101
|
Tran HB, Macowan MG, Abdo A, Donnelley M, Parsons D, Hodge S. Enhanced inflammasome activation and reduced sphingosine-1 phosphate S1P signalling in a respiratory mucoobstructive disease model. JOURNAL OF INFLAMMATION-LONDON 2020; 17:16. [PMID: 32336954 PMCID: PMC7175514 DOI: 10.1186/s12950-020-00248-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/14/2020] [Indexed: 12/28/2022]
Abstract
Background Inflammasomes and sphingosine-1-phosphate (S1P) signalling are increasingly subject to intensive research in human diseases. We hypothesize that in respiratory muco-obstructive diseases, mucus obstruction enhances NLRP3 inflammasome activation and dysregulated S1P signalling. Methods Lung tissues from mice overexpressing the beta-unit of the epithelial sodium channel (βENaC) and their littermate controls were examined by histology, immunofluorescence and confocal microscopy, followed by ImageJ quantitative analysis. Results Lower airways in βENaC mice showed patchy patterns of mucus obstruction and neutrophil-dominant infiltrations. In contrast to a ubiquitous distribution of TNFα specks, significantly (p < 0.05) increased specks of bronchiolar NLRP3, IL-1β, and IgG in the βENaC mouse lungs were localized to the vicinity of mucus obstruction sites. Bright Spinster homologue 2 (SPNS2) at the epithelial apex and positive correlation with sphingosine kinase 1 (SPHK1) (R2 = 0.640; p < 0.001) supported the normal bronchial epithelium as an active generator of extracellular S1P. SPNS2 in βENaC mice was sharply reduced (38%, p < 0.05) and lost apical localization at sites of mucus obstruction. A significant (34%; p < 0.01) decrease in epithelial SPHK2 was also noted at mucus obstruction sites. Conclusion These results support that mucus obstruction may enhance NLRP3 inflammasome activation and dysregulated S1P signaling.
Collapse
Affiliation(s)
- Hai B Tran
- 1Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,2Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Matthew G Macowan
- 1Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,2Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Adrian Abdo
- 2Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Martin Donnelley
- 2Adelaide Medical School, University of Adelaide, Adelaide, Australia.,3Respiratory and Sleep Medicine, Women's and Children's Hospital, Adelaide, Australia.,4Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - David Parsons
- 2Adelaide Medical School, University of Adelaide, Adelaide, Australia.,3Respiratory and Sleep Medicine, Women's and Children's Hospital, Adelaide, Australia.,4Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Sandra Hodge
- 1Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, Australia.,2Adelaide Medical School, University of Adelaide, Adelaide, Australia
| |
Collapse
|
102
|
Cottrill KA, Farinha CM, McCarty NA. The bidirectional relationship between CFTR and lipids. Commun Biol 2020; 3:179. [PMID: 32313074 PMCID: PMC7170930 DOI: 10.1038/s42003-020-0909-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 03/23/2020] [Indexed: 02/08/2023] Open
Abstract
Cystic Fibrosis (CF) is the most common life-shortening genetic disease among Caucasians, resulting from mutations in the gene encoding the Cystic Fibrosis Transmembrane conductance Regulator (CFTR). While work to understand this protein has resulted in new treatment strategies, it is important to emphasize that CFTR exists within a complex lipid bilayer - a concept largely overlooked when performing structural and functional studies. In this review we discuss cellular lipid imbalances in CF, mechanisms by which lipids affect membrane protein activity, and the specific impact of detergents and lipids on CFTR function.
Collapse
Affiliation(s)
- Kirsten A Cottrill
- Molecular and Systems Pharmacology PhD Program, Emory University, Atlanta, GA, USA
| | - Carlos M Farinha
- Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Nael A McCarty
- Molecular and Systems Pharmacology PhD Program, Emory University, Atlanta, GA, USA.
