou LG, Koniar BL, Mysz MA, Rich TP, Ingbar DH, Schumacher RJ. A Preclinical Safety Study of Thyroid Hormone Instilled into the Lungs of Healthy Rats-an Investigational Therapy for ARDS. J Pharmacol Exp Ther 2020; 376:74-83. [PMID: 33127750 DOI: 10.1124/jpet.120.000060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe, life-threatening form of respiratory failure characterized by pulmonary edema, inflammation, and hypoxemia due to reduced alveolar fluid clearance (AFC). Alveolar fluid clearance is required for recovery and effective gas exchange, and higher rates of AFC are associated with reduced mortality. Thyroid hormones play multiple roles in lung function, and L-3,5,3'-triiodothyronine (T3) has multiple effects on lung alveolar type II cells. T3 enhances AFC in normal adult rat lungs when administered intramuscularly and in normal or hypoxia-injured lungs when given intratracheally. The safety of a commercially available formulation of liothyronine sodium (synthetic T3) administered intratracheally was assessed in an Investigational New Drug Application-enabling toxicology study in healthy rats. Instillation of the commercial formulation of T3 without modification rapidly caused tracheal injury and often mortality. Intratracheal instillation of T3 that was reformulated and brought to a neutral pH at the maximum feasible dose of 2.73 µg T3 in 300 µl for 5 consecutive days had no clinically relevant T3-related adverse clinical, histopathologic, or clinical pathology findings. There were no unscheduled deaths that could be attributed to the reformulated T3 or control articles, no differences in the lung weights, and no macroscopic or microscopic findings considered to be related to treatment with T3. This preclinical safety study has paved the way for a phase I/II study to determine the safety and tolerability of a T3 formulation delivered into the lungs of patients with ARDS, including coronavirus disease 2019-associated ARDS, and to measure the effect on extravascular lung water in these patients. SIGNIFICANCE STATEMENT: There is growing interest in treating lung disease with thyroid hormone [triiodothyronine (T3)] in pulmonary edema and acute respiratory distress syndrome (ARDS). However, there is not any published experience on the impact of direct administration of T3 into the lung. An essential step is to determine the safety of multiple doses of T3 administered in a relevant animal species. This study enabled Food and Drug Administration approval of a phase I/II clinical trial of T3 instillation in patients with ARDS, including coronavirus disease 2019-associated ARDS (T3-ARDS ClinicalTrials.gov Identifier NCT04115514).
Collapse
Affiliation(s)
- Craig M Flory
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| | - Beverly J Norris
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| | - Nicole A Larson
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| | - Lia G Coicou
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| | - Brenda L Koniar
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| | - Margaret A Mysz
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| | - Timothy P Rich
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| | - David H Ingbar
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| | - Robert J Schumacher
- Pulmonology, Allergy, Critical Care and Sleep Medicine Division (T.P.R., D.H.I.), Center for Translational Medicine (C.M.F., B.J.N., N.A.L., L.G.C., B.L.K., M.A.M., R.J.S.), University of Minnesota, Minneapolis, Minnesota and Department of Pulmonary, Critical Care, Sleep Medicine and Respiratory Care, Essentia Health, Duluth, Minnesota (T.P.R.)
| |
Collapse
|
39
|
Melo-Narváez MC, Stegmayr J, Wagner DE, Lehmann M. Lung regeneration: implications of the diseased niche and ageing. Eur Respir Rev 2020; 29:29/157/200222. [DOI: 10.1183/16000617.0222-2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022] Open
Abstract
Most chronic and acute lung diseases have no cure, leaving lung transplantation as the only option. Recent work has improved our understanding of the endogenous regenerative capacity of the lung and has helped identification of different progenitor cell populations, as well as exploration into inducing endogenous regeneration through pharmaceutical or biological therapies. Additionally, alternative approaches that aim at replacing lung progenitor cells and their progeny through cell therapy, or whole lung tissue through bioengineering approaches, have gained increasing attention. Although impressive progress has been made, efforts at regenerating functional lung tissue are still ineffective. Chronic and acute lung diseases are most prevalent in the elderly and alterations in progenitor cells with ageing, along with an increased inflammatory milieu, present major roadblocks for regeneration. Multiple cellular mechanisms, such as cellular senescence and mitochondrial dysfunction, are aberrantly regulated in the aged and diseased lung, which impairs regeneration. Existing as well as new human in vitro models are being developed, improved and adapted in order to study potential mechanisms of lung regeneration in different contexts. This review summarises recent advances in understanding endogenous as well as exogenous regeneration and the development of in vitro models for studying regenerative mechanisms.
