1
|
Zheng Z, Zong Y, Ma Y, Tian Y, Pang Y, Zhang C, Gao J. Glucagon-like peptide-1 receptor: mechanisms and advances in therapy. Signal Transduct Target Ther 2024; 9:234. [PMID: 39289339 PMCID: PMC11408715 DOI: 10.1038/s41392-024-01931-z] [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: 02/18/2024] [Revised: 06/17/2024] [Accepted: 07/16/2024] [Indexed: 09/19/2024] Open
Abstract
The glucagon-like peptide-1 (GLP-1) receptor, known as GLP-1R, is a vital component of the G protein-coupled receptor (GPCR) family and is found primarily on the surfaces of various cell types within the human body. This receptor specifically interacts with GLP-1, a key hormone that plays an integral role in regulating blood glucose levels, lipid metabolism, and several other crucial biological functions. In recent years, GLP-1 medications have become a focal point in the medical community due to their innovative treatment mechanisms, significant therapeutic efficacy, and broad development prospects. This article thoroughly traces the developmental milestones of GLP-1 drugs, from their initial discovery to their clinical application, detailing the evolution of diverse GLP-1 medications along with their distinct pharmacological properties. Additionally, this paper explores the potential applications of GLP-1 receptor agonists (GLP-1RAs) in fields such as neuroprotection, anti-infection measures, the reduction of various types of inflammation, and the enhancement of cardiovascular function. It provides an in-depth assessment of the effectiveness of GLP-1RAs across multiple body systems-including the nervous, cardiovascular, musculoskeletal, and digestive systems. This includes integrating the latest clinical trial data and delving into potential signaling pathways and pharmacological mechanisms. The primary goal of this article is to emphasize the extensive benefits of using GLP-1RAs in treating a broad spectrum of diseases, such as obesity, cardiovascular diseases, non-alcoholic fatty liver disease (NAFLD), neurodegenerative diseases, musculoskeletal inflammation, and various forms of cancer. The ongoing development of new indications for GLP-1 drugs offers promising prospects for further expanding therapeutic interventions, showcasing their significant potential in the medical field.
Collapse
Affiliation(s)
- Zhikai Zheng
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Yiyang Ma
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yucheng Tian
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
2
|
Ex Vivo Pulmonary Oedema after In Vivo Blast-Induced Rat Lung Injury: Time Dependency, Blast Intensity and Beta-2 Adrenergic Receptor Role. Biomedicines 2022; 10:biomedicines10112930. [PMID: 36428498 PMCID: PMC9687465 DOI: 10.3390/biomedicines10112930] [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: 09/01/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Objective: Current treatments for blast-induced lung injury are limited to supportive procedures including mechanical ventilation. The study aimed to investigate the role of post-trauma-induced oedema generation in the function of time and trauma intensity and the probable role of beta 2-adrenergic receptors (β2-ARs) agonists on pulmonary oedema. The study is conducted using an ex vivo model after an experimental in vivo blast-induced thorax trauma in rats. Methods: Rats were randomised and divided into two groups, blast and sham. The blast group were anaesthetised and exposed to the blast wave (3.16 ± 0.43 bar) at a distance of 3.5 cm from the thorax level. The rats were sacrificed 10 min after the blast, the lungs explanted and treated with terbutaline, formoterol, propranolol or amiloride to assess the involvement of sodium transport. Other groups of rats were exposed to distances of 5 and 7 cm from the thorax to reduce the intensity of the injury. Further, one group of rats was studied after 180 min and one after 360 min after a 3.5 cm blast injury. Sham controls were exposed to identical procedures except for receiving blast overpressure. Results: Lung injury and oedema generation depended on time after injury and injury intensity. Perfusion with amiloride resulted in a further increase in oedema formation as indicated by weight gain (p < 0.001), diminished tidal volume (Tv) (p < 0.001), and increased airway resistance (p < 0.001). Formoterol caused a significant increase in the Tv (p < 0.001) and a significant decrease in the airway resistance (p < 0.01), while the lung weight was not influenced. Trauma-related oedema was significantly reduced by terbutaline in terms of lung weight gain (p < 0.01), Tv (p < 0.001), and airway resistance (p < 0.01) compared to control blast-injured lungs. Terbutaline-induced effects were completely blocked by the β-receptor antagonist propranolol (p < 0.05). Similarly, amiloride, which was added to terbutaline perfusion, reversed terbutaline-induced weight gain reduction (p < 0.05). Conclusions: β2-adrenoceptor stimulation had a beneficial impact by amiloride-dependent sodium and therefore, fluid transport mechanisms on the short-term ex vivo oedema generation in a trauma-induced in vivo lung injury of rats.
Collapse
|
3
|
Baloglu E, Velineni K, Ermis-Kaya E, Mairbäurl H. Hypoxia Aggravates Inhibition of Alveolar Epithelial Na-Transport by Lipopolysaccharide-Stimulation of Alveolar Macrophages. Int J Mol Sci 2022; 23:ijms23158315. [PMID: 35955448 PMCID: PMC9368968 DOI: 10.3390/ijms23158315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammation and hypoxia impair alveolar barrier tightness, inhibit Na- and fluid reabsorption, and cause edema. We tested whether stimulated alveolar macrophages affect alveolar Na-transport and whether hypoxia aggravates the effects of inflammation, and tested for involved signaling pathways. Primary rat alveolar type II cells (rA2) were co-cultured with rat alveolar macrophages (NR8383) or treated with NR8383-conditioned media after stimulation with lipopolysaccharide (LPS; 1 µg/mL) and exposed to normoxia and hypoxia (1.5% O2). LPS caused a fast, transient increase in TNFα and IL-6 mRNA in macrophages and a sustained increase in inducible nitric oxide synthase (NOS2) mRNA in macrophages and in rA2 cells resulting in elevated nitrite levels and secretion of TNF-α and IL-6 into culture media. In normoxia, 24 h of LPS treated NR8383 decreased the transepithelial electrical resistance (TEER) of co-cultures, of amiloride-sensitive short circuit current (ISCΔamil); whereas Na/K-ATPase activity was not affected. Inhibition was also seen with conditioned media from LPS-stimulated NR8383 on rA2, but was less pronounced after dialysis to remove small molecules and nitrite. The effect of LPS-stimulated macrophages on TEER and Na-transport was fully prevented by the iNOS-inhibitor L-NMMA applied to co-cultures and to rA2 mono-cultures. Hypoxia in combination with LPS-stimulated NR8383 totally abolished TEER and ISCΔamil. These results indicate that the LPS-stimulation of alveolar macrophages impairs alveolar epithelial Na-transport by NO-dependent mechanisms, where part of the NO is produced by rA2 induced by signals from LPS stimulated alveolar macrophages.
Collapse
Affiliation(s)
- Emel Baloglu
- Department of Medical Pharmacology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
- Translational Lung Research Center Heidelberg (TLRC-H), Part of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany; (K.V.); (E.E.-K.)
| | - Kalpana Velineni
- Translational Lung Research Center Heidelberg (TLRC-H), Part of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany; (K.V.); (E.E.-K.)
| | - Ezgi Ermis-Kaya
- Translational Lung Research Center Heidelberg (TLRC-H), Part of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany; (K.V.); (E.E.-K.)
| | - Heimo Mairbäurl
- Translational Lung Research Center Heidelberg (TLRC-H), Part of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany; (K.V.); (E.E.-K.)
- Medical Clinic VII, Sports Medicine, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Translational Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +49-6221-56-39329
| |
Collapse
|
4
|
Zhou W, Yu T, Hua Y, Hou Y, Ding Y, Nie H. Effects of Hypoxia on Respiratory Diseases: Perspective View of Epithelial Ion Transport. Am J Physiol Lung Cell Mol Physiol 2022; 323:L240-L250. [PMID: 35819839 DOI: 10.1152/ajplung.00065.2022] [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: 12/15/2022] Open
Abstract
The balance of gas exchange and lung ventilation is essential for the maintenance of body homeostasis. There are many ion channels and transporters in respiratory epithelial cells, including epithelial sodium channel, Na,K-ATPase, cystic fibrosis transmembrane conductance regulator, and some transporters. These ion channels/transporters maintain the capacity of liquid layer on the surface of respiratory epithelial cells, and provide an immune barrier for the respiratory system to clear off foreign pathogens. However, in some harmful external environment and/or pathological conditions, the respiratory epithelium is prone to hypoxia, which would destroy the ion transport function of the epithelium and unbalance the homeostasis of internal environment, triggering a series of pathological reactions. Many respiratory diseases associated with hypoxia manifest an increased expression of hypoxia-inducible factor-1, which mediates the integrity of the epithelial barrier and affects epithelial ion transport function. It is important to study the relationship between hypoxia and ion transport function, whereas the mechanism of hypoxia-induced ion transport dysfunction in respiratory diseases is not clear. This review focuses on the relationship of hypoxia and respiratory diseases, as well as dysfunction of ion transport and tight junctions in respiratory epithelial cells under hypoxia.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yu Hua
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
5
|
Baloglu E, Nonnenmacher G, Seleninova A, Berg L, Velineni K, Ermis-Kaya E, Mairbäurl H. The role of hypoxia-induced modulation of alveolar epithelial Na +- transport in hypoxemia at high altitude. Pulm Circ 2020; 10:50-58. [PMID: 33110497 PMCID: PMC7557693 DOI: 10.1177/2045894020936662] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022] Open
Abstract
Reabsorption of excess alveolar fluid is driven by vectorial Na+-transport across alveolar epithelium, which protects from alveolar flooding and facilitates gas exchange. Hypoxia inhibits Na+-reabsorption in cultured cells and in-vivo by decreasing activity of epithelial Na+-channels (ENaC), which impairs alveolar fluid clearance. Inhibition also occurs during in-vivo hypoxia in humans and laboratory animals. Signaling mechanisms that inhibit alveolar reabsorption are poorly understood. Because cellular adaptation to hypoxia is regulated by hypoxia-inducible transcription factors (HIF), we tested whether HIFs are involved in decreasing Na+-transport in hypoxic alveolar epithelium. Expression of HIFs was suppressed in cultured rat primary alveolar epithelial cells (AEC) with shRNAs. Hypoxia (1.5% O2, 24 h) decreased amiloride-sensitive transepithelial Na+-transport, decreased the mRNA expression of α-, β-, and γ-ENaC subunits, and reduced the amount of αβγ-ENaC subunits in the apical plasma membrane. Silencing HIF-2α partially prevented impaired fluid reabsorption in hypoxic rats and prevented the hypoxia-induced decrease in α- but not the βγ-subunits of ENaC protein expression resulting in a less active form of ENaC in hypoxic AEC. Inhibition of alveolar reabsorption also caused pulmonary vasoconstriction in ventilated rats. These results indicate that a HIF-2α-dependent decrease in Na+-transport in hypoxic alveolar epithelium decreases alveolar reabsorption. Because susceptibles to high-altitude pulmonary edema (HAPE) have decreased Na+-transport even in normoxia, inhibition of alveolar reabsorption by hypoxia at high altitude might further impair alveolar gas exchange. Thus, aggravated hypoxemia might further enhance hypoxic pulmonary vasoconstriction and might subsequently cause HAPE.
Collapse
Affiliation(s)
- Emel Baloglu
- Department of Pharmacology, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey.,Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany
| | | | - Anna Seleninova
- Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany
| | - Lena Berg
- Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany
| | - Kalpana Velineni
- Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany
| | - Ezgi Ermis-Kaya
- Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany
| | - Heimo Mairbäurl
- Translational Lung Research Center Heidelberg (TLRC), Heidelberg, Germany.,Translational Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
6
|
Wang M, Guo X, Zhao H, Lv J, Wang H, An Y. Adenosine A 2B receptor activation stimulates alveolar fluid clearance through alveolar epithelial sodium channel via cAMP pathway in endotoxin-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2020; 318:L787-L800. [PMID: 32129084 DOI: 10.1152/ajplung.00195.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Clinical studies have established that the capacity of removing excess fluid from alveoli is impaired in most patients with acute respiratory distress syndrome. Impaired alveolar fluid clearance (AFC) correlates with poor outcomes. Adenosine A2B receptor (A2BAR) has the lowest affinity with adenosine among four adenosine receptors. It is documented that A2BAR can activate adenylyl cyclase (AC) resulting in elevated cAMP. Based on the understanding that cAMP is a key regulator of epithelial sodium channel (ENaC), which is the limited step in sodium transport, we hypothesized that A2BAR signaling may affect AFC in acute lung injury (ALI) through regulating ENaC via cAMP, thus attenuating pulmonary edema. To address this, we utilized pharmacological approaches to determine the role of A2BAR in AFC in rats with endotoxin-induced lung injury and further focused on the mechanisms in vitro. We observed elevated pulmonary A2BAR level in rats with ALI and the similar upregulation in alveolar epithelial cells exposed to LPS. A2BAR stimulation significantly attenuated pulmonary edema during ALI, an effect that was associated with enhanced AFC and increased ENaC expression. The regulatory effects of A2BAR on ENaC-α expression were further verified in cultured alveolar epithelial type II (ATII) cells. More importantly, activation of A2BAR dramatically increased amiloride-sensitive Na+ currents in ATII cells. Moreover, we observed that A2BAR activation stimulated cAMP accumulation, whereas the cAMP inhibitor abolished the regulatory effect of A2BAR on ENaC-α expression, suggesting that A2BAR activation regulates ENaC-α expression via cAMP-dependent mechanism. Together, these findings suggest that signaling through alveolar epithelial A2BAR promotes alveolar fluid balance during endotoxin-induced ALI by regulating ENaC via cAMP pathway, raising the hopes for treatment of pulmonary edema due to ALI.
Collapse
Affiliation(s)
- Mengnan Wang
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Xiaoxia Guo
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Huiying Zhao
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Jie Lv
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Huixia Wang
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Youzhong An
- Department of Critical Care Medicine, Peking University People's Hospital, Beijing, China
| |
Collapse
|
7
|
Efficacy of Quercetin as a potent sensitizer of β2-AR in combating the impairment of fluid clearance in lungs of rats under hypoxia. Respir Physiol Neurobiol 2020; 273:103334. [DOI: 10.1016/j.resp.2019.103334] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/08/2019] [Accepted: 10/20/2019] [Indexed: 12/12/2022]
|
8
|
The Hen or the Egg: Impaired Alveolar Oxygen Diffusion and Acute High-altitude Illness? Int J Mol Sci 2019; 20:ijms20174105. [PMID: 31443549 PMCID: PMC6747186 DOI: 10.3390/ijms20174105] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/18/2019] [Accepted: 08/20/2019] [Indexed: 01/11/2023] Open
Abstract
Individuals ascending rapidly to altitudes >2500 m may develop symptoms of acute mountain sickness (AMS) within a few hours of arrival and/or high-altitude pulmonary edema (HAPE), which occurs typically during the first three days after reaching altitudes above 3000-3500 m. Both diseases have distinct pathologies, but both present with a pronounced decrease in oxygen saturation of hemoglobin in arterial blood (SO2). This raises the question of mechanisms impairing the diffusion of oxygen (O2) across the alveolar wall and whether the higher degree of hypoxemia is in causal relationship with developing the respective symptoms. In an attempt to answer these questions this article will review factors affecting alveolar gas diffusion, such as alveolar ventilation, the alveolar-to-arterial O2-gradient, and balance between filtration of fluid into the alveolar space and its clearance, and relate them to the respective disease. The resultant analysis reveals that in both AMS and HAPE the main pathophysiologic mechanisms are activated before aggravated decrease in SO2 occurs, indicating that impaired alveolar epithelial function and the resultant diffusion limitation for oxygen may rather be a consequence, not the primary cause, of these altitude-related illnesses.
Collapse
|
9
|
Dagenais A, Desjardins J, Shabbir W, Roy A, Filion D, Sauvé R, Berthiaume Y. Loss of barrier integrity in alveolar epithelial cells downregulates ENaC expression and activity via Ca 2+ and TRPV4 activation. Pflugers Arch 2018; 470:1615-1631. [PMID: 30088081 DOI: 10.1007/s00424-018-2182-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 06/14/2018] [Accepted: 07/09/2018] [Indexed: 01/08/2023]
Abstract
The epithelial Na channel (ENaC) plays an essential role in lung physiology by modulating the amount of liquid lining the respiratory epithelium. Here, we tested the effect of breaking alveolar epithelial cell barrier integrity on ENaC expression and function. We found that either mechanical wounding by scratching the monolayer or disruption of tight junction with EDTA induced a ~ 50% decrease of α,β and γENaC mRNA expression and an 80% reduction of ENaC short-circuit current (Isc) at 6 h. Scratching the cell monolayer generated a Ca2+ wave that spread from the margin of the scratch to distant cells. Pretreatment with BAPTA-AM, an intracellular Ca2+ chelator, abolished the effect of mechanical wounding and EDTA on αENaC mRNA expression, suggesting that [Ca2+]i is important for this modulation. We tested the hypothesis that a mechanosensitive channel such as TRPV4, a cationic channel known to increase [Ca2+]i, could mediate this effect. Activation of the channel with the TRPV4 specific agonist GSK-1016790A (GSK) decreased αENAC mRNA expression and almost completely abolished ENaC Isc. Pretreatment of alveolar epithelial cells with HC-067047 (HC0), a specific TRPV4 antagonist, reduced the extent of αENAC mRNA downregulation by mechanical wounding and EDTA. Altogether, our results suggest that mechanical stress induced by wounding or TRPV4-mediated loss of tight junction increases [Ca2+]i and elicits a Ca2+ wave that affects ENaC expression and function away from the site of injury. These data are important to better understand how Ca2+ signaling affects lung liquid clearance in injured lungs.
Collapse
Affiliation(s)
- André Dagenais
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada.
- Département de médecine, Université de Montréal, Montreal, Quebec, Canada.
| | - Julie Desjardins
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada
| | - Waheed Shabbir
- Institute of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Antoine Roy
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada
| | - Dominic Filion
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada
| | - Rémy Sauvé
- Département de pharmacologie et physiologie, Université de Montréal, Montreal, Quebec, Canada
| | - Yves Berthiaume
- Institut de recherches cliniques de Montréal, 110 Avenue des Pins Ouest, Montreal, Quebec, H2W 1R7, Canada
- Département de médecine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
10
|
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
|
11
|
Magnani ND, Dada LA, Sznajder JI. Ubiquitin-proteasome signaling in lung injury. Transl Res 2018; 198:29-39. [PMID: 29752900 PMCID: PMC6986356 DOI: 10.1016/j.trsl.2018.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/15/2018] [Accepted: 04/16/2018] [Indexed: 12/21/2022]
Abstract
Cell homeostasis requires precise coordination of cellular proteins function. Ubiquitination is a post-translational modification that modulates protein half-life and function and is tightly regulated by ubiquitin E3 ligases and deubiquitinating enzymes. Lung injury can progress to acute respiratory distress syndrome that is characterized by an inflammatory response and disruption of the alveolocapillary barrier resulting in alveolar edema accumulation and hypoxemia. Ubiquitination plays an important role in the pathobiology of acute lung injury as it regulates the proteins modulating the alveolocapillary barrier and the inflammatory response. Better understanding of the signaling pathways regulated by ubiquitination may lead to novel therapeutic approaches by targeting specific elements of the ubiquitination pathways.
Collapse
Affiliation(s)
- Natalia D Magnani
- Pulmonary and Critical Care Division, Northwestern Feinberg School of Medicine, Chicago, Illinois
| | - Laura A Dada
- Pulmonary and Critical Care Division, Northwestern Feinberg School of Medicine, Chicago, Illinois
| | - Jacob I Sznajder
- Pulmonary and Critical Care Division, Northwestern Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
12
|
Intermittent Hypoxia Increases the Severity of Bleomycin-Induced Lung Injury in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1240192. [PMID: 29725493 PMCID: PMC5872634 DOI: 10.1155/2018/1240192] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 01/03/2018] [Accepted: 01/10/2018] [Indexed: 01/02/2023]
Abstract
Background Severe obstructive sleep apnea (OSA) with chronic intermittent hypoxia (IH) is common in idiopathic pulmonary fibrosis (IPF). Here, we evaluated the impact of IH on bleomycin- (BLM-) induced pulmonary fibrosis in mice. Methods C57BL/6J mice received intratracheal BLM or saline and were exposed to IH (40 cycles/hour; FiO2 nadir: 6%; 8 hours/day) or intermittent air (IA). In the four experimental groups, we evaluated (i) survival; (ii) alveolar inflammation, pulmonary edema, lung oxidative stress, and antioxidant enzymes; (iii) lung cell apoptosis; and (iv) pulmonary fibrosis. Results Survival at day 21 was lower in the BLM-IH group (p < 0.05). Pulmonary fibrosis was more severe at day 21 in BLM-IH mice, as assessed by lung collagen content (p = 0.02) and histology. At day 4, BLM-IH mice developed a more severe neutrophilic alveolitis, (p < 0.001). Lung oxidative stress was observed, and superoxide dismutase and glutathione peroxidase expression was decreased in BLM-IH mice (p < 0.05 versus BLM-IA group). At day 8, pulmonary edema was observed and lung cell apoptosis was increased in the BLM-IH group. Conclusion These results show that exposure to chronic IH increases mortality, lung inflammation, and lung fibrosis in BLM-treated mice. This study raises the question of the worsening impact of severe OSA in IPF patients.
Collapse
|
13
|
Hamacher J, Hadizamani Y, Borgmann M, Mohaupt M, Männel DN, Moehrlen U, Lucas R, Stammberger U. Cytokine-Ion Channel Interactions in Pulmonary Inflammation. Front Immunol 2018; 8:1644. [PMID: 29354115 PMCID: PMC5758508 DOI: 10.3389/fimmu.2017.01644] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
The lungs conceptually represent a sponge that is interposed in series in the bodies’ systemic circulation to take up oxygen and eliminate carbon dioxide. As such, it matches the huge surface areas of the alveolar epithelium to the pulmonary blood capillaries. The lung’s constant exposure to the exterior necessitates a competent immune system, as evidenced by the association of clinical immunodeficiencies with pulmonary infections. From the in utero to the postnatal and adult situation, there is an inherent vital need to manage alveolar fluid reabsorption, be it postnatally, or in case of hydrostatic or permeability edema. Whereas a wealth of literature exists on the physiological basis of fluid and solute reabsorption by ion channels and water pores, only sparse knowledge is available so far on pathological situations, such as in microbial infection, acute lung injury or acute respiratory distress syndrome, and in the pulmonary reimplantation response in transplanted lungs. The aim of this review is to discuss alveolar liquid clearance in a selection of lung injury models, thereby especially focusing on cytokines and mediators that modulate ion channels. Inflammation is characterized by complex and probably time-dependent co-signaling, interactions between the involved cell types, as well as by cell demise and barrier dysfunction, which may not uniquely determine a clinical picture. This review, therefore, aims to give integrative thoughts and wants to foster the unraveling of unmet needs in future research.
Collapse
Affiliation(s)
- Jürg Hamacher
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Internal Medicine V - Pneumology, Allergology, Respiratory and Environmental Medicine, Faculty of Medicine, Saarland University, Saarbrücken, Germany.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Yalda Hadizamani
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Michèle Borgmann
- Internal Medicine and Pneumology, Lindenhofspital, Bern, Switzerland.,Lungen- und Atmungsstiftung Bern, Bern, Switzerland
| | - Markus Mohaupt
- Internal Medicine, Sonnenhofspital Bern, Bern, Switzerland
| | | | - Ueli Moehrlen
- Paediatric Visceral Surgery, Universitäts-Kinderspital Zürich, Zürich, Switzerland
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Vascular Biology Center, Medical College of Georgia, Augusta, GA, United States
| | - Uz Stammberger
- Lungen- und Atmungsstiftung Bern, Bern, Switzerland.,Novartis Institutes for Biomedical Research, Translational Clinical Oncology, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
14
|
Nag S, Resnick A. Stabilization of hypoxia inducible factor by cobalt chloride can alter renal epithelial transport. Physiol Rep 2017; 5:5/24/e13531. [PMID: 29263117 PMCID: PMC5742700 DOI: 10.14814/phy2.13531] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 01/10/2023] Open
Abstract
Given the importance of the transcriptional regulator hypoxia-inducible factor-1 (HIF-1) for adaptive hypoxia responses, we examined the effect of stabilized HIF-1α on renal epithelial permeability and directed sodium transport. This study was motivated by histological analysis of cystic kidneys showing increased expression levels of HIF-1α and HIF-2α We hypothesize that compression induced localized ischemia-hypoxia of normal epithelia near a cyst leads to local stabilization of HIF-1α, leading to altered transepithelial transport that encourages cyst expansion. We found that stabilized HIF-1α alters both transcellular and paracellular transport through renal epithelial monolayers in a manner consistent with secretory behavior, indicating localized ischemia-hypoxia may lead to altered salt and water transport through kidney epithelial monolayers. A quantity of 100 μmol/L Cobalt chloride (CoCl2) was used acutely to stabilize HIF-1α in confluent cultures of mouse renal epithelia. We measured increased transepithelial permeability and decreased transepithelial resistance (TER) when HIF-1α was stabilized. Most interestingly, we measured a change in the direction of sodium current, most likely corresponding to abnormal secretory function, supporting our positive-feedback hypothesis.
Collapse
Affiliation(s)
- Subhra Nag
- Department of Biology, Geology and Environmental Sciences, Cleveland State University, Cleveland, Ohio
| | - Andrew Resnick
- Department of Biology, Geology and Environmental Sciences, Cleveland State University, Cleveland, Ohio .,Department of Physics Cleveland State University, Cleveland, Ohio.,Center for Gene Regulation in Health and Disease, Cleveland State University, Cleveland, Ohio
| |
Collapse
|
15
|
Bernard O, Jeny F, Uzunhan Y, Dondi E, Terfous R, Label R, Sutton A, Larghero J, Vanneaux V, Nunes H, Boncoeur E, Planès C, Dard N. Mesenchymal stem cells reduce hypoxia-induced apoptosis in alveolar epithelial cells by modulating HIF and ROS hypoxic signaling. Am J Physiol Lung Cell Mol Physiol 2017; 314:L360-L371. [PMID: 29167125 DOI: 10.1152/ajplung.00153.2017] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Distal lung diseases, such as pulmonary fibrosis or acute lung injury, are commonly associated with local alveolar hypoxia that may be deleterious through the stimulation of alveolar epithelial cell (AEC) apoptosis. In various murine models of alveolar injury, administration of allogenic human mesenchymal stem cells (hMSCs) exerts an overall protective paracrine effect, limiting lung inflammation and fibrosis. However, the precise mechanisms on lung cells themselves remain poorly understood. Here, we investigated whether hMSC-conditioned medium (hMSC-CM) would protect AECs from hypoxia-induced apoptosis and explored the mechanisms involved in this cytoprotective effect. Exposure of rat primary AECs to hypoxia (1.5% O2 for 24 h) resulted in hypoxia-inducible factor (HIF)-1α protein stabilization, partly dependent on reactive oxygen species (ROS) accumulation, and in a twofold increase in AEC apoptosis that was prevented by the HIF inhibitor 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl-indazole and the antioxidant drug N-acetyl cysteine. Incubation of AECs with hMSC-CM significantly reduced hypoxia-induced apoptosis. hMSC-CM decreased HIF-1α protein expression, as well as ROS accumulation through an increase in antioxidant enzyme activities. Expression of Bnip3 and CHOP, two proapoptotic targets of HIF-1α and ROS pathways, respectively, was suppressed by hMSC-CM, while Bcl-2 expression was restored. The paracrine protective effect of hMSC was partly dependent on keratinocyte growth factor and hepatocyte growth factor secretion, preventing ROS and HIF-1α accumulation.
Collapse
Affiliation(s)
- Olivier Bernard
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France
| | - Florence Jeny
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France.,Assistance publique-Hôpitaux de Paris, Hôpital Avicenne, Bobigny, France
| | - Yurdagül Uzunhan
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France.,Assistance publique-Hôpitaux de Paris, Hôpital Avicenne, Bobigny, France
| | - Elisabetta Dondi
- Institut National de la Santé et de la Recherche Médicale, UMR 978, Bobigny, France
| | - Rahma Terfous
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France
| | - Rabab Label
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France
| | - Angela Sutton
- Institut National de la Santé et de la Recherche Médicale, UMR 1148, Laboratory for Vascular Translational Science, UFR Santé Médecine et Biologie Humaine, Université Paris 13, Sorbonne Paris Cité, Groupe Biothérapies et Glycoconjugués, Bobigny, France
| | - Jérôme Larghero
- AP-HP, Hôpital Saint Louis, Unité de Thérapie Cellulaire et Centre d'Investigation Clinique de Biothérapies, Paris, France, Université Paris Diderot, Sorbonne Paris Cité, Paris , France
| | - Valérie Vanneaux
- AP-HP, Hôpital Saint Louis, Unité de Thérapie Cellulaire et Centre d'Investigation Clinique de Biothérapies, Paris, France, Université Paris Diderot, Sorbonne Paris Cité, Paris , France
| | - Hilario Nunes
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France.,Assistance publique-Hôpitaux de Paris, Hôpital Avicenne, Bobigny, France
| | - Emilie Boncoeur
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France
| | - Carole Planès
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France.,Assistance publique-Hôpitaux de Paris, Hôpital Avicenne, Bobigny, France
| | - Nicolas Dard
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, EA 2363, Bobigny, France
| |
Collapse
|
16
|
Vadász I, Sznajder JI. Gas Exchange Disturbances Regulate Alveolar Fluid Clearance during Acute Lung Injury. Front Immunol 2017; 8:757. [PMID: 28725223 PMCID: PMC5495863 DOI: 10.3389/fimmu.2017.00757] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/15/2017] [Indexed: 01/07/2023] Open
Abstract
Disruption of the alveolar-capillary barrier and accumulation of pulmonary edema, if not resolved, result in poor alveolar gas exchange leading to hypoxia and hypercapnia, which are hallmarks of acute lung injury and the acute respiratory distress syndrome (ARDS). Alveolar fluid clearance (AFC) is a major function of the alveolar epithelium and is mediated by the concerted action of apically-located Na+ channels [epithelial Na+ channel (ENaC)] and the basolateral Na,K-ATPase driving vectorial Na+ transport. Importantly, those patients with ARDS who cannot clear alveolar edema efficiently have worse outcomes. While hypoxia can be improved in most cases by O2 supplementation and mechanical ventilation, the use of lung protective ventilation settings can lead to further CO2 retention. Whether the increase in CO2 concentrations has deleterious or beneficial effects have been a topic of significant controversy. Of note, both low O2 and elevated CO2 levels are sensed by the alveolar epithelium and by distinct and specific molecular mechanisms impair the function of the Na,K-ATPase and ENaC thereby inhibiting AFC and leading to persistence of alveolar edema. This review discusses recent discoveries on the sensing and signaling events initiated by hypoxia and hypercapnia and the relevance of these results in identification of potential novel therapeutic targets in the treatment of ARDS.
Collapse
Affiliation(s)
- István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Jacob I Sznajder
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
17
|
Trac PT, Thai TL, Linck V, Zou L, Greenlee M, Yue Q, Al-Khalili O, Alli AA, Eaton AF, Eaton DC. Alveolar nonselective channels are ASIC1a/α-ENaC channels and contribute to AFC. Am J Physiol Lung Cell Mol Physiol 2017; 312:L797-L811. [PMID: 28283476 DOI: 10.1152/ajplung.00379.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/13/2022] Open
Abstract
A thin fluid layer in alveoli is normal and results from a balance of fluid entry and fluid uptake by transepithelial salt and water reabsorption. Conventional wisdom suggests the reabsorption is via epithelial Na+ channels (ENaC), but if all Na+ reabsorption were via ENaC, then amiloride, an ENaC inhibitor, should block alveolar fluid clearance (AFC). However, amiloride blocks only half of AFC. The reason for failure to block is clear from single-channel measurements from alveolar epithelial cells: ENaC channels are observed, but another channel is present at the same frequency that is nonselective for Na+ over K+, has a larger conductance, and has shorter open and closed times. These two channel types are known as highly selective channels (HSC) and nonselective cation channels (NSC). HSC channels are made up of three ENaC subunits since knocking down any of the subunits reduces HSC number. NSC channels contain α-ENaC since knocking down α-ENaC reduces the number of NSC (knocking down β- or γ-ENaC has no effect on NSC, but the molecular composition of NSC channels remains unclear). We show that NSC channels consist of at least one α-ENaC and one or more acid-sensing ion channel 1a (ASIC1a) proteins. Knocking down either α-ENaC or ASIC1a reduces both NSC and HSC number, and no NSC channels are observable in single-channel patches on lung slices from ASIC1a knockout mice. AFC is reduced in knockout mice, and wet wt-to-dry wt ratio is increased, but the percentage increase in wet wt-to-dry wt ratio is larger than expected based on the reduction in AFC.
Collapse
Affiliation(s)
- Phi T Trac
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| | - Tiffany L Thai
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| | - Valerie Linck
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| | - Li Zou
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| | - Megan Greenlee
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| | - Qiang Yue
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| | - Otor Al-Khalili
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| | - Abdel A Alli
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida
| | - Amity F Eaton
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| | - Douglas C Eaton
- Department of Physiology and Center for Cell and Molecular Signaling, Emory University School of Medicine, Atlanta, Georgia; and
| |
Collapse
|
18
|
Taylor NE, Baker SE, Olson TP, Lalande S, Johnson BD, Snyder EM. Albuterol Improves Alveolar-Capillary Membrane Conductance in Healthy Humans. CLINICAL MEDICINE INSIGHTS-CIRCULATORY RESPIRATORY AND PULMONARY MEDICINE 2016; 10:19-25. [PMID: 27773996 PMCID: PMC5063752 DOI: 10.4137/ccrpm.s30251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 06/02/2016] [Accepted: 06/07/2016] [Indexed: 11/05/2022]
Abstract
BACKGROUND Beta-2 adrenergic receptors (β2ARs) are located throughout the body including airway and alveolar cells. The β2ARs regulate lung fluid clearance through a variety of mechanisms including ion transport on alveolar cells and relaxation of the pulmonary lymphatics. We examined the effect of an inhaled β2-agonist (albuterol) on alveolar-capillary membrane conductance (DM) and pulmonary capillary blood volume (VC) in healthy humans. METHODS We assessed the diffusing capacity of the lungs for carbon monoxide (DLCO) and nitric oxide (DLNO) at baseline, 30 minutes, and 60 minutes following nebulized albuterol (2.5 mg, diluted in 3 mL normal saline) in 45 healthy subjects. Seventeen subjects repeated these measures following nebulized normal saline (age = 27 ± 9 years, height = 165 ± 21 cm, weight = 68 ± 12 kg, BMI = 26 ± 9 kg/m2). Cardiac output (Q), heart rate, systemic vascular resistance (SVR), blood pressure, oxygen saturation, forced expiratory volume at one-second (FEV1), and forced expiratory flow at 50% of forced vital capacity (FEF50) were assessed at baseline, 30 minutes, and 60 minutes following the administration of albuterol or saline. RESULTS Albuterol resulted in a decrease in SVR, and an increase in Q, FEV1, and FEF50 compared to saline controls. Albuterol also resulted in a decrease in VC at 60 minutes post albuterol. Both albuterol and normal saline resulted in no change in DLCO or DM when assessed alone, but a significant increase was observed in DM when accounting for changes in VC. CONCLUSION These data suggest that nebulized albuterol improves pulmonary function in healthy humans, while nebulization of both albuterol and saline results in an increase in DM/VC.
Collapse
Affiliation(s)
- Natalie E Taylor
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sarah E Baker
- Research Fellow, Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | - Thomas P Olson
- Assistant Professor of Medicine, Consultant, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Sophie Lalande
- Assistant Professor, Department of Kinesiology, University of Toledo, OH, USA
| | - Bruce D Johnson
- Professor of Medicine and Physiology, Consultant, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Eric M Snyder
- Assistant Professor, School of Kinesiology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
19
|
Krause NC, Kutsche HS, Santangelo F, DeLeon ER, Dittrich NP, Olson KR, Althaus M. Hydrogen sulfide contributes to hypoxic inhibition of airway transepithelial sodium absorption. Am J Physiol Regul Integr Comp Physiol 2016; 311:R607-17. [DOI: 10.1152/ajpregu.00177.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/13/2016] [Indexed: 01/23/2023]
Abstract
In lung epithelial cells, hypoxia decreases the expression and activity of sodium-transporting molecules, thereby reducing the rate of transepithelial sodium absorption. The mechanisms underlying the sensing of hypoxia and subsequent coupling to sodium-transporting molecules remain unclear. Hydrogen sulfide (H2S) has recently been recognized as a cellular signaling molecule whose intracellular concentrations critically depend on oxygen levels. Therefore, it was questioned whether endogenously produced H2S contributes to hypoxic inhibition of sodium transport. In electrophysiological Ussing chamber experiments, hypoxia was established by decreasing oxygen concentrations in the chambers. Hypoxia concentration dependently and reversibly decreased amiloride-sensitive sodium absorption by cultured H441 monolayers and freshly dissected porcine tracheal epithelia due to inhibition of basolateral Na+/K+-ATPase. Exogenous application of H2S by the sulfur salt Na2S mimicked the effect of hypoxia and inhibited amiloride-sensitive sodium absorption by both tissues in an oxygen-dependent manner. Hypoxia increased intracellular concentrations of H2S and decreased the concentration of polysulfides. Pretreatment with the cystathionine-γ-lyase inhibitor d/l-propargylglycine (PAG) decreased hypoxic inhibition of sodium transport by H441 monolayers, whereas inhibition of cystathionine-β-synthase (with aminooxy-acetic acid; AOAA) or 3-mercaptopyruvate sulfurtransferase (with aspartate) had no effect. Inhibition of all of these H2S-generating enzymes with a combination of AOAA, PAG, and aspartate decreased the hypoxic inhibition of sodium transport by H441 cells and pig tracheae and decreased H2S production by tracheae. These data suggest that airway epithelial cells endogenously produce H2S during hypoxia, and this contributes to hypoxic inhibition of transepithelial sodium absorption.
Collapse
Affiliation(s)
- Nicole C. Krause
- Institute for Animal Physiology, Justus-Liebig-University, Giessen, Germany; and
| | - Hanna S. Kutsche
- Institute for Animal Physiology, Justus-Liebig-University, Giessen, Germany; and
| | - Fabrizio Santangelo
- Institute for Animal Physiology, Justus-Liebig-University, Giessen, Germany; and
| | - Eric R. DeLeon
- Department of Physiology, Indiana University School of Medicine-South Bend, South Bend, Indiana
| | - Nikolaus P. Dittrich
- Institute for Animal Physiology, Justus-Liebig-University, Giessen, Germany; and
| | - Kenneth R. Olson
- Department of Physiology, Indiana University School of Medicine-South Bend, South Bend, Indiana
| | - Mike Althaus
- Institute for Animal Physiology, Justus-Liebig-University, Giessen, Germany; and
| |
Collapse
|
20
|
Davieds B, Gross J, Berger MM, Baloğlu E, Bärtsch P, Mairbäurl H. Inhibition of alveolar Na transport and LPS causes hypoxemia and pulmonary arterial vasoconstriction in ventilated rats. Physiol Rep 2016; 4:e12985. [PMID: 27670411 PMCID: PMC5037927 DOI: 10.14814/phy2.12985] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 08/31/2016] [Indexed: 01/11/2023] Open
Abstract
Oxygen diffusion across the alveolar wall is compromised by low alveolar oxygen but also by pulmonary edema, and leads to hypoxemia and hypoxic pulmonary vasoconstriction (HPV). To test, whether inhibition of alveolar fluid reabsorption results in an increased pulmonary arterial pressure and whether this effect enhances HPV, we established a model, where anesthetized rats were ventilated with normoxic (21% O2) and hypoxic (13.5% O2) gas received aerosolized amiloride and lipopolisaccharide (LPS) to inhibit alveolar fluid reabsorption. Right ventricular systolic pressure (RVsP) was measured as an indicator of pulmonary arterial pressure. Oxygen pressure (PaO2) and saturation (SaO2) in femoral arterial blood served as indicator of oxygen diffusion across the alveolar wall. Aerosolized amiloride and bacterial LPS decreased PaO2 and SaO2 and increased RVsP even when animals were ventilated with normoxic gas. Ventilation with hypoxic gas decreased PaO2 by 35 mmHg and increased RVsP by 10 mmHg. However, combining hypoxia with amiloride and LPS did not aggravate the decrease in PaO2 and SaO2 and had no effect on the increase in RVsP relative to hypoxia alone. There was a direct relation between SaO2 and PaO2 and the RVsP under all experimental conditions. Two hours but not 1 h exposure to aerosolized amiloride and LPS in normoxia as well as hypoxia increased the lung wet-to-dry-weight ratio indicating edema formation. Together these findings indicate that inhibition of alveolar reabsorption causes pulmonary edema, impairs oxygen diffusion across the alveolar wall, and leads to an increased pulmonary arterial pressure.
Collapse
Affiliation(s)
- Bodo Davieds
- Medical Clinic VII, Sports Medicine, University of Heidelberg, Heidelberg, Germany
| | - Julian Gross
- Medical Clinic VII, Sports Medicine, University of Heidelberg, Heidelberg, Germany
| | - Marc M Berger
- Department of Anesthesiology, Perioperative and General Critical Care Medicine Salzburg General Hospital Paracelsus Medical University, Salzburg, Austria
| | - Emel Baloğlu
- Medical Clinic VII, Sports Medicine, University of Heidelberg, Heidelberg, Germany Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany Department of Pharmacology, Acibadem University, Istanbul, Turkey
| | - Peter Bärtsch
- Medical Clinic VII, Sports Medicine, University of Heidelberg, Heidelberg, Germany
| | - Heimo Mairbäurl
- Medical Clinic VII, Sports Medicine, University of Heidelberg, Heidelberg, Germany Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
21
|
Uzunhan Y, Bernard O, Marchant D, Dard N, Vanneaux V, Larghero J, Gille T, Clerici C, Valeyre D, Nunes H, Boncoeur E, Planès C. Mesenchymal stem cells protect from hypoxia-induced alveolar epithelial-mesenchymal transition. Am J Physiol Lung Cell Mol Physiol 2016; 310:L439-51. [DOI: 10.1152/ajplung.00117.2015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 12/22/2015] [Indexed: 01/13/2023] Open
Abstract
Administration of bone marrow-derived human mesenchymal stem cells (hMSC) reduces lung inflammation, fibrosis, and mortality in animal models of lung injury, by a mechanism not completely understood. We investigated whether hMSC would prevent epithelial-mesenchymal transition (EMT) induced by hypoxia in primary rat alveolar epithelial cell (AEC). In AEC cultured on semipermeable filters, prolonged hypoxic exposure (1.5% O2 for up to 12 days) induced phenotypic changes consistent with EMT, i.e., a change in cell morphology, a decrease in transepithelial resistance (Rte) and in the expression of epithelial markers [zonula occludens-1 (ZO-1), E-cadherin, AQP-5, TTF-1], together with an increase in mesenchymal markers [vimentin, α-smooth muscle actin (α-SMA)]. Expression of transcription factors driving EMT such as SNAIL1, ZEB1, and TWIST1 increased after 2, 24, and 48 h of hypoxia, respectively. Hypoxia also induced TGF-β1 mRNA expression and the secretion of active TGF-β1 in apical medium, and the expression of connective tissue growth factor (CTGF), two inducers of EMT. Coculture of AEC with hMSC partially prevented the decrease in Rte and in ZO-1, E-cadherin, and TTF-1 expression, and the increase in vimentin expression induced by hypoxia. It also abolished the increase in TGF-β1 expression and in TGF-β1-induced genes ZEB1, TWIST1, and CTGF. Finally, incubation with human recombinant KGF at a concentration similar to what was measured in hMSC-conditioned media restored the expression of TTF-1 and prevented the increase in TWIST1, TGF-β1, and CTGF in hypoxic AEC. Our results indicate that hMSC prevent hypoxia-induced alveolar EMT through the paracrine modulation of EMT signaling pathways and suggest that this effect is partly mediated by KGF.
Collapse
Affiliation(s)
- Yurdagül Uzunhan
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
- AP-HP, Hôpital Avicenne, Bobigny, France
| | - Olivier Bernard
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
| | - Dominique Marchant
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
| | - Nicolas Dard
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
| | - Valérie Vanneaux
- AP-HP, Hôpital Saint Louis, Unité de Thérapie Cellulaire et CIC de Biothérapies, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Jérôme Larghero
- AP-HP, Hôpital Saint Louis, Unité de Thérapie Cellulaire et CIC de Biothérapies, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Thomas Gille
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
- AP-HP, Hôpital Avicenne, Bobigny, France
| | - Christine Clerici
- Université Paris Diderot, Sorbonne Paris Cité, Inserm U1152, Paris, France; and
- AP-HP, Hôpital Bichat, Paris, France
| | - Dominique Valeyre
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
- AP-HP, Hôpital Avicenne, Bobigny, France
| | - Hilario Nunes
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
- AP-HP, Hôpital Avicenne, Bobigny, France
| | - Emilie Boncoeur
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
| | - Carole Planès
- Université Paris 13, Sorbonne Paris Cité, Laboratoire Hypoxie & Poumon, Bobigny, France
- AP-HP, Hôpital Avicenne, Bobigny, France
| |
Collapse
|
22
|
Chronic Hypoxemia in Children With Congenital Heart Defect Impairs Airway Epithelial Sodium Transport. Pediatr Crit Care Med 2016; 17:45-52. [PMID: 26509813 DOI: 10.1097/pcc.0000000000000568] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Ambient hypoxia impairs the airway epithelial Na transport, which is crucial in lung edema reabsorption. Whether chronic systemic hypoxemia affects airway Na transport has remained largely unknown. We have therefore investigated whether chronic systemic hypoxemia in children with congenital heart defect affects airway epithelial Na transport, Na transporter-gene expression, and short-term lung edema accumulation. DESIGN Prospective, observational study. SETTING Tertiary care medical center responsible for nationwide pediatric cardiac surgery. PATIENTS Ninety-nine children with congenital heart defect or acquired heart disease (age range, 6 d to 14.8 yr) were divided into three groups based on their level of preoperative systemic hypoxemia: 1) normoxemic patients (SpO2% ≥ 95%; n = 44), 2) patients with cyanotic congenital heart defect and moderate hypoxemia (SpO2 86-94%; n = 16), and 3) patients with cyanotic congenital heart defect and profound systemic hypoxemia (SpO2 ≤ 85%; n = 39). MEASUREMENTS AND MAIN RESULTS Nasal transepithelial potential difference served as a surrogate measure for epithelial Na transport of the respiratory tract. Profoundly hypoxemic patients had 29% lower basal nasal transepithelial potential difference (p = 0.02) and 55% lower amiloride-sensitive nasal transepithelial potential difference (p = 0.0003) than normoxemic patients. In profoundly hypoxemic patients, nasal epithelial messenger RNA expressions of two airway Na transporters (amiloride-sensitive epithelial Na channel and β1- Na-K-ATPase) were not attenuated, but instead α1-Na-K-ATPase messenger RNA levels were higher (p = 0.03) than in the normoxemic patients, indicating that posttranscriptional factors may impair airway Na transport. The chest radiograph lung edema score increased after congenital cardiac surgery in profoundly hypoxemic patients (p = 0.0004) but not in patients with normoxemia or moderate hypoxemia. CONCLUSIONS The impaired airway epithelial amiloride-sensitive Na transport activity in profoundly hypoxemic children with cyanotic congenital heart defect may hinder defense against lung edema after cardiac surgery.
Collapse
|
23
|
Janér C, Pitkänen OM, Süvari L, Turpeinen U, Palojärvi A, Andersson S, Helve O. Duration of gestation and mode of delivery affect the genes of transepithelial sodium transport in pulmonary adaptation. Neonatology 2015; 107:27-33. [PMID: 25301528 DOI: 10.1159/000363729] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/19/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUND Respiratory distress due to inadequate lung liquid clearance is a significant problem in infants delivered late preterm or early term, especially by elective cesarean delivery (CD). Lung liquid clearance depends on epithelial ion transport and in animals is induced by glucocorticoids. OBJECTIVES In newborn late preterm and term infants to study airway epithelial gene expressions of epithelial sodium channel (ENaC), and the serum and glucocorticoid-inducible kinase 1 (SGK1), and their association with cortisol, mode of delivery, and gestational age (GA). METHODS Infants were delivered at 35(0/7)-41(6/7) weeks. Cortisol in umbilical cord plasma was analyzed with liquid chromatography-tandem mass spectrometry. ENaC and SGK1 mRNAs in airway epithelial cells obtained within 3 h and at 1 day postnatally were quantified with real-time PCR. RESULTS ENaC and SGK1 mRNAs were significantly lower in late preterm and early term infants than in those ≥ 39(0/7) weeks. Significant correlations existed between both ENaC and SGK1 and cord cortisol and GA. In term infants, SGK1 mRNA at 1.5 h was higher after vaginal delivery than elective CD. CONCLUSIONS In late preterm and early term infants, low expression of ENaC and SGK1 may parallel insufficient lung liquid clearance predisposing to respiratory distress. Lower SGK1 expression after term CD could translate into insufficient sodium and lung liquid absorption. The findings demonstrate a central role for cortisol in regulation of ENaC and potentially perinatal sodium and lung liquid clearance.
Collapse
Affiliation(s)
- Cecilia Janér
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | | | | | | | | | | |
Collapse
|
24
|
Lazrak A, Jurkuvenaite A, Ness EC, Zhang S, Woodworth BA, Muhlebach MS, Stober VP, Lim YP, Garantziotis S, Matalon S. Inter-α-inhibitor blocks epithelial sodium channel activation and decreases nasal potential differences in ΔF508 mice. Am J Respir Cell Mol Biol 2014; 50:953-62. [PMID: 24303840 DOI: 10.1165/rcmb.2013-0215oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Increased activity of lung epithelial sodium channels (ENaCs) contributes to the pathophysiology of cystic fibrosis (CF) by increasing the rate of epithelial lining fluid reabsorption. Inter-α-inhibitor (IαI), a serum protease inhibitor, may decrease ENaC activity by preventing its cleavage by serine proteases. High concentrations of IαI were detected in the bronchoalveolar lavage fluid (BALF) of children with CF and lower airway diseases. IαI decreased amiloride-sensitive (IENaC) but not cAMP-activated Cl(-) currents across confluent monolayers of rat ATII, and mouse nasal epithelial cells grew in primary culture by 45 and 25%, respectively. Changes in IENaC by IαI in ATII cells were accompanied by increased levels of uncleaved (immature) surface α-ENaC. IαI increased airway surface liquid depth overlying murine nasal epithelial cells to the same extent as amiloride, consistent with ENaC inhibition. Incubation of lung slices from C57BL/6, those lacking phenylalanine at position 508 (∆F508), or CF transmembrane conductance regulator knockout mice with IαI for 3 hours decreased the open probability of their ENaC channels by 50%. ∆F508 mice had considerably higher levels the amiloride-sensitive fractions of ENaC nasal potential difference (ENaC-NPD) than wild-type littermates and only background levels of IαI in their BALF. A single intranasal instillation of IαI decreased their ENaC-NPD 24 hours later by 25%. In conclusion, we show that IαI is present in the BALF of children with CF, is an effective inhibitor of ENaC proteolysis, and decreases ENaC activity in lung epithelial cells of ∆F508 mice.
Collapse
|
25
|
Gille T, Randrianarison-Pellan N, Goolaerts A, Dard N, Uzunhan Y, Ferrary E, Hummler E, Clerici C, Planès C. Hypoxia-induced inhibition of epithelial Na(+) channels in the lung. Role of Nedd4-2 and the ubiquitin-proteasome pathway. Am J Respir Cell Mol Biol 2014; 50:526-37. [PMID: 24093724 DOI: 10.1165/rcmb.2012-0518oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transepithelial sodium transport via alveolar epithelial Na(+) channels (ENaC) and Na(+),K(+)-ATPase constitutes the driving force for removal of alveolar edema fluid. Alveolar hypoxia associated with pulmonary edema may impair ENaC activity and alveolar Na(+) absorption through a decrease of ENaC subunit expression at the apical membrane of alveolar epithelial cells (AECs). Here, we investigated the mechanism(s) involved in this process in vivo in the β-Liddle mouse strain mice carrying a truncation of β-ENaC C-terminus abolishing the interaction between β-ENaC and the ubiquitin protein-ligase Nedd4-2 that targets the channel for endocytosis and degradation and in vitro in rat AECs. Hypoxia (8% O2 for 24 h) reduced amiloride-sensitive alveolar fluid clearance by 69% in wild-type mice but had no effect in homozygous mutated β-Liddle littermates. In vitro, acute exposure of AECs to hypoxia (0.5-3% O2 for 1-6 h) rapidly decreased transepithelial Na(+) transport as assessed by equivalent short-circuit current Ieq and the amiloride-sensitive component of Na(+) current across the apical membrane, reflecting ENaC activity. Hypoxia induced a decrease of ENaC subunit expression in the apical membrane of AECs with no change in intracellular expression and induced a 2-fold increase in α-ENaC polyubiquitination. Hypoxic inhibition of amiloride-sensitive Ieq was fully prevented by preincubation with the proteasome inhibitors MG132 and lactacystin or with the antioxidant N-acetyl-cysteine. Our data strongly suggest that Nedd4-2-mediated ubiquitination of ENaC leading to endocytosis and degradation of apical Na(+) channels is a key feature of hypoxia-induced inhibition of transepithelial alveolar Na(+) transport.
Collapse
Affiliation(s)
- Thomas Gille
- 1 Université Paris 13, Sorbonne Paris Cité, Laboratoire Réponses Cellulaires et Fonctionnelles à l'Hypoxie (EA 2363), Bobigny, France
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Goolaerts A, Pellan-Randrianarison N, Larghero J, Vanneaux V, Uzunhan Y, Gille T, Dard N, Planès C, Matthay MA, Clerici C. Conditioned media from mesenchymal stromal cells restore sodium transport and preserve epithelial permeability in an in vitro model of acute alveolar injury. Am J Physiol Lung Cell Mol Physiol 2014; 306:L975-85. [PMID: 24682451 DOI: 10.1152/ajplung.00242.2013] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) or their media (MSC-M) were reported to reverse acute lung injury (ALI)-induced decrease of alveolar fluid clearance. To determine the mechanisms by which MSC-M exert their beneficial effects, an in vitro model of alveolar epithelial injury was created by exposing primary rat alveolar epithelial cells (AECs) to hypoxia (3% O2) plus cytomix, a combination of IL-1β, TNF-α, and IFN-γ. MSC-M were collected from human MSCs exposed for 12 h to either normoxia (MSC-M) or to hypoxia plus cytomix (HCYT-MSC-M). This latter condition was used to model the effect of alveolar inflammation and hypoxia on paracrine secretion of MSCs in the injured lung. Comparison of paracrine soluble factors in MSC media showed that the IL-1 receptor antagonist and prostaglandin E2 were markedly increased while keratinocyte growth factor (KGF) was twofold lower in HCYT-MSC-M compared with MSC-M. In AECs, hypoxia plus cytomix increased protein permeability, reduced amiloride-sensitive short-circuit current (AS-Isc), and also decreased the number of α-epithelial sodium channel (α-ENaC) subunits in the apical membrane. To test the effects of MSC media, MSC-M and HCYT-MSC-M were added for an additional 12 h to AECs exposed to hypoxia plus cytomix. MSC-M and HCYT-MSC-M completely restored epithelial permeability to normal. MSC-M, but not HCYT-MSC-M, significantly prevented the hypoxia plus cytomix-induced decrease of ENaC activity and restored apical α-ENaC channels. Interestingly, KGF-deprived MSC-M were unable to restore amiloride-sensitive sodium transport, indicating a possible role for KGF in the beneficial effect of MSC-M. These results indicate that MSC-M may be a preferable therapeutic option for ALI.
Collapse
Affiliation(s)
- Arnaud Goolaerts
- Institut National de la Santé et de la Recherche Médicale, U773, Paris, France
| | - Nadia Pellan-Randrianarison
- Institut National de la Santé et de la Recherche Médicale, U773, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France
| | - Jérôme Larghero
- Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France; AP-HP, Hôpital Saint Louis, Unité de Thérapie Cellulaire et CIC de Biothérapies, Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Valérie Vanneaux
- Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France; AP-HP, Hôpital Saint Louis, Unité de Thérapie Cellulaire et CIC de Biothérapies, Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Yurdagül Uzunhan
- Université Paris 13, Sorbonne Paris Cité, EA2363, Bobigny, France; AP-HP, Hôpital Avicenne, Bobigny, France; and
| | - Thomas Gille
- Université Paris 13, Sorbonne Paris Cité, EA2363, Bobigny, France; AP-HP, Hôpital Avicenne, Bobigny, France; and
| | - Nicolas Dard
- Université Paris 13, Sorbonne Paris Cité, EA2363, Bobigny, France
| | - Carole Planès
- Université Paris 13, Sorbonne Paris Cité, EA2363, Bobigny, France; AP-HP, Hôpital Avicenne, Bobigny, France; and
| | - Michael A Matthay
- Institut National de la Santé et de la Recherche Médicale, U773, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France
| | - Christine Clerici
- Institut National de la Santé et de la Recherche Médicale, U773, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France; AP-HP, Hôpital Bichat, Paris, France
| |
Collapse
|
27
|
Alli AA, Brewer EM, Montgomery DS, Ghant MS, Eaton DC, Brown LA, Helms MN. Chronic ethanol exposure alters the lung proteome and leads to mitochondrial dysfunction in alveolar type 2 cells. Am J Physiol Lung Cell Mol Physiol 2014; 306:L1026-35. [PMID: 24682449 DOI: 10.1152/ajplung.00287.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The lungs can undergo irreversible damage from chronic alcohol consumption. Herein, we developed an animal model predisposed for edematous lung injury following chronic ingestion of alcohol to better understand the etiology of alcohol-related disorders. Using animal modeling, alongside high-throughput proteomic and microarray assays, we identified changes in lung protein and transcript in mice and rats, respectively, following chronic alcohol ingestion or a caloric control diet. Liquid chromatography-mass spectrometry identified several mitochondrial-related proteins in which the expression was upregulated following long-term alcohol ingestion in mice. Consistent with these observations, rat gene chip microarray analysis of alveolar cells obtained from animals maintained on a Lieber-DeCarli liquid alcohol diet confirmed significant changes in mitochondrial-related transcripts in the alcohol lung. Transmission electron microscopy revealed significant changes in the mitochondrial architecture in alcohol mice, particularly following lipopolysaccharide exposure. Chronic alcohol ingestion was also shown to worsen mitochondrial respiration, mitochondrial membrane polarization, and NAD(+)-to-NADH ratios in alveolar type 2 cells. In summary, our studies show causal connection between chronic alcohol ingestion and mitochondrial dysfunction, albeit the specific role of each of the mitochondrial-related proteins and transcripts identified in our study requires additional study.
Collapse
Affiliation(s)
- Abdel A Alli
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia
| | - Elizabeth M Brewer
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia; and
| | | | - Marcus S Ghant
- Department of Biological Sciences, Clark Atlanta University, Atlanta, Georgia
| | - Douglas C Eaton
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia; and
| | - Lou Ann Brown
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia; and
| | - My N Helms
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia; and
| |
Collapse
|
28
|
Zhu YG, Hao Q, Monsel A, Feng XM, Lee JW. Adult stem cells for acute lung injury: remaining questions and concerns. Respirology 2014; 18:744-56. [PMID: 23578018 DOI: 10.1111/resp.12093] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/02/2013] [Indexed: 12/22/2022]
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome remains a major cause of morbidity and mortality in hospitalized patients. The pathophysiology of ALI involves complex interactions between the inciting event, such as pneumonia, sepsis or aspiration, and the host immune response resulting in lung protein permeability, impaired resolution of pulmonary oedema, an intense inflammatory response in the injured alveolus and hypoxemia. In multiple preclinical studies, adult stem cells have been shown to be therapeutic due to both the ability to mitigate injury and inflammation through paracrine mechanisms and perhaps to regenerate tissue by virtue of their multi-potency. These characteristics have stimulated intensive research efforts to explore the possibility of using stem or progenitor cells for the treatment of lung injury. A variety of stem or progenitor cells have been isolated, characterized and tested experimentally in preclinical animal models of ALI. However, questions remain concerning the optimal dose, route and the adult stem or progenitor cell to use. Here, the current mechanisms underlying the therapeutic effect of stem cells in ALI as well as the questions that will arise as clinical trials for ALI are planned are reviewed.
Collapse
Affiliation(s)
- Ying-Gang Zhu
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
29
|
TGF-β directs trafficking of the epithelial sodium channel ENaC which has implications for ion and fluid transport in acute lung injury. Proc Natl Acad Sci U S A 2013; 111:E374-83. [PMID: 24324142 DOI: 10.1073/pnas.1306798111] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
TGF-β is a pathogenic factor in patients with acute respiratory distress syndrome (ARDS), a condition characterized by alveolar edema. A unique TGF-β pathway is described, which rapidly promoted internalization of the αβγ epithelial sodium channel (ENaC) complex from the alveolar epithelial cell surface, leading to persistence of pulmonary edema. TGF-β applied to the alveolar airspaces of live rabbits or isolated rabbit lungs blocked sodium transport and caused fluid retention, which--together with patch-clamp and flow cytometry studies--identified ENaC as the target of TGF-β. TGF-β rapidly and sequentially activated phospholipase D1, phosphatidylinositol-4-phosphate 5-kinase 1α, and NADPH oxidase 4 (NOX4) to produce reactive oxygen species, driving internalization of βENaC, the subunit responsible for cell-surface stability of the αβγENaC complex. ENaC internalization was dependent on oxidation of βENaC Cys(43). Treatment of alveolar epithelial cells with bronchoalveolar lavage fluids from ARDS patients drove βENaC internalization, which was inhibited by a TGF-β neutralizing antibody and a Tgfbr1 inhibitor. Pharmacological inhibition of TGF-β signaling in vivo in mice, and genetic ablation of the nox4 gene in mice, protected against perturbed lung fluid balance in a bleomycin model of lung injury, highlighting a role for both proximal and distal components of this unique ENaC regulatory pathway in lung fluid balance. These data describe a unique TGF-β-dependent mechanism that regulates ion and fluid transport in the lung, which is not only relevant to the pathological mechanisms of ARDS, but might also represent a physiological means of acutely regulating ENaC activity in the lung and other organs.
Collapse
|
30
|
Abstract
Oxygen-sensing pathways have been extensively explored in the context of homeostatic responses to hypoxic episodes; however, little is known of their involvement in the morphogenesis of respiratory structures (mitochondria, placenta, lung) during development in utero. This review identifies four essential loci where oxygen signalling pathways may cue the development of respiratory structures as: (i). mitochondrial biogenesis coupled with muted oxidative function dependent on the hypoxia-sustained production of NO; (ii). the generation of oxygen gradients which drive trophoblast differentiation and the formation of the chorionic gas exchange interface of the placenta; (iii). the proliferation and epithelial/endothelial differentiation of mesenchyme during the initiation of lung morphogenesis; and (iv). the regulation of epithelial fluid secretion/absorption in the lung. The identification of these oxygen-regulated developmental stages clarifies the close association between oxygen availability, reactive oxygen species and the morphogenesis of gas exchange structures and bears with it the implication that these pathways set the scope for aerobic metabolic performance throughout life.
Collapse
Affiliation(s)
- Stephen C Land
- Maternal and Child Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
| |
Collapse
|
31
|
Paracrine activity of stem cells in therapy for acute lung injury and adult respiratory distress syndrome. J Trauma Acute Care Surg 2013; 74:1351-6. [PMID: 23609289 DOI: 10.1097/ta.0b013e318283d942] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
32
|
Paracrine activity of stem cells in therapy for acute lung injury and adult respiratory distress syndrome. J Trauma Acute Care Surg 2013. [DOI: 10.1097/01586154-201305000-00024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Bhattacharya J, Matthay MA. Regulation and repair of the alveolar-capillary barrier in acute lung injury. Annu Rev Physiol 2013; 75:593-615. [PMID: 23398155 DOI: 10.1146/annurev-physiol-030212-183756] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Considerable progress has been made in understanding the basic mechanisms that regulate fluid and protein exchange across the endothelial and epithelial barriers of the lung under both normal and pathological conditions. Clinically relevant lung injury occurs most commonly from severe viral and bacterial infections, aspiration syndromes, and severe shock. The mechanisms of lung injury have been identified in both experimental and clinical studies. Recovery from lung injury requires the reestablishment of an intact endothelial barrier and a functional alveolar epithelial barrier capable of secreting surfactant and removing alveolar edema fluid. Repair mechanisms include the participation of endogenous progenitor cells in strategically located niches in the lung. Novel treatment strategies include the possibility of cell-based therapy that may reduce the severity of lung injury and enhance lung repair.
Collapse
Affiliation(s)
- Jahar Bhattacharya
- Division of Pulmonary Allergy and Critical Care, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| | | |
Collapse
|
34
|
Tan CD, Smolenski RT, Harhun MI, Patel HK, Ahmed SG, Wanisch K, Yáñez-Muñoz RJ, Baines DL. AMP-activated protein kinase (AMPK)-dependent and -independent pathways regulate hypoxic inhibition of transepithelial Na+ transport across human airway epithelial cells. Br J Pharmacol 2013; 167:368-82. [PMID: 22509822 DOI: 10.1111/j.1476-5381.2012.01993.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE Pulmonary transepithelial Na(+) transport is reduced by hypoxia, but in the airway the regulatory mechanisms remain unclear. We investigated the role of AMPK and ROS in the hypoxic regulation of apical amiloride-sensitive Na(+) channels and basolateral Na(+) K(+) ATPase activity. EXPERIMENTAL APPROACH H441 human airway epithelial cells were used to examine the effects of hypoxia on Na(+) transport, AMP : ATP ratio and AMPK activity. Lentiviral constructs were used to modify cellular AMPK abundance and activity; pharmacological agents were used to modify cellular ROS. KEY RESULTS AMPK was activated by exposure to 3% or 0.2% O(2) for 60 min in cells grown in submerged culture or when fluid (0.1 mL·cm(-2) ) was added to the apical surface of cells grown at the air-liquid interface. Only 0.2% O(2) activated AMPK in cells grown at the air-liquid interface. AMPK activation was associated with elevation of cellular AMP:ATP ratio and activity of the upstream kinase LKB1. Hypoxia inhibited basolateral ouabain-sensitive I(sc) (I(ouabain) ) and apical amiloride-sensitive Na(+) conductance (G(Na+) ). Modification of AMPK activity prevented the effect of hypoxia on I(ouabain) (Na(+) K(+) ATPase) but not apical G(Na+) . Scavenging of superoxide and inhibition of NADPH oxidase prevented the effect of hypoxia on apical G(Na+) (epithelial Na(+) channels). CONCLUSIONS AND IMPLICATIONS Hypoxia activates AMPK-dependent and -independent pathways in airway epithelial cells. Importantly, these pathways differentially regulate apical Na(+) channels and basolateral Na(+) K(+) ATPase activity to decrease transepithelial Na(+) transport. Luminal fluid potentiated the effect of hypoxia and activated AMPK, which could have important consequences in lung disease conditions.
Collapse
Affiliation(s)
- C D Tan
- Pharmacology and Cell Physiology Research Group, Division of Biomedical Sciences, St George's University of London, Cranmer Terrace, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ji W, Fu J, Nie H, Xue X. Expression and activity of epithelial sodium channel in hyperoxia-induced bronchopulmonary dysplasia in neonatal rats. Pediatr Int 2012; 54:735-42. [PMID: 22591391 DOI: 10.1111/j.1442-200x.2012.03662.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND The aim of the present study was to investigate the expression and activity of epithelial sodium channel (ENaC) in hyperoxia-induced bronchopulmonary dysplasia (BPD) in neonatal rats. METHODS Neonatal rats were exposed to hyperoxia to establish BPD models (control group was exposed to air), lung water was measured and Western blot was applied to detect the expression of three homologous subunits: α-, β- and γ-ENaC in the lung tissues. Furthermore, ATII cells were isolated from neonatal rats, and primarily cultured under normoxic or hyperoxic conditions. The ENaC expression was also examined in these cells. In addition, the amiloride-sensitive Na(+) currents induced by hyperoxia were recorded using the whole-cell patch clamp technique. RESULTS The α-ENaC expression was increased after 5 days of hyperoxia in rat lung tissues, whereas not after 1, 3 and 7 days. ATII cells showed α-ENaC expression was reduced after 1 and 2 days' hyperoxia, but no change after 3 days. In contrast, β- and γ-ENaC expression was increased after hyperoxia in both in vivo and in vitro experiments. The amiloride-sensitive Na(+) currents in hyperoxia-exposed ATII cells were also increased, which was consistent with the upregulated expression of β- and γ-ENaC. CONCLUSION Hyperoxia upregulates the expression of ENaC, especially β- and γ-ENaC subunits, in both neonatal rat lung tissues and ATII cells. Hyperoxia also enhanced the activity of ENaC in neonatal rat ATII cells. Dysfunctional transport of Na(+) may not be a key factor involving pulmonary edema at the early stage of BPD.
Collapse
Affiliation(s)
- Weihua Ji
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | | | | | | |
Collapse
|
36
|
Matthay MA, Ware LB, Zimmerman GA. The acute respiratory distress syndrome. J Clin Invest 2012; 122:2731-40. [PMID: 22850883 DOI: 10.1172/jci60331] [Citation(s) in RCA: 1364] [Impact Index Per Article: 104.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The acute respiratory distress syndrome (ARDS) is an important cause of acute respiratory failure that is often associated with multiple organ failure. Several clinical disorders can precipitate ARDS, including pneumonia, sepsis, aspiration of gastric contents, and major trauma. Physiologically, ARDS is characterized by increased permeability pulmonary edema, severe arterial hypoxemia, and impaired carbon dioxide excretion. Based on both experimental and clinical studies, progress has been made in understanding the mechanisms responsible for the pathogenesis and the resolution of lung injury, including the contribution of environmental and genetic factors. Improved survival has been achieved with the use of lung-protective ventilation. Future progress will depend on developing novel therapeutics that can facilitate and enhance lung repair.
Collapse
Affiliation(s)
- Michael A Matthay
- Cardiovascular Research Institute and Departments of Medicine and Anesthesia, UCSF, San Francisco, CA, USA.
| | | | | |
Collapse
|
37
|
Abstract
The number of patients listed for lung transplantation largely exceeds the number of available transplantable organs because of both a shortage of organ donors and a low utilization rate of donor lungs. Normothermic ex vivo lung perfusion (EVLP) is a method that maintains the organ in physiologically protective conditions outside the body during preservation, and shows great promise to increase utilization of donor lungs by allowing more accurate evaluation, as well as treatment and repair, of damaged donor lungs prior to transplantation. This article will cover the rationale, technical details and results of experimental and clinical studies with EVLP. The significant potential applications of EVLP in lung transplantation, lung regeneration and oncology are discussed.
Collapse
|
38
|
Fronius M, Clauss WG, Althaus M. Why Do We have to Move Fluid to be Able to Breathe? Front Physiol 2012; 3:146. [PMID: 22661953 PMCID: PMC3357553 DOI: 10.3389/fphys.2012.00146] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 05/01/2012] [Indexed: 12/11/2022] Open
Abstract
The ability to breathe air represents a fundamental step in vertebrate evolution that was accompanied by several anatomical and physiological adaptations. The morphology of the air-blood barrier is highly conserved within air-breathing vertebrates. It is formed by three different plies, which are represented by the alveolar epithelium, the basal lamina, and the endothelial layer. Besides these conserved morphological elements, another common feature of vertebrate lungs is that they contain a certain amount of fluid that covers the alveolar epithelium. The volume and composition of the alveolar fluid is regulated by transepithelial ion transport mechanisms expressed in alveolar epithelial cells. These transport mechanisms have been reviewed extensively. Therefore, the present review focuses on the properties and functional significance of the alveolar fluid. How does the fluid enter the alveoli? What is the fate of the fluid in the alveoli? What is the function of the alveolar fluid in the lungs? The review highlights the importance of the alveolar fluid, its volume and its composition. Maintenance of the fluid volume and composition within certain limits is critical to facilitate gas exchange. We propose that the alveolar fluid is an essential element of the air-blood barrier. Therefore, it is appropriate to refer to this barrier as being formed by four plies, namely (1) the thin fluid layer covering the apical membrane of the epithelial cells, (2) the epithelial cell layer, (3) the basal membrane, and (4) the endothelial cells.
Collapse
Affiliation(s)
- Martin Fronius
- Molecular Cell Physiology, Institute of Animal Physiology, Justus-Liebig-University Giessen Giessen, Germany
| | | | | |
Collapse
|
39
|
Faria D, Lentze N, Almaça J, Luz S, Alessio L, Tian Y, Martins JP, Cruz P, Schreiber R, Rezwan M, Farinha CM, Auerbach D, Amaral MD, Kunzelmann K. Regulation of ENaC biogenesis by the stress response protein SERP1. Pflugers Arch 2012; 463:819-27. [PMID: 22526458 DOI: 10.1007/s00424-012-1091-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Revised: 02/12/2012] [Accepted: 02/21/2012] [Indexed: 11/29/2022]
Abstract
Cystic fibrosis lung disease is caused by reduced Cl(-) secretion along with enhanced Na(+) absorption, leading to reduced airway surface liquid and compromised mucociliary clearance. Therapeutic strategies have been developed to activate cystic fibrosis transmembrane conductance regulator (CFTR) or to overcome enhanced Na(+) absorption by the epithelial Na(+) channel (ENaC). In a split-ubiquitin-based two-hybrid screening, we identified stress-associated ER protein 1 (SERP1)/ribosome-associated membrane protein 4 as a novel interacting partner for the ENaC β-subunit. SERP1 is induced during cell stress and interacts with the molecular chaperone calnexin, thus controlling early biogenesis of membrane proteins. ENaC activity was measured in the human airway epithelial cell lines H441 and A549 and in voltage clamp experiments with ENaC-overexpressing Xenopus oocytes. We found that expression of SERP1 strongly inhibits amiloride-sensitive Na(+) transport. SERP1 coimmunoprecipitated and colocalized with βENaC in the endoplasmic reticulum, together with the chaperone calnexin. In contrast to the inhibitory effects on ENaC, SERP1 appears to promote expression of CFTR. Taken together, SERP1 is a novel cochaperone and regulator of ENaC expression.
Collapse
Affiliation(s)
- Diana Faria
- Department of Physiology, University of Regensburg, Regensburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Lee JW, Fang X, Krasnodembskaya A, Howard JP, Matthay MA. Concise review: Mesenchymal stem cells for acute lung injury: role of paracrine soluble factors. Stem Cells 2011; 29:913-9. [PMID: 21506195 DOI: 10.1002/stem.643] [Citation(s) in RCA: 318] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Morbidity and mortality have declined only modestly in patients with clinical acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), despite extensive research into the pathophysiology. Current treatment remains primarily supportive with lung-protective ventilation and a fluid conservative strategy. Pharmacologic therapies that reduce the severity of lung injury in preclinical models have not yet been translated to effective clinical treatment options. Consequently, further research in translational therapies is needed. Cell-based therapy with mesenchymal stem cells (MSCs) is one attractive new therapeutic approach. MSCs have the capacity to secrete multiple paracrine factors that can regulate endothelial and epithelial permeability, decrease inflammation, enhance tissue repair, and inhibit bacterial growth. This review will focus on recent studies, which support the potential therapeutic use of MSCs in ALI/ARDS, with an emphasis on the role of paracrine soluble factors.
Collapse
Affiliation(s)
- Jae W Lee
- Department of Anesthesiology, University of California San Francisco, California 94143, USA.
| | | | | | | | | |
Collapse
|
42
|
Janér C, Pitkänen OM, Helve O, Andersson S. Airway expression of the epithelial sodium channel α-subunit correlates with cortisol in term newborns. Pediatrics 2011; 128:e414-21. [PMID: 21768316 DOI: 10.1542/peds.2011-0167] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Glucocorticoids have profound effects on lung maturation and function. In in vitro and animal models, they induce epithelial sodium channels (ENaCs) in the airway epithelium, a process that is important to perinatal lung fluid clearance. OBJECTIVE The objective of this study was to determine whether, in newborn infants, airway ENaC expression is associated with cortisol concentrations. METHODS Cord blood, saliva, and cells from nasal epithelium were obtained from 69 infants delivered at term. Epithelial and saliva sampling was repeated 3 times: <3, 22 to 29, and 40 to 54 hours postnatally. Cortisol, thyrotropin, and free triiodothyronine concentrations were measured with immunoassays, and expression of α-ENaC and β-ENaC was quantified with real-time reverse-transcriptase polymerase chain reaction. RESULTS Expression of α-ENaC <30 minutes postnatally correlated with cord plasma cortisol in infants delivered by elective cesarean delivery. In addition, in the total study population <2 hours postnatally, α-ENaC expression correlated with salivary cortisol concentrations. β-ENaC expression, in contrast, showed no association with cortisol concentrations. A significant decrease in β-ENaC expression during the first postnatal day was revealed, whereas timing of the peak in α-ENaC expression seemed to depend on mode of delivery. CONCLUSIONS These results support a role in humans for endogenous glucocorticoids in the regulation of airway ion transport. This finding may be a physiologic mechanism mediating pulmonary adaptation in the newborn infant.
Collapse
Affiliation(s)
- Cecilia Janér
- Children's Hospital, Pediatric Research Center, Helsinki University Central Hospital, Biomedicum Helsinki 2 U E104b, Helsinki, Finland.
| | | | | | | |
Collapse
|
43
|
Clerici C. The challenge of modeling human acute respiratory distress syndrome: a new model of lung injury due to sepsis with impaired alveolar edema fluid removal. Am J Physiol Lung Cell Mol Physiol 2011; 301:L20-2. [DOI: 10.1152/ajplung.00126.2011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Christine Clerici
- Université Paris Diderot, Sorbonne Paris Cité, Department of Physiology, F:75013 Paris, France; Institut National de la Santé et de la Recherche Médicale, U773, CRB3, F:75018 Paris; and AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| |
Collapse
|
44
|
Baloglu E, Reingruber T, Bärtsch P, Mairbäurl H. β2-Adrenergics in hypoxia desensitize receptors but blunt inhibition of reabsorption in rat lungs. Am J Respir Cell Mol Biol 2011; 45:1059-68. [PMID: 21562317 DOI: 10.1165/rcmb.2010-0273oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Alveolar edema and decreased inspired Po(2) decrease the oxygen supply to alveolar epithelia, impairing β(2)-adrenergic receptor (β2AR) signaling and alveolar reabsorption. β2AR agonists potently stimulate alveolar reabsorption. Thus, hypoxia impairs a major defense mechanism that provides protection from alveolar edema. Because in vivo data on the combined effects of prolonged hypoxia and β2AR agonist treatment on β2AR signaling are sparse, we tested whether in vivo hypoxia augments the inactivation of β2AR during prolonged stimulation. Rats were exposed to normoxia (N) and hypoxia (8% O(2); H), and were also treated with terbutaline (T; 2.5 mg/kg, intraperitoneal, twice daily) or saline (S) for 4 days. β2AR signaling was studied in alveolar epithelial (ATII) cells and in whole-lung tissue from treated rats. The terbutaline-stimulated formation of cyclic adenosine monophosphate was decreased by approximately 40% in whole lung and in ATII cells of NT, HS, and HT. The effects were not additive. The β2AR number was increased in HS, but decreased in NT and HT. Treatment increased the G-protein-coupled receptor kinase 2 protein in the plasma membranes of ATII cells, but did not affect G proteins. In vivo hypoxia significantly decreased total and amiloride-sensitive alveolar fluid reabsorption, which was prevented by acute alveolar treatment and 4 days of systemic terbutaline treatment. The αENaC (subunit of epithelial Na channels) protein in plasma membranes was increased in HT, without effects on mRNA. These results indicate that prolonged alveolar hypoxia and treatment with terbutaline impaired β2AR signaling in alveolar epithelia and in whole lungs, and this signaling was not further impaired by hypoxia. Despite impaired β2AR signaling, treatment with terbutaline for 4 days prevented the inhibition of alveolar reabsorption caused by in vivo hypoxia.
Collapse
Affiliation(s)
- Emel Baloglu
- Medical Clinic VII, Sports Medicine, University Hospital Heidelberg, University of Heidelberg, Germany
| | | | | | | |
Collapse
|
45
|
Dodrill MW, Beezhold DH, Meighan T, Kashon ML, Fedan JS. Lipopolysaccharide increases Na+,K+-pump, but not ENaC, expression in guinea-pig airway epithelium. Eur J Pharmacol 2011; 651:176-86. [DOI: 10.1016/j.ejphar.2010.10.088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 10/18/2010] [Accepted: 10/31/2010] [Indexed: 10/18/2022]
|
46
|
Amiloride-sensitive sodium channels and pulmonary edema. Pulm Med 2010; 2011:830320. [PMID: 21637371 PMCID: PMC3100597 DOI: 10.1155/2011/830320] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 12/01/2010] [Indexed: 01/11/2023] Open
Abstract
The development of pulmonary edema can be considered as a combination of alveolar flooding via increased fluid filtration, impaired alveolar-capillary barrier integrity, and disturbed resolution due to decreased alveolar fluid clearance. An important mechanism regulating alveolar fluid clearance is sodium transport across the alveolar epithelium. Transepithelial sodium transport is largely dependent on the activity of sodium channels in alveolar epithelial cells. This paper describes how sodium channels contribute to alveolar fluid clearance under physiological conditions and how deregulation of sodium channel activity might contribute to the pathogenesis of lung diseases associated with pulmonary edema. Furthermore, sodium channels as putative molecular targets for the treatment of pulmonary edema are discussed.
Collapse
|
47
|
Husted RF, Lu H, Sigmund RD, Stokes JB. Oxygen regulation of the epithelial Na channel in the collecting duct. Am J Physiol Renal Physiol 2010; 300:F412-24. [PMID: 21123494 DOI: 10.1152/ajprenal.00245.2010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The PO(2) within the kidney changes dramatically from cortex to medulla. The present experiments examined the effect of changing PO(2) on epithelial Na channel (ENaC)-mediated Na transport in the collecting duct using the mpkCCD-c14 cell line. Decreasing ambient O(2) concentration from 20 to 8% decreased ENaC activity by 40%; increasing O(2) content to 40% increased ENaC activity by 50%. The O(2) effect required several hours to develop and was not mimicked by the acid pH that developed in monolayers incubated in low-O(2) medium. Corticosteroids increased ENaC activity at each O(2) concentration; there was no interaction. The pathways for O(2) and steroid regulation of ENaC are different since O(2) did not substantially affect Sgk1, α-ENaC, Gilz, or Usp2-45 mRNA levels, genes involved in steroid-mediated ENaC regulation. The regulation of ENaC activity by these levels of O(2) appears not to be mediated by changes in hypoxia-inducible factor-1α or -2α activity or a change in AMP kinase activity. Changes in O(2) concentration had minimal effect on α- or γ-ENaC mRNA and protein levels; there were moderate effects on β-ENaC levels. However, 40% O(2) induced substantially greater total β- and γ-ENaC on the apical surface compared with 8% O(2); both subunits demonstrated a greater increase in the mature forms. The α-ENaC subunit was difficult to detect on the apical surface, perhaps because our antibodies do not recognize the major mature form. These results identify a mechanism of ENaC regulation that may be important in different regions of the kidney and in responses to changes in dietary NaCl.
Collapse
Affiliation(s)
- Russell F Husted
- Fraternal Order of Eagles Diabetes Research Center, Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
48
|
Mac Sweeney R, Fischer H, McAuley DF. Nasal potential difference to detect Na+ channel dysfunction in acute lung injury. Am J Physiol Lung Cell Mol Physiol 2010; 300:L305-18. [PMID: 21112943 DOI: 10.1152/ajplung.00223.2010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Pulmonary fluid clearance is regulated by the active transport of Na(+) and Cl(-) through respiratory epithelial ion channels. Ion channel dysfunction contributes to the pathogenesis of various pulmonary fluid disorders including high-altitude pulmonary edema (HAPE) and neonatal respiratory distress syndrome (RDS). Nasal potential difference (NPD) measurement allows an in vivo investigation of the functionality of these channels. This technique has been used for the diagnosis of cystic fibrosis, the archetypal respiratory ion channel disorder, for over a quarter of a century. NPD measurements in HAPE and RDS suggest constitutive and acquired dysfunction of respiratory epithelial Na(+) channels. Acute lung injury (ALI) is characterized by pulmonary edema due to alveolar epithelial-interstitial-endothelial injury. NPD measurement may enable identification of critically ill ALI patients with a susceptible phenotype of dysfunctional respiratory Na(+) channels and allow targeted therapy toward Na(+) channel function.
Collapse
Affiliation(s)
- R Mac Sweeney
- Respiratory Medicine Research Programme, Centre for Infection and Immunity, Queen’s University, Belfast, Northern Ireland
| | | | | |
Collapse
|
49
|
Hee L, Dinudom A, Mitchell AJ, Grau GE, Cook DI, Hunt NH, Ball HJ. Reduced activity of the epithelial sodium channel in malaria-induced pulmonary oedema in mice. Int J Parasitol 2010; 41:81-8. [PMID: 20816846 PMCID: PMC7125784 DOI: 10.1016/j.ijpara.2010.07.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Revised: 07/22/2010] [Accepted: 07/23/2010] [Indexed: 11/05/2022]
Abstract
Lung complications during malaria infection can range from coughs and impairments in gas transfer to the development of acute respiratory distress syndrome (ARDS). Infecting C57BL/6 mice with Plasmodium berghei K173 strain (PbK) resulted in pulmonary oedema, capillaries congested with leukocytes and infected red blood cells (iRBCs), and leukocyte infiltration into the lungs. This new model of malaria-associated lung pathology, without any accompanying cerebral complications, allows the investigation of mechanisms leading to the lung disease. The activity of the amiloride-sensitive epithelial sodium channel (ENaC) in alveolar epithelial cells is decreased by several respiratory tract pathogens and this is suggested to contribute to pulmonary oedema. We show that PbK, a pathogen that remains in the circulation, also decreased the activity and expression of ENaC, suggesting that infectious agents can have indirect effects on ENaC activity in lung epithelial cells. The reduced ENaC activity may contribute to the pulmonary oedema induced by PbK malaria.
Collapse
Affiliation(s)
- Leia Hee
- Discipline of Pathology, School of Medical Sciences and Bosch Institute, University of Sydney, NSW 2006, Australia
| | | | | | | | | | | | | |
Collapse
|
50
|
Dodrill MW, Fedan JS. Lipopolysaccharide hyperpolarizes guinea pig airway epithelium by increasing the activities of the epithelial Na(+) channel and the Na(+)-K(+) pump. Am J Physiol Lung Cell Mol Physiol 2010; 299:L550-8. [PMID: 20639350 DOI: 10.1152/ajplung.00123.2010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Earlier, we found that systemic administration of lipopolysaccharide (LPS; 4 mg/kg) hyperpolarized the transepithelial potential difference (V(t)) of tracheal epithelium in the isolated, perfused trachea (IPT) of the guinea pig 18 h after injection. As well, LPS increased the hyperpolarization component of the response to basolateral methacholine, and potentiated the epithelium-derived relaxing factor-mediated relaxation responses to hyperosmolar solutions applied to the apical membrane. We hypothesized that LPS stimulates the transepithelial movement of Na(+) via the epithelial sodium channel (ENaC)/Na(+)-K(+) pump axis, leading to hyperpolarization of V(t). LPS increased the V(t)-depolarizing response to amiloride (10 μM), i.e., offset the effect of LPS, indicating that Na(+) transport activity was increased. The functional activity of ENaC was measured in the IPT after short-circuiting the Na(+)-K(+) pump with basolateral amphotericin B (7.5 μM). LPS had no effect on the hyperpolarization response to apical trypsin (100 U/ml) in the Ussing chamber, indicating that channel-activating proteases are not involved in the LPS-induced activation of ENaC. To assess Na(+)-K(+) pump activity in the IPT, ENaC was short-circuited with apical amphotericin B. The greater V(t) in the presence of amphotericin B in tracheas from LPS-treated animals compared with controls revealed that LPS increased Na(+)-K(+) pump activity. This finding was confirmed in the Ussing chamber by inhibiting the Na(+)-K(+) pump via extracellular K(+) removal, loading the epithelium with Na(+), and observing a greater hyperpolarization response to K(+) restoration. Together, the findings of this study reveal that LPS hyperpolarizes the airway epithelium by increasing the activities of ENaC and the Na(+)-K(+) pump.
Collapse
Affiliation(s)
- Michael W Dodrill
- Department of Basic Pharmaceutical Sciences, Robert C. Byrd Health Sciences Center, West Virginia University, USA
| | | |
Collapse
|