- Department of Pediatrics and Children's Healthcare of Atlanta, Center for Cystic Fibrosis and Airways Disease Research, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
103
|
Age-Dependent Progression in Lung Pathophysiology can be Prevented by Restoring Fatty Acid and Ceramide Imbalance in Cystic Fibrosis. Lung 2020; 198:459-469. [DOI: 10.1007/s00408-020-00353-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/02/2020] [Indexed: 11/29/2022]
|
104
|
McElvaney OJ, Zaslona Z, Becker-Flegler K, Palsson-McDermott EM, Boland F, Gunaratnam C, Gulbins E, O'Neill LA, Reeves EP, McElvaney NG. Specific Inhibition of the NLRP3 Inflammasome as an Antiinflammatory Strategy in Cystic Fibrosis. Am J Respir Crit Care Med 2020; 200:1381-1391. [PMID: 31454256 DOI: 10.1164/rccm.201905-1013oc] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Rationale: Cystic fibrosis (CF) pulmonary disease is characterized by chronic infection with Pseudomonas aeruginosa and sustained neutrophil-dominant inflammation. The lack of effective antiinflammatory therapies for people with CF (PWCF) represents a significant challenge.Objectives: To identify altered immunometabolism in the CF neutrophil and investigate the feasibility of specific inhibition of the NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome as a CF antiinflammatory strategy in vivo.Methods: Key markers of increased aerobic glycolysis, known as a Warburg effect, including cytosolic PKM2 (pyruvate kinase M2), phosphorylated PKM2, succinate, HIF-1α (hypoxia-inducible factor-1α), lactate, and the IL-1β precursor pro-IL-1β, as well as caspase-1 activity and processing of pro-IL-1β to IL-1β by the NLRP3 inflammasome, were measured in neutrophils from blood and airway secretions from healthy control subjects (n = 12), PWCF (n = 16), and PWCF after double-lung transplantation (n = 6). The effects of specific inhibition of NLRP3 on airway inflammation and bacterial clearance in a murine CF model were subsequently assessed in vivo.Measurements and Main Results: CF neutrophils display increased aerobic glycolysis in the systemic circulation. This effect is driven by low-level endotoxemia, unaffected by CFTR (cystic fibrosis transmembrane conductance regulator) modulation, and resolves after transplant. The increased pro-IL-1β produced is processed to its mature active form in the LPS-rich CF lung by the NLRP3 inflammasome via caspase-1. Specific NLRP3 inhibition in vivo with MCC950 inhibited IL-1β in the lungs of CF mice (P < 0.0001), resulting in significantly reduced airway inflammation and improved Pseudomonas clearance (P < 0.0001).Conclusions: CF neutrophil immunometabolism is altered in response to inflammation. NLRP3 inflammasome inhibition may have an antiinflammatory and anti-infective role in CF.
Collapse
Affiliation(s)
- Oliver J McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, and.,Cystic Fibrosis Unit, Beaumont Hospital, Dublin, Ireland
| | - Zbigniew Zaslona
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; and
| | | | - Eva M Palsson-McDermott
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; and
| | - Fiona Boland
- Division of Biostatistics and Population Health Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Erich Gulbins
- Department of Molecular Biology, University Duisburg-Essen, Essen, Germany
| | - Luke A O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland; and
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Department of Medicine, and
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, and.,Cystic Fibrosis Unit, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
105
|
Comparative lipidomics of 5-Fluorouracil-sensitive and -resistant colorectal cancer cells reveals altered sphingomyelin and ceramide controlled by acid sphingomyelinase (SMPD1). Sci Rep 2020; 10:6124. [PMID: 32273521 PMCID: PMC7145850 DOI: 10.1038/s41598-020-62823-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/14/2020] [Indexed: 11/08/2022] Open
Abstract
5-Fluorouracil (5-FU) is a chemotherapeutic drug widely used to treat colorectal cancer. 5-FU is known to gradually lose its efficacy in treating colorectal cancer following the acquisition of resistance. We investigated the mechanism of 5-FU resistance using comprehensive lipidomic approaches. We performed lipidomic analysis on 5-FU–resistant (DLD-1/5-FU) and -sensitive (DLD-1) colorectal cancer cells using MALDI-MS and LC-MRM-MS. In particular, sphingomyelin (SM) species were significantly up-regulated in 5-FU–resistant cells in MALDI-TOF analysis. Further, we quantified sphingolipids including SM and Ceramide (Cer) using Multiple Reaction Monitoring (MRM), as they play a vital role in drug resistance. We found that 5-FU resistance in DLD-1/5-FU colorectal cancer cells was mainly associated with SM increase and Cer decrease, which are controlled by acid sphingomyelinase (SMPD1). In addition, reduction of SMPD1 expression was confirmed by LC-MRM-MS analysis and the effect of SMPD1 in drug resistance was assessed by treating DLD-1 cells with siRNA-SMPD1. Furthermore, clinical colorectal cancer data set analysis showed that down-regulation of SMPD1 was associated with resistance to chemotherapy regimens that include 5-FU. Thus, from our study, we propose that SM/Cer and SMPD1 are new potential target molecules for therapeutic strategies to overcome 5-FU resistance.
Collapse
|
106
|
Hadas Y, Vincek AS, Youssef E, Żak MM, Chepurko E, Sultana N, Sharkar MTK, Guo N, Komargodski R, Kurian AA, Kaur K, Magadum A, Fargnoli A, Katz MG, Hossain N, Kenigsberg E, Dubois NC, Schadt E, Hajjar R, Eliyahu E, Zangi L. Altering Sphingolipid Metabolism Attenuates Cell Death and Inflammatory Response After Myocardial Infarction. Circulation 2020; 141:916-930. [PMID: 31992066 PMCID: PMC7135928 DOI: 10.1161/circulationaha.119.041882] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 02/06/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Sphingolipids have recently emerged as a biomarker of recurrence and mortality after myocardial infarction (MI). The increased ceramide levels in mammalian heart tissues during acute MI, as demonstrated by several groups, is associated with higher cell death rates in the left ventricle and deteriorated cardiac function. Ceramidase, the only enzyