1
|
Mthunzi L, Islam MN, Gusarova GA, Bhattacharya S, Karolewski B, Bhattacharya J. Macrophage-specific lipid nanoparticle therapy blocks the lung's mechanosensitive immunity due to macrophage-epithelial interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.24.541735. [PMID: 37292826 PMCID: PMC10245918 DOI: 10.1101/2023.05.24.541735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The lung's mechanosensitive immune response, which occurs when pulmonary alveoli are overstretched, is a major impediment to ventilation therapy for hypoxemic respiratory failure. The cause is not known. We tested the hypothesis that alveolar stretch causes stretch of alveolar macrophages (AMs), leading to the immune response. In lungs viewed by optical imaging, sessile AMs expressed gap junctional protein connexin-43 (Cx43), and they communicated with the alveolar epithelium through gap junctions. Alveolar hyperinflation increased Ca 2+ in the AMs but did not stretch the AMs. The Ca 2+ response, and concomitant TNFα secretion by AMs were blocked in mice with AM-specific deletion of Cx43. The AM responses, as also lung injury due to mechanical ventilation at high tidal volume, were inhibited by AM-specific delivery of lipid nanoparticles containing Xestospongin C, which blocked the induced Ca 2+ increases. We conclude, Cx43- and Ca 2+ -dependent AM-epithelial interactions determine the lung's mechanosensitive immunity, providing a basis for therapy for ventilator- induced lung injury.
Collapse
|
2
|
Hook JL. A role for TMEM63 in the lung. J Clin Invest 2024; 134:e178948. [PMID: 38426500 PMCID: PMC10904035 DOI: 10.1172/jci178948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Surfactants are essential for breathing. Although major progress has been made in the past half century toward an understanding of surfactant secretion mechanisms, the identity of the mechanosensor that couples breathing to surfactant secretion has remained elusive. In this issue of the JCI, Chen, Li, and colleagues provide evidence that the mechanosensor is the transmembrane 63 (TMEM63) ion channel. These findings open new avenues for future research into lung mechanobiology.
Collapse
Affiliation(s)
- Jaime L. Hook
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine and
- Global Health and Emerging Pathogens Institute, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
3
|
Abstract
Pulmonary surfactant is a critical component of lung function in healthy individuals. It functions in part by lowering surface tension in the alveoli, thereby allowing for breathing with minimal effort. The prevailing thinking is that low surface tension is attained by a compression-driven squeeze-out of unsaturated phospholipids during exhalation, forming a film enriched in saturated phospholipids that achieves surface tensions close to zero. A thorough review of past and recent literature suggests that the compression-driven squeeze-out mechanism may be erroneous. Here, we posit that a surfactant film enriched in saturated lipids is formed shortly after birth by an adsorption-driven sorting process and that its composition does not change during normal breathing. We provide biophysical evidence for the rapid formation of an enriched film at high surfactant concentrations, facilitated by adsorption structures containing hydrophobic surfactant proteins. We examine biophysical evidence for and against the compression-driven squeeze-out mechanism and propose a new model for surfactant function. The proposed model is tested against existing physiological and pathophysiological evidence in neonatal and adult lungs, leading to ideas for biophysical research, that should be addressed to establish the physiological relevance of this new perspective on the function of the mighty thin film that surfactant provides.
Collapse
Affiliation(s)
- Fred Possmayer
- Department of Biochemistry, Western University, London, Ontario N6A 3K7, Canada
- Department of Obstetrics/Gynaecology, Western University, London, Ontario N6A 3K7, Canada
| | - Yi Y Zuo
- Department of Mechanical Engineering, University of Hawaii at Manon, Honolulu, Hawaii 96822, United States
- Department of Pediatrics, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii 96826, United States
| | - Ruud A W Veldhuizen
- Department of Physiology & Pharmacology, Western University, London, Ontario N6A 5C1, Canada
- Department of Medicine, Western University, London, Ontario N6A 3K7, Canada
- Lawson Health Research Institute, London, Ontario N6A 4V2, Canada
| | - Nils O Petersen
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2G2, Canada
- Department of Chemistry, Western University, London, Ontario N6A 5B7, Canada
| |
Collapse
|
4
|
Xu W, Ma X, Wang Q, Ye J, Wang N, Ye Z, Chen T. GCN5L1 regulates pulmonary surfactant production by modulating lamellar body biogenesis and trafficking in mouse alveolar epithelial cells. Cell Mol Biol Lett 2023; 28:90. [PMID: 37936104 PMCID: PMC10631113 DOI: 10.1186/s11658-023-00506-0] [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: 06/05/2023] [Accepted: 10/25/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND The pulmonary surfactant that lines the air-liquid surface within alveoli is a protein-lipid mixture essential for gas exchange. Surfactant lipids and proteins are synthesized and stored in the lamellar body (LB) before being secreted from alveolar type II (AT2) cells. The molecular and cellular mechanisms that regulate these processes are incompletely understood. We previously identified an essential role of general control of amino acid synthesis 5 like 1 (GCN5L1) and the biogenesis of lysosome-related organelle complex 1 subunit 1 (BLOS1) in surfactant system development in zebrafish. Here, we explored the role of GCN5L1 in pulmonary surfactant regulation. METHOD GCN5L1 knockout cell lines were generated with the CRISPR/Cas9 system. Cell viability was analyzed by MTT assay. Released surfactant proteins were measured by ELISA. Released surfactant lipids were measured based on coupled enzymatic reactions. Gene overexpression was mediated through lentivirus. The RNA levels were detected through RNA-sequencing (RNA-seq) and quantitative reverse transcription (qRT)- polymerase chain reaction (PCR). The protein levels were detected through western blotting. The cellular localization was analyzed by immunofluorescence. Morphology of the lamellar body was analyzed through transmission electron microscopy (TEM), Lysotracker staining, and BODIPY phosphatidylcholine labeling. RESULTS Knocking out GCN5L1 in MLE-12 significantly decreased the release of surfactant proteins and lipids. We detected the downregulation of some surfactant-related genes and misregulation of the ROS-Erk-Foxo1-Cebpα axis in mutant cells. Modulating the activity of the axis or reconstructing the mitochondrial expression of GCN5L1 could partially restore the expression of these surfactant-related genes. We further showed that MLE-12 cells contained many LB-like organelles that were lipid enriched and positive for multiple LB markers. These organelles were smaller in size and accumulated in the absence of GCN5L1, indicating both biogenesis and trafficking defects. Accumulated endogenous surfactant protein (SP)-B or exogenously expressed SP-B/SP-C in adenosine triphosphate-binding cassette transporterA3 (ABCA3)-positive organelles was detected in mutant cells. GCN5L1 localized to the mitochondria and LBs. Reconstruction of mitochondrial GCN5L1 expression rescued the organelle morphology but failed to restore the trafficking defect and surfactant release, indicating specific roles associated with different subcellular localizations. CONCLUSIONS In summary, our study identified GCN5L1 as a new regulator of pulmonary surfactant that plays a role in the biogenesis and positioning/trafficking of surfactant-containing LBs.
Collapse
Affiliation(s)
- Wenqin Xu
- Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, China
| | - Xiaocui Ma
- Henan Clinical Research Center of Childhood Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Qing Wang
- Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, China
| | - Jingjing Ye
- Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, China
| | - Nengqian Wang
- Department of Pediatrics, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Zhenzhen Ye
- Department of Pediatrics, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Tianbing Chen
- Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, China.
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wannan Medical College, Wuhu, China.
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Wannan Medical College, Wuhu, China.
| |
Collapse
|
5
|
Tang S, De Jesus AC, Chavez D, Suthakaran S, Moore SK, Suthakaran K, Homami S, Rathnasinghe R, May AJ, Schotsaert M, Britto CJ, Bhattacharya J, Hook JL. Rescue of alveolar wall liquid secretion blocks fatal lung injury due to influenza-staphylococcal coinfection. J Clin Invest 2023; 133:e163402. [PMID: 37581936 PMCID: PMC10541650 DOI: 10.1172/jci163402] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/10/2023] [Indexed: 08/17/2023] Open
Abstract
Secondary lung infection by inhaled Staphylococcus aureus (SA) is a common and lethal event for individuals infected with influenza A virus (IAV). How IAV disrupts host defense to promote SA infection in lung alveoli, where fatal lung injury occurs, is not known. We addressed this issue using real-time determinations of alveolar responses to IAV in live, intact, perfused lungs. Our findings show that IAV infection blocked defensive alveolar wall liquid (AWL) secretion and induced airspace liquid absorption, thereby reversing normal alveolar liquid dynamics and inhibiting alveolar clearance of inhaled SA. Loss of AWL secretion resulted from inhibition of the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel in the alveolar epithelium, and airspace liquid absorption was caused by stimulation of the alveolar epithelial Na+ channel (ENaC). Loss of AWL secretion promoted alveolar stabilization of inhaled SA, but rescue of AWL secretion protected against alveolar SA stabilization and fatal SA-induced lung injury in IAV-infected mice. These findings reveal a central role for AWL secretion in alveolar defense against inhaled SA and identify AWL inhibition as a critical mechanism of IAV lung pathogenesis. AWL rescue may represent a new therapeutic approach for IAV-SA coinfection.
Collapse
Affiliation(s)
- Stephanie Tang
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
- Graduate School of Biomedical Sciences
| | - Ana Cassandra De Jesus
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Deebly Chavez
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Sayahi Suthakaran
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
- Graduate School of Biomedical Sciences
| | - Sarah K.L. Moore
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Keshon Suthakaran
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
| | - Sonya Homami
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
- Graduate School of Biomedical Sciences
| | - Raveen Rathnasinghe
- Graduate School of Biomedical Sciences
- Global Health and Emerging Pathogens Institute, Department of Microbiology
| | - Alison J. May
- Department of Cell, Developmental and Regenerative Biology
- Department of Otolaryngology, and
- Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Schotsaert
- Global Health and Emerging Pathogens Institute, Department of Microbiology
| | - Clemente J. Britto
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jahar Bhattacharya
- Departments of Medicine and Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
| | - Jaime L. Hook
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine
- Global Health and Emerging Pathogens Institute, Department of Microbiology
| |
Collapse
|
6
|
The mitochondrial calcium uniporter of pulmonary type 2 cells determines severity of acute lung injury. Nat Commun 2022; 13:5837. [PMID: 36192486 PMCID: PMC9529882 DOI: 10.1038/s41467-022-33543-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 09/21/2022] [Indexed: 11/28/2022] Open
Abstract
Acute Lung Injury (ALI) due to inhaled pathogens causes high mortality. Underlying mechanisms are inadequately understood. Here, by optical imaging of live mouse lungs we show that a key mechanism is the viability of cytosolic Ca2+ buffering by the mitochondrial Ca2+ uniporter (MCU) in the lung’s surfactant-secreting, alveolar type 2 cells (AT2). The buffering increased mitochondrial Ca2+ and induced surfactant secretion in wild-type mice, but not in mice with AT2-specific MCU knockout. In the knockout mice, ALI due to intranasal LPS instillation caused severe pulmonary edema and mortality, which were mitigated by surfactant replenishment prior to LPS instillation, indicating surfactant’s protective effect against alveolar edema. In wild-type mice, intranasal LPS, or Pseudomonas aeruginosa decreased AT2 MCU. Loss of MCU abrogated buffering. The resulting mortality was reduced by spontaneous recovery of MCU expression, or by MCU replenishment. Enhancement of AT2 mitochondrial buffering, hence endogenous surfactant secretion, through MCU replenishment might be a therapy against ALI. Acute lung injury caused by inhalation of pathogens leads to mortality, but the mechanisms are unclear. Here, the authors show in mice that that loss of the mitochondrial calcium uniporter (MCU) of alveolar type 2 cells (AT2) impaired mitochondrial Ca2+ buffering and surfactant secretion, and increased mortality, in response to LPS instillation, suggesting the MCU as a potential therapeutic target in ALI.
Collapse
|
7
|
Sanches Santos Rizzo Zuttion M, Moore SKL, Chen P, Beppu AK, Hook JL. New Insights into the Alveolar Epithelium as a Driver of Acute Respiratory Distress Syndrome. Biomolecules 2022; 12:biom12091273. [PMID: 36139112 PMCID: PMC9496395 DOI: 10.3390/biom12091273] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
The alveolar epithelium serves as a barrier between the body and the external environment. To maintain efficient gas exchange, the alveolar epithelium has evolved to withstand and rapidly respond to an assortment of inhaled, injury-inducing stimuli. However, alveolar damage can lead to loss of alveolar fluid barrier function and exuberant, non-resolving inflammation that manifests clinically as acute respiratory distress syndrome (ARDS). This review discusses recent discoveries related to mechanisms of alveolar homeostasis, injury, repair, and regeneration, with a contemporary emphasis on virus-induced lung injury. In addition, we address new insights into how the alveolar epithelium coordinates injury-induced lung inflammation and review maladaptive lung responses to alveolar damage that drive ARDS and pathologic lung remodeling.
Collapse
Affiliation(s)
- Marilia Sanches Santos Rizzo Zuttion
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sarah Kathryn Littlehale Moore
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter Chen
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew Kota Beppu
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jaime Lynn Hook
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence:
| |
Collapse
|
8
|
Ali M, Zhang X, LaCanna R, Tomar D, Elrod JW, Tian Y. MICU1-dependent mitochondrial calcium uptake regulates lung alveolar type 2 cell plasticity and lung regeneration. JCI Insight 2022; 7:154447. [PMID: 35050901 PMCID: PMC8876408 DOI: 10.1172/jci.insight.154447] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022] Open
Abstract
Lung alveolar type 2 (AT2) cells are progenitors for alveolar type 1 (AT1) cells. Although many factors regulate AT2 cell plasticity, the role of mitochondrial calcium (mCa2+) uptake in controlling AT2 cells remains unclear. We previously identified that the miR-302 family supports lung epithelial progenitor cell proliferation and less differentiated phenotypes during development. Here, we report that a sustained elevation of miR-302 in adult AT2 cells decreases AT2-to-AT1 cell differentiation during the Streptococcus pneumoniae–induced lung injury repair. We identified that miR-302 targets and represses the expression of mitochondrial Ca2+ uptake 1 (MICU1), which regulates mCa2+ uptake through the mCa2+ uniporter channel by acting as a gatekeeper at low cytosolic Ca2+ levels. Our results reveal a marked increase in MICU1 protein expression and decreased mCa2+ uptake during AT2-to-AT1 cell differentiation in the adult lung. Deletion of Micu1 in AT2 cells reduces AT2-to-AT1 cell differentiation during steady-state tissue maintenance and alveolar epithelial regeneration after bacterial pneumonia. These studies indicate that mCa2+ uptake is extensively modulated during AT2-to-AT1 cell differentiation and that MICU1-dependent mCa2+ uniporter channel gating is a prominent mechanism modulating AT2-to-AT1 cell differentiation.
Collapse
Affiliation(s)
- Mir Ali
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Xiaoying Zhang
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Ryan LaCanna
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Dhanendra Tomar
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - John W Elrod
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| | - Ying Tian
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, United States of America
| |
Collapse
|
9
|
Gusarova GA, Das SR, Islam MN, Westphalen K, Jin G, Shmarakov IO, Li L, Bhattacharya S, Bhattacharya J. Actin fence therapy with exogenous V12Rac1 protects against acute lung injury. JCI Insight 2021; 6:135753. [PMID: 33749665 PMCID: PMC8026177 DOI: 10.1172/jci.insight.135753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/11/2021] [Indexed: 02/05/2023] Open
Abstract
High mortality in acute lung injury (ALI) results from sustained proinflammatory signaling by alveolar receptors, such as TNF-α receptor type 1 (TNFR1). Factors that determine the sustained signaling are not known. Unexpectedly, optical imaging of live alveoli revealed a major TNF-α–induced surge of alveolar TNFR1 due to a Ca2+-dependent mechanism that decreased the cortical actin fence. Mouse mortality due to inhaled LPS was associated with cofilin activation, actin loss, and the TNFR1 surge. The constitutively active form of the GTPase, Rac1 (V12Rac1), given intranasally (i.n.) as a noncovalent construct with a cell-permeable peptide, enhanced alveolar filamentous actin (F-actin) and blocked the TNFR1 surge. V12Rac1 also protected against ALI-induced mortality resulting from i.n. instillation of LPS or of Pseudomonas aeruginosa. We propose a potentially new therapeutic paradigm in which actin enhancement by exogenous Rac1 strengthens the alveolar actin fence, protecting against proinflammatory receptor hyperexpression, and therefore blocking ALI.
Collapse
Affiliation(s)
- Galina A Gusarova
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Shonit R Das
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Mohammad N Islam
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Kristin Westphalen
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Guangchun Jin
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | - Li Li
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Sunita Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Pediatrics, and
| | - Jahar Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Physiology and Cellular Biophysics, Vagelos College of Physicians and Surgeons of Columbia University, New York, New York, USA
| |
Collapse
|
10
|
Islam MN, Gusarova GA, Das SR, Li L, Monma E, Anjaneyulu M, Owusu-Ansah E, Bhattacharya S, Bhattacharya J. The Mitochondrial Calcium Uniporter of Pulmonary Type 2 Cells Determines Severity of ARDS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33469582 DOI: 10.1101/2021.01.18.427173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Acute lung immunity to inhaled pathogens elicits defensive pneumonitis that may convert to the Acute Respiratory Distress Syndrome (ARDS), causing high mortality. Mechanisms underlying the conversion are not understood, but are of intense interest because of the ARDS-induced mortality in the ongoing Covid-19 pandemic. Here, by optical imaging of live lungs we show that key to the lethality is the functional status of mitochondrial Ca2+ buffering across the mitochondrial Ca2+ uniporter (MCU) in the alveolar type 2 cells (AT2), which protect alveolar stability. In mice subjected to ARDS by airway exposure to lipopolysaccharide (LPS), or to Pseudomonas aeruginosa, there was marked loss of MCU expression in AT2. The ability of mice to survive ARDS depended on the extent to which the MCU expression recovered, indicating that the viability of Ca2+ buffering by AT2 mitochondria critically determines ARDS severity. Mitochondrial transfer to enhance AT2 MCU expression might protect against ARDS.
Collapse
|
11
|
Nguyen TL, Perlman CE. Sulforhodamine B and exogenous surfactant effects on alveolar surface tension under acute respiratory distress syndrome conditions. J Appl Physiol (1985) 2020; 129:1505-1513. [PMID: 32969780 DOI: 10.1152/japplphysiol.00422.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In the acute respiratory distress syndrome (ARDS), alveolar surface tension, T, may be elevated. Elevated T should increase ventilation-induced lung injury. Exogenous surfactant therapy, intended to lower T, has not reduced mortality. Sulforhodamine B (SRB) might, alternatively, be used to lower T. We test whether substances suspected of elevating T in ARDS raise T in the lungs and test the abilities of exogenous surfactant and SRB to reduce T. In isolated rat lungs, we micropuncture a surface alveolus and instill a solution of a purported T-raising substance: control saline, cell debris, secretory phospholipase A2 (sPLA2), acid, or mucins. We test each substance alone; with albumin, to model proteinaceous edema liquid; with albumin and exogenous surfactant; and with albumin and SRB. We determine T in situ in the lungs by combining servo-nulling pressure measurement with confocal microscopy and applying the Laplace relation. With control saline, albumin does not alter T, additional surfactant raises T, and additional SRB lowers T. The experimental substances, without or with albumin, raise T. Excepting under aspiration conditions, addition of surfactant or SRB lowers T. Exogenous surfactant activity is concentration and ventilation dependent. Sulforhodamine B, which could be delivered intravascularly, holds promise as an alternative therapeutic.NEW & NOTEWORTHY In the acute respiratory distress syndrome (ARDS), lowering surface tension, T, should reduce ventilation injury yet exogenous surfactant has not reduced mortality. We show with direct T determination in isolated lungs that substances suggested to elevate T in ARDS indeed raise T, and exogenous surfactant reduces T. Further, we extend our previous finding that sulforhodamine B (SRB) reduces T below normal in healthy lungs and show that SRB, too, reduces T under ARDS conditions.
Collapse
Affiliation(s)
- Tam L Nguyen
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| | - Carrie E Perlman
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, New Jersey
| |
Collapse
|
12
|
Guenthart BA, O'Neill JD, Kim J, Queen D, Chicotka S, Fung K, Simpson M, Donocoff R, Salna M, Marboe CC, Cunningham K, Halligan SP, Wobma HM, Hozain AE, Romanov A, Vunjak-Novakovic G, Bacchetta M. Regeneration of severely damaged lungs using an interventional cross-circulation platform. Nat Commun 2019; 10:1985. [PMID: 31064987 PMCID: PMC6504972 DOI: 10.1038/s41467-019-09908-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 04/01/2019] [Indexed: 12/13/2022] Open
Abstract
The number of available donor organs limits lung transplantation, the only lifesaving therapy for the increasing population of patients with end-stage lung disease. A prevalent etiology of injury that renders lungs unacceptable for transplantation is gastric aspiration, a deleterious insult to the pulmonary epithelium. Currently, severely damaged donor lungs cannot be salvaged with existing devices or methods. Here we report the regeneration of severely damaged lungs repaired to meet transplantation criteria by utilizing an interventional cross-circulation platform in a clinically relevant swine model of gastric aspiration injury. Enabled by cross-circulation with a living swine, prolonged extracorporeal support of damaged lungs results in significant improvements in lung function, cellular regeneration, and the development of diagnostic tools for non-invasive organ evaluation and repair. We therefore propose that the use of an interventional cross-circulation platform could enable recovery of otherwise unsalvageable lungs and thus expand the donor organ pool.
Collapse
Affiliation(s)
- Brandon A Guenthart
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA.,Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - John D O'Neill
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Jinho Kim
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA.,Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, USA
| | - Dawn Queen
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Scott Chicotka
- Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Kenmond Fung
- Department of Clinical Perfusion, Columbia University Medical Center, Columbia University, New York, NY, 1003, USA
| | - Michael Simpson
- Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Rachel Donocoff
- Institute of Comparative Medicine, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Michael Salna
- Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Charles C Marboe
- Department of Pathology and Cell Biology, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Katherine Cunningham
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Susan P Halligan
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Holly M Wobma
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Ahmed E Hozain
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA.,Department of Surgery, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Alexander Romanov
- Institute of Comparative Medicine, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA. .,Department of Medicine, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA.
| | - Matthew Bacchetta
- Department of Biomedical Engineering, Columbia University Medical Center, Columbia University, New York, NY, 10032, USA. .,Department of Thoracic and Cardiovascular Surgery, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
13
|
Tamò L, Hibaoui Y, Kallol S, Alves MP, Albrecht C, Hostettler KE, Feki A, Rougier JS, Abriel H, Knudsen L, Gazdhar A, Geiser T. Generation of an alveolar epithelial type II cell line from induced pluripotent stem cells. Am J Physiol Lung Cell Mol Physiol 2018; 315:L921-L932. [DOI: 10.1152/ajplung.00357.2017] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Differentiation of primary alveolar type II epithelial cells (AEC II) to AEC type I in culture is a major barrier in the study of the alveolar epithelium in vitro. The establishment of an AEC II cell line derived from induced pluripotent stem cells (iPSC) represents a novel opportunity to study alveolar epithelial cell biology, for instance, in the context of lung injury, fibrosis, and repair. In the present study, we generated long-lasting AEC II from iPSC (LL-iPSC-AEC II). LL-iPSC-AEC II displayed morphological characteristics of AEC II, including growth in a cobblestone monolayer, the presence of lamellar bodies, and microvilli, as shown by electron microscopy. Also, LL-iPSC-AEC II expressed AEC type II proteins, such as cytokeratin, surfactant protein C, and LysoTracker DND 26 (a marker for lamellar bodies). Furthermore, the LL-iPSC-AEC II exhibited functional properties of AEC II by an increase of transepithelial electrical resistance over time, secretion of inflammatory mediators in biologically relevant quantities (IL-6 and IL-8), and efficient in vitro alveolar epithelial wound repair. Consistent with the AEC II phenotype, the cell line showed the ability to uptake and release surfactant protein B, to secrete phospholipids, and to differentiate into AEC type I. In summary, we established a long-lasting, but finite AEC type II cell line derived from iPSC as a novel cellular model to study alveolar epithelial cell biology in lung health and disease.
Collapse
Affiliation(s)
- Luca Tamò
- Department of Pulmonary Medicine, University Hospital Bern, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Youssef Hibaoui
- Department of Gynecology and Obstetrics, University Hospital Geneva, Geneva, Switzerland
| | - Sampada Kallol
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Marco P. Alves
- Department of Pediatric Pneumology, University Hospital Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Institute of Virology and Immunology, Bern, Switzerland
| | - Christiane Albrecht
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- Swiss National Center of Competence in Research, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Katrin E. Hostettler
- Clinics of Respiratory Medicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Anis Feki
- Department of Gynecology and Obstetrics, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | | | - Hugues Abriel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
- Swiss National Center of Competence in Research, National Centre of Competence in Research TransCure, University of Bern, Bern, Switzerland
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
14
|
Culture of human alveolar epithelial type II cells by sprouting. Respir Res 2018; 19:204. [PMID: 30340591 PMCID: PMC6195695 DOI: 10.1186/s12931-018-0906-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 10/01/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Type II alveolar epithelial cells (AT2) play a pivotal role in maintaining the integrity and function of the alveoli. Only recently, the role of impaired epithelial repair mechanisms after injury in the pathogenesis of idiopathic pulmonary fibrosis has been demonstrated, and has shifted the AT2 cell in the focus of interest. Therefore, using primary human AT2 cells instead of cell lines for in vitro experiments has become desirable. Several groups have developed methods to isolate human AT2 cells applying tissue digestion and consecutive filtration in their protocols. Here we present a technique to isolate primary human AT2 cells by sprouting directly from peripheral human lung tissue. METHODS Epithelial cell cultures were established from lung tissue obtained from patients undergoing diagnostic or therapeutic video-assisted thoracoscopic surgery or undergoing flexible bronchoscopy with transbronchial biopsy. Lung tissue was cut into small pieces and those were placed into cell culture flasks containing supplemented epithelial growth medium for cell sprouting. Cells were characterized by immunofluorescence stainings for E-cadherin, pan-cytokeratin, surfactant protein C (SP-C), and for lysotracker; fluorescent surfactant associated protein B (SP-B) uptake and secretion was assessed by live cell imaging; RNA levels of SP-A, SP-B, SP-C, and SP-D were determined by real-time PCR; Electron microscopy was used to search for the presence of lamellar bodies. RESULTS Sprouting of cells started two to four days after the start of culture. Epithelial differentiation was confirmed by positive staining for E-cadherin and pan-cytokeratin. Further characterization demonstrated positivity for the AT2 cell marker SP-C and for lysotracker which selectively labels lamellar bodies in cultured AT2 cells. The up-take and release of SP-B, a mechanism described for AT2 cells only, was demonstrated by live cell imaging. Real-time RT-PCR showed mRNA expression of all four surfactant proteins with highest levels for SP-B. The presence of lamellar bodies was demonstrated by electron microscopy. CONCLUSIONS This study describes a novel method for isolating AT2 cells from human adult lung tissue by sprouting. The characterization of the cultured AT2 cells complies with current criteria for an alveolar type 2 cell phenotype. Compared to current protocols for the culture of AT2 cells, isolating the cells by sprouting is simple, avoids proteolytic tissue digestion, and has the advantage to be successful even from as few tissue as attained from a transbronchial forceps biopsy.
Collapse
|
15
|
Hook JL, Islam MN, Parker D, Prince AS, Bhattacharya S, Bhattacharya J. Disruption of staphylococcal aggregation protects against lethal lung injury. J Clin Invest 2018; 128:1074-1086. [PMID: 29431734 DOI: 10.1172/jci95823] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 01/04/2018] [Indexed: 01/23/2023] Open
Abstract
Infection by Staphylococcus aureus strain USA300 causes tissue injury, multiorgan failure, and high mortality. However, the mechanisms by which the bacteria adhere to, then stabilize on, mucosal surfaces before causing injury remain unclear. We addressed these issues through the first real-time determinations of USA300-alveolar interactions in live lungs. We found that within minutes, inhaled USA300 established stable, self-associated microaggregates in niches at curved, but not at flat, regions of the alveolar wall. The microaggregates released α-hemolysin toxin, causing localized alveolar injury, as indicated by epithelial dye loss, mitochondrial depolarization, and cytosolic Ca2+ increase. Spread of cytosolic Ca2+ through intercellular gap junctions to adjoining, uninfected alveoli caused pulmonary edema. Systemic pretreatment with vancomycin, a USA300-cidal antibiotic, failed to protect mice infected with inhaled WT USA300. However, vancomycin pretreatment markedly abrogated mortality in mice infected with mutant USA300 that lacked the aggregation-promoting factor PhnD. We interpret USA300-induced mortality as having resulted from rapid bacterial aggregation in alveolar niches. These findings indicate, for the first time to our knowledge, that alveolar microanatomy is critical in promoting the aggregation and, hence, in causing USA300-induced alveolar injury. We propose that in addition to antibiotics, strategies for bacterial disaggregation may constitute novel therapy against USA300-induced lung injury.
Collapse
Affiliation(s)
- Jaime L Hook
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | - Mohammad N Islam
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine
| | | | | | - Sunita Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Pediatrics, and
| | - Jahar Bhattacharya
- Lung Biology Laboratory, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine.,Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
16
|
Brown K, Filuta A, Ludwig MG, Seuwen K, Jaros J, Vidal S, Arora K, Naren AP, Kandasamy K, Parthasarathi K, Offermanns S, Mason RJ, Miller WE, Whitsett JA, Bridges JP. Epithelial Gpr116 regulates pulmonary alveolar homeostasis via Gq/11 signaling. JCI Insight 2017; 2:93700. [PMID: 28570277 DOI: 10.1172/jci.insight.93700] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/02/2017] [Indexed: 12/27/2022] Open
Abstract
Pulmonary function is dependent upon the precise regulation of alveolar surfactant. Alterations in pulmonary surfactant concentrations or function impair ventilation and cause tissue injury. Identification of the molecular pathways that sense and regulate endogenous alveolar surfactant concentrations, coupled with the ability to pharmacologically modulate them both positively and negatively, would be a major therapeutic advance for patients with acute and chronic lung diseases caused by disruption of surfactant homeostasis. The orphan adhesion GPCR GPR116 (also known as Adgrf5) is a critical regulator of alveolar surfactant concentrations. Here, we show that human and mouse GPR116 control surfactant secretion and reuptake in alveolar type II (AT2) cells by regulating guanine nucleotide-binding domain α q and 11 (Gq/11) signaling. Synthetic peptides derived from the ectodomain of GPR116 activated Gq/11-dependent inositol phosphate conversion, calcium mobilization, and cortical F-actin stabilization to inhibit surfactant secretion. AT2 cell-specific deletion of Gnaq and Gna11 phenocopied the accumulation of surfactant observed in Gpr116-/- mice. These data provide proof of concept that GPR116 is a plausible therapeutic target to modulate endogenous alveolar surfactant pools to treat pulmonary diseases associated with surfactant dysfunction.
Collapse
Affiliation(s)
- Kari Brown
- Department of Pediatrics, Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Alyssa Filuta
- Department of Pediatrics, Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Julian Jaros
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Solange Vidal
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Kavisha Arora
- Department of Pediatrics, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Anjaparavanda P Naren
- Department of Pediatrics, Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Kathirvel Kandasamy
- Department of Physiology, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - Kaushik Parthasarathi
- Department of Physiology, University of Tennessee Health Sciences Center, Memphis, Tennessee, USA
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Robert J Mason
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
| | - William E Miller
- Department of Molecular Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jeffrey A Whitsett
- Department of Pediatrics, Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - James P Bridges
- Department of Pediatrics, Perinatal Institute, Section of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
17
|
O’Neill JD, Guenthart BA, Kim J, Chicotka S, Queen D, Fung K, Marboe C, Romanov A, Huang SXL, Chen YW, Snoeck HW, Bacchetta M, Vunjak-Novakovic G. Cross-circulation for extracorporeal support and recovery of the lung. Nat Biomed Eng 2017. [DOI: 10.1038/s41551-017-0037] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
18
|
Malacrida L, Astrada S, Briva A, Bollati-Fogolín M, Gratton E, Bagatolli LA. Spectral phasor analysis of LAURDAN fluorescence in live A549 lung cells to study the hydration and time evolution of intracellular lamellar body-like structures. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1858:2625-2635. [PMID: 27480804 PMCID: PMC5045802 DOI: 10.1016/j.bbamem.2016.07.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 10/21/2022]
Abstract
Using LAURDAN spectral imaging and spectral phasor analysis we concurrently studied the growth and hydration state of subcellular organelles (lamellar body-like, LB-like) from live A549 lung cancer cells at different post-confluence days. Our results reveal a time dependent two-step process governing the size and hydration of these intracellular LB-like structures. Specifically, a first step (days 1 to 7) is characterized by an increase in their size, followed by a second one (days 7 to 14) where the organelles display a decrease in their global hydration properties. Interestingly, our results also show that their hydration properties significantly differ from those observed in well-characterized artificial lamellar model membranes, challenging the notion that a pure lamellar membrane organization is present in these organelles at intracellular conditions. Finally, these LB-like structures show a significant increase in their hydration state upon secretion, suggesting a relevant role of entropy during this process.
Collapse
Affiliation(s)
- Leonel Malacrida
- Área de Investigación Respiratoria, Departamento de Fisiopatología, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Uruguay; Unidad de Bioquímica y Proteómica Analítica, Institut Pasteur de Montevideo, Uruguay; Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California at Irvine, Irvine, CA, USA.
| | - Soledad Astrada
- Unidad de Biología Celular, Institut Pasteur de Montevideo, Uruguay
| | - Arturo Briva
- Área de Investigación Respiratoria, Departamento de Fisiopatología, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Uruguay
| | | | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Biomedical Engineering Department, University of California at Irvine, Irvine, CA, USA
| | - Luis A Bagatolli
- MEMPHYS - Center for Biomembrane Physics, University of Southern Denmark, Odense M, Denmark.
| |
Collapse
|
19
|
Schwingshackl A, Roan E, Teng B, Waters CM. TREK-1 Regulates Cytokine Secretion from Cultured Human Alveolar Epithelial Cells Independently of Cytoskeletal Rearrangements. PLoS One 2015; 10:e0126781. [PMID: 26001192 PMCID: PMC4441361 DOI: 10.1371/journal.pone.0126781] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/28/2015] [Indexed: 01/15/2023] Open
Abstract
Background TREK-1 deficient alveolar epithelial cells (AECs) secrete less IL-6, more MCP-1, and contain less F-actin. Whether these alterations in cytokine secretion and F-actin content are related remains unknown. We now hypothesized that cytokine secretion from TREK-1-deficient AECs was regulated by cytoskeletal rearrangements. Methods We determined F-actin and α-tubulin contents of control, TREK-1-deficient and TREK-1-overexpressing human A549 cells by confocal microscopy and western blotting, and measured IL-6 and MCP-1 levels using real-time PCR and ELISA. Results Cytochalasin D decreased the F-actin content of control cells. Jasplakinolide increased the F-actin content of TREK-1 deficient cells, similar to the effect of TREK-1 overexpression in control cells. Treatment of control and TREK-1 deficient cells with TNF-α, a strong stimulus for IL-6 and MCP-1 secretion, had no effect on F-actin structures. The combination of TNF-α+cytochalasin D or TNF-α+jasplakinolide had no additional effect on the F-actin content or architecture when compared to cytochalasin D or jasplakinolide alone. Although TREK-1 deficient AECs contained less F-actin at baseline, quantified biochemically, they contained more α-tubulin. Exposure to nocodazole disrupted α-tubulin filaments in control and TREK-1 deficient cells, but left the overall amount of α-tubulin unchanged. Although TNF-α had no effect on the F-actin or α-tubulin contents, it increased IL-6 and MCP-1 production and secretion from control and TREK-1 deficient cells. IL-6 and MCP-1 secretions from control and TREK-1 deficient cells after TNF-α+jasplakinolide or TNF-α+nocodazole treatment was similar to the effect of TNF-α alone. Interestingly, cytochalasin D decreased TNF-α-induced IL-6 but not MCP-1 secretion from control but not TREK-1 deficient cells. Conclusion Although cytochalasin D, jasplakinolide and nocodazole altered the F-actin and α-tubulin structures of control and TREK-1 deficient AEC, the changes in cytokine secretion from TREK-1 deficient cells cannot be explained by cytoskeletal rearrangements in these cells.
Collapse
Affiliation(s)
- Andreas Schwingshackl
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States of America
- * E-mail:
| | - Esra Roan
- Department of Biomedical Engineering, University of Memphis, Memphis, TN, United States of America
| | - Bin Teng
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Christopher M. Waters
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, United States of America
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States of America
| |
Collapse
|
20
|
Otulakowski G, Engelberts D, Gusarova GA, Bhattacharya J, Post M, Kavanagh BP. Hypercapnia attenuates ventilator-induced lung injury via a disintegrin and metalloprotease-17. J Physiol 2014; 592:4507-21. [PMID: 25085885 DOI: 10.1113/jphysiol.2014.277616] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Hypercapnic acidosis, common in mechanically ventilated patients, has been reported to exert both beneficial and harmful effects in models of lung injury. Understanding its effects at the molecular level may provide insight into mechanisms of injury and protection. The aim of this study was to establish the effects of hypercapnic acidosis on mitogen‐activated protein kinase (MAPK) activation, and determine the relevant signalling pathways. p44/42 MAPK activation in a murine model of ventilator‐induced lung injury (VILI) correlated with injury and was reduced in hypercapnia. When cultured rat alveolar epithelial cells were subjected to cyclic stretch, activation of p44/42 MAPK was dependent on epidermal growth factor receptor (EGFR) activity and on shedding of EGFR ligands; exposure to 12% CO2 without additional buffering blocked ligand shedding, as well as EGFR and p44/42 MAPK activation. The EGFR ligands are known substrates of the matrix metalloprotease ADAM17, suggesting stretch activates and hypercapnic acidosis blocks stretch‐mediated activation of ADAM17. This was corroborated in the isolated perfused mouse lung, where elevated CO2 also inhibited stretch‐activated shedding of the ADAM17 substrate TNFR1 from airway epithelial cells. Finally, in vivo confirmation was obtained in a two‐hit murine model of VILI where pharmacological inhibition of ADAM17 reduced both injury and p44/42 MAPK activation. Thus, ADAM17 is an important proximal mediator of VILI; its inhibition is one mechanism of hypercapnic protection and may be a target for clinical therapy.
Collapse
Affiliation(s)
- Gail Otulakowski
- Physiology and Experimental Medicine Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Canada
| | - Doreen Engelberts
- Physiology and Experimental Medicine Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Canada
| | - Galina A Gusarova
- Departments of Medicine and Physiology, Columbia University, New York, NY, USA
| | - Jahar Bhattacharya
- Departments of Medicine and Physiology, Columbia University, New York, NY, USA
| | - Martin Post
- Physiology and Experimental Medicine Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Canada
| | - Brian P Kavanagh
- Physiology and Experimental Medicine Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Canada Departments of Critical Care Medicine and Anaesthesia, Hospital for Sick Children, University of Toronto, Toronto, Canada
| |
Collapse
|