1
|
Guggemos J, Fuller SJ, Skarratt KK, Mayer B, Schneider EM. Loss-of-function/gain-of-function polymorphisms of the ATP sensitive P2X7R influence sepsis, septic shock, pneumonia, and survival outcomes. Front Immunol 2024; 15:1352789. [PMID: 38966639 PMCID: PMC11222724 DOI: 10.3389/fimmu.2024.1352789] [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: 12/08/2023] [Accepted: 02/07/2024] [Indexed: 07/06/2024] Open
Abstract
Introduction Extracellular ATP (eATP) released from damaged cells activates the P2X7 receptor (P2X7R) ion channel on the surface of surrounding cells, resulting in calcium influx, potassium efflux and inflammasome activation. Inherited changes in the P2X7R gene (P2RX7) influence eATP induced responses. Single nucleotide polymorphisms (SNPs) of P2RX7 influence both function and signaling of the receptor, that in addition to ion flux includes pathogen control and immunity. Methods Subjects (n = 105) were admitted to the ICU at the University Hospital Ulm, Germany between June 2018 and August 2019. Of these, subjects with a diagnosis of sepsis (n = 75), were also diagnosed with septic shock (n = 24), and/or pneumonia (n = 42). Subjects with pneumonia (n = 43) included those without sepsis (n = 1), sepsis without shock (n = 29) and pneumonia with septic shock (n = 13). Out of the 75 sepsis/septic shock patients, 33 patients were not diagnosed with pneumonia. Controls (n = 30) were recruited to the study from trauma patients and surgical patients without sepsis, septic shock, or pneumonia. SNP frequencies were determined for 16 P2RX7 SNPs known to affect P2X7R function, and association studies were performed between frequencies of these SNPs in sepsis, septic shock, and pneumonia compared to controls. Results The loss-of-function (LOF) SNP rs17525809 (T253C) was found more frequently in patients with septic shock, and non-septic trauma patients when compared to sepsis. The LOF SNP rs2230911 (C1096G) was found to be more frequent in patients with sepsis and septic shock than in non-septic trauma patients. The frequencies of these SNPs were even higher in sepsis and septic patients with pneumonia. The current study also confirmed a previous study by our group that showed a five SNP combination that included the GOF SNPs rs208294 (C489T) and rs2230912 (Q460R) that was designated #21211 was associated with increased odds of survival in severe sepsis. Discussion The results found an association between expression of LOF P2RX7 SNPs and presentation to the ICU with sepsis, and septic shock compared to control ICU patients. Furthermore, frequencies of LOF SNPs were found to be higher in sepsis patients with pneumonia compared to those without pneumonia. In addition, a five SNP GOF combination was associated with increased odds of survival in severe sepsis. These results suggest that P2RX7 is required to control infection in pneumonia and that inheritance of LOF variants increases the risk of sepsis when associated with pneumonia. This study confirms that P2RX7 genotyping in pneumonia may identify patients at risk of developing sepsis. The study also identifies P2X7R as a target in sepsis associated with an excessive immune response in subjects with GOF SNP combinations.
Collapse
Affiliation(s)
- Johanna Guggemos
- Clinic for Anesthesiology and Intensive Care Medicine, Ulm University Hospital, Ulm, Germany
| | - Stephen J. Fuller
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW, Australia
- Department of Haematology, Nepean Hospital, Penrith, NSW, Australia
| | - Kristen K. Skarratt
- Nepean Clinical School, Faculty of Medicine and Health, The University of Sydney, Kingswood, NSW, Australia
- Department of Haematology, Nepean Hospital, Penrith, NSW, Australia
| | - Benjamin Mayer
- Institute for Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - E. Marion Schneider
- Clinic for Anesthesiology and Intensive Care Medicine, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
2
|
Sierra-Marquez J, Schaller L, Sassenbach L, Ramírez-Fernández A, Alt P, Rissiek B, Zimmer B, Schredelseker J, Hector J, Stähler T, Koch-Nolte F, Staab-Weijnitz CA, Dietrich A, Kopp R, Nicke A. Different localization of P2X4 and P2X7 receptors in native mouse lung - lack of evidence for a direct P2X4-P2X7 receptor interaction. Front Immunol 2024; 15:1425938. [PMID: 38953020 PMCID: PMC11215518 DOI: 10.3389/fimmu.2024.1425938] [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: 04/30/2024] [Accepted: 05/28/2024] [Indexed: 07/03/2024] Open
Abstract
Introduction P2X receptors are a family of homo- and heterotrimeric cation channels gated by extracellular ATP. The P2X4 and P2X7 subunits show overlapping expression patterns and have been involved in similar physiological processes, such as pain and inflammation as well as various immune cell functions. While formation of P2X2/P2X3 heterotrimers produces a distinct pharmacological phenotype and has been well established, functional identification of a P2X4/P2X7 heteromer has been difficult and evidence for and against a physical association has been found. Most of this evidence stems, however, from in vitro model systems. Methods Here, we used a P2X7-EGFP BAC transgenic mouse model as well as P2X4 and P2X7 knock-out mice to re-investigate a P2X4-P2X7 interaction in mouse lung by biochemical and immunohistochemical experiments as well as quantitative expression analysis. Results No detectable amounts of P2X4 could be co-purified from mouse lung via P2X7-EGFP. In agreement with these findings, immuno-histochemical analysis using a P2X7-specific nanobody revealed only limited overlap in the cellular and subcellular localizations of P2X4 and P2X7 in both the native lung tissue and primary cells. Comparison of P2X4 and P2X7 transcript and protein levels in the respective gene-deficient and wild type mice showed no mutual interrelation between their expression levels in whole lungs. However, a significantly reduced P2rx7 expression was found in alveolar macrophages of P2rx4 -/- mice. Discussion In summary, our detailed analysis of the cellular and subcellular P2X4 and P2X7 localization and expression does not support a physiologically relevant direct association of P2X4 and P2X7 subunits or receptors in vivo.
Collapse
Affiliation(s)
- Juan Sierra-Marquez
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lena Schaller
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Lukas Sassenbach
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Antonio Ramírez-Fernández
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Philipp Alt
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Béla Zimmer
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Johann Schredelseker
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Partner Site Munich Heart Alliance, Munich, Germany
| | - Julia Hector
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Tobias Stähler
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia A. Staab-Weijnitz
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), Member of the German Center for Lung Research (DZL), Germany
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Robin Kopp
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
3
|
Lindo J, Nogueira C, Soares R, Cunha N, Almeida MR, Rodrigues L, Coelho P, Rodrigues F, Cunha RA, Gonçalves T. Genetic Polymorphisms of P2RX7 but Not of ADORA2A Are Associated with the Severity of SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:6135. [PMID: 38892324 PMCID: PMC11173306 DOI: 10.3390/ijms25116135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
SARS-CoV-2 infection ranges from mild to severe presentations, according to the intensity of the aberrant inflammatory response. Purinergic receptors dually control the inflammatory response: while adenosine A2A receptors (A2ARs) are anti-inflammatory, ATP P2X7 receptors (P2X7Rs) exert pro-inflammatory effects. The aim of this study was to assess if there were differences in allelic and genotypic frequencies of a loss-of-function SNP of ADORA2A (rs2298383) and a gain-of-function single nucleotide polymorphism (SNP) of P2RX7 (rs208294) in the severity of SARS-CoV-2-associated infection. Fifty-five individuals were enrolled and categorized according to the severity of the infection. Endpoint genotyping was performed in blood cells to screen for both SNPs. The TT genotype (vs. CT + CC) and the T allele (vs. C allele) of P2RX7 SNP were found to be associated with more severe forms of COVID-19, whereas the association between ADORA2A SNP and the severity of infection was not significantly different. The T allele of P2RX7 SNP was more frequent in people with more than one comorbidity and with cardiovascular conditions and was associated with colorectal cancer. Our findings suggest a more prominent role of P2X7R rather than of A2AR polymorphisms in SARS-CoV-2 infection, although larger population-based studies should be performed to validate our conclusions.
Collapse
Affiliation(s)
- Jorge Lindo
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Célia Nogueira
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Rui Soares
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
- Clinical Pathology Service, Portuguese Oncology Institute of Coimbra, 3004-011 Coimbra, Portugal;
| | - Nuno Cunha
- Clinical Pathology Service, Portuguese Oncology Institute of Coimbra, 3004-011 Coimbra, Portugal;
| | - Maria Rosário Almeida
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Lisa Rodrigues
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Patrícia Coelho
- IPCB/ESALD—Instituto Politécnico de Castelo Branco, Escola Superior de Saúde Dr. Lopes Dias, SPRINT-IPCB—Sport Physical Activity and Health Research & Innovation Center, 6000-767 Castelo Branco, Portugal; (P.C.); (F.R.)
| | - Francisco Rodrigues
- IPCB/ESALD—Instituto Politécnico de Castelo Branco, Escola Superior de Saúde Dr. Lopes Dias, SPRINT-IPCB—Sport Physical Activity and Health Research & Innovation Center, 6000-767 Castelo Branco, Portugal; (P.C.); (F.R.)
| | - Rodrigo A. Cunha
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| | - Teresa Gonçalves
- FMUC—Faculty of Medicine, University Coimbra, 3004-504 Coimbra, Portugal; (J.L.); (C.N.); (R.S.); (M.R.A.)
- CNC-UC—Center for Neuroscience and Cell Biology, University Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
4
|
Wei C, Fu M, Zhang H, Yao B. How is the P2X7 receptor signaling pathway involved in epileptogenesis? Neurochem Int 2024; 173:105675. [PMID: 38211839 DOI: 10.1016/j.neuint.2024.105675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
Epilepsy, a condition characterized by spontaneous recurrent epileptic seizures, is among the most prevalent neurological disorders. This disorder is estimated to affect approximately 70 million people worldwide. Although antiseizure medications are considered the first-line treatments for epilepsy, most of the available antiepileptic drugs are not effective in nearly one-third of patients. This calls for the development of more effective drugs. Evidence from animal models and epilepsy patients suggests that strategies that interfere with the P2X7 receptor by binding to adenosine triphosphate (ATP) are potential treatments for this patient population. This review describes the role of the P2X7 receptor signaling pathways in epileptogenesis. We highlight the genes, purinergic signaling, Pannexin1, glutamatergic signaling, adenosine kinase, calcium signaling, and inflammatory response factors involved in the process, and conclude with a synopsis of these key connections. By unraveling the intricate interplay between P2X7 receptors and epileptogenesis, this review provides ideas for designing potent clinical therapies that will revolutionize both prevention and treatment for epileptic patients.