Collapse
|
40
|
Rogers CJ, Harman RJ, Bunnell BA, Schreiber MA, Xiang C, Wang FS, Santidrian AF, Minev BR. Rationale for the clinical use of adipose-derived mesenchymal stem cells for COVID-19 patients. J Transl Med 2020; 18:203. [PMID: 32423449 PMCID: PMC7232924 DOI: 10.1186/s12967-020-02380-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 05/14/2020] [Indexed: 02/08/2023] Open
Abstract
In late 2019, a novel coronavirus (SARS-CoV-2) emerged in Wuhan, capital city of Hubei province in China. Cases of SARS-CoV-2 infection quickly grew by several thousand per day. Less than 100 days later, the World Health Organization declared that the rapidly spreading viral outbreak had become a global pandemic. Coronavirus disease 2019 (COVID-19) is typically associated with fever and respiratory symptoms. It often progresses to severe respiratory distress and multi-organ failure which carry a high mortality rate. Older patients or those with medical comorbidities are at greater risk for severe disease. Inflammation, pulmonary edema and an over-reactive immune response can lead to hypoxia, respiratory distress and lung damage. Mesenchymal stromal/stem cells (MSCs) possess potent and broad-ranging immunomodulatory activities. Multiple in vivo studies in animal models and ex vivo human lung models have demonstrated the MSC's impressive capacity to inhibit lung damage, reduce inflammation, dampen immune responses and aid with alveolar fluid clearance. Additionally, MSCs produce molecules that are antimicrobial and reduce pain. Upon administration by the intravenous route, the cells travel directly to the lungs where the majority are sequestered, a great benefit for the treatment of pulmonary disease. The in vivo safety of local and intravenous administration of MSCs has been demonstrated in multiple human clinical trials, including studies of acute respiratory distress syndrome (ARDS). Recently, the application of MSCs in the context of ongoing COVID-19 disease and other viral respiratory illnesses has demonstrated reduced patient mortality and, in some cases, improved long-term pulmonary function. Adipose-derived stem cells (ASC), an abundant type of MSC, are proposed as a therapeutic option for the treatment of COVID-19 in order to reduce morbidity and mortality. Additionally, when proven to be safe and effective, ASC treatments may reduce the demand on critical hospital resources. The ongoing COVID-19 outbreak has resulted in significant healthcare and socioeconomic burdens across the globe. There is a desperate need for safe and effective treatments. Cellular based therapies hold great promise for the treatment of COVID-19. This literature summary reviews the scientific rationale and need for clinical studies of adipose-derived stem cells and other types of mesenchymal stem cells in the treatment of patients who suffer with COVID-19.
Collapse
Affiliation(s)
| | | | - Bruce A. Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA USA
| | - Martin A. Schreiber
- Department of Surgery, Oregon Health and Science University, Portland, OR USA
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Fu-Sheng Wang
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center, Beijing, 100039 China
| | | | - Boris R. Minev
- Calidi Biotherapeutics, Inc., San Diego, CA USA
- Department of Radiation Medicine and Applied Sciences, Moores UCSD Cancer Center, San Diego, CA USA
| |
Collapse
|
41
|
Kryvenko V, Wessendorf M, Morty RE, Herold S, Seeger W, Vagin O, Dada LA, Sznajder JI, Vadász I. Hypercapnia Impairs Na,K-ATPase Function by Inducing Endoplasmic Reticulum Retention of the β-Subunit of the Enzyme in Alveolar Epithelial Cells. Int J Mol Sci 2020; 21:E1467. [PMID: 32098115 PMCID: PMC7073107 DOI: 10.3390/ijms21041467] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 01/02/2023] Open
Abstract
Alveolar edema, impaired alveolar fluid clearance, and elevated CO2 levels (hypercapnia) are hallmarks of the acute respiratory distress syndrome (ARDS). This study investigated how hypercapnia affects maturation of the Na,K-ATPase (NKA), a key membrane transporter, and a cell adhesion molecule involved in the resolution of alveolar edema in the endoplasmic reticulum (ER). Exposure of human alveolar epithelial cells to elevated CO2 concentrations caused a significant retention of NKA-β in the ER and, thus, decreased levels of the transporter in the Golgi apparatus. These effects were associated with a marked reduction of the plasma membrane (PM) abundance of the NKA-α/β complex as well as a decreased total and ouabain-sensitive ATPase activity. Furthermore, our study revealed that the ER-retained NKA-β subunits were only partially assembled with NKA α-subunits, which suggests that hypercapnia modifies the ER folding environment. Moreover, we observed that elevated CO2 levels decreased intracellular ATP production and increased ER protein and, particularly, NKA-β oxidation. Treatment with α-ketoglutaric acid (α-KG), which is a metabolite that has been shown to increase ATP levels and rescue mitochondrial function in hypercapnia-exposed cells, attenuated the deleterious effects of elevated CO2 concentrations and restored NKA PM abundance and function. Taken together, our findings provide new insights into the regulation of NKA in alveolar epithelial cells by elevated CO2 levels, which may lead to the development of new therapeutic approaches for patients with ARDS and hypercapnia.
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany; (V.K.); (M.W.); (R.E.M.); (S.H.); (W.S.)
- The Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
| | - Miriam Wessendorf
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany; (V.K.); (M.W.); (R.E.M.); (S.H.); (W.S.)
| | - Rory E. Morty
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany; (V.K.); (M.W.); (R.E.M.); (S.H.); (W.S.)
- The Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Susanne Herold
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany; (V.K.); (M.W.); (R.E.M.); (S.H.); (W.S.)
- The Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
| | - Werner Seeger
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany; (V.K.); (M.W.); (R.E.M.); (S.H.); (W.S.)
- The Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Olga Vagin
- Department of Physiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA;
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Laura A. Dada
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (L.A.D.); (J.I.S.)
| | - Jacob I. Sznajder
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (L.A.D.); (J.I.S.)
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), 35392 Giessen, Germany; (V.K.); (M.W.); (R.E.M.); (S.H.); (W.S.)
- The Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
| |
Collapse
|
42
|
Ba F, Zhou X, Zhang Y, Wu C, Xu S, Wu L, Li J, Yin Y, Gu X. Lipoxin A4 ameliorates alveolar fluid clearance disturbance in lipopolysaccharide-induced lung injury via aquaporin 5 and MAPK signaling pathway. J Thorac Dis 2019; 11:3599-3608. [PMID: 31559067 DOI: 10.21037/jtd.2019.08.86] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background A characteristic of acute lung injury (ALI) is the inflammatory damage of alveolar fluid transport. Lipoxins are endogenous lipids involving in the resolution of inflammation. It is found that lipoxin A4 (LXA4) has the distinct properties to improve the anti-edema and pro-resolution function in inflammation. Since aquaporins (AQPs) have essential roles in the integrity of barrier function during fluid transport, especially AQP5 in the maintaining of the epithelium permeability, the current study is aimed to evaluate the potential role of LXA4 in regulating alveolar fluid clearance (AFC) during fluid transport and the corresponding change of AQP5 in the lung. Methods ALI was induced by the lipopolysaccharide (LPS) intraperitoneal injection, and LXA4 treatment was given 8 hours after LPS administration. We investigated changes in the capacity of AFC, pro-inflammatory cytokine concentrations in bronchoalveolar lavage fluid (BALF) and the severity of ALI. Then AQP5 expression in lung tissue and potential regulatory pathways in LPS-induced ALI was explored. Results LXA4 treatment was found to inhibit AFC capacity, inflammatory cytokine release, partially, alleviate ALI severity, and restored AQP5 expression partially. Additionally, we found that LXA4 played a protective role by the inhibition of the phosphorylation of p38 and JNK. Conclusions In summary, our results suggest that LXA4 plays a protective role in lipopolysaccharide-induced ALI by restoring AFC capacity and upregulating AQP5 expression and inhibiting the phosphorylation of p38 and JNK. These findings suggest potential new mechanism of LXA4 as anti-inflammation therapy for the impairment of alveolar fluid transport in ALI.
Collapse
Affiliation(s)
- Fang Ba
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Xiaoming Zhou
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yingqi Zhang
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Cen Wu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Shenqian Xu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Liqin Wu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jiayang Li
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yan Yin
- Institute of Respiratory Disease, First Hospital of China Medical University, Shenyang 110004, China
| | - Xiu Gu
- Department of Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
43
|
Englert JA, Bobba C, Baron RM. Integrating molecular pathogenesis and clinical translation in sepsis-induced acute respiratory distress syndrome. JCI Insight 2019; 4:e124061. [PMID: 30674720 PMCID: PMC6413834 DOI: 10.1172/jci.insight.124061] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Sepsis-induced acute respiratory distress syndrome (ARDS) has high morbidity and mortality and arises after lung infection or infection at extrapulmonary sites. An aberrant host response to infection leads to disruption of the pulmonary alveolar-capillary barrier, resulting in lung injury characterized by hypoxemia, inflammation, and noncardiogenic pulmonary edema. Despite increased understanding of the molecular biology underlying sepsis-induced ARDS, there are no targeted pharmacologic therapies for this devastating condition. Here, we review the molecular underpinnings of sepsis-induced ARDS with a focus on relevant clinical and translational studies that point toward novel therapeutic strategies.
Collapse
Affiliation(s)
- Joshua A. Englert
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher Bobba
- Division of Pulmonary, Critical Care and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Rebecca M. Baron
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
44
|
Loy H, Kuok DIT, Hui KPY, Choi MHL, Yuen W, Nicholls JM, Peiris JSM, Chan MCW. Therapeutic Implications of Human Umbilical Cord Mesenchymal Stromal Cells in Attenuating Influenza A(H5N1) Virus-Associated Acute Lung Injury. J Infect Dis 2019; 219:186-196. [PMID: 30085072 PMCID: PMC6306016 DOI: 10.1093/infdis/jiy478] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Background Highly pathogenic avian influenza viruses can cause severe forms of acute lung injury (ALI) in humans, where pulmonary flooding leads to respiratory failure. The therapeutic benefits of bone marrow mesenchymal stromal cells (MSCs) have been demonstrated in a model of ALI due to influenza A(H5N1) virus. However, clinical translation is impractical and limited by a decline in efficacy as the age of the donor increases. Umbilical cord MSCs (UC-MSCs) are easier to obtain by comparison, and their primitive source may offer more-potent therapeutic effects. Methods Here we investigate the therapeutic efficacy of UC-MSCs on the mechanisms of pulmonary edema formation and alveolar fluid clearance and protein permeability of A(H5N1)-infected human alveolar epithelial cells. UC-MSCs were also tested in a mouse model of influenza ALI. Results We found that UC-MSCs were effective in restoring impaired alveolar fluid clearance and protein permeability of A(H5N1)-infected human alveolar epithelial cells. UC-MSCs consistently outperformed bone marrow MSCs, partly because of greater growth factor secretion of angiopoietin 1 and hepatocyte growth factor. Conditioned UC-MSC medium and UC-MSC exosomes were also able to recapitulate these effects. However, UC-MSCs only slightly improved survival of A(H5N1)-infected mice. Conclusions Our results suggest that UC-MSCs are effective in restoring alveolar fluid clearance and protein permeability in A(H5N1)-associated ALI and confer functional in addition to practical advantages over conventional bone marrow MSCs.
Collapse
Affiliation(s)
- Hayley Loy
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Denise I T Kuok
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kenrie P Y Hui
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Miranda H L Choi
- Healthbaby Biotech, Hong Kong Special Administrative Region, China
| | - W Yuen
- Healthbaby Biotech, Hong Kong Special Administrative Region, China
| | - John M Nicholls
- Department of Pathology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - J S Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Michael C W Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
45
|
Lai YC, Wang L, Gladwin MT. Insights into the pulmonary vascular complications of heart failure with preserved ejection fraction. J Physiol 2018; 597:1143-1156. [PMID: 30549058 DOI: 10.1113/jp275858] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 11/19/2018] [Indexed: 12/21/2022] Open
Abstract
Pulmonary hypertension in the setting of heart failure with preserved ejection fraction (PH-HFpEF) is a growing public health problem that is increasing in prevalence. While PH-HFpEF is defined by a high mean pulmonary artery pressure, high left ventricular end-diastolic pressure and a normal ejection fraction, some HFpEF patients develop PH in the presence of pulmonary vascular remodelling with a high transpulmonary pressure gradient or pulmonary vascular resistance. Ageing, increased left atrial pressure and stiffness, mitral regurgitation, as well as features of metabolic syndrome, which include obesity, diabetes and hypertension, are recognized as risk factors for PH-HFpEF. Qualitative studies have documented that patients with PH-HFpEF develop more severe symptoms than those with HFpEF and are associated with more significant exercise intolerance, frequent hospitalizations, right heart failure and reduced survival. Currently, there are no effective therapies for PH-HFpEF, although a number of candidate drugs are being evaluated, including soluble guanylate cyclase stimulators, phosphodiesterase type 5 inhibitors, sodium nitrite and endothelin receptor antagonists. In this review we attempt to provide an updated overview of recent findings pertaining to the pulmonary vascular complications in HFpEF in terms of clinical definitions, epidemiology and pathophysiology. Mechanisms leading to pulmonary vascular remodelling in HFpEF, a summary of pre-clinical models of HFpEF and PH-HFpEF, and new candidate therapeutic strategies for the treatment of PH-HFpEF are summarized.
Collapse
Affiliation(s)
- Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Longfei Wang
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.,The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
46
|
Oliveira VR, Uriarte JJ, Falcones B, Zin WA, Navajas D, Farré R, Almendros I. Escherichia coli lipopolysaccharide induces alveolar epithelial cell stiffening. J Biomech 2018; 83:315-318. [PMID: 30527389 DOI: 10.1016/j.jbiomech.2018.11.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/23/2018] [Accepted: 11/24/2018] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Application of lipopolysaccharide (LPS) is a widely employed model to mimic acute respiratory distress syndrome (ARDS). Available data regarding LPS-induced biomechanical changes on pulmonary epithelial cells are limited only to P. aeruginosa LPS. Considering that LPS from different bacteria could promote a specific mechanical response in epithelial cells, we aim to assess the effect of E. coli LPS, widely employed as a model of ARDS, in the biomechanics of alveolar epithelial cells. METHODS Young's modulus (E) of alveolar epithelial cells (A549) was measured by atomic force microscopy every 5 min throughout 60 min of experiment after treatment with LPS from E. coli (100 μg/mL). The percentage of cells presenting actin stress fibers (F-actin staining) was also evaluated. Control cells were treated with culture medium and the values obtained were compared with LPS-treated cells for each time-point. RESULTS Application of LPS induced significant increase in E after 20 min (77%) till 60 min (104%) in comparison to controls. Increase in lung epithelial cell stiffness induced by LPS was associated with a higher number of cells presenting cytoskeletal remodeling. CONCLUSIONS The observed effects of E. coli LPS on alveolar epithelial cells suggest that this widely-used LPS is able to promote a quick formation of actin stress fibers and stiffening cells, thereby facilitating the disruption of the pulmonary epithelial barrier.
Collapse
Affiliation(s)
- Vinícius Rosa Oliveira
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Laboratório de Fisiologia da Respiração, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juan José Uriarte
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias CIBERES, Madrid, Spain
| | - Bryan Falcones
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Walter Araujo Zin
- Laboratório de Fisiologia da Respiração, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel Navajas
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias CIBERES, Madrid, Spain; Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias CIBERES, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer IDIBAPS, Barcelona, Spain
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias CIBERES, Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer IDIBAPS, Barcelona, Spain.
| |
Collapse
|
47
|
Wang Q, Yan SF, Hao Y, Jin SW. Specialized Pro-resolving Mediators Regulate Alveolar Fluid Clearance during Acute Respiratory Distress Syndrome. Chin Med J (Engl) 2018; 131:982-989. [PMID: 29664060 PMCID: PMC5912066 DOI: 10.4103/0366-6999.229890] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Objective Acute respiratory distress syndrome (ARDS) is an acute and lethal clinical syndrome that is characterized by the injury of alveolar epithelium, which impairs active fluid transport in the lung, and impedes the reabsorption of edema fluid from the alveolar space. This review aimed to discuss the role of pro-resolving mediators on the regulation of alveolar fluid clearance (AFC) in ARDS. Data Sources Articles published up to September 2017 were selected from the PubMed, with the keywords of "alveolar fluid clearance" or "lung edema" or "acute lung injury" or "acute respiratory distress syndrome", and "specialized pro-resolving mediators" or "lipoxin" or "resolvin" or "protectin" or "maresin" or "alveolar epithelial cells" or "aspirin-triggered lipid mediators" or "carbon monoxide and heme oxygenase" or "annexin A1". Study Selection We included all relevant articles published up to September 2017, with no limitation of study design. Results Specialized pro-resolving mediators (SPMs), as the proinflammatory mediators, not only upregulated epithelial sodium channel, Na,K-ATPase, cystic fibrosis transmembrane conductance regulator (CFTR), and aquaporins levels, but also improved Na,K-ATPase activity to promote AFC in ARDS. In addition to the direct effects on ion channels and pumps of the alveolar epithelium, the SPMs also inhibited the inflammatory cytokine expression and improved the alveolar epithelial cell repair to enhance the AFC in ARDS. Conclusions The present review discusses a novel mechanism for pulmonary edema fluid reabsorption. SPMs might provide new opportunities to design "reabsorption-targeted" therapies with high degrees of precision in controlling ALI/ARDS.
Collapse
Affiliation(s)
- Qian Wang
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Song-Fan Yan
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yu Hao
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Sheng-Wei Jin
- Department of Anesthesia and Critical Care, Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
48
|
Schwede M, Wilfong EM, Zemans RL, Lee PJ, dos Santos C, Fang X, Matthay MA. Effects of bone marrow-derived mesenchymal stromal cells on gene expression in human alveolar type II cells exposed to TNF-α, IL-1β, and IFN-γ. Physiol Rep 2018; 6:e13831. [PMID: 30136410 PMCID: PMC6105627 DOI: 10.14814/phy2.13831] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/17/2018] [Accepted: 07/21/2018] [Indexed: 01/15/2023] Open
Abstract
The acute respiratory distress syndrome (ARDS) is common in critically ill patients and has a high mortality rate. Mesenchymal stromal cells (MSCs) have demonstrated therapeutic potential in animal models of ARDS, and their benefits occur in part through interactions with alveolar type II (ATII) cells. However, the effects that MSCs have on human ATII cells have not been well studied. Using previously published microarray data, we performed genome-wide differential gene expression analyses of human ATII cells that were (1) unstimulated, (2) exposed to proinflammatory cytokines (CytoMix), or (3) exposed to proinflammatory cytokines plus MSCs. Findings were validated by qPCR. Alveolar type II cells differentially expressed hundreds of genes when exposed either to proinflammatory cytokines or to proinflammatory cytokines plus MSCs. Stimulation with proinflammatory cytokines increased expression of inflammatory genes and downregulated genes related to surfactant function and alveolar fluid clearance. Some of these changes, including expression of some cytokines and genes related to surfactant, were reversed by exposure to MSCs. In addition, MSCs induced upregulation of other potentially beneficial genes, such as those related to extracellular matrix remodeling. We confirmed several of these gene expression changes by qPCR. Thus, ATII cells downregulate genes associated with surfactant and alveolar fluid clearance when exposed to inflammatory cytokines, and mesenchymal stromal cells partially reverse many of these gene expression changes.
Collapse
Affiliation(s)
- Matthew Schwede
- Department of MedicineUniversity of CaliforniaSan FranciscoCalifornia
| | - Erin M. Wilfong
- Division of Allergy, Pulmonary and Critical Care MedicineDepartment of MedicineVanderbilt University Medical CenterNashvilleTennessee
| | - Rachel L. Zemans
- Division of Pulmonary and Critical Care MedicineUniversity of Michigan Medical SchoolAnn ArborMichigan
- Cellular and Molecular Biology ProgramUniversity of Michigan Medical SchoolAnn ArborMichigan
| | - Patty J. Lee
- Section of PulmonaryCritical Care & Sleep MedicineYale University School of MedicineNew HavenConnecticut
| | - Claudia dos Santos
- Interdepartmental Division of Critical Care MedicineSt. Michael's HospitalTorontoOntarioCanada
- Division of RespirologyDepartment of MedicineSt. Michael's HospitalTorontoOntarioCanada
- Li Ka Shing Knowledge InstituteTorontoOntarioCanada
| | - Xiaohui Fang
- Cardiovascular Research InstituteUniversity of CaliforniaSan FranciscoSan FranciscoCalifornia
| | - Michael A. Matthay
- Department of MedicineUniversity of CaliforniaSan FranciscoCalifornia
- Cardiovascular Research InstituteUniversity of CaliforniaSan FranciscoSan FranciscoCalifornia
- Department of AnesthesiaUniversity of CaliforniaSan FranciscoSan FranciscoCalifornia
| |
Collapse
|
49
|
Association of Genetic Variation in the Epithelial Sodium Channel Gene with Urinary Sodium Excretion and Blood Pressure. Nutrients 2018; 10:nu10050612. [PMID: 29757959 PMCID: PMC5986492 DOI: 10.3390/nu10050612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/10/2018] [Accepted: 05/11/2018] [Indexed: 11/17/2022] Open
Abstract
This study was performed to investigate whether genetic variation in the epithelial sodium channel (ENaC) is associated with 24-h urinary sodium excretion and blood pressure. A total of 3345 participants of the KoGES_Ansan and Ansung study were eligible for this study. Genomic DNA samples were isolated from peripheral blood and genotyped on the Affymetrix Genome-Wide Human SNP Array 5.0. Thirty-four single nucleotide polymorphisms (SNPs) were extracted for gene regions (SCNN1A, SCNN1B, and SCNN1G) as additive components by using Plink. Twenty-four-hour sodium excretions were estimated from spot urine samples using the Tanaka formula. The general linear model (GLM) was applied to assess the association between SNPs and urinary sodium excretion or blood pressure. In the SCNN1G gene, six SNPs (rs4073291, rs12934362, rs7404408, rs4494543, rs5735, and rs6497657) were significantly different in 24-h urinary sodium excretion according to gene variants. However, no difference was found in blood pressure among participants with gene variants of ENaC. Our finding indicated that 24-h urinary sodium excretions were different according to variants of the SCNN1G gene in large samples. Further studies to replicate these findings are warranted.
Collapse
|
50
|
Lecuona E, Sznajder JI. Stretching to Understand How Proteostasis and the Unfolded Protein Response Regulate Lung Injury. Am J Respir Cell Mol Biol 2018; 57:143-144. [PMID: 28762764 DOI: 10.1165/rcmb.2017-0170ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Emilia Lecuona
- 1 Pulmonary and Critical Care Medicine Northwestern University Chicago, Illinois
| | - Jacob I Sznajder
- 1 Pulmonary and Critical Care Medicine Northwestern University Chicago, Illinois
| |
Collapse
|