1
|
MacKenzie B, Mahavadi P, Jannini-Sa YAP, Creyns B, Coelho AL, Espindola M, Ruppert C, Hötzenecker K, Hogaboam C, Guenther A. Pre-clinical proof-of-concept of anti-fibrotic activity of caveolin-1 scaffolding domain peptide LTI-03 in ex vivo precision cut lung slices from patients with Idiopathic Pulmonary Fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.589970. [PMID: 38712072 PMCID: PMC11071419 DOI: 10.1101/2024.04.24.589970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Rationale: While rodent lung fibrosis models are routinely used to evaluate novel antifibrotics, these models have largely failed to predict clinical efficacy of novel drug candidates for Idiopathic Pulmonary Fibrosis (IPF). Moreover, single target therapeutic strategies for IPF have failed and current multi-target standard of care drugs are not curative. Caveolin-1 (CAV-1) is an integral membrane protein, which, via its caveolin scaffolding domain (CSD), interacts with caveolin binding domains (CBD). CAV-1 regulates homeostasis, and its expression is decreased in IPF lungs. LTI-03 is a seven amino acid peptide derived from the CSD and formulated for dry powder inhalation; it was well tolerated in normal volunteers ( NCT04233814 ) and a safety trial is underway in IPF patients ( NCT05954988 ). Objectives: Anti-fibrotic efficacy of LTI-03 and other CSD peptides has been observed in IPF lung monocultures, and rodent pulmonary, dermal, and heart fibrosis models. This study aimed to characterize progressive fibrotic activity in IPF PCLS explants and to evaluate the antifibrotic effects of LTI-03 and nintedanib in this model. Methods: First, CBD regions were identified in IPF signaling proteins using in silico analysis. Then, IPF PCLS (n=8) were characterized by COL1A1 immunostaining, multiplex immunoassays, and bulk RNA sequencing following treatment every 12hrs with LTI-03 at 0.5, 3.0, or 10 μM; nintedanib at 0.1 μM or 1 μM; or control peptide (CP) at 10 μM. Measurements and Main Results: CBDs were present in proteins implicated in IPF, including VEGFR, FGFR and PDGFR. Increased expression of profibrotic mediators indicated active fibrotic activity in IPF PCLS over five days. LTI-03 dose dependently decreased COL1A1 staining, and like nintedanib, decreased profibrotic proteins and transcripts. Unlike nintedanib, LTI-03 did not induce cellular necrosis signals. Conclusion: IPF PCLS explants demonstrate molecular activity indicative of fibrosis during 5 days in culture and LTI-03 broadly attenuated pro-fibrotic proteins and pathways, further supporting the potential therapeutic effectiveness of LTI-03 for IPF.
Collapse
|
2
|
Kryvenko V, Vadász I. Alveolar-capillary endocytosis and trafficking in acute lung injury and acute respiratory distress syndrome. Front Immunol 2024; 15:1360370. [PMID: 38533500 PMCID: PMC10963603 DOI: 10.3389/fimmu.2024.1360370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/29/2024] [Indexed: 03/28/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is associated with high morbidity and mortality but lacks specific therapeutic options. Diverse endocytic processes play a key role in all phases of acute lung injury (ALI), including the initial insult, development of respiratory failure due to alveolar flooding, as a consequence of altered alveolar-capillary barrier function, as well as in the resolution or deleterious remodeling after injury. In particular, clathrin-, caveolae-, endophilin- and glycosylphosphatidyl inositol-anchored protein-mediated endocytosis, as well as, macropinocytosis and phagocytosis have been implicated in the setting of acute lung damage. This manuscript reviews our current understanding of these endocytic pathways and subsequent intracellular trafficking in various phases of ALI, and also aims to identify potential therapeutic targets for patients with ARDS.
Collapse
Affiliation(s)
- Vitalii Kryvenko
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- The Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| | - István Vadász
- Department of Internal Medicine, Justus Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
- The Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Institute for Lung Health (ILH), Giessen, Germany
| |
Collapse
|
3
|
Borek I, Birnhuber A, Voelkel NF, Marsh LM, Kwapiszewska G. The vascular perspective on acute and chronic lung disease. J Clin Invest 2023; 133:e170502. [PMID: 37581311 PMCID: PMC10425217 DOI: 10.1172/jci170502] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
The pulmonary vasculature has been frequently overlooked in acute and chronic lung diseases, such as acute respiratory distress syndrome (ARDS), pulmonary fibrosis (PF), and chronic obstructive pulmonary disease (COPD). The primary emphasis in the management of these parenchymal disorders has largely revolved around the injury and aberrant repair of epithelial cells. However, there is increasing evidence that the vascular endothelium plays an active role in the development of acute and chronic lung diseases. The endothelial cell network in the capillary bed and the arterial and venous vessels provides a metabolically highly active barrier that controls the migration of immune cells, regulates vascular tone and permeability, and participates in the remodeling processes. Phenotypically and functionally altered endothelial cells, and remodeled vessels, can be found in acute and chronic lung diseases, although to different degrees, likely because of disease-specific mechanisms. Since vascular remodeling is associated with pulmonary hypertension, which worsens patient outcomes and survival, it is crucial to understand the underlying vascular alterations. In this Review, we describe the current knowledge regarding the role of the pulmonary vasculature in the development and progression of ARDS, PF, and COPD; we also outline future research directions with the hope of facilitating the development of mechanism-based therapies.
Collapse
Affiliation(s)
- Izabela Borek
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Norbert F. Voelkel
- Pulmonary Medicine Department, University of Amsterdam Medical Centers, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Leigh M. Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, German Lung Center (DZL), Cardiopulmonary Institute, Giessen, Germany
| |
Collapse
|
4
|
Garcia-Vilanova A, Olmo-Fontánez AM, Moliva JI, Allué-Guardia A, Singh H, Merritt RE, Maselli DJ, Peters JI, Restrepo BI, Wang Y, Schlesinger LS, Turner J, Weintraub ST, Torrelles JB. The Aging Human Lung Mucosa: A Proteomics Study. J Gerontol A Biol Sci Med Sci 2022; 77:1969-1974. [PMID: 35460553 PMCID: PMC9536443 DOI: 10.1093/gerona/glac091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Indexed: 11/13/2022] Open
Abstract
The older adult population, estimated to double by 2050, is at increased risk of respiratory infections and other pulmonary diseases. Biochemical changes in the lung alveolar lining fluid (ALF) and in alveolar compartment cells can alter local immune responses as we age, generating opportunities for invading pathogens to establish successful infections. Indeed, the lung alveolar space of older adults is a pro-inflammatory, pro-oxidative, dysregulated environment that remains understudied. We performed an exploratory, quantitative proteomic profiling of the soluble proteins present in ALF, developing insight into molecular fingerprints, pathways, and regulatory networks that characterize the alveolar space in old age, comparing it to that of younger individuals. We identified 457 proteins that were significantly differentially expressed in older adult ALF, including increased production of matrix metalloproteinases, markers of cellular senescence, antimicrobials, and proteins of neutrophilic granule origin, among others, suggesting that neutrophils in the lungs of older adults could be potential contributors to the dysregulated alveolar environment with increasing age. Finally, we describe a hypothetical regulatory network mediated by the serum response factor that could explain the neutrophilic profile observed in the older adult population.
Collapse
Affiliation(s)
- Andreu Garcia-Vilanova
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Angélica M Olmo-Fontánez
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Integrated Biomedical Sciences Program, The University of Texas Health Science Center, San Antonio, Texas, USA
| | - Juan I Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anna Allué-Guardia
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Harjinder Singh
- Division of Pulmonary and Critical Care Medicine, School of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| | - Robert E Merritt
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Diego J Maselli
- Division of Pulmonary and Critical Care Medicine, School of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| | - Jay I Peters
- Division of Pulmonary and Critical Care Medicine, School of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| | | | - Yufeng Wang
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, UTSA, San Antonio, Texas, USA
| | - Larry S Schlesinger
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Joanne Turner
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, UT Health San Antonio, San Antonio, Texas, USA
| | - Jordi B Torrelles
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
5
|
Contribution of Adenosine in the Physiological Changes and Injuries Secondary to Exposure to Extreme Oxygen Pressure in Healthy Subjects. Biomedicines 2022; 10:biomedicines10092059. [PMID: 36140160 PMCID: PMC9495509 DOI: 10.3390/biomedicines10092059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/04/2022] [Accepted: 08/16/2022] [Indexed: 12/05/2022] Open
Abstract
Climbers and aviators are exposed to severe hypoxia at high altitudes, whereas divers are exposed to hyperoxia at depth. The aim of this study was to report changes in the adenosinergic system induced by exposure to extreme oxygen partial pressures. At high altitudes, the increased adenosine concentration contributes to brain protection against hypoxia through various mechanisms such as stimulation of glycogenolysis for ATP production, reduction in neuronal energy requirements, enhancement in 2,3-bisphosphoglycerate production, and increase in cerebral blood flow secondary to vasodilation of cerebral arteries. In the context of mountain illness, the increased level of A2AR expression leads to glial dysfunction through neuroinflammation and is involved in the pathogenesis of neurological disorders. Nonetheless, a high level of adenosine concentration can protect against high-altitude pulmonary edema via a decrease in pulmonary arterial pressure. The adenosinergic system is also involved in the acclimatization phenomenon induced by prolonged exposure to altitude hypoxia. During hyperoxic exposure, decreased extracellular adenosine and low A2A receptor expression contribute to vasoconstriction. The resulting decrease in cerebral blood flow is considered a preventive phenomenon against cerebral oxygen toxicity through the decrease in oxygen delivery to the brain. With regard to lung oxygen toxicity, hyperoxia leads to an increase in extracellular adenosine, which acts to preserve pulmonary barrier function. Changes in the adenosinergic system induced by exposure to extreme oxygen partial pressures frequently have a benefit in decreasing the risk of adverse effects.
Collapse
|
6
|
Parinandi N, Gerasimovskaya E, Verin A. Editorial: Molecular mechanisms of lung endothelial permeability. Front Physiol 2022; 13:976873. [PMID: 35936898 PMCID: PMC9355505 DOI: 10.3389/fphys.2022.976873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 01/16/2023] Open
Affiliation(s)
- Narasimham Parinandi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Evgenia Gerasimovskaya
- Division of Critical Care Medicine, Department of Pediatrics, University of Colorado Denver, Aurora, CO, United States
| | - Alexander Verin
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States,*Correspondence: Alexander Verin,
| |
Collapse
|
7
|
Chen D, Sun J, Zhu J, Ding X, Lan T, Wang X, Wu W, Ou Z, Zhu L, Ding P, Wang H, Luo L, Xiang R, Wang X, Qiu J, Wang S, Li H, Chai C, Liang L, An F, Zhang L, Han L, Zhu Y, Wang F, Yuan Y, Wu W, Sun C, Lu H, Wu J, Sun X, Zhang S, Sahu SK, Liu P, Xia J, Zhang L, Chen H, Fang D, Zeng Y, Wu Y, Cui Z, He Q, Jiang S, Ma X, Feng W, Xu Y, Li F, Liu Z, Chen L, Chen F, Jin X, Qiu W, Wang T, Li Y, Xing X, Yang H, Xu Y, Hua Y, Liu Y, Liu H, Xu X. Single cell atlas for 11 non-model mammals, reptiles and birds. Nat Commun 2021; 12:7083. [PMID: 34873160 PMCID: PMC8648889 DOI: 10.1038/s41467-021-27162-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 09/18/2021] [Indexed: 01/08/2023] Open
Abstract
The availability of viral entry factors is a prerequisite for the cross-species transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Large-scale single-cell screening of animal cells could reveal the expression patterns of viral entry genes in different hosts. However, such exploration for SARS-CoV-2 remains limited. Here, we perform single-nucleus RNA sequencing for 11 non-model species, including pets (cat, dog, hamster, and lizard), livestock (goat and rabbit), poultry (duck and pigeon), and wildlife (pangolin, tiger, and deer), and investigated the co-expression of ACE2 and TMPRSS2. Furthermore, cross-species analysis of the lung cell atlas of the studied mammals, reptiles, and birds reveals core developmental programs, critical connectomes, and conserved regulatory circuits among these evolutionarily distant species. Overall, our work provides a compendium of gene expression profiles for non-model animals, which could be employed to identify potential SARS-CoV-2 target cells and putative zoonotic reservoirs.
Collapse
Affiliation(s)
| | - Jian Sun
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Jiacheng Zhu
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiangning Ding
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianming Lan
- BGI-Shenzhen, Shenzhen, 518083, China
- Department of Biology, University of Copenhagen, DK-2100, Copenhagen, Denmark
| | - Xiran Wang
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | | | - Zhihua Ou
- BGI-Shenzhen, Shenzhen, 518083, China
| | | | - Peiwen Ding
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haoyu Wang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lihua Luo
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rong Xiang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoling Wang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiaying Qiu
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiyou Wang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haimeng Li
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chaochao Chai
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Langchao Liang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fuyu An
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou, 510520, China
| | - Le Zhang
- College of Wildlife Resources Northeast Forestry University, Harbin, 150040, China
| | - Lei Han
- College of Wildlife Resources Northeast Forestry University, Harbin, 150040, China
| | - Yixin Zhu
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | | | - Wendi Wu
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Chengcheng Sun
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haorong Lu
- China National Genebank, BGI-Shenzhen, Shenzhen, 518120, China
- Shenzhen Key Laboratory of Environmental Microbial Genomics and Application, BGI-Shenzhen, Shenzhen, 518120, China
| | - Jihong Wu
- Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
- Key Laboratory of Myopia, Ministry of Health, Shanghai, China
| | - Xinghuai Sun
- Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
- Key Laboratory of Myopia, Ministry of Health, Shanghai, China
| | - Shenghai Zhang
- Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, China
- Key Laboratory of Myopia, Ministry of Health, Shanghai, China
| | | | - Ping Liu
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Jun Xia
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Lijing Zhang
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haixia Chen
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Yuying Zeng
- BGI-Shenzhen, Shenzhen, 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiquan Wu
- HIV and AIDS Malignancy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1868, USA
| | - Zehua Cui
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | - Qian He
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China
| | | | - Xiaoyan Ma
- Department of Biochemistry, University of Cambridge, Cambridge, CB21QW, UK
| | | | - Yan Xu
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Fang Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lei Chen
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Fang Chen
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen, 518083, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tianjiao Wang
- Institute of Special Animal and Plant Sciences (ISAPS) of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Yang Li
- Institute of Special Animal and Plant Sciences (ISAPS) of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Xiumei Xing
- Institute of Special Animal and Plant Sciences (ISAPS) of Chinese Academy of Agricultural Sciences, Changchun, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, 518083, China
- Guangdong Provincial Academician Workstation of BGI Synthetic Genomics, BGI-Shenzhen, Shenzhen, 518120, China
| | - Yanchun Xu
- College of Wildlife Resources Northeast Forestry University, Harbin, 150040, China
- College of Wildlife and Protected Areas, Northeast Forestry University, No. 26, Hexing Road, Xiangfang District, Harbin, 150040, China
| | - Yan Hua
- Guangdong Provincial Key Laboratory of Silviculture, Protection and Utilization, Guangdong Academy of Forestry, Guangzhou, 510520, China.
| | - Yahong Liu
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
| | - Huan Liu
- BGI-Shenzhen, Shenzhen, 518083, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Agricultural Genomics, BGI-Shenzhen, Shenzhen, 518083, China.
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, 518083, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, 518083, Shenzhen, China.
| |
Collapse
|
8
|
Liu Y, Garron TM, Chang Q, Su Z, Zhou C, Gong EC, Zheng J, Yin Y, Ksiazek T, Brasel T, Jin Y, Boor P, Comer JE, Gong B. Cell-type apoptosis in lung during SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33398280 DOI: 10.1101/2020.12.23.424254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The SARS-CoV-2 pandemic has inspired renewed interest in understanding the fundamental pathology of acute respiratory distress syndrome (ARDS) following infection because fatal COVID-19 cases are commonly linked to respiratory failure due to ARDS. The pathologic alteration known as diffuse alveolar damage in endothelial and epithelial cells is a critical feature of acute lung injury in ARDS. However, the pathogenesis of ARDS following SRAS-CoV-2 infection remains largely unknown. In the present study, we examined apoptosis in post-mortem lung sections from COVID-19 patients and lung tissues from a non-human primate model of SARS-CoV-2 infection, in a cell-type manner, including type 1 and 2 alveolar cells and vascular endothelial cells (ECs), macrophages, and T cells. Multiple-target immunofluorescence (IF) assays and western blotting suggest both intrinsic and extrinsic apoptotic pathways are activated during SARS-CoV-2 infection. Furthermore, we observed that SARS-CoV-2 fails to induce apoptosis in human bronchial epithelial cells (i.e., BEAS2B cells) and primary human umbilical vein endothelial cells (HUVECs), which are refractory to SARS-CoV-2 infection. However, infection of co-cultured Vero cells and HUVECs or Vero cells and BEAS2B cells with SARS-CoV-2 induced apoptosis in both Vero cells and HUVECs/BEAS2B cells, but did not alter the permissiveness of HUVECs or BEAS2B cells to the virus. Post-exposure treatment of the co-culture of Vero cells and HUVECs with an EPAC1-specific activator ameliorated apoptosis in HUVECs. These findings may help to delineate a novel insight into the pathogenesis of ARDS following SARS-CoV-2 infection.
Collapse
|
9
|
Ni K, Wang C, Carnino JM, Jin Y. The Evolving Role of Caveolin-1: A Critical Regulator of Extracellular Vesicles. Med Sci (Basel) 2020; 8:medsci8040046. [PMID: 33158117 PMCID: PMC7712126 DOI: 10.3390/medsci8040046] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/26/2020] [Accepted: 10/30/2020] [Indexed: 12/24/2022] Open
Abstract
Emerging evidence suggests that extracellular vesicles (EVs) play an essential role in mediating intercellular communication and inter-organ crosstalk both at normal physiological conditions and in the pathogenesis of human diseases. EV cargos are made up of a broad spectrum of molecules including lipids, proteins, and nucleic acids such as DNA, RNA, and microRNAs. The complex EV cargo composition is cell type-specific. A dynamic change in EV cargos occurs along with extracellular stimuli and a change in the pathophysiological status of the host. Currently, the underlying mechanisms by which EVs are formed and EV cargos are selected in the absence and presence of noxious stimuli and pathogens remain incompletely explored. The term EVs refers to a heterogeneous group of vesicles generated via different mechanisms. Some EVs are formed via direct membrane budding, while the others are produced through multivesicular bodies (MVBs) or during apoptosis. Despite the complexity of EV formation and EV cargo selection, recent studies suggest that caveolin-1, a well-known structural protein of caveolae, regulates the formation and cargo selection of some EVs, such as microvesicles (MVs). In this article, we will review the current understanding of this emerging and novel role of cav-1.
Collapse
Affiliation(s)
| | | | | | - Yang Jin
- Correspondence: ; Tel.: +1-617-358-1356; Fax: +1-617-536-8093
| |
Collapse
|
10
|
Yan F, Su L, Chen X, Wang X, Gao H, Zeng Y. Molecular regulation and clinical significance of caveolin-1 methylation in chronic lung diseases. Clin Transl Med 2020; 10:151-160. [PMID: 32508059 PMCID: PMC7240871 DOI: 10.1002/ctm2.2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 02/27/2020] [Accepted: 02/27/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic lung diseases represent a largely global burden whose pathogenesis remains largely unknown. Research increasingly suggests that epigenetic modifications, especially DNA methylation, play a mechanistic role in chronic lung diseases. DNA methylation can affect gene expression and induce various diseases. Of the caveolae in plasma membrane of cell, caveolin-1 (Cav-1) is a crucial structural constituent involved in many important life activities. With the increasingly advanced progress of genome-wide methylation sequencing technologies, the important impact of Cav-1 DNA methylation has been discovered. The present review overviews the biological characters, functions, and structure of Cav-1; epigenetic modifications of Cav-1 in health and disease; expression and regulation of Cav-1 DNA methylation in the respiratory system and its significance; as well as clinical potential as disease-specific biomarker and targets for early diagnosis and therapy.
Collapse
Affiliation(s)
- Furong Yan
- Clinical Center for Molecular Diagnosis and TherapySecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Lili Su
- Clinical Center for Molecular Diagnosis and TherapySecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Xiaoyang Chen
- Department of Pulmonary and Critical Care MedicineRespiratory Medicine Center of Fujian ProvinceSecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Xiangdong Wang
- Clinical Center for Molecular Diagnosis and TherapySecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Hongzhi Gao
- Clinical Center for Molecular Diagnosis and TherapySecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Yiming Zeng
- Department of Pulmonary and Critical Care MedicineRespiratory Medicine Center of Fujian ProvinceSecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| |
Collapse
|
11
|
Lin X, Barravecchia M, Matthew Kottmann R, Sime P, Dean DA. Caveolin-1 gene therapy inhibits inflammasome activation to protect from bleomycin-induced pulmonary fibrosis. Sci Rep 2019; 9:19643. [PMID: 31873099 PMCID: PMC6928213 DOI: 10.1038/s41598-019-55819-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 11/30/2019] [Indexed: 01/04/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a devastating and fatal disease and characterized by increased deposition of extracellular matrix proteins and scar formation in the lung, resulting from alveolar epithelial damage and accumulation of inflammatory cells. Evidence suggests that Caveolin-1 (Cav-1), a major component of caveolae which regulates cell signaling and endocytosis, is a potential target to treat fibrotic diseases, although the mechanisms and responsible cell types are unclear. We show that Cav-1 expression was downregulated both in alveolar epithelial type I cells in bleomycin-injured mouse lungs and in lung sections from IPF patients. Increased expression of IL-1β and caspase-1 has been observed in IPF patients, indicating inflammasome activation associated with IPF. Gene transfer of a plasmid expressing Cav-1 using transthoracic electroporation reduced infiltration of neutrophils and monocytes/macrophages and protected from subsequent bleomycin-induced pulmonary fibrosis. Overexpression of Cav-1 suppressed bleomycin- or silica-induced activation of caspase-1 and maturation of pro-IL-1β to secrete cleaved IL-1β both in mouse lungs and in primary type I cells. These results demonstrate that gene transfer of Cav-1 downregulates inflammasome activity and protects from subsequent bleomycin-mediated pulmonary fibrosis. This indicates a pivotal regulation of Cav-1 in inflammasome activity and suggests a novel therapeutic strategy for patients with IPF.
Collapse
Affiliation(s)
- Xin Lin
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Michael Barravecchia
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - R Matthew Kottmann
- Department of Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - Patricia Sime
- Department of Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA
| | - David A Dean
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, 14642, USA.
| |
Collapse
|
12
|
Zuniga-Hertz JP, Patel HH. The Evolution of Cholesterol-Rich Membrane in Oxygen Adaption: The Respiratory System as a Model. Front Physiol 2019; 10:1340. [PMID: 31736773 PMCID: PMC6828933 DOI: 10.3389/fphys.2019.01340] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022] Open
Abstract
The increase in atmospheric oxygen levels imposed significant environmental pressure on primitive organisms concerning intracellular oxygen concentration management. Evidence suggests the rise of cholesterol, a key molecule for cellular membrane organization, as a cellular strategy to restrain free oxygen diffusion under the new environmental conditions. During evolution and the increase in organismal complexity, cholesterol played a pivotal role in the establishment of novel and more complex functions associated with lipid membranes. Of these, caveolae, cholesterol-rich membrane domains, are signaling hubs that regulate important in situ functions. Evolution resulted in complex respiratory systems and molecular response mechanisms that ensure responses to critical events such as hypoxia facilitated oxygen diffusion and transport in complex organisms. Caveolae have been structurally and functionally associated with respiratory systems and oxygen diffusion control through their relationship with molecular response systems like hypoxia-inducible factors (HIF), and particularly as a membrane-localized oxygen sensor, controlling oxygen diffusion balanced with cellular physiological requirements. This review will focus on membrane adaptations that contribute to regulating oxygen in living systems.
Collapse
Affiliation(s)
- Juan Pablo Zuniga-Hertz
- Department of Anesthesiology, VA San Diego Healthcare System, University of California, San Diego, San Diego, CA, United States
| | - Hemal H Patel
- Department of Anesthesiology, VA San Diego Healthcare System, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
13
|
Ammonium lauryl sulfate-induced apoptotic cell death may be due to mitochondrial dysfunction triggered by caveolin-1. Toxicol In Vitro 2019; 57:132-142. [DOI: 10.1016/j.tiv.2019.02.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 01/23/2019] [Accepted: 02/25/2019] [Indexed: 11/22/2022]
|
14
|
Oliveira SDS, Chen J, Castellon M, Mao M, Raj JU, Comhair S, Erzurum S, Silva CLM, Machado RF, Bonini MG, Minshall RD. Injury-Induced Shedding of Extracellular Vesicles Depletes Endothelial Cells of Cav-1 (Caveolin-1) and Enables TGF-β (Transforming Growth Factor-β)-Dependent Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2019; 39:1191-1202. [PMID: 30943774 PMCID: PMC7297129 DOI: 10.1161/atvbaha.118.312038] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Objective- To determine whether pulmonary arterial hypertension is associated with endothelial cell (EC)-Cav-1 (caveolin-1) depletion, EC-derived extracellular vesicle cross talk with macrophages, and proliferation of Cav-1 depleted ECs via TGF-β (transforming growth factor-β) signaling. Approach and Results- Pulmonary vascular disease was induced in Sprague-Dawley rats by exposure to a single injection of VEGFRII (vascular endothelial growth factor receptor II) antagonist SU5416 (Su) followed by hypoxia (Hx) plus normoxia (4 weeks each-HxSu model) and in WT (wild type; Tie2.Cre-; Cav1 lox/lox) and EC- Cav1-/- (Tie2.Cre+; Cav1 fl/fl) mice (Hx: 4 weeks). We observed reduced lung Cav-1 expression in the HxSu rat model in association with increased Cav-1+ extracellular vesicle shedding into the circulation. Whereas WT mice exposed to hypoxia exhibited increased right ventricular systolic pressure and pulmonary microvascular thickening compared with the group maintained in normoxia, the remodeling was further increased in EC- Cav1-/- mice indicating EC Cav-1 expression protects against hypoxia-induced pulmonary hypertension. Depletion of EC Cav-1 was associated with reduced BMPRII (bone morphogenetic protein receptor II) expression, increased macrophage-dependent TGF-β production, and activation of pSMAD2/3 signaling in the lung. In vitro, in the absence of Cav-1, eNOS (endothelial NO synthase) dysfunction was implicated in the mechanism of EC phenotype switching. Finally, reduced expression of EC Cav-1 in lung histological sections from human pulmonary arterial hypertension donors was associated with increased plasma concentration of Cav-1, extracellular vesicles, and TGF-β, indicating Cav-1 may be a plasma biomarker of vascular injury and key determinant of TGF-β-induced pulmonary vascular remodeling. Conclusions- EC Cav-1 depletion occurs, in part, via Cav-1+ extracellular vesicle shedding into the circulation, which contributes to increased TGF-β signaling, EC proliferation, vascular remodeling, and pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Suellen D S Oliveira
- From the Department of Anesthesiology (S.D.S.O., M.C., R.D.M.), University of Illinois at Chicago
| | - Jiwang Chen
- Department of Medicine (J.C., M.M., R.F.M., M.G.B.), University of Illinois at Chicago
- Research Resources Center Cardiovascular Research Core (J.C., M.C.), University of Illinois at Chicago
| | - Maricela Castellon
- From the Department of Anesthesiology (S.D.S.O., M.C., R.D.M.), University of Illinois at Chicago
- Research Resources Center Cardiovascular Research Core (J.C., M.C.), University of Illinois at Chicago
| | - Mao Mao
- Department of Medicine (J.C., M.M., R.F.M., M.G.B.), University of Illinois at Chicago
| | - J Usha Raj
- Department of Pediatrics (J.U.R.), University of Illinois at Chicago
| | - Suzy Comhair
- Lerner Research Institute (S.C., S.E.), Cleveland Clinic Foundation, OH
| | - Serpil Erzurum
- Lerner Research Institute (S.C., S.E.), Cleveland Clinic Foundation, OH
| | - Claudia L M Silva
- Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil (C.L.M.S.)
| | - Roberto F Machado
- Department of Medicine (J.C., M.M., R.F.M., M.G.B.), University of Illinois at Chicago
| | - Marcelo G Bonini
- Department of Medicine (J.C., M.M., R.F.M., M.G.B.), University of Illinois at Chicago
| | - Richard D Minshall
- From the Department of Anesthesiology (S.D.S.O., M.C., R.D.M.), University of Illinois at Chicago
- Department of Pharmacology (R.D.M.), University of Illinois at Chicago
| |
Collapse
|
15
|
Vogel ER, Manlove LJ, Kuipers I, Thompson MA, Fang YH, Freeman MR, Britt RD, Faksh A, Yang B, Prakash YS, Pabelick CM. Caveolin-1 scaffolding domain peptide prevents hyperoxia-induced airway remodeling in a neonatal mouse model. Am J Physiol Lung Cell Mol Physiol 2019; 317:L99-L108. [PMID: 31042080 DOI: 10.1152/ajplung.00111.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Reactive airway diseases are significant sources of pulmonary morbidity in neonatal and pediatric patients. Supplemental oxygen exposure in premature infants contributes to airway diseases such as asthma and promotes development of airway remodeling, characterized by increased airway smooth muscle (ASM) mass and extracellular matrix (ECM) deposition. Decreased plasma membrane caveolin-1 (CAV1) expression has been implicated in airway disease and may contribute to airway remodeling and hyperreactivity. Here, we investigated the impact of clinically relevant moderate hyperoxia (50% O2) on airway remodeling and caveolar protein expression in a neonatal mouse model. Within 12 h of birth, litters of B6129SF2J mice were randomized to room air (RA) or 50% hyperoxia exposure for 7 days with or without caveolin-1 scaffolding domain peptide (CSD; caveolin-1 mimic; 10 µl, 0.25 mM daily via intraperitoneal injection) followed by 14 days of recovery in normoxia. Moderate hyperoxia significantly increased airway reactivity and decreased pulmonary compliance at 3 wk. Histologic assessment demonstrated airway wall thickening and increased ASM mass following hyperoxia. RNA from isolated ASM demonstrated significant decreases in CAV1 and cavin-1 in hyperoxia-exposed animals while cavin-3 was increased. Supplementation with intraperitoneal CSD mitigated both the physiologic and histologic changes observed with hyperoxia. Overall, these data show that moderate hyperoxia is detrimental to developing airway and may predispose to airway reactivity and remodeling. Loss of CAV1 is one mechanism through which hyperoxia produces these deleterious effects. Supplementation of CAV1 using CSD or similar analogs may represent a new therapeutic avenue for blunting hyperoxia-induced pulmonary damage in neonates.
Collapse
Affiliation(s)
- Elizabeth R Vogel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Logan J Manlove
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota
| | - Ine Kuipers
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota
| | - Michael A Thompson
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota
| | - Yun-Hua Fang
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Michelle R Freeman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota
| | - Rodney D Britt
- Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Arij Faksh
- Department of Obstetrics and Gynecology, Mayo Clinic , Rochester, Minnesota
| | - Binxia Yang
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| | - Christina M Pabelick
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic , Rochester, Minnesota.,Department of Physiology and Biomedical Engineering, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
16
|
Chignalia AZ, Isbatan A, Patel M, Ripper R, Sharlin J, Shosfy J, Borlaug BA, Dull RO. Pressure-dependent NOS activation contributes to endothelial hyperpermeability in a model of acute heart failure. Biosci Rep 2018; 38:BSR20181239. [PMID: 30355657 PMCID: PMC6250809 DOI: 10.1042/bsr20181239] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 10/11/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022] Open
Abstract
Aims: Acute increases in left ventricular end diastolic pressure (LVEDP) can induce pulmonary edema (PE). The mechanism(s) for this rapid onset edema may involve more than just increased fluid filtration. Lung endothelial cell permeability is regulated by pressure-dependent activation of nitric oxide synthase (NOS). Herein, we demonstrate that pressure-dependent NOS activation contributes to vascular failure and PE in a model of acute heart failure (AHF) caused by hypertension.Methods and results: Male Sprague-Dawley rats were anesthetized and mechanically ventilated. Acute hypertension was induced by norepinephrine (NE) infusion and resulted in an increase in LVEDP and pulmonary artery pressure (Ppa) that were associated with a rapid fall in PaO2, and increases in lung wet/dry ratio and injury scores. Heart failure (HF) lungs showed increased nitrotyrosine content and ROS levels. L-NAME pretreatment mitigated the development of PE and reduced lung ROS concentrations to sham levels. Apocynin (Apo) pretreatment inhibited PE. Addition of tetrahydrobiopterin (BH4) to AHF rats lung lysates and pretreatment of AHF rats with folic acid (FA) prevented ROS production indicating endothelial NOS (eNOS) uncoupling.Conclusion: Pressure-dependent NOS activation leads to acute endothelial hyperpermeability and rapid PE by an increase in NO and ROS in a model of AHF. Acute increases in pulmonary vascular pressure, without NOS activation, was insufficient to cause significant PE. These results suggest a clinically relevant role of endothelial mechanotransduction in the pathogenesis of AHF and further highlights the concept of active barrier failure in AHF. Therapies targetting the prevention or reversal of endothelial hyperpermeability may be a novel therapeutic strategy in AHF.
Collapse
Affiliation(s)
- Andreia Z Chignalia
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, U.S.A.
| | - Ayman Isbatan
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
| | - Milan Patel
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
| | - Richard Ripper
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
- Research and Development Service, Jesse Brown Veterans Affairs Medical Center, 820 S Damen Ave., Chicago, IL 60612, U.S.A
| | - Jordan Sharlin
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
| | - Joelle Shosfy
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic and Foundation, 200 First St SW, Rochester, MN 55905, U.S.A
| | - Randal O Dull
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
- Department of Anesthesiology, University of Arizona College of Medicine and Banner-University Medical Center, Tucson, AZ 85724, U.S.A
| |
Collapse
|
17
|
Sago CD, Lokugamage MP, Lando GN, Djeddar N, Shah NN, Syed C, Bryksin AV, Dahlman JE. Modifying a Commonly Expressed Endocytic Receptor Retargets Nanoparticles in Vivo. NANO LETTERS 2018; 18:7590-7600. [PMID: 30216729 PMCID: PMC6426696 DOI: 10.1021/acs.nanolett.8b03149] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanoparticles are often targeted to receptors expressed on specific cells, but few receptors are (i) highly expressed on one cell type and (ii) involved in endocytosis. One unexplored alternative is manipulating an endocytic gene expressed on multiple cell types; an ideal gene would inhibit delivery to cell type A more than cell type B, promoting delivery to cell type B. This would require a commonly expressed endocytic gene to alter nanoparticle delivery in a cell type-dependent manner in vivo; whether this can occur is unknown. Based on its microenvironmental regulation, we hypothesized Caveolin 1 (Cav1) would exert cell type-specific effects on nanoparticle delivery. Fluorescence was not sensitive enough to investigate this question, and as a result, we designed a platform named QUANT to study nanoparticle biodistribution. QUANT is 108× more sensitive than fluorescence and can be multiplexed. By measuring how 226 lipid nanoparticles (LNPs) delivered nucleic acids to multiple cell types in vivo in wild-type and Cav1 knockout mice, we found Cav1 altered delivery in a cell-type specific manner. Cav1 knockout did not alter LNP delivery to lung and kidney macrophages but substantially reduced LNP delivery to Kupffer cells, which are liver-resident macrophages. These data suggest caveolin-mediated endocytosis of nanomedicines by macrophages varies with tissue type. These results suggest manipulating receptors expressed on multiple cell types can tune drug delivery.
Collapse
|
18
|
Zhang XF, Yang Y, Yang XY, Tong Q. RETRACTED: LEF-1 gene silencing inhibits pulmonary vascular remodeling and occurrence of pulmonary arterial hypertension through the β-catenin signaling pathway. Biomed Pharmacother 2018; 108:817-827. [PMID: 30372893 DOI: 10.1016/j.biopha.2018.08.118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/15/2018] [Accepted: 08/23/2018] [Indexed: 12/22/2022] Open
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the authors as the validity of the pulmonary vascular remodeling indicators cannot be guaranteed. The authors tried post publication to reproduce the results of the cell proliferation and cell aging, however they were not able to confirm the data that was presented by the article.
Collapse
Affiliation(s)
- Xian-Feng Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Jilin University, Changchun130021, PR China
| | - Yang Yang
- Department of Cardiology, The First Affiliated Hospital of Jilin University, Changchun130021, PR China
| | - Xin-Yu Yang
- Department of Cardiology, The First Affiliated Hospital of Jilin University, Changchun130021, PR China
| | - Qian Tong
- Department of Cardiology, The First Affiliated Hospital of Jilin University, Changchun130021, PR China.
| |
Collapse
|
19
|
Jin LG, Zeng S, Sun XQ, Wu C, Chen JL, Cui M, Pang QF. Deletion 101 residue at caveolin-1 scaffolding domain peptides impairs the ability of increasing heme oxygenase-1 activity. Int Immunopharmacol 2018; 63:137-144. [PMID: 30092496 DOI: 10.1016/j.intimp.2018.07.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/02/2018] [Accepted: 07/23/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Resident alveolar macrophages (AMs) are activated and release proinflammatory mediators and chemokines during acute lung injury. We have previous reported that caveolin-1 (Cav-1) scaffolding domain (CSD) peptide inhibited the proinflammatory cytokines expression by up-regulating heme oxygenase-1 (HO-1) activity. In this study, we aimed to investigate the effect of residue R101 in CSD peptide on the activity of HO-1 in AMs. METHODS The binding mode between HO-1 and CSD peptides (WT CSD and Δ101 CSD truncation peptides) was analyzed and the free energy was calculated. The inflammatory genes and M1/M2macrophage polarization-associated genes expression were measured by real-time PCR. The activities of HO-1 were determined by the spectrophotometical method. Western blot analyzed the content of Cav-1, HO-1, IκB and MAPK signals (phosphorylated ERK, JNK and p38 MAPK). RESULTS Δ101CSD peptide could bind to HO-1 protein and to disrupt the interaction of HO-1 and Cav-1. However, Δ101CSD peptide had lower activity of HO-1 in LPS-treated AMs compared with WT CSD. The expression of IL-1β and MCP-1 and NO content were decreased by WT CSD peptide in LPS treated AMs. However, only MCP-1 expression and NO content were downregulated byΔ101CSD peptide. Meanwhile, compared with those in LPS + hemin + WT CSD group, the mRNA expression of TNF-α, Cd86, IL-12b and NOS2 significantly increased while expression of IL10, Arg1 and CD163 significantly decreased in LPS + hemin + Δ101CSD group. The effect of WT CSD peptide on the inhibition of MAPK signaling pathway were stronger than Δ101 CSD peptide evidenced by the level of phosphorylated ERK, JNK and p38 MAPK. CONCLUSION Deletion of residue R101 impairs the ability of CSD peptide to increase HO-1 activity and to dampen inflammatory response in LPS-challenged rat AMs.
Collapse
Affiliation(s)
- Liu-Gen Jin
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China; The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Si Zeng
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China
| | - Xue-Qian Sun
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chen Wu
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jun-Liang Chen
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Meng Cui
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qing-Feng Pang
- Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
20
|
Schilling JM, Head BP, Patel HH. Caveolins as Regulators of Stress Adaptation. Mol Pharmacol 2018; 93:277-285. [PMID: 29358220 DOI: 10.1124/mol.117.111237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/19/2018] [Indexed: 12/21/2022] Open
Abstract
Caveolins have been recognized over the past few decades as key regulators of cell physiology. They are ubiquitously expressed and regulate a number of processes that ultimately impact efficiency of cellular processes. Though not critical to life, they are central to stress adaptation in a number of organs. The following review will focus specifically on the role of caveolin in stress adaptation in the heart, brain, and eye, three organs that are susceptible to acute and chronic stress and that show as well declining function with age. In addition, we consider some novel molecular mechanisms that may account for this stress adaptation and also offer potential to drive the future of caveolin research.
Collapse
Affiliation(s)
- Jan M Schilling
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| | - Brian P Head
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| | - Hemal H Patel
- Veterans Administration San Diego Healthcare System and Department of Anesthesiology, UCSD School of Medicine, San Diego, California
| |
Collapse
|
21
|
Parthasarathi K. The Pulmonary Vascular Barrier: Insights into Structure, Function, and Regulatory Mechanisms. MOLECULAR AND FUNCTIONAL INSIGHTS INTO THE PULMONARY VASCULATURE 2018; 228:41-61. [DOI: 10.1007/978-3-319-68483-3_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
22
|
Udayantha HMV, Bathige SDNK, Priyathilaka TT, Lee S, Kim MJ, Lee J. Identification and characterization of molluscan caveolin-1 ortholog from Haliotis discus discus: Possible involvement in embryogenesis and host defense mechanism against pathogenic stress. Gene Expr Patterns 2017; 27:85-92. [PMID: 29128397 DOI: 10.1016/j.gep.2017.11.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/13/2017] [Accepted: 11/06/2017] [Indexed: 02/05/2023]
Abstract
Caveolins are principal membrane proteins of caveolae that play a central role in signal transduction, substrate transport, and membrane trafficking in various cell types. Numerous studies have reported the crucial role of caveolin-1 (CAV1) in response to invading microbes; yet, very little is known about molluscan CAV1. In this study, we identified and characterized CAV1 ortholog from the disk abalone, Haliotis discus discus (HdCAV1). The cDNA sequence of HdCAV1 is 826 bp long and encodes a 127-amino acid polypeptide. Characteristic caveolin superfamily domain (Glu3 - Lys126) and two possible transmembrane domains (Cys48 - Tyr67 and Ile103 - Phe120) were identified in the HdCAV1 protein. Homology analysis revealed that HdCAV1 shared higher identity (>47%) with molluscans, but lower identity with other species. Phylogenetic tree constructed by the neighbor-joining (NJ) method revealed a distinct evolutionary pathway for molluscans. Transcriptional analysis by SYBR Green qPCR showed the highest expression of HdCAV1 mRNA in late veliger stage, as compared to that in other embryonic developmental stages of disk abalone. In adult animals, gill tissue showed highest HdCAV1 transcript levels under normal physiological condition. Stimulations with two bacteria (Vibrio parahaemolyticus and Listeria monocytogenes), viral hemorrhagic septicemia virus, and two pathogen-associated molecular patterns (LPS and poly I:C) significantly modulated the expression of HdCAV1 transcripts. Collectively, these data suggest that CAV1 plays an important role in embryogenesis and host immune defense in disk abalone.
Collapse
Affiliation(s)
- H M V Udayantha
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Department of Fisheries and Aquaculture, Faculty of Fisheries and Marine Sciences and Technology, University of Ruhuna, Matara, Sri Lanka
| | - S D N K Bathige
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Sri Lanka Institute of Nanotechnology (SLINTEC), Nanotechnology and Science Park, Mahenwatta, Pitipana, Homagama, Sri Lanka
| | - Thanthrige Thiunuwan Priyathilaka
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - Sukkyoung Lee
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea
| | - Myoung-Jin Kim
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province 63243, Republic of Korea.
| |
Collapse
|
23
|
Morris CJ, Aljayyoussi G, Mansour O, Griffiths P, Gumbleton M. Endocytic Uptake, Transport and Macromolecular Interactions of Anionic PAMAM Dendrimers within Lung Tissue. Pharm Res 2017; 34:2517-2531. [PMID: 28616685 PMCID: PMC5736778 DOI: 10.1007/s11095-017-2190-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/22/2017] [Indexed: 12/13/2022]
Abstract
Purpose Polyamidoamine (PAMAM) dendrimers are a promising class of nanocarrier with applications in both small and large molecule drug delivery. Here we report a comprehensive evaluation of the uptake and transport pathways that contribute to the lung disposition of dendrimers. Methods Anionic PAMAM dendrimers and control dextran probes were applied to an isolated perfused rat lung (IPRL) model and lung epithelial monolayers. Endocytosis pathways were examined in primary alveolar epithelial cultures by confocal microscopy. Molecular interactions of dendrimers with protein and lipid lung fluid components were studied using small angle neutron scattering (SANS). Results Dendrimers were absorbed across the intact lung via a passive, size-dependent transport pathway at rates slower than dextrans of similar molecular sizes. SANS investigations of concentration-dependent PAMAM transport in the IPRL confirmed no aggregation of PAMAMs with either albumin or dipalmitoylphosphatidylcholine lung lining fluid components. Distinct endocytic compartments were identified within primary alveolar epithelial cells and their functionality in the rapid uptake of fluorescent dendrimers and model macromolecular probes was confirmed by co-localisation studies. Conclusions PAMAM dendrimers display favourable lung biocompatibility but modest lung to blood absorption kinetics. These data support the investigation of dendrimer-based carriers for controlled-release drug delivery to the deep lung. Electronic supplementary material The online version of this article (doi:10.1007/s11095-017-2190-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher J Morris
- School of Pharmacy, University of East Anglia, Norwich Research Park, NR4 7TJ, UK.
| | - Ghaith Aljayyoussi
- Cardiff School of Pharmacy & Pharmaceutical Sciences, Redwood Building, Cardiff, CF10 3NB, UK
| | - Omar Mansour
- Department of Pharmaceutical, Chemical and Environmental Science, University of Greenwich, Medway Campus, Kent, ME4 4TB, UK
| | - Peter Griffiths
- Department of Pharmaceutical, Chemical and Environmental Science, University of Greenwich, Medway Campus, Kent, ME4 4TB, UK
| | - Mark Gumbleton
- Cardiff School of Pharmacy & Pharmaceutical Sciences, Redwood Building, Cardiff, CF10 3NB, UK.
| |
Collapse
|
24
|
Oliveira SDS, Castellon M, Chen J, Bonini MG, Gu X, Elliott MH, Machado RF, Minshall RD. Inflammation-induced caveolin-1 and BMPRII depletion promotes endothelial dysfunction and TGF-β-driven pulmonary vascular remodeling. Am J Physiol Lung Cell Mol Physiol 2017; 312:L760-L771. [PMID: 28188225 DOI: 10.1152/ajplung.00484.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/11/2017] [Accepted: 02/05/2017] [Indexed: 12/14/2022] Open
Abstract
Endothelial cell (EC) activation and vascular injury are hallmark features of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Caveolin-1 (Cav-1) is highly expressed in pulmonary microvascular ECs and plays a key role in maintaining vascular homeostasis. The aim of this study was to determine if the lung inflammatory response to Escherichia coli lipopolysaccharide (LPS) promotes priming of ECs via Cav-1 depletion and if this contributes to the onset of pulmonary vascular remodeling. To test the hypothesis that depletion of Cav-1 primes ECs to respond to profibrotic signals, C57BL6 wild-type (WT) mice (Tie2.Cre-;Cav1fl/fl ) were exposed to nebulized LPS (10 mg; 1 h daily for 4 days) and compared with EC-specific Cav1-/- (Tie2.Cre+;Cav1fl/fl ). After 96 h of LPS exposure, total lung Cav-1 and bone morphogenetic protein receptor type II (BMPRII) expression were reduced in WT mice. Moreover, plasma albumin leakage, infiltration of immune cells, and levels of IL-6/IL-6R and transforming growth factor-β (TGF-β) were elevated in both LPS-treated WT and EC-Cav1-/- mice. Finally, EC-Cav1-/- mice exhibited a modest increase in microvascular thickness basally and even more so on exposure to LPS (96 h). EC-Cav1-/- mice and LPS-treated WT mice exhibited reduced BMPRII expression and endothelial nitric oxide synthase uncoupling, which along with increased TGF-β promoted TGFβRI-dependent SMAD-2/3 phosphorylation. Finally, human lung sections from patients with ARDS displayed reduced EC Cav-1 expression, elevated TGF-β levels, and severe pulmonary vascular remodeling. Thus EC Cav-1 depletion, oxidative stress-mediated reduction in BMPRII expression, and enhanced TGF-β-driven SMAD-2/3 signaling promote pulmonary vascular remodeling in inflamed lungs.
Collapse
Affiliation(s)
- Suellen D S Oliveira
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois
| | - Maricela Castellon
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois.,Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Jiwang Chen
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Marcelo G Bonini
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Xiaowu Gu
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Michael H Elliott
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Roberto F Machado
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Richard D Minshall
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois; .,Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
25
|
Vogel ER, Britt RD, Faksh A, Kuipers I, Pandya H, Prakash YS, Martin RJ, Pabelick CM. Moderate hyperoxia induces extracellular matrix remodeling by human fetal airway smooth muscle cells. Pediatr Res 2017; 81:376-383. [PMID: 27925619 PMCID: PMC5309184 DOI: 10.1038/pr.2016.218] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/19/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Premature infants are at increased risk for airway diseases, such as wheezing and asthma, because of early exposure to risk factors including hyperoxia. As in adult asthma, airway remodeling and increased extracellular matrix (ECM) deposition is involved. METHODS We assessed the impact of 24-72 h of moderate hyperoxia (50%) on human fetal airway smooth muscle (fASM) ECM deposition through western blot, modified in-cell western, and zymography techniques. RESULTS Hyperoxia exposure significantly increased collagen I and collagen III deposition, increased pro- and cleaved matrix metalloproteinase 9 (MMP9) activity, and decreased endogenous MMP inhibitor, TIMP1, expression. Hyperoxia-induced change in caveolin-1 (CAV1) expression was assessed as a potential mechanism for the changes in ECM deposition. CAV1 expression was decreased following hyperoxia. Supplementation of CAV1 activity with caveolar scaffolding domain (CSD) peptide abrogated the hyperoxia-mediated ECM changes. CONCLUSION These results demonstrate that moderate hyperoxia enhances ECM deposition in developing airways by altering the balance between MMPs and their inhibitors (TIMPs), and by increasing collagen deposition. These effects are partly mediated by a hyperoxia-induced decrease in CAV1 expression. In conjunction with prior data demonstrating increased fASM proliferation with hyperoxia, these data further demonstrate that hyperoxia is an important instigator of remodeling in developing airways.
Collapse
Affiliation(s)
- Elizabeth R. Vogel
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Rodney D. Britt
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Arij Faksh
- Department of Obstetrics and Gynecology (Division of Maternal Fetal Medicine), Mayo Clinic, Rochester, MN, USA
| | - Ine Kuipers
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | - Hitesh Pandya
- Department of Pediatrics, University of Leicester, Leicester, England, UK
| | - YS Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Richard J. Martin
- Department of Pediatrics (Division of Neonatology), Rainbow-Babies Children’s Hospital, Case Western Reserve University, Cleveland, OH, USA
| | - Christina M. Pabelick
- Department of Anesthesiology, Mayo Clinic, Rochester, MN, USA,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA,Corresponding Author: Christina M. Pabelick, MD, Professor of Anesthesiology and Physiology, 4-184 W Jos SMH, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, 507-255-7481, 507-255-7300 (fax),
| |
Collapse
|
26
|
Bone marrow-derived macrophages exclusively expressed caveolin-2: The role of inflammatory activators and hypoxia. Immunobiology 2015. [DOI: 10.1016/j.imbio.2015.06.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
27
|
Han B, Tiwari A, Kenworthy AK. Tagging strategies strongly affect the fate of overexpressed caveolin-1. Traffic 2015; 16:417-38. [PMID: 25639341 PMCID: PMC4440517 DOI: 10.1111/tra.12254] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 12/24/2014] [Accepted: 12/24/2014] [Indexed: 01/01/2023]
Abstract
Caveolin-1 (Cav1) is the primary scaffolding protein of caveolae, flask-shaped invaginations of the plasma membrane thought to function in endocytosis, mechanotransduction, signaling and lipid homeostasis. A significant amount of our current knowledge about caveolins and caveolae is derived from studies of transiently overexpressed, C-terminally tagged caveolin proteins. However, how different tags affect the behavior of ectopically expressed Cav1 is still largely unknown. To address this question, we performed a comparative analysis of the subcellular distribution, oligomerization state and detergent resistance of transiently overexpressed Cav1 labeled with three different C-terminal tags (EGFP, mCherry and myc). We show that addition of fluorescent protein tags enhances the aggregation and/or degradation of both wild-type Cav1 and an oligomerization defective P132L mutant. Strikingly, complexes formed by overexpressed Cav1 fusion proteins excluded endogenous Cav1 and Cav2, and the properties of native caveolins were largely preserved even when abnormal aggregates were present in cells. These findings suggest that differences in tagging strategies may be a source of variation in previously published studies of Cav1 and that overexpressed Cav1 may exert functional effects outside of caveolae. They also highlight the need for a critical re-evaluation of current knowledge based on transient overexpression of tagged Cav1.
Collapse
Affiliation(s)
- Bing Han
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of MedicineNashville, TN, USA
| | - Ajit Tiwari
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of MedicineNashville, TN, USA
| | - Anne K Kenworthy
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of MedicineNashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of MedicineNashville, TN, USA
- Epithelial Biology Program, Vanderbilt University School of MedicineNashville, TN, USA
- Chemical and Physical Biology Program, Vanderbilt UniversityNashville, TN, USA
| |
Collapse
|
28
|
|
29
|
Hofmann A, Gosemann JH, Takahashi T, Friedmacher F, Duess JW, Puri P. Imbalance of caveolin-1 and eNOS expression in the pulmonary vasculature of experimental diaphragmatic hernia. ACTA ACUST UNITED AC 2014; 101:341-6. [PMID: 25078423 DOI: 10.1002/bdrb.21117] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/12/2014] [Indexed: 01/17/2023]
Abstract
BACKGROUND Caveolin-1 (Cav-1) exerts major regulatory functions on intracellular signaling pathways originating at the plasma membrane. Cav-1 is a key regulator in adverse lung remodeling and the development of pulmonary hypertension (PH) regulating vasomotor tone through its ability to reduce nitric oxide (NO) production. This low-output endothelial NO synthase (eNOS) derived NO maintains normal pulmonary vascular homeostasis. Cav-1 deficiency leads to increased bioavailability of NO, which has been linked to increased nitrosative stress. Inhibition of eNOS reduced oxidant production and reversed PH, supporting the concept that Cav-1 regulation of eNOS activity is crucial to endothelial homeostasis in lungs. We designed this study to investigate the hypothesis that expression of Cav-1 is downregulated while eNOS expression is upregulated by the pulmonary endothelium in the nitrofen-induced congenital diaphragmatic hernia (CDH). METHODS Pregnant rats were exposed to nitrofen or vehicle on day 9.5 (D9.5). Fetuses were sacrificed on D21 and divided into nitrofen and control groups. Quantitative real-time polymerase chain reaction, Western blotting, and confocal immunofluorescence were performed to determine pulmonary gene expression levels and protein expression of Cav-1 and eNOS. RESULTS Pulmonary Cav-1 gene expression levels were significantly decreased, while eNOS gene expression was significantly increased in nitrofen-induced CDH(+). Western blotting and confocal microscopy revealed decreased pulmonary Cav-1 protein expression, while eNOS protein expression was increased in CDH(+) compared to controls. CONCLUSION The striking evidence of markedly decreased gene and protein expression of Cav-1 with concurrently increased eNOS gene and protein expression in the pulmonary vasculature suggests that activation of eNOS secondary to Cav-1 deficiency may play an important role in the pathogenesis of PH in the nitrofen-induced CDH.
Collapse
Affiliation(s)
- Alejandro Hofmann
- National Children's Research Centre, Our Lady's Children's Hospital, Gate 5, Dublin, Ireland
| | | | | | | | | | | |
Collapse
|
30
|
Piegeler T, Dull RO, Hu G, Castellon M, Chignalia AZ, Koshy RG, Votta-Velis EG, Borgeat A, Schwartz DE, Beck-Schimmer B, Minshall RD. Ropivacaine attenuates endotoxin plus hyperinflation-mediated acute lung injury via inhibition of early-onset Src-dependent signaling. BMC Anesthesiol 2014; 14:57. [PMID: 25097454 PMCID: PMC4112848 DOI: 10.1186/1471-2253-14-57] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 07/08/2014] [Indexed: 11/18/2022] Open
Abstract
Background Acute lung injury (ALI) is associated with high mortality due to the lack of effective therapeutic strategies. Mechanical ventilation itself can cause ventilator-induced lung injury. Pulmonary vascular barrier function, regulated in part by Src kinase-dependent phosphorylation of caveolin-1 and intercellular adhesion molecule-1 (ICAM-1), plays a crucial role in the development of protein-/neutrophil-rich pulmonary edema, the hallmark of ALI. Amide-linked local anesthetics, such as ropivacaine, have anti-inflammatory properties in experimental ALI. We hypothesized ropivacaine may attenuate inflammation in a “double-hit” model of ALI triggered by bacterial endotoxin plus hyperinflation via inhibition of Src-dependent signaling. Methods C57BL/6 (WT) and ICAM-1−/− mice were exposed to either nebulized normal saline (NS) or lipopolysaccharide (LPS, 10 mg) for 1 hour. An intravenous bolus of 0.33 mg/kg ropivacaine or vehicle was followed by mechanical ventilation with normal (7 ml/kg, NTV) or high tidal volume (28 ml/kg, HTV) for 2 hours. Measures of ALI (excess lung water (ELW), extravascular plasma equivalents, permeability index, myeloperoxidase activity) were assessed and lungs were homogenized for Western blot analysis of phosphorylated and total Src, ICAM-1 and caveolin-1. Additional experiments evaluated effects of ropivacaine on LPS-induced phosphorylation/expression of Src, ICAM-1 and caveolin-1 in human lung microvascular endothelial cells (HLMVEC). Results WT mice treated with LPS alone showed a 49% increase in ELW compared to control animals (p = 0.001), which was attenuated by ropivacaine (p = 0.001). HTV ventilation alone increased measures of ALI even more than LPS, an effect which was not altered by ropivacaine. LPS plus hyperinflation (“double-hit”) increased all ALI parameters (ELW, EVPE, permeability index, MPO activity) by 3–4 fold compared to control, which were again decreased by ropivacaine. Western blot analyses of lung homogenates as well as HLMVEC treated in culture with LPS alone showed a reduction in Src activation/expression, as well as ICAM-1 expression and caveolin-1 phosphorylation. In ICAM-1−/− mice, neither addition of LPS to HTV ventilation alone nor ropivacaine had an effect on the development of ALI. Conclusions Ropivacaine may be a promising therapeutic agent for treating the cause of pulmonary edema by blocking inflammatory Src signaling, ICAM-1 expression, leukocyte infiltration, and vascular hyperpermeability.
Collapse
Affiliation(s)
- Tobias Piegeler
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA ; Institute of Anesthesiology, University Hospital Zurich, Zurich, Switzerland
| | - Randal O Dull
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA ; Department of Pharmacology, University of Illinois Hospital > Health Sciences System, Chicago, IL, USA ; Department of Bioengineering, University of Illinois Hospital > Health Sciences System, Chicago, IL, USA
| | - Guochang Hu
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA ; Department of Pharmacology, University of Illinois Hospital > Health Sciences System, Chicago, IL, USA
| | - Maricela Castellon
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA ; Department of Pharmacology, University of Illinois Hospital > Health Sciences System, Chicago, IL, USA
| | - Andreia Z Chignalia
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA
| | - Ruben G Koshy
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA
| | - E Gina Votta-Velis
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA ; Department of Anesthesiology, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Alain Borgeat
- Department of Anesthesiology, Balgrist University Hospital, Zurich, Switzerland
| | - David E Schwartz
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA
| | | | - Richard D Minshall
- Department of Anesthesiology, University of Illinois Hospital > Health Sciences System, 835 S. Wolcott Ave (m/c 868), Chicago, IL 60612, USA ; Department of Pharmacology, University of Illinois Hospital > Health Sciences System, Chicago, IL, USA ; Department of Bioengineering, University of Illinois Hospital > Health Sciences System, Chicago, IL, USA ; Center for Lung and Vascular Biology, University of Illinois Hospital > Health Sciences System, Chicago, IL, USA
| |
Collapse
|
31
|
Royce SG, Le Saux CJ. Role of caveolin-1 in asthma and chronic inflammatory respiratory diseases. Expert Rev Respir Med 2014; 8:339-47. [PMID: 24742020 DOI: 10.1586/17476348.2014.905915] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Caveolin-1 (Cav-1) is the major protein present in invaginations of the plasma membrane of cells known as caveolae. Cav-1 is expressed in numerous resident and inflammatory cells implicated in the pathogenesis of asthma and chronic inflammatory respiratory diseases including chronic obstructive pulmonary disease. A remarkable repertoire of functions has been identified for Cav-1 and these extend to, and have relevance to, asthma and chronic inflammatory respiratory diseases. Important processes influenced by Cav-1 include inflammation, fibrosis, smooth muscle contractility, regulation of apoptosis and cell senescence as well as epithelial barrier function and homeostasis. A better understanding of Cav-1 may be useful in developing new therapies for chronic inflammatory respiratory diseases.
Collapse
Affiliation(s)
- Simon G Royce
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | | |
Collapse
|
32
|
Mundy DI, Lopez AM, Posey KS, Chuang JC, Ramirez CM, Scherer PE, Turley SD. Impact of the loss of caveolin-1 on lung mass and cholesterol metabolism in mice with and without the lysosomal cholesterol transporter, Niemann-Pick type C1. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:995-1002. [PMID: 24747682 DOI: 10.1016/j.bbalip.2014.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 04/08/2014] [Accepted: 04/11/2014] [Indexed: 11/17/2022]
Abstract
Caveolin-1 (Cav-1) is a major structural protein in caveolae in the plasma membranes of many cell types, particularly endothelial cells and adipocytes. Loss of Cav-1 function has been implicated in multiple diseases affecting the cardiopulmonary and central nervous systems, as well as in specific aspects of sterol and lipid metabolism in the liver and intestine. Lungs contain an exceptionally high level of Cav-1. Parameters of cholesterol metabolism in the lung were measured, initially in Cav-1-deficient mice (Cav-1(-/-)), and subsequently in Cav-1(-/-) mice that also lacked the lysosomal cholesterol transporter Niemann-Pick C1 (Npc1) (Cav-1(-/-):Npc1(-/-)). In 50-day-old Cav-1(-/-) mice fed a low- or high-cholesterol chow diet, the total cholesterol concentration (mg/g) in the lungs was marginally lower than in the Cav-1(+/+) controls, but due to an expansion in their lung mass exceeding 30%, whole-lung cholesterol content (mg/organ) was moderately elevated. Lung mass (g) in the Cav-1(-/-):Npc1(-/-) mice (0.356±0.022) markedly exceeded that in their Cav-1(+/+):Npc1(+/+) controls (0.137±0.009), as well as in their Cav-1(-/-):Npc1(+/+) (0.191±0.013) and Cav-1(+/+):Npc1(-/-) (0.213±0.022) littermates. The corresponding lung total cholesterol contents (mg/organ) in mice of these genotypes were 6.74±0.17, 0.71±0.05, 0.96±0.05 and 3.12±0.43, respectively, with the extra cholesterol in the Cav-1(-/-):Npc1(-/-) and Cav-1(+/+):Npc1(-/-) mice being nearly all unesterified (UC). The exacerbation of the Npc1 lung phenotype and increase in the UC level in the Cav-1(-/-):Npc1(-/-) mice imply a regulatory role of Cav-1 in pulmonary cholesterol metabolism when lysosomal sterol transport is disrupted.
Collapse
Affiliation(s)
- Dorothy I Mundy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA.
| | - Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA.
| | - Kenneth S Posey
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA.
| | - Jen-Chieh Chuang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA.
| | - Charina M Ramirez
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA.
| | - Philipp E Scherer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA.
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, USA.
| |
Collapse
|
33
|
Thompson MA, Prakash YS, Pabelick CM. The role of caveolae in the pathophysiology of lung diseases. Expert Rev Respir Med 2013; 8:111-22. [PMID: 24308657 DOI: 10.1586/17476348.2014.855610] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Caveolae are flask-shaped plasma membrane invaginations formed by constitutive caveolin proteins and regulatory cavin proteins. Caveolae harbor a range of signaling components such as receptors, ion channels and regulatory molecules. There is now increasing evidence that caveolins and cavins play an important role in a variety of diseases. However, the mechanisms by which these caveolar proteins affect lung health and disease are still under investigation, with emerging data suggesting complex roles in disease pathophysiology. This review summarizes the current state of understanding of how caveolar proteins contribute to lung structure and function and how their altered expression and/or function can influence lung diseases.
Collapse
|
34
|
Bakhshi FR, Mao M, Shajahan AN, Piegeler T, Chen Z, Chernaya O, Sharma T, Elliott WM, Szulcek R, Bogaard HJ, Comhair S, Erzurum S, van Nieuw Amerongen GP, Bonini MG, Minshall RD. Nitrosation-dependent caveolin 1 phosphorylation, ubiquitination, and degradation and its association with idiopathic pulmonary arterial hypertension. Pulm Circ 2013; 3:816-30. [PMID: 25006397 PMCID: PMC4070841 DOI: 10.1086/674753] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 09/18/2013] [Indexed: 01/15/2023] Open
Abstract
In the present study, we tested the hypothesis that chronic inflammation and oxidative/nitrosative stress induce caveolin 1 (Cav-1) degradation, providing an underlying mechanism of endothelial cell activation/dysfunction and pulmonary vascular remodeling in patients with idiopathic pulmonary arterial hypertension (IPAH). We observed reduced Cav-1 protein despite increased Cav-1 messenger RNA expression and also endothelial nitric oxide synthase (eNOS) hyperphosphorylation in human pulmonary artery endothelial cells (PAECs) from patients with IPAH. In control human lung endothelial cell cultures, tumor necrosis factor α-induced nitric oxide (NO) production and S-nitrosation (SNO) of Cav-1 Cys-156 were associated with Src displacement and activation, Cav-1 Tyr-14 phosphorylation, and destabilization of Cav-1 oligomers within 5 minutes that could be blocked by eNOS or Src inhibition. Prolonged stimulation (72 hours) with NO donor DETANONOate reduced oligomerized and total Cav-1 levels by 40%-80%, similar to that observed in IPAH patient-derived PAECs. NO donor stimulation of endothelial cells for >72 hours, which was associated with sustained Src activation and Cav-1 phosphorylation, ubiquitination, and degradation, was blocked by NOS inhibitor L-NAME, Src inhibitor PP2, and proteosomal inhibitor MG132. Thus, chronic inflammation, sustained eNOS and Src signaling, and Cav-1 degradation may be important causal factors in the development of IPAH by promoting PAEC dysfunction/activation via sustained oxidative/nitrosative stress.
Collapse
Affiliation(s)
- Farnaz R. Bakhshi
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Mao Mao
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ayesha N. Shajahan
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tobias Piegeler
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Zhenlong Chen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Olga Chernaya
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tiffany Sharma
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - W. Mark Elliott
- Pulmonary Division, James Hogg Research Centre Biobank, University of British Columbia, Vancouver, Canada
| | - Robert Szulcek
- Department of Physiology, Institute for Cardiovascular Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
- Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Harm Jan Bogaard
- Department of Pulmonology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Suzy Comhair
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Serpil Erzurum
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Geerten P. van Nieuw Amerongen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Physiology, Institute for Cardiovascular Research, Vrije Universiteit (VU) University Medical Center, Amsterdam, The Netherlands
| | - Marcelo G. Bonini
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Medicine, Section of Cardiology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois, USA
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
35
|
Marí M, Morales A, Colell A, García-Ruiz C, Kaplowitz N, Fernández-Checa JC. Mitochondrial glutathione: features, regulation and role in disease. Biochim Biophys Acta Gen Subj 2012; 1830:3317-28. [PMID: 23123815 DOI: 10.1016/j.bbagen.2012.10.018] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 10/12/2012] [Accepted: 10/23/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Mitochondria are the powerhouse of mammalian cells and the main source of reactive oxygen species (ROS) associated with oxygen consumption. In addition, they also play a strategic role in controlling the fate of cells through regulation of death pathways. Mitochondrial ROS production fulfills a signaling role through regulation of redox pathways, but also contributes to mitochondrial damage in a number of pathological states. SCOPE OF REVIEW Mitochondria are exposed to the constant generation of oxidant species, and yet the organelle remains functional due to the existence of an armamentarium of antioxidant defense systems aimed to repair oxidative damage, of which mitochondrial glutathione (mGSH) is of particular relevance. Thus, the aim of the review is to cover the regulation of mGSH and its role in disease. MAJOR CONCLUSIONS Cumulating evidence over recent years has demonstrated the essential role for mGSH in mitochondrial physiology and disease. Despite its high concentration in the mitochondrial matrix, mitochondria lack the enzymes to synthesize GSH de novo, so that mGSH originates from cytosolic GSH via transport through specific mitochondrial carriers, which exhibit sensitivity to membrane dynamics. Depletion of mGSH sensitizes cells to stimuli leading to oxidative stress such as TNF, hypoxia or amyloid β-peptide, thereby contributing to disease pathogenesis. GENERAL SIGNIFICANCE Understanding the regulation of mGSH may provide novel insights to disease pathogenesis and toxicity and the opportunity to design therapeutic targets of intervention in cell death susceptibility and disease. This article is part of a Special Issue entitled Cellular functions of glutathione.
Collapse
Affiliation(s)
- Montserrat Marí
- Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona, Spain.
| | | | | | | | | | | |
Collapse
|