Collapse
Affiliation(s)
- Caichuan Wei
- Department of Pediatrics, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuchang District, Wuhan, Hubei Province 430060, China
| | - Miaoying Fu
- Department of Pediatrics, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuchang District, Wuhan, Hubei Province 430060, China
| | - Haiju Zhang
- Department of Pediatrics, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuchang District, Wuhan, Hubei Province 430060, China
| | - Baozhen Yao
- Department of Pediatrics, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuchang District, Wuhan, Hubei Province 430060, China.
| |
Collapse
|
5
|
Janho dit Hreich S, Juhel T, Hofman P, Vouret-Craviari V. Protocol for Evaluating In Vivo the Activation of the P2RX7 Immunomodulator. Biol Proced Online 2023; 25:1. [PMID: 36600200 PMCID: PMC9811721 DOI: 10.1186/s12575-022-00188-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND P2RX7 is a purinergic receptor with pleiotropic activities that is activated by high levels of extracellular ATP that are found in inflamed tissues. P2RX7 has immunomodulatory and anti-tumor proprieties and is therefore a therapeutic target for various diseases. Several compounds are developed to either inhibit or enhance its activation. However, studying their effect on P2RX7's activities is limited to in vitro and ex vivo studies that require the use of unphysiological media that could affect its activation. Up to now, the only way to assess the activity of P2RX7 modulators on the receptor in vivo was in an indirect manner. RESULTS We successfully developed a protocol allowing the detection of P2RX7 activation in vivo in lungs of mice, by taking advantage of its unique macropore formation ability. The protocol is based on intranasal delivery of TO-PRO™-3, a non-permeant DNA intercalating dye, and fluorescence measurement by flow cytometry. We show that ATP enhances TO-PRO™-3 fluorescence mainly in lung immune cells of mice in a P2RX7-dependant manner. CONCLUSIONS The described approach has allowed the successful analysis of P2RX7 activity directly in the lungs of WT and transgenic C57BL6 mice. The provided detailed guidelines and recommendations will support the use of this protocol to study the potency of pharmacologic or biologic compounds targeting P2RX7.
Collapse
Affiliation(s)
- Serena Janho dit Hreich
- grid.463830.a0000 0004 8340 3111Université Côte d’Azur, CNRS, INSERM, IRCAN, 28 avenue de Valombrose, 06108 Nice, France ,grid.464719.90000 0004 0639 4696FHU OncoAge, Pasteur Hospital, 30 voie Romaine, 06001 Nice, France
| | - Thierry Juhel
- grid.463830.a0000 0004 8340 3111Université Côte d’Azur, CNRS, INSERM, IRCAN, 28 avenue de Valombrose, 06108 Nice, France
| | - Paul Hofman
- grid.463830.a0000 0004 8340 3111Université Côte d’Azur, CNRS, INSERM, IRCAN, 28 avenue de Valombrose, 06108 Nice, France ,grid.464719.90000 0004 0639 4696FHU OncoAge, Pasteur Hospital, 30 voie Romaine, 06001 Nice, France ,grid.464719.90000 0004 0639 4696Laboratory of Clinical and Experimental Pathology and Biobank, Pasteur Hospital, 30 voie Romaine, 06001 Nice, France ,grid.464719.90000 0004 0639 4696Hospital-Related Biobank (BB-0033-00025), Pasteur Hospital, 30 voie Romaine, 06001 Nice, France
| | - Valérie Vouret-Craviari
- grid.463830.a0000 0004 8340 3111Université Côte d’Azur, CNRS, INSERM, IRCAN, 28 avenue de Valombrose, 06108 Nice, France ,grid.464719.90000 0004 0639 4696FHU OncoAge, Pasteur Hospital, 30 voie Romaine, 06001 Nice, France
| |
Collapse
|
6
|
Moreira A, Tovar M, Smith AM, Lee GC, Meunier JA, Cheema Z, Moreira A, Winter C, Mustafa SB, Seidner S, Findley T, Garcia JGN, Thébaud B, Kwinta P, Ahuja SK. Development of a peripheral blood transcriptomic gene signature to predict bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L76-L87. [PMID: 36472344 PMCID: PMC9829478 DOI: 10.1152/ajplung.00250.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/27/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common lung disease of extreme prematurity, yet mechanisms that associate with or identify neonates with increased susceptibility for BPD are largely unknown. Combining artificial intelligence with gene expression data is a novel approach that may assist in better understanding mechanisms underpinning chronic lung disease and in stratifying patients at greater risk for BPD. The objective of this study is to develop an early peripheral blood transcriptomic signature that can predict preterm neonates at risk for developing BPD. Secondary analysis of whole blood microarray data from 97 very low birth weight neonates on day of life 5 was performed. BPD was defined as positive pressure ventilation or oxygen requirement at 28 days of age. Participants were randomly assigned to a training (70%) and testing cohort (30%). Four gene-centric machine learning models were built, and their discriminatory abilities were compared with gestational age or birth weight. This study adheres to the transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD) statement. Neonates with BPD (n = 62 subjects) exhibited a lower median gestational age (26.0 wk vs. 30.0 wk, P < 0.01) and birth weight (800 g vs. 1,280 g, P < 0.01) compared with non-BPD neonates. From an initial pool (33,252 genes/patient), 4,523 genes exhibited a false discovery rate (FDR) <1%. The area under the receiver operating characteristic curve (AUC) for predicting BPD utilizing gestational age or birth weight was 87.8% and 87.2%, respectively. The machine learning models, using a combination of five genes, revealed AUCs ranging between 85.8% and 96.1%. Pathways integral to T cell development and differentiation were associated with BPD. A derived five-gene whole blood signature can accurately predict BPD in the first week of life.
Collapse
Affiliation(s)
- Alvaro Moreira
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Miriam Tovar
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Alisha M Smith
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Grace C Lee
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- Pharmacotherapy Education and Research Center, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- College of Pharmacy, The University of Texas at Austin, Austin, Texas
| | - Justin A Meunier
- Veterans Administration Research Center for AIDS and HIV-1 Infection and Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Zoya Cheema
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Axel Moreira
- Division of Critical Care, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Caitlyn Winter
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Shamimunisa B Mustafa
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Steven Seidner
- Department of Pediatrics, Neonatology Regenerative and Precision Medicine Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
| | - Tina Findley
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, Texas
| | - Joe G N Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona
| | - Bernard Thébaud
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Department of Pediatrics, Children's Hospital of Eastern Ontario (CHEO) and CHEO Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Przemko Kwinta
- Neonatal Intensive Care Unit, Department of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Sunil K Ahuja
- Veterans Administration Center for Personalized Medicine, South Texas Veterans Health Care System, San Antonio, Texas
- The Foundation for Advancing Veterans' Health Research, South Texas Veterans Health Care System, San Antonio, Texas
- Department of Microbiology, Immunology & Molecular Genetics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| |
Collapse
|
7
|
Hasan D, Shono A, van Kalken CK, van der Spek PJ, Krenning EP, Kotani T. A novel definition and treatment of hyperinflammation in COVID-19 based on purinergic signalling. Purinergic Signal 2021; 18:13-59. [PMID: 34757513 PMCID: PMC8578920 DOI: 10.1007/s11302-021-09814-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/18/2021] [Indexed: 12/15/2022] Open
Abstract
Hyperinflammation plays an important role in severe and critical COVID-19. Using inconsistent criteria, many researchers define hyperinflammation as a form of very severe inflammation with cytokine storm. Therefore, COVID-19 patients are treated with anti-inflammatory drugs. These drugs appear to be less efficacious than expected and are sometimes accompanied by serious adverse effects. SARS-CoV-2 promotes cellular ATP release. Increased levels of extracellular ATP activate the purinergic receptors of the immune cells initiating the physiologic pro-inflammatory immune response. Persisting viral infection drives the ATP release even further leading to the activation of the P2X7 purinergic receptors (P2X7Rs) and a severe yet physiologic inflammation. Disease progression promotes prolonged vigorous activation of the P2X7R causing cell death and uncontrolled ATP release leading to cytokine storm and desensitisation of all other purinergic receptors of the immune cells. This results in immune paralysis with co-infections or secondary infections. We refer to this pathologic condition as hyperinflammation. The readily available and affordable P2X7R antagonist lidocaine can abrogate hyperinflammation and restore the normal immune function. The issue is that the half-maximal effective concentration for P2X7R inhibition of lidocaine is much higher than the maximal tolerable plasma concentration where adverse effects start to develop. To overcome this, we selectively inhibit the P2X7Rs of the immune cells of the lymphatic system inducing clonal expansion of Tregs in local lymph nodes. Subsequently, these Tregs migrate throughout the body exerting anti-inflammatory activities suppressing systemic and (distant) local hyperinflammation. We illustrate this with six critically ill COVID-19 patients treated with lidocaine.
Collapse
Affiliation(s)
| | - Atsuko Shono
- Department of Anaesthesiology and Critical Care Medicine, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| | | | - Peter J van der Spek
- Department of Pathology & Clinical Bioinformatics, Erasmus MC, Erasmus Universiteit Rotterdam, 3015 CE, Rotterdam, The Netherlands
| | | | - Toru Kotani
- Department of Anaesthesiology and Critical Care Medicine, School of Medicine, Showa University, Tokyo, 142-8666, Japan
| |
Collapse
|
8
|
Leão Batista Simões J, Fornari Basso H, Cristine Kosvoski G, Gavioli J, Marafon F, Elias Assmann C, Barbosa Carvalho F, Dulce Bagatini M. Targeting purinergic receptors to suppress the cytokine storm induced by SARS-CoV-2 infection in pulmonary tissue. Int Immunopharmacol 2021; 100:108150. [PMID: 34537482 PMCID: PMC8435372 DOI: 10.1016/j.intimp.2021.108150] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/26/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022]
Abstract
The etiological agent of coronavirus disease (COVID-19) is the new member of the Coronaviridae family, a severe acute respiratory syndrome coronavirus 2 virus (SARS-CoV-2), responsible for the pandemic that is plaguing the world. The single-stranded RNA virus is capable of infecting the respiratory tract, by binding the spike (S) protein on its viral surface to receptors for the angiotensin II-converting enzyme (ACE2), highly expressed in the pulmonary tissue, enabling the interaction of the virus with alveolar epithelial cells promoting endocytosis and replication of viral material. The infection triggers the activation of the immune system, increased purinergic signaling, and the release of cytokines as a defense mechanism, but the response can become exaggerated and prompt the so-called “cytokine storm”, developing cases such as severe acute respiratory syndrome (SARS). This is characterized by fever, cough, and difficulty breathing, which can progress to pneumonia, failure of different organs and death. Thus, the present review aims to compile and correlate the mechanisms involved between the immune and purinergic systems with COVID-19, since the modulation of purinergic receptors, such as A2A, A2B, and P2X7 expressed by immune cells, seems to be effective as a promising therapy, to reduce the severity of the disease, as well as aid in the treatment of acute lung diseases and other cases of generalized inflammation.
Collapse
Affiliation(s)
| | | | | | - Jullye Gavioli
- Medical School, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Filomena Marafon
- Postgraduate Program in Biochemistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Charles Elias Assmann
- Postgraduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | | | | |
Collapse
|
9
|
Diem K, Fauler M, Fois G, Hellmann A, Winokurow N, Schumacher S, Kranz C, Frick M. Mechanical stretch activates piezo1 in caveolae of alveolar type I cells to trigger ATP release and paracrine stimulation of surfactant secretion from alveolar type II cells. FASEB J 2020; 34:12785-12804. [PMID: 32744386 DOI: 10.1096/fj.202000613rrr] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/12/2022]
Abstract
Secretion of pulmonary surfactant in the alveoli of the lungs is essential to maintain lung function. Stretching of alveoli during lung inflation is the main trigger for surfactant secretion. Yet, the molecular mechanisms how mechanical distension of alveoli results in surfactant secretion are still elusive. The alveolar epithelium consists of alveolar epithelial type I (ATI) and surfactant secreting type II (ATII) cells. ATI, but not ATII cells, express caveolae, small plasma membrane invaginations that can respond to plasma membrane stresses and serve mechanotransductive roles. Within this study, we investigated the role of caveolae as mechanosensors in the alveolus. We generated a human caveolin-1 knockout ATI cell (hAELVicav-/- ) using CRISPR/Cas9. Wildtype (hAELViwt ) and hAELVicav-/- cells grown on flexible membranes responded to increasing stretch amplitudes with rises in intracellular Ca2+ . The response was less frequent and started at higher stretch amplitudes in hAELVicav-/- cells. Stretch-induced Ca2+ -signals depended on Ca2+ -entry via piezo1 channels, localized within caveolae in hAELViwt and primary ATI cells. Ca2+ -entry via piezo1 activated pannexin-1 hemichannels resulting in ATP release from ATI cells. ATP release was reduced in hAELVicav-/- cells. In co-cultures resembling the alveolar epithelium, released ATP stimulated Ca2+ signals and surfactant secretion from neighboring ATII cells when co-cultured with hAELViwt but not hAELVicav-/- cells. In summary, we propose that caveolae in ATI cells are mechanosensors within alveoli regulating stretch-induced surfactant secretion from ATII cells.
Collapse
Affiliation(s)
- Kathrin Diem
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Andreas Hellmann
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Natalie Winokurow
- Institute of Molecular and Cellular Anatomy, Ulm University, Ulm, Germany
| | - Stefan Schumacher
- Institute of Molecular and Cellular Anatomy, Ulm University, Ulm, Germany
| | - Christine Kranz
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
10
|
Resolving the Ionotropic P2X4 Receptor Mystery Points Towards a New Therapeutic Target for Cardiovascular Diseases. Int J Mol Sci 2020; 21:ijms21145005. [PMID: 32679900 PMCID: PMC7404342 DOI: 10.3390/ijms21145005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022] Open
Abstract
Adenosine triphosphate (ATP) is a primordial versatile autacoid that changes its role from an intracellular energy saver to a signaling molecule once released to the extracellular milieu. Extracellular ATP and its adenosine metabolite are the main activators of the P2 and P1 purinoceptor families, respectively. Mounting evidence suggests that the ionotropic P2X4 receptor (P2X4R) plays pivotal roles in the regulation of the cardiovascular system, yet further therapeutic advances have been hampered by the lack of selective P2X4R agonists. In this review, we provide the state of the art of the P2X4R activity in the cardiovascular system. We also discuss the role of P2X4R activation in kidney and lungs vis a vis their interplay to control cardiovascular functions and dysfunctions, including putative adverse effects emerging from P2X4R activation. Gathering this information may prompt further development of selective P2X4R agonists and its translation to the clinical practice.
Collapse
|
11
|
Wirsching E, Fauler M, Fois G, Frick M. P2 Purinergic Signaling in the Distal Lung in Health and Disease. Int J Mol Sci 2020; 21:E4973. [PMID: 32674494 PMCID: PMC7404078 DOI: 10.3390/ijms21144973] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
The distal lung provides an intricate structure for gas exchange in mammalian lungs. Efficient gas exchange depends on the functional integrity of lung alveoli. The cells in the alveolar tissue serve various functions to maintain alveolar structure, integrity and homeostasis. Alveolar epithelial cells secrete pulmonary surfactant, regulate the alveolar surface liquid (ASL) volume and, together with resident and infiltrating immune cells, provide a powerful host-defense system against a multitude of particles, microbes and toxicants. It is well established that all of these cells express purinergic P2 receptors and that purinergic signaling plays important roles in maintaining alveolar homeostasis. Therefore, it is not surprising that purinergic signaling also contributes to development and progression of severe pathological conditions like pulmonary inflammation, acute lung injury/acute respiratory distress syndrome (ALI/ARDS) and pulmonary fibrosis. Within this review we focus on the role of P2 purinergic signaling in the distal lung in health and disease. We recapitulate the expression of P2 receptors within the cells in the alveoli, the possible sources of ATP (adenosine triphosphate) within alveoli and the contribution of purinergic signaling to regulation of surfactant secretion, ASL volume and composition, as well as immune homeostasis. Finally, we summarize current knowledge of the role for P2 signaling in infectious pneumonia, ALI/ARDS and idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
| | | | | | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (E.W.); (M.F.); (G.F.)
| |
Collapse
|
12
|
Sehlmeyer K, Ruwisch J, Roldan N, Lopez-Rodriguez E. Alveolar Dynamics and Beyond - The Importance of Surfactant Protein C and Cholesterol in Lung Homeostasis and Fibrosis. Front Physiol 2020; 11:386. [PMID: 32431623 PMCID: PMC7213507 DOI: 10.3389/fphys.2020.00386] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Surfactant protein C (SP-C) is an important player in enhancing the interfacial adsorption of lung surfactant lipid films to the alveolar air-liquid interface. Doing so, surface tension drops down enough to stabilize alveoli and the lung, reducing the work of breathing. In addition, it has been shown that SP-C counteracts the deleterious effect of high amounts of cholesterol in the surfactant lipid films. On its side, cholesterol is a well-known modulator of the biophysical properties of biological membranes and it has been proven that it activates the inflammasome pathways in the lung. Even though the molecular mechanism is not known, there are evidences suggesting that these two molecules may interplay with each other in order to keep the proper function of the lung. This review focuses in the role of SP-C and cholesterol in the development of lung fibrosis and the potential pathways in which impairment of both molecules leads to aberrant lung repair, and therefore impaired alveolar dynamics. From molecular to cellular mechanisms to evidences in animal models and human diseases. The evidences revised here highlight a potential SP-C/cholesterol axis as target for the treatment of lung fibrosis.
Collapse
Affiliation(s)
- Kirsten Sehlmeyer
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
| | - Jannik Ruwisch
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
| | - Nuria Roldan
- Alveolix AG and ARTORG Center, University of Bern, Bern, Switzerland
| | - Elena Lopez-Rodriguez
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover, Member of the German Centre for Lung Research, Hanover, Germany
- Institute of Functional Anatomy, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
13
|
Le TTT, Berg NK, Harting MT, Li X, Eltzschig HK, Yuan X. Purinergic Signaling in Pulmonary Inflammation. Front Immunol 2019; 10:1633. [PMID: 31379836 PMCID: PMC6646739 DOI: 10.3389/fimmu.2019.01633] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/01/2019] [Indexed: 12/21/2022] Open
Abstract
Purine nucleotides and nucleosides are at the center of biologic reactions. In particular, adenosine triphosphate (ATP) is the fundamental energy currency of cellular activity and adenosine has been demonstrated to play essential roles in human physiology and pathophysiology. In this review, we examine the role of purinergic signaling in acute and chronic pulmonary inflammation, with emphasis on ATP and adenosine. ATP is released into extracellular space in response to cellular injury and necrosis. It is then metabolized to adenosine monophosphate (AMP) via ectonucleoside triphosphate diphosphohydrolase-1 (CD39) and further hydrolyzed to adenosine via ecto-5'-nucleotidase (CD73). Adenosine signals via one of four adenosine receptors to exert pro- or anti-inflammatory effects. Adenosine signaling is terminated by intracellular transport by concentrative or equilibrative nucleoside transporters (CNTs and ENTs), deamination to inosine by adenosine deaminase (ADA), or phosphorylation back into AMP via adenosine kinase (AK). Pulmonary inflammatory and hypoxic conditions lead to increased extracellular ATP, adenosine diphosphate (ADP) and adenosine levels, which translates to increased adenosine signaling. Adenosine signaling is central to the pulmonary injury response, leading to various effects on inflammation, repair and remodeling processes that are either tissue-protective or tissue destructive. In the acute setting, particularly through activation of adenosine 2A and 2B receptors, adenosine signaling serves an anti-inflammatory, tissue-protective role. However, excessive adenosine signaling in the chronic setting promotes pro-inflammatory, tissue destructive effects in chronic pulmonary inflammation.
Collapse
Affiliation(s)
- Thanh-Thuy T. Le
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Nathaniel K. Berg
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Matthew T. Harting
- Department of Pediatric Surgery, McGovern Medical School, Children's Memorial Hermann Hospital, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiangyun Li
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
- Department of Anesthesiology, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China
| | - Holger K. Eltzschig
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Xiaoyi Yuan
- Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
14
|
Benzaquen J, Heeke S, Janho Dit Hreich S, Douguet L, Marquette CH, Hofman P, Vouret-Craviari V. Alternative splicing of P2RX7 pre-messenger RNA in health and diseases: Myth or reality? Biomed J 2019; 42:141-154. [PMID: 31466708 PMCID: PMC6717933 DOI: 10.1016/j.bj.2019.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 12/21/2022] Open
Abstract
Alternative splicing (AS) tremendously increases the use of genetic information by generating protein isoforms that differ in protein-protein interactions, catalytic activity and/or subcellular localization. This review is not dedicated to AS in general, but rather we focus our attention on AS of P2RX7 pre-mRNA. Whereas P2RX7 mRNA is expressed by virtually all eukaryotic mammalian cells, the expression of this channel receptor is restrained to certain cells. When expressed at the cell membrane, P2RX7 controls downstream events including release of inflammatory molecules, phagocytosis, cell proliferation and death and metabolic events. Therefore, P2RX7 is an important actor of health and diseases. In this review, we summarize the general mechanisms leading to AS. Further, we recapitulate our current knowledge concerning the functional regions in P2RX7, identified at the genetic or exonic levels, and how AS may affect the expression of these regions. Finally, the potential of P2RX7 splice variants to control the fate of cancer cells is discussed.
Collapse
Affiliation(s)
- Jonathan Benzaquen
- University of Cote d'Azur, CNRS, INSERM, IRCAN, Nice, France; FHU OncoAge, Nice, France
| | - Simon Heeke
- University of Cote d'Azur, CNRS, INSERM, IRCAN, Nice, France; Laboratory of Clinical and Experimental Pathology and Biobank, Pasteur Hospital, Nice, France; FHU OncoAge, Nice, France
| | | | | | - Charles Hugo Marquette
- University of Cote d'Azur, CNRS, INSERM, IRCAN, Nice, France; FHU OncoAge, Nice, France; University of Cote d'Azur, CHU de Nice, Department of Pulmonary Medicine, FHU OncoAge, Nice, France
| | - Paul Hofman
- University of Cote d'Azur, CNRS, INSERM, IRCAN, Nice, France; Laboratory of Clinical and Experimental Pathology and Biobank, Pasteur Hospital, Nice, France; Hospital-Related Biobank (BB-0033-00025), Pasteur Hospital, Nice, France; FHU OncoAge, Nice, France
| | | |
Collapse
|
15
|
Miron VV, Bottari NB, Assmann CE, Stefanello N, da Costa P, Pelinson LP, Reichert KP, da Silva AD, Lopes TF, da Cruz IBM, Sévigny J, Morsch VM, Schetinger MRC, Cardoso AM. Physical exercise prevents alterations in purinergic system and oxidative status in lipopolysaccharide-induced sepsis in rats. J Cell Biochem 2018; 120:3232-3242. [PMID: 30230598 DOI: 10.1002/jcb.27590] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/07/2018] [Indexed: 12/11/2022]
Abstract
Sepsis is a generalized infection that involves alterations in inflammatory parameters, oxidant status, and purinergic signaling in many tissues. Physical exercise has emerged as a tool to prevent this disease because of its anti-inflammatory and antioxidant properties. Thus, in this study, we investigated the effects of physical exercise on preventing alterations in purinergic system components, oxidative stress, and inflammatory parameters in lipopolysaccharide (LPS)-induced sepsis in rats. Male Wistar rats were divided into four groups: control, exercise (EX), LPS, and EX+LPS. The resisted physical exercise was performed for 12 weeks on a ladder with 1 m height. After 72 hours of the last exercise session, the animals received 2.5 mg/kg of LPS for induction of sepsis, and after 24 hours, lungs and blood samples were collected for analysis. The results showed that the exercise protocol used was able to prevent, in septic animals: (1) the increase in body temperature; (2) the increase of lipid peroxidation and reactive species levels in the lung, (3) the increase in adenosine triphosphate levels in serum; (4) the change in the activity of the enzymes ectonucleotidases in lymphocytes, partially; (5) the change in the density of purinergic enzymes and receptors in the lung, and (6) the increase of IL-6 and IL-1β gene expression. Our results revealed the involvement of purinergic signaling and oxidative damage in the mechanisms by which exercise prevents sepsis aggravations. Therefore, the regular practice of physical exercise is encouraged as a better way to prepare the body against sepsis complications.
Collapse
Affiliation(s)
- Vanessa Valéria Miron
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Nathieli Bianchin Bottari
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Charles Elias Assmann
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Naiara Stefanello
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Pauline da Costa
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Luana Paula Pelinson
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Karine Paula Reichert
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Anielen Dutra da Silva
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Thauan Faccin Lopes
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Ivana Beatrice Mânica da Cruz
- Morphology Department, Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Centre de recherche du CHU de Québec-Université Laval, Québec City, Québec, Canada
| | - Vera Maria Morsch
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Maria Rosa Chitolina Schetinger
- Department of Biochemistry and Molecular Biology, Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Andréia Machado Cardoso
- Federal University of Fronteira Sul, Academic Coordination, Medicine, Campus Chapecó, Chapecó, Santa Catarina, Brazil
| |
Collapse
|
16
|
A feasibility study into adenosine triphosphate measurement in exhaled breath condensate: a potential bedside method to monitor alveolar deformation. Purinergic Signal 2018; 14:215-221. [PMID: 29752619 PMCID: PMC6107466 DOI: 10.1007/s11302-018-9607-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/24/2018] [Indexed: 12/13/2022] Open
Abstract
Recent research suggested an important role for pulmonary extracellular adenosine triphosphate (ATP) in the development of ventilation-induced lung injury. This injury is induced by mechanical deformation of alveolar epithelial cells, which in turn release ATP to the extracellular space. Measuring extracellular ATP in exhaled breath condensate (EBC) may be a non-invasive biomarker for alveolar deformation. Here, we study the feasibility of bedside ATP measurement in EBC. We measured ATP levels in EBC in ten subjects before and after an exercise test, which increases respiratory parameters and alveolar deformation. EBC lactate concentrations were measured as a dilution marker. We found a significant increase in ATP levels in EBC (before 73 RLU [IQR 50–209] versus after 112 RLU [IQR 86–203]; p value 0.047), and the EBC ATP-to-EBC lactate ratio increased as well (p value 0.037). We present evidence that bedside measurement of ATP in EBC is feasible and that ATP levels in EBC increase after exercise. Future research should measure ATP levels in EBC during mechanical ventilation as a potential biomarker for alveolar deformation.
Collapse
|
17
|
Hasan D, Satalin J, van der Zee P, Kollisch-Singule M, Blankman P, Shono A, Somhorst P, den Uil C, Meeder H, Kotani T, Nieman GF. Excessive Extracellular ATP Desensitizes P2Y2 and P2X4 ATP Receptors Provoking Surfactant Impairment Ending in Ventilation-Induced Lung Injury. Int J Mol Sci 2018; 19:ijms19041185. [PMID: 29652806 PMCID: PMC5979391 DOI: 10.3390/ijms19041185] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/16/2022] Open
Abstract
Stretching the alveolar epithelial type I (AT I) cells controls the intercellular signaling for the exocytosis of surfactant by the AT II cells through the extracellular release of adenosine triphosphate (ATP) (purinergic signaling). Extracellular ATP is cleared by extracellular ATPases, maintaining its homeostasis and enabling the lung to adapt the exocytosis of surfactant to the demand. Vigorous deformation of the AT I cells by high mechanical power ventilation causes a massive release of extracellular ATP beyond the clearance capacity of the extracellular ATPases. When extracellular ATP reaches levels >100 μM, the ATP receptors of the AT II cells become desensitized and surfactant impairment is initiated. The resulting alteration in viscoelastic properties and in alveolar opening and collapse time-constants leads to alveolar collapse and the redistribution of inspired air from the alveoli to the alveolar ducts, which become pathologically dilated. The collapsed alveoli connected to these dilated alveolar ducts are subject to a massive strain, exacerbating the ATP release. After reaching concentrations >300 μM extracellular ATP acts as a danger-associated molecular pattern, causing capillary leakage, alveolar space edema, and further deactivation of surfactant by serum proteins. Decreasing the tidal volume to 6 mL/kg or less at this stage cannot prevent further lung injury.
Collapse
Affiliation(s)
- Djo Hasan
- Mobile Intensive Care Unit Zuid-West Nederland, 3062 NW Rotterdam, The Netherlands.
- Department of Surgery, Erasmus MC, Erasmus Universiteit Rotterdam, 3015 CE Rotterdam, The Netherlands.
| | - Joshua Satalin
- Department of Surgery, Upstate Medical University, Syracuse, NY 13210, USA.
| | - Philip van der Zee
- Adult Intensive Care Unit, Erasmus MC, Erasmus Universiteit Rotterdam, 3015 CE Rotterdam, The Netherlands.
| | | | - Paul Blankman
- Department of Anesthesiology, Universitair Medisch Centrum Utrecht, 3584 CX Utrecht, The Netherlands.
| | - Atsuko Shono
- Department of Anesthesiology, Shimane University, Izumo, Shimane Prefecture 693-0021, Japan.
| | - Peter Somhorst
- Adult Intensive Care Unit, Erasmus MC, Erasmus Universiteit Rotterdam, 3015 CE Rotterdam, The Netherlands.
| | - Corstiaan den Uil
- Adult Intensive Care Unit, Erasmus MC, Erasmus Universiteit Rotterdam, 3015 CE Rotterdam, The Netherlands.
- Department of Cardiology, Erasmus MC, Erasmus Universiteit Rotterdam, 3062 PA Rotterdam, The Netherlands.
| | - Han Meeder
- Mobile Intensive Care Unit Zuid-West Nederland, 3062 NW Rotterdam, The Netherlands.
- Adult Intensive Care Unit, Erasmus MC, Erasmus Universiteit Rotterdam, 3015 CE Rotterdam, The Netherlands.
| | - Toru Kotani
- Department of Anesthesiology and Critical Care Medicine, Showa University, School of Medicine, Tokyo 142-8666, Japan.
| | - Gary F Nieman
- Department of Surgery, Upstate Medical University, Syracuse, NY 13210, USA.
| |
Collapse
|
18
|
Savio LEB, de Andrade Mello P, da Silva CG, Coutinho-Silva R. The P2X7 Receptor in Inflammatory Diseases: Angel or Demon? Front Pharmacol 2018; 9:52. [PMID: 29467654 PMCID: PMC5808178 DOI: 10.3389/fphar.2018.00052] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
Under physiological conditions, adenosine triphosphate (ATP) is present at low levels in the extracellular milieu, being massively released by stressed or dying cells. Once outside the cells, ATP and related nucleotides/nucleoside generated by ectonucleotidases mediate a high evolutionary conserved signaling system: the purinergic signaling, which is involved in a variety of pathological conditions, including inflammatory diseases. Extracellular ATP has been considered an endogenous adjuvant that can initiate inflammation by acting as a danger signal through the activation of purinergic type 2 receptors-P2 receptors (P2Y G-protein coupled receptors and P2X ligand-gated ion channels). Among the P2 receptors, the P2X7 receptor is the most extensively studied from an immunological perspective, being involved in both innate and adaptive immune responses. P2X7 receptor activation induces large-scale ATP release via its intrinsic ability to form a membrane pore or in association with pannexin hemichannels, boosting purinergic signaling. ATP acting via P2X7 receptor is the second signal to the inflammasome activation, inducing both maturation and release of pro-inflammatory cytokines, such as IL-1β and IL-18, and the production of reactive nitrogen and oxygen species. Furthermore, the P2X7 receptor is involved in caspases activation, as well as in apoptosis induction. During adaptive immune response, P2X7 receptor modulates the balance between the generation of T helper type 17 (Th17) and T regulatory (Treg) lymphocytes. Therefore, this receptor is involved in several inflammatory pathological conditions. In infectious diseases and cancer, P2X7 receptor can have different and contrasting effects, being an angel or a demon depending on its level of activation, cell studied, type of pathogen, and severity of infection. In neuroinflammatory and neurodegenerative diseases, P2X7 upregulation and function appears to contribute to disease progression. In this review, we deeply discuss P2X7 receptor dual function and its pharmacological modulation in the context of different pathologies, and we also highlight the P2X7 receptor as a potential target to treat inflammatory related diseases.
Collapse
Affiliation(s)
- Luiz E B Savio
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paola de Andrade Mello
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Cleide Gonçalves da Silva
- Division of Vascular Surgery, Department of Surgery, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
19
|
Fois G, Winkelmann VE, Bareis L, Staudenmaier L, Hecht E, Ziller C, Ehinger K, Schymeinsky J, Kranz C, Frick M. ATP is stored in lamellar bodies to activate vesicular P2X 4 in an autocrine fashion upon exocytosis. J Gen Physiol 2017; 150:277-291. [PMID: 29282210 PMCID: PMC5806682 DOI: 10.1085/jgp.201711870] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/12/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022] Open
Abstract
P2X4 receptor activation facilitates secretion of pulmonary surfactant from secretory vesicles called lamellar bodies in alveolar epithelial cells. Fois et al. reveal that P2X4 receptors on the lamellar body membranes are activated by ATP stored within the vesicles themselves upon vesicle exocytosis. Vesicular P2X4 receptors are known to facilitate secretion and activation of pulmonary surfactant in the alveoli of the lungs. P2X4 receptors are expressed in the membrane of lamellar bodies (LBs), large secretory lysosomes that store lung surfactant in alveolar type II epithelial cells, and become inserted into the plasma membrane after exocytosis. Subsequent activation of P2X4 receptors by adenosine triphosphate (ATP) results in local fusion-activated cation entry (FACE), facilitating fusion pore dilation, surfactant secretion, and surfactant activation. Despite the importance of ATP in the alveoli, and hence lung function, the origin of ATP in the alveoli is still elusive. In this study, we demonstrate that ATP is stored within LBs themselves at a concentration of ∼1.9 mM. ATP is loaded into LBs by the vesicular nucleotide transporter but does not activate P2X4 receptors because of the low intraluminal pH (5.5). However, the rise in intravesicular pH after opening of the exocytic fusion pore results in immediate activation of vesicular P2X4 by vesicular ATP. Our data suggest a new model in which agonist (ATP) and receptor (P2X4) are located in the same intracellular compartment (LB), protected from premature degradation (ATP) and activation (P2X4), and ideally placed to ensure coordinated and timely receptor activation as soon as fusion occurs to facilitate surfactant secretion.
Collapse
Affiliation(s)
- Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Lara Bareis
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Elena Hecht
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Charlotte Ziller
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | | | - Jürgen Schymeinsky
- Immunology and Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Christine Kranz
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
20
|
Wu X, Ren J, Chen G, Wu L, Song X, Li G, Deng Y, Wang G, Gu G, Li J. Systemic blockade of P2X7 receptor protects against sepsis-induced intestinal barrier disruption. Sci Rep 2017; 7:4364. [PMID: 28663567 PMCID: PMC5491531 DOI: 10.1038/s41598-017-04231-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 05/11/2017] [Indexed: 02/07/2023] Open
Abstract
Sepsis, during which the intestinal epithelial barrier is frequently disrupted, remains a challenging and life-threatening problem in clinical practice. The P2X7 receptor (P2X7R) is a non-selective adenosine triphosphate-gated cation channel present in macrophages that is involved in inflammatory responses. However, little is known about the role of P2X7R in macrophages during sepsis-induced intestinal barrier disruption. In this study, mice were treated with the P2X7R antagonist A740003 or the agonist BzATP by intra-peritoneal injection after the induction of gut-origin sepsis. The survival rates, inflammatory responses, intestinal barrier integrity, macrophage marker expression, and ERK and NF-κB activities were evaluated. Intestinal macrophages were also isolated and studied after exposure to Brilliant Blue G or BzATP. We found that a systemic P2X7R blockade downregulated sepsis-induced inflammatory responses and attenuated intestinal barrier dysfunction based on the evidence that mice in the A740003-treated group exhibited alleviated pro-inflammatory cytokine synthesis, intestinal hyperpermeability, epithelial apoptosis rates and tight junction damage compared with the septic mice. These changes were partly mediated by the inhibition of M1 macrophages activation via ERK/NF-κB pathways. Our data presented herein show that a P2X7R blockade could be a potential therapeutic target for the treatment of sepsis-induced intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Xiuwen Wu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianan Ren
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Guopu Chen
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Wu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xian Song
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guanwei Li
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Youming Deng
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Gefei Wang
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guosheng Gu
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jieshou Li
- Department of Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
21
|
Hasan D, Blankman P, Nieman GF. Purinergic signalling links mechanical breath profile and alveolar mechanics with the pro-inflammatory innate immune response causing ventilation-induced lung injury. Purinergic Signal 2017; 13:363-386. [PMID: 28547381 PMCID: PMC5563293 DOI: 10.1007/s11302-017-9564-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/26/2017] [Indexed: 02/06/2023] Open
Abstract
Severe pulmonary infection or vigorous cyclic deformation of the alveolar epithelial type I (AT I) cells by mechanical ventilation leads to massive extracellular ATP release. High levels of extracellular ATP saturate the ATP hydrolysis enzymes CD39 and CD73 resulting in persistent high ATP levels despite the conversion to adenosine. Above a certain level, extracellular ATP molecules act as danger-associated molecular patterns (DAMPs) and activate the pro-inflammatory response of the innate immunity through purinergic receptors on the surface of the immune cells. This results in lung tissue inflammation, capillary leakage, interstitial and alveolar oedema and lung injury reducing the production of surfactant by the damaged AT II cells and deactivating the surfactant function by the concomitant extravasated serum proteins through capillary leakage followed by a substantial increase in alveolar surface tension and alveolar collapse. The resulting inhomogeneous ventilation of the lungs is an important mechanism in the development of ventilation-induced lung injury. The high levels of extracellular ATP and the upregulation of ecto-enzymes and soluble enzymes that hydrolyse ATP to adenosine (CD39 and CD73) increase the extracellular adenosine levels that inhibit the innate and adaptive immune responses rendering the host susceptible to infection by invading microorganisms. Moreover, high levels of extracellular adenosine increase the expression, the production and the activation of pro-fibrotic proteins (such as TGF-β, α-SMA, etc.) followed by the establishment of lung fibrosis.
Collapse
Affiliation(s)
- Djo Hasan
- Department of Adult ICU, University Hospital Erasmus MC Rotterdam, 's-Gravendijkwal 230 3015 CE, Rotterdam, the Netherlands.
| | - Paul Blankman
- Department of Adult ICU, University Hospital Erasmus MC Rotterdam, 's-Gravendijkwal 230 3015 CE, Rotterdam, the Netherlands
| | - Gary F Nieman
- Department of Surgery, Upstate Medical University, 750 E Adams St, Syracuse, NY, 13210, USA
| |
Collapse
|
22
|
Mishra A, Guo Y, Zhang L, More S, Weng T, Chintagari NR, Huang C, Liang Y, Pushparaj S, Gou D, Breshears M, Liu L. A Critical Role for P2X7 Receptor-Induced VCAM-1 Shedding and Neutrophil Infiltration during Acute Lung Injury. THE JOURNAL OF IMMUNOLOGY 2016; 197:2828-37. [PMID: 27559050 DOI: 10.4049/jimmunol.1501041] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/25/2016] [Indexed: 01/23/2023]
Abstract
Pulmonary neutrophils are the initial inflammatory cells that are recruited during lung injury and are crucial for innate immunity. However, pathological recruitment of neutrophils results in lung injury. The objective of this study is to determine whether the novel neutrophil chemoattractant, soluble VCAM-1 (sVCAM-1), recruits pathological levels of neutrophils to injury sites and amplifies lung inflammation during acute lung injury. The mice with P2X7 receptor deficiency, or treated with a P2X7 receptor inhibitor or anti-VCAM-1 Abs, were subjected to a clinically relevant two-hit LPS and mechanical ventilation-induced acute lung injury. Neutrophil infiltration and lung inflammation were measured. Neutrophil chemotactic activities were determined by a chemotaxis assay. VCAM-1 shedding and signaling pathways were assessed in isolated lung epithelial cells. Ab neutralization of sVCAM-1 or deficiency or antagonism of P2X7R reduced neutrophil infiltration and proinflammatory cytokine levels. The ligands for sVCAM-1 were increased during acute lung injury. sVCAM-1 had neutrophil chemotactic activities and activated alveolar macrophages. VCAM-1 is released into the alveolar airspace from alveolar epithelial type I cells through P2X7 receptor-mediated activation of the metalloproteinase ADAM-17. In conclusion, sVCAM-1 is a novel chemoattractant for neutrophils and an activator for alveolar macrophages. Targeting sVCAM-1 provides a therapeutic intervention that could block pathological neutrophil recruitment, without interfering with the physiological recruitment of neutrophils, thus avoiding the impairment of host defenses.
Collapse
Affiliation(s)
- Amarjit Mishra
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078
| | - Yujie Guo
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078; Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078
| | - Li Zhang
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078
| | - Sunil More
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078; Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078
| | - Tingting Weng
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078
| | - Narendranath Reddy Chintagari
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078
| | - Chaoqun Huang
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078; Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078
| | - Yurong Liang
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078; Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078
| | - Samuel Pushparaj
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078; Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China; and
| | - Melanie Breshears
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078; Department of Pathobiology, Oklahoma State University, Stillwater, OK 74078
| | - Lin Liu
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078; Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078;
| |
Collapse
|
23
|
Platelet-derived Wnt antagonist Dickkopf-1 is implicated in ICAM-1/VCAM-1-mediated neutrophilic acute lung inflammation. Blood 2015; 126:2220-9. [PMID: 26351298 DOI: 10.1182/blood-2015-02-622233] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/20/2015] [Indexed: 12/14/2022] Open
Abstract
Neutrophil infiltration represents the early acute inflammatory response in acute lung injury. The recruitment of neutrophils from the peripheral blood across the endothelial-epithelial barrier into the alveolar airspace is highly regulated by the adhesion molecules on alveolar epithelial cells (AECs). Wnt/β-catenin signaling is involved in the progression of inflammatory lung diseases including asthma, emphysema, and pulmonary fibrosis. However, the function of Wnt/β-catenin signaling in acute lung inflammation is unknown. Here, we identified platelet-derived Dickkopf-1 (Dkk1) as the major Wnt antagonist contributing to the suppression of Wnt/β-catenin signaling in AECs during acute lung inflammation. Intratracheal administration of Wnt3a or an antibody capable of neutralizing Dkk1 inhibited neutrophil influx into the alveolar airspace of injured lungs. Activation of Wnt/β-catenin signaling in AECs attenuated intercellular adhesion molecule 1 (ICAM-1)/vascular cell adhesion molecule 1 (VCAM-1)-mediated adhesion of both macrophages and neutrophils to AECs. Our results suggest a role for Wnt/β-catenin signaling in modulating the inflammatory response, and a functional communication between platelets and AECs during acute lung inflammation. Targeting Wnt/β-catenin signaling and the communication between platelets and AECs therefore represents potential therapeutic strategies to limit the damage of acute pulmonary inflammation.
Collapse
|
24
|
KOLOMAZNIK M, ZILA I, KOPINCOVA J, MOKRA D, CALKOVSKA A. Changes in Lung Surfactant Proteins in Rats With Lipopolysaccharide--Induced Fever. Physiol Res 2014; 63:S619-28. [DOI: 10.33549/physiolres.932928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The study was designed to prove the hypothesis that lipopolysaccharide (LPS)-induced fever elicits the changes in surfactant specific proteins, potentially related to thermal tachypnea. In adult rats fever was induced by intraperitoneal administration of LPS at a dose 100 µg/kg of body weight; control group received saline. Respiratory parameters, arterial blood gases and pH and colonic body temperature (BT) were recorded. Five hours later, surfactant proteins (SP) A, B, C and D were evaluated in bronchoalveolar lavage fluid (BALF) and lung tissue (LT). LPS evoked monophasic thermic response (at 300 min 38.7±0.2 vs. 36.4±0.3 °C, P0.05) and an increase in minute ventilation due to changes in breathing rate and tidal volume. LPS-instilled animals had higher levels of SP-A and SP-D in LT (P0.05 and 0.01), and higher SP-D in BALF (P0.01) than controls. SP-B increased in LT and SP-C in BALF of animals with LPS (both P0.05 vs. controls). The changes in all surfactant specific proteins are present in LPS-induced fever. Alterations of proteins related to local immune mechanisms (SP-A, SP-D) are probably a part of general inflammatory response to pyrogen. Changes in proteins related to surface activity (SP-B and SP-C) might reflect the effort of the body to stabilize the lungs in thermal challenge.
Collapse
Affiliation(s)
| | | | | | | | - A. CALKOVSKA
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| |
Collapse
|
25
|
Robinson LE, Shridar M, Smith P, Murrell-Lagnado RD. Plasma membrane cholesterol as a regulator of human and rodent P2X7 receptor activation and sensitization. J Biol Chem 2014; 289:31983-31994. [PMID: 25281740 PMCID: PMC4231676 DOI: 10.1074/jbc.m114.574699] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
P2X7 receptors are nonselective cation channels gated by high extracellular ATP, but with sustained activation, receptor sensitization occurs, whereby the intrinsic pore dilates, making the cell permeable to large organic cations, which eventually leads to cell death. P2X7 receptors associate with cholesterol-rich lipid rafts, but it is unclear how this affects the properties of the receptor channel. Here we show that pore-forming properties of human and rodent P2X7 receptors are sensitive to perturbations of cholesterol levels. Acute depletion of cholesterol with 5 mm methyl-β-cyclodextrin (MCD) caused a substantial increase in the rate of agonist-evoked pore formation, as measured by the uptake of ethidium dye, whereas cholesterol loading inhibited this process. Patch clamp analysis of P2X7 receptor currents carried by Na+ and N-methyl-d-glucamine (NMDG+) showed enhanced activation and current facilitation following cholesterol depletion. This contrasts with the inhibitory effect of methyl-β-cyclodextrin reported for other P2X subtypes. Mutational analysis suggests the involvement of an N-terminal region and a proximal C-terminal region that comprises multiple cholesterol recognition amino acid consensus (CRAC) motifs, in the cholesterol sensitivity of channel gating. These results reveal cholesterol as a negative regulator of P2X7 receptor pore formation, protecting cells from P2X7-mediated cell death.
Collapse
Affiliation(s)
- Lucy E Robinson
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Mitesh Shridar
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Philip Smith
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom
| | - Ruth D Murrell-Lagnado
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, United Kingdom.
| |
Collapse
|
26
|
Guo Y, Mishra A, Weng T, Chintagari NR, Wang Y, Zhao C, Huang C, Liu L. Wnt3a mitigates acute lung injury by reducing P2X7 receptor-mediated alveolar epithelial type I cell death. Cell Death Dis 2014; 5:e1286. [PMID: 24922070 PMCID: PMC4611727 DOI: 10.1038/cddis.2014.254] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 04/05/2014] [Accepted: 04/14/2014] [Indexed: 02/07/2023]
Abstract
Acute lung injury (ALI) is characterized by pulmonary endothelial and epithelial cell damage, and loss of the alveolar–capillary barrier. We have previously shown that P2X7 receptor (P2X7R), a cell death receptor, is specifically expressed in alveolar epithelial type I cells (AEC I). In this study, we hypothesized that P2X7R-mediated purinergic signaling and its interaction with Wnt/β-catenin signaling contributes to AEC I death. We examined the effect of P2X7R agonist 2′-3′-O-(4-benzoylbenzoyl)-ATP (BzATP) and Wnt agonist Wnt3a on AEC I death in vitro and in vivo. We also assessed the therapeutic potential of Wnt3a in a clinically relevant ALI model of intratracheal lipopolysaccharide (LPS) exposure in ventilated mice. We found that the activation of P2X7R by BzATP caused the death of AEC I by suppressing Wnt/β-catenin signaling through stimulating glycogen synthase kinase-3β (GSK-3β) and proteasome. On the other hand, the activation of Wnt/β-catenin signaling by Wnt3a, GSK-3β inhibitor, or proteasome inhibitor blocked the P2X7R-mediated cell death. More importantly, Wnt3a attenuated the AEC I damage caused by intratracheal instillation of BzATP in rats or LPS in ventilated mice. Our results suggest that Wnt3a overrides the effect of P2X7R on the Wnt/β-catenin signaling to prevent the AEC I death and restrict the severity of ALI.
Collapse
Affiliation(s)
- Y Guo
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratorym, Oklahoma State University, Stillwater, OK 74078, USA
| | - A Mishra
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratorym, Oklahoma State University, Stillwater, OK 74078, USA
| | - T Weng
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratorym, Oklahoma State University, Stillwater, OK 74078, USA
| | - N R Chintagari
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratorym, Oklahoma State University, Stillwater, OK 74078, USA
| | - Y Wang
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratorym, Oklahoma State University, Stillwater, OK 74078, USA
| | - C Zhao
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratorym, Oklahoma State University, Stillwater, OK 74078, USA
| | - C Huang
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratorym, Oklahoma State University, Stillwater, OK 74078, USA
| | - L Liu
- Department of Physiological Sciences, Center for Veterinary Health Sciences, Lundberg-Kienlen Lung Biology and Toxicology Laboratorym, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
27
|
P2X7 receptor antagonism inhibits p38 mitogen-activated protein kinase activation and ameliorates neuronal apoptosis after subarachnoid hemorrhage in rats. Crit Care Med 2014; 41:e466-74. [PMID: 23963136 DOI: 10.1097/ccm.0b013e31829a8246] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVES Brilliant blue G, a selective P2X7 receptor antagonist, exhibits neuroprotective properties. This study examined whether brilliant blue G treatment ameliorates early brain injury after experimental subarachnoid hemorrhage, specifically via inhibiting p38 mitogen-activated protein kinase-related proapoptotic pathways. DESIGN Controlled in vivo laboratory study. SETTING Animal research laboratory. SUBJECTS One hundred fifty-four adult male Sprague-Dawley rats weighing 280-320 g. INTERVENTIONS Subarachnoid hemorrhage was induced in rats by endovascular perforation. Experiment 1 implemented sham-operated rats (sham) and subarachnoid hemorrhage animals, which received vehicle (subarachnoid hemorrhage + vehicle), brilliant blue G (subarachnoid hemorrhage + brilliant blue G), or brilliant blue G plus 2'(3')-O-(4-Benzoylbenzoyl)adenosine 5'-triphosphate (BzATP) (subarachnoid hemorrhage + brilliant blue G + BzATP). The animals were intraperitoneally treated with brilliant blue G (30 mg/kg) at 30 minutes after subarachnoid hemorrhage. BzATP (50 μg/rat), a P2X7 receptor agonist, was intracerebroventricularly administered. Experiment 2 implemented sham-operated rats (sham) and subarachnoid hemorrhage animals, which received vehicle (subarachnoid hemorrhage + vehicle), scramble small interfering RNA (subarachnoid hemorrhage + scramble small interfering RNA), or P2X7 receptor small interfering RNA (subarachnoid hemorrhage + P2X7 receptor small interfering RNA). Subarachnoid hemorrhage grading, neurobehavioral score, and brain edema were evaluated at 24 and 72 hours after surgery. The expression of phosphorylated p38 mitogen-activated protein kinase, phosphorylated extracellular signal-regulated kinases, phosphorylated c-Jun N-terminal kinases, P2X7 receptor, Bcl-2, and cleaved caspase-3 in the left cerebral hemisphere were determined by Western blot. Neuronal apoptosis was examined by double immunofluorescence staining using P2X7 receptor, terminal deoxynucleotidyl transferase-mediated uridine 5'-triphosphate-biotin nick end-labeling, and neuronal nuclei. MEASUREMENTS AND MAIN RESULTS Brilliant blue G significantly improved neurobehavioral function and ameliorated brain water content at 24 and 72 hours after subarachnoid hemorrhage. BzATP reversed these treatment effects. Brilliant blue G attenuated neuronal apoptosis in the subcortex, which was associated with decreased expression of phosphorylated p38 mitogen-activated protein kinase and cleaved caspase-3 and an increased expression of Bcl-2 in the left cerebral hemisphere. The beneficial effects of P2X7 receptor small interfering RNA were also mediated by a p38 mitogen-activated protein kinase pathway. CONCLUSIONS Inhibition of P2X7 receptor by brilliant blue G or P2X7 receptor small interfering RNA can prevent early brain injury via p38 mitogen-activated protein kinase after subarachnoid hemorrhage.
Collapse
|
28
|
Bridges JP, Ludwig MG, Mueller M, Kinzel B, Sato A, Xu Y, Whitsett JA, Ikegami M. Orphan G protein-coupled receptor GPR116 regulates pulmonary surfactant pool size. Am J Respir Cell Mol Biol 2013; 49:348-57. [PMID: 23590306 DOI: 10.1165/rcmb.2012-0439oc] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pulmonary surfactant levels within the alveoli are tightly regulated to maintain lung volumes and promote efficient gas exchange across the air/blood barrier. Quantitative and qualitative abnormalities in surfactant are associated with severe lung diseases in children and adults. Although the cellular and molecular mechanisms that control surfactant metabolism have been studied intensively, the critical molecular pathways that sense and regulate endogenous surfactant levels within the alveolus have not been identified and constitute a fundamental knowledge gap in the field. In this study, we demonstrate that expression of an orphan G protein-coupled receptor, GPR116, in the murine lung is developmentally regulated, reaching maximal levels 1 day after birth, and is highly expressed on the apical surface of alveolar type I and type II epithelial cells. To define the physiological role of GPR116 in vivo, mice with a targeted mutation of the Gpr116 locus, Gpr116(Δexon17), were generated. Gpr116(Δexon17) mice developed a profound accumulation of alveolar surfactant phospholipids at 4 weeks of age (12-fold) that was further increased at 20 weeks of age (30-fold). Surfactant accumulation in Gpr116(Δexon17) mice was associated with increased saturated phosphatidylcholine synthesis at 4 weeks and the presence of enlarged, lipid-laden macrophages, neutrophilia, and alveolar destruction at 20 weeks. mRNA microarray analyses indicated that P2RY2, a purinergic receptor known to mediate surfactant secretion, was induced in Gpr116(Δexon17) type II cells. Collectively, these data support the concept that GPR116 functions as a molecular sensor of alveolar surfactant lipid pool sizes by regulating surfactant secretion.
Collapse
Affiliation(s)
- James P Bridges
- Perinatal Institute, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Miklavc P, Thompson KE, Frick M. A new role for P2X4 receptors as modulators of lung surfactant secretion. Front Cell Neurosci 2013; 7:171. [PMID: 24115920 PMCID: PMC3792447 DOI: 10.3389/fncel.2013.00171] [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: 08/05/2013] [Accepted: 09/12/2013] [Indexed: 12/17/2022] Open
Abstract
In recent years, P2X receptors have attracted increasing attention as regulators of exocytosis and cellular secretion. In various cell types, P2X receptors have been found to stimulate vesicle exocytosis directly via Ca(2+) influx and elevation of the intracellular Ca(2+) concentration. Recently, a new role for P2X4 receptors as regulators of secretion emerged. Exocytosis of lamellar bodies (LBs), large storage organelles for lung surfactant, results in a local, fusion-activated Ca(2+) entry (FACE) in alveolar type II epithelial cells. FACE is mediated via P2X4 receptors that are located on the limiting membrane of LBs and inserted into the plasma membrane upon exocytosis of LBs. The localized Ca(2+) influx at the site of vesicle fusion promotes fusion pore expansion and facilitates surfactant release. In addition, this inward-rectifying cation current across P2X4 receptors mediates fluid resorption from lung alveoli. It is hypothesized that the concomitant reduction in the alveolar lining fluid facilitates insertion of surfactant into the air-liquid interphase thereby "activating" it. These findings constitute a novel role for P2X4 receptors in regulating vesicle content secretion as modulators of the secretory output during the exocytic post-fusion phase.
Collapse
Affiliation(s)
- Pika Miklavc
- Institute of General Physiology, University of Ulm Ulm, Germany
| | | | | |
Collapse
|
30
|
Fujisawa S, Romin Y, Barlas A, Petrovic LM, Turkekul M, Fan N, Xu K, Garcia AR, Monette S, Klimstra DS, Erinjeri JP, Solomon SB, Manova-Todorova K, Sofocleous CT. Evaluation of YO-PRO-1 as an early marker of apoptosis following radiofrequency ablation of colon cancer liver metastases. Cytotechnology 2013; 66:259-73. [PMID: 24065619 DOI: 10.1007/s10616-013-9565-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 04/10/2013] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Radiofrequency (RF) ablation (RFA) is a minimally invasive treatment for colorectal-cancer liver metastases (CLM) in selected nonsurgical patients. Unlike surgical resection, RFA is not followed by routine pathological examination of the target tumor and the surrounding liver tissue. The aim of this study was the evaluation of apoptotic events after RFA. Specifically, we evaluated YO-PRO-1 (YP1), a green fluorescent DNA marker for cells with compromised plasma membrane, as a potential, early marker of cell death. YP1 was applied on liver tissue adherent on the RF electrode used for CLM ablation, as well as on biopsy samples from the center and the margin of the ablation zone as depicted by dynamic CT immediately after RFA. Normal pig and mouse liver tissues were used for comparison. The same samples were also immunostained for fragmented DNA (TUNEL assay) and for active mitochondria (anti-OxPhos antibody). YP1 was also used simultaneously with propidium iodine (PI) to stain mouse liver and samples from ablated CLM. Following RFA of human CLM, more than 90 % of cells were positive for YP1. In nonablated, dissected pig and mouse liver however, we found similar YP1 signals (93.1 % and 65 %, respectively). In samples of intact mouse liver parenchyma, there was a significantly smaller proportion of YP1 positive cells (22.7 %). YP1 and PI staining was similar for ablated CLM. However in dissected normal mouse liver there was initial YP1 positivity and complete absence of the PI signal and only later there was PI signal. CONCLUSION This is the first time that YP1 was applied in liver parenchymal tissue (rather than cell culture). The results suggest that YP1 is a very sensitive marker of early cellular events reflecting an early and widespread plasma membrane injury that allows YP1 penetration into the cells.
Collapse
Affiliation(s)
- Sho Fujisawa
- Molecular Cytology Core Facility, Memorial Sloan-Kettering Cancer, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The distal airways are covered with a heterogeneous layer of cells known as the alveolar epithelium. Alveolar epithelial cells provide the major barrier between the airspace and fluid filled tissue compartments. As such, regulation of the alveolar epithelium is critical to maintain a healthy lung and for optimal gas exchange. In this chapter, we discuss functional roles for alveolar epithelial cells with particular emphasis on intercellular junctions and communication. As a thin layer of cells directly exposed to atmospheric oxygen, alveoli are particularly sensitive to oxidant insults. Alcohol significantly diminishes the normal antioxidant reserves of the alveolar epithelium, thereby rendering it sensitized for an exaggerated damage response to acute and chronic injuries. The effects of alcohol on alveolar epithelia are discussed along with open questions and potential therapeutic targets to prevent the pathophysiology of alcoholic lung disease.
Collapse
|
32
|
Mishra A. New insights of P2X7 receptor signaling pathway in alveolar functions. J Biomed Sci 2013; 20:26. [PMID: 23634990 PMCID: PMC3652760 DOI: 10.1186/1423-0127-20-26] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 04/29/2013] [Indexed: 12/20/2022] Open
Abstract
Purinergic P2X7 receptor (P2X7R), an ATP-gated cation channel, is unique among all other family members because of its ability to respond to various stimuli and to modulate pro-inflammatory signaling. The activation of P2X7R in immune cells is absolutely required for mature interleukin -1beta (IL-1beta) and IL-18 production and release. Lung alveoli are lined by the structural alveolar epithelial type I (AEC I) and alveolar epithelial type II cells (AEC II). AEC I plays important roles in alveolar barrier protection and fluid homeostasis whereas AEC II synthesizes and secrete surfactant and prevents alveoli from collapse. Earlier studies indicated that purinergic P2X7 receptors were specifically expressed in AEC I. However, their implication in alveolar functions has not been explored. This paper reviews two important signaling pathways of P2X7 receptors in surfactant homeostatsis and Acute Lung Injury (ALI). Thus, P2X7R resides at the critical nexus of alveolar pathophysiology.
Collapse
Affiliation(s)
- Amarjit Mishra
- National Institute of Health, 10 Center Dr, Bldg No, 10, Bethesda, MD 20892, USA.
| |
Collapse
|
33
|
Pulmonary surfactant preserves viability of alveolar type II cells exposed to polymyxin B in vitro. PLoS One 2013; 8:e62105. [PMID: 23620808 PMCID: PMC3631157 DOI: 10.1371/journal.pone.0062105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 03/18/2013] [Indexed: 01/23/2023] Open
Abstract
Background Exogenous surfactant derived from animal lungs is applied for treatment of surfactant deficiency. By means of its rapid spreading properties, it could transport pharmaceutical agents to the terminal air spaces. The antimicrobial peptide Polymyxin B (PxB) is used as a topical antibiotic for inhalation therapy. Whereas it has been shown that PxB mixed with surfactant is not inhibiting surface activity while antimicrobiotic activity is preserved, little is known concerning the effects on synthesis of endogenous surfactant in alveolar type II cells (ATIIC). Objective To investigate ATIIC viability and surfactant-exocytosis depending on PxB and/or surfactant exposure. Methods ATIIC were isolated from rat lungs as previously described and were cultivated for 48 h. After incubation for a period of 1–5 h with either PxB (0.05 or 0.1 mg/ml), modified porcine surfactant (5 or 10 mg/ml) or mixtures of both, viability and exocytosis (spontanously and after stimulation) were determined by fluorescence staining of intracellular surfactant. Results PxB 0.1 mg/ml, but not porcine surfactant or porcine surfactant plus PxB reduces ATIIC-viability. Only PxB alone, but not in combination with porcine surfactant, rapidly reduces fluorescence in ATIIC at maximum within 3 h, indicating stimulation of exocytosis. Subsequent ionomycin-stimulation does not further increase exocytosis of PxB incubated ATIIC. In presence of surfactant, stimulating effects of PxB and ionomycin on exocytosis are reduced. Conclusion PxB alone shows negative effects on ATIIC, which are counterbalanced in mixtures with surfactant. So far, our studies found no results discouraging the concept of a combined treatment with PxB and surfactant mixtures.
Collapse
|
34
|
Thompson KE, Korbmacher JP, Hecht E, Hobi N, Wittekindt OH, Dietl P, Kranz C, Frick M. Fusion-activated cation entry (FACE) via P2X₄ couples surfactant secretion and alveolar fluid transport. FASEB J 2013; 27:1772-83. [PMID: 23307836 DOI: 10.1096/fj.12-220533] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Two fundamental mechanisms within alveoli are essential for lung function: regulated fluid transport and secretion of surfactant. Surfactant is secreted via exocytosis of lamellar bodies (LBs) in alveolar type II (ATII) cells. We recently reported that LB exocytosis results in fusion-activated cation entry (FACE) via P2X₄ receptors on LBs. We propose that FACE, in addition to facilitating surfactant secretion, modulates alveolar fluid transport. Correlative fluorescence and atomic force microscopy revealed that FACE-dependent water influx correlated with individual fusion events in rat primary ATII cells. Moreover, ATII cell monolayers grown at air-liquid interface exhibited increases in short-circuit current (Isc) on stimulation with ATP or UTP. Both are potent agonists for LB exocytosis, but only ATP activates FACE. ATP, not UTP, elicited additional fusion-dependent increases in Isc. Overexpressing dominant-negative P2X₄ abrogated this effect by ∼50%, whereas potentiating P2X4 lead to ∼80% increase in Isc. Finally, we monitored changes in alveolar surface liquid (ASL) on ATII monolayers by confocal microscopy. Only stimulation with ATP, not UTP, led to a significant, fusion-dependent, 20% decrease in ASL, indicating apical-to-basolateral fluid transport across ATII monolayers. Our data support the first direct link between LB exocytosis, regulation of surfactant secretion, and transalveolar fluid resorption via FACE.
Collapse
|
35
|
Van Scott MR, Chandler J, Olmstead S, Brown JM, Mannie M. Airway Anatomy, Physiology, and Inflammation. THE TOXICANT INDUCTION OF IRRITANT ASTHMA, RHINITIS, AND RELATED CONDITIONS 2013. [PMCID: PMC7122617 DOI: 10.1007/978-1-4614-9044-9_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
36
|
Agassandian M, Mallampalli RK. Surfactant phospholipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:612-25. [PMID: 23026158 DOI: 10.1016/j.bbalip.2012.09.010] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 09/07/2012] [Accepted: 09/16/2012] [Indexed: 12/16/2022]
Abstract
Pulmonary surfactant is essential for life and is composed of a complex lipoprotein-like mixture that lines the inner surface of the lung to prevent alveolar collapse at the end of expiration. The molecular composition of surfactant depends on highly integrated and regulated processes involving its biosynthesis, remodeling, degradation, and intracellular trafficking. Despite its multicomponent composition, the study of surfactant phospholipid metabolism has focused on two predominant components, disaturated phosphatidylcholine that confers surface-tension lowering activities, and phosphatidylglycerol, recently implicated in innate immune defense. Future studies providing a better understanding of the molecular control and physiological relevance of minor surfactant lipid components are needed. This article is part of a Special Issue entitled Phospholipids and Phospholipid Metabolism.
Collapse
Affiliation(s)
- Marianna Agassandian
- Department of Medicine, Acute Lung Injury Center of Excellence, the University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
37
|
Burnstock G, Brouns I, Adriaensen D, Timmermans JP. Purinergic signaling in the airways. Pharmacol Rev 2012; 64:834-68. [PMID: 22885703 DOI: 10.1124/pr.111.005389] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Evidence for a significant role and impact of purinergic signaling in normal and diseased airways is now beyond dispute. The present review intends to provide the current state of knowledge of the involvement of purinergic pathways in the upper and lower airways and lungs, thereby differentiating the involvement of different tissues, such as the epithelial lining, immune cells, airway smooth muscle, vasculature, peripheral and central innervation, and neuroendocrine system. In addition to the vast number of well illustrated functions for purinergic signaling in the healthy respiratory tract, increasing data pointing to enhanced levels of ATP and/or adenosine in airway secretions of patients with airway damage and respiratory diseases corroborates the emerging view that purines act as clinically important mediators resulting in either proinflammatory or protective responses. Purinergic signaling has been implicated in lung injury and in the pathogenesis of a wide range of respiratory disorders and diseases, including asthma, chronic obstructive pulmonary disease, inflammation, cystic fibrosis, lung cancer, and pulmonary hypertension. These ostensibly enigmatic actions are based on widely different mechanisms, which are influenced by the cellular microenvironment, but especially the subtypes of purine receptors involved and the activity of distinct members of the ectonucleotidase family, the latter being potential protein targets for therapeutic implementation.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Royal Free Campus, London, UK.
| | | | | | | |
Collapse
|
38
|
Weng T, Mishra A, Guo Y, Wang Y, Su L, Huang C, Zhao C, Xiao X, Liu L. Regulation of lung surfactant secretion by microRNA-150. Biochem Biophys Res Commun 2012; 422:586-9. [PMID: 22595456 DOI: 10.1016/j.bbrc.2012.05.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 05/08/2012] [Indexed: 11/25/2022]
Abstract
P2X7 receptor (P2X7R) is a purinergic ion-channel receptor. We have previously shown that the activation of P2X7R in alveolar type I cells stimulates surfactant secretion in alveolar type II cells. In this study, we determined whether miR-150 regulates P2X7R-mediated surfactant secretion. The miR-150 expression level in alveolar type II cells was much higher than alveolar type I cells, which was inversely correlated with the P2X7R protein level. An adenovirus expressing miR-150 significantly reduced the P2X7R protein expression in E10 cells, an alveolar type I cell line. Furthermore, pre-treatment of E10 cells with the adenovirus reduced the surfactant secretion induced by E10 cell conditioned medium. Our study demonstrates that miR-150 regulates surfactant secretion through P2X7R.
Collapse
Affiliation(s)
- Tingting Weng
- Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dietl P, Haller T, Frick M. Spatio-temporal aspects, pathways and actions of Ca(2+) in surfactant secreting pulmonary alveolar type II pneumocytes. Cell Calcium 2012; 52:296-302. [PMID: 22591642 DOI: 10.1016/j.ceca.2012.04.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Revised: 04/11/2012] [Accepted: 04/18/2012] [Indexed: 01/16/2023]
Abstract
The type II cell of the pulmonary alveolus is a polarized epithelial cell that secretes surfactant into the alveolar space by regulated exocytosis of lamellar bodies (LBs). This process consists of multiple sequential steps and is correlated to elevations of the cytoplasmic Ca(2+) concentration ([Ca(2+)](c)) required for extended periods of secretory activity. Both chemical (purinergic) and mechanical (cell stretch or exposure to an air-liquid interface) stimuli give rise to complex Ca(2+) signals (such as Ca(2+) peaks, spikes and plateaus) that differ in shape, origin and spatio-temporal behavior. This review summarizes current knowledge about Ca(2+) channels, including vesicular P2X4 purinoceptors, in type II cells and associated signaling cascades within the alveolar microenvironment, and relates stimulus-dependent activation of these pathways with distinct stages of surfactant secretion, including pre- and postfusion stages of LB exocytosis.
Collapse
Affiliation(s)
- Paul Dietl
- Institute of General Physiology, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany.
| | | | | |
Collapse
|
40
|
|
41
|
Briva A, Santos C, Malacrida L, Rocchiccioli F, Soto J, Angulo M, Batthyany C, Cairoli E, Piriz H. Adenosine triphosphate-dependent calcium signaling during ventilator-induced lung injury is amplified by hypercapnia. Exp Lung Res 2011; 37:471-81. [PMID: 21870898 DOI: 10.3109/01902148.2011.598217] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Adenosine triphosphate (ATP) is released by alveolar epithelial cells during ventilator-induced lung injury (VILI) and regulates fluid transport across epithelia. High CO(2) levels are observed in patients with "permissive hypercapnia," which inhibits alveolar fluid reabsorption (AFR) in alveolar epithelial cells. The authors set out to determine whether VILI affects AFR and whether the purinergic pathway is modulated in cells exposed to hypercapnia. Control group was compared against VILI (tidal volume [Vt] = 35 mL/kg, zero positive end-expiratory pressure [PEEP]) and protective ventilation (Vt = 6 mL/kg, PEEP = 10 cm H(2)O) groups. Lung mechanics, histology, and AFR were evaluated. Alveolar epithelial cells (AECs) were loaded with Fura 2-AM to measure intracellular calcium in the presence ATP (10 μM) at 5% or 10% CO(2) as compared with baseline. High tidal volume ventilation impairs lung mechanics and AFR. Hypercapnia (HC) increases intracellular calcium levels in response to ATP stimulation. HC + ATP is the most detrimental combination decreasing AFR. Purinergic signaling in AECs is modulated by high CO(2) levels via increased cytosolic calcium. The authors reason that this modulation may play a role in the impairment of alveolar epithelial functions induced by hypercapnia.
Collapse
Affiliation(s)
- Arturo Briva
- Departamento de Fisiopatologia, Hospital de Clinicas, Montevideo, Uruguay.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Fusion-activated Ca2+ entry via vesicular P2X4 receptors promotes fusion pore opening and exocytotic content release in pneumocytes. Proc Natl Acad Sci U S A 2011; 108:14503-8. [PMID: 21844344 DOI: 10.1073/pnas.1101039108] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ca(2+) is considered a key element in multiple steps during regulated exocytosis. During the postfusion phase, an elevated cytoplasmic Ca(2+) concentration ([Ca(2+)])(c) leads to fusion pore dilation. In neurons and neuroendocrine cells, this results from activation of voltage-gated Ca(2+) channels in the plasma membrane. However, these channels are activated in the prefusion stage, and little is known about Ca(2+) entry mechanisms during the postfusion stage. This may be particularly important for slow and nonexcitable secretory cells. We recently described a "fusion-activated" Ca(2+) entry (FACE) mechanism in alveolar type II (ATII) epithelial cells. FACE follows initial fusion pore opening with a delay of 200-500 ms. The site, molecular mechanisms, and functions of this mechanism remain unknown, however. Here we show that vesicle-associated Ca(2+) channels mediate FACE. Using RT-PCR, Western blot analysis, and immunofluorescence, we demonstrate that P2X(4) receptors are expressed on exocytotic vesicles known as lamellar bodies (LBs). Electrophysiological, pharmacological, and genetic data confirm that FACE is mediated via these vesicular P2X(4) receptors. Furthermore, analysis of fluorophore diffusion into and out of individual vesicles after exocytotic fusion provides evidence that FACE regulates postfusion events of LB exocytosis via P2X(4). Fusion pore dilation was clearly correlated with the amplitude of FACE, and content release from fused LBs was accelerated in fusions followed by FACE. Based on these findings, we propose a model for regulation of the exocytotic postfusion phase in nonexcitable cells in which Ca(2+) influx via vesicular Ca(2+) channels regulates fusion pore expansion and vesicle content release.
Collapse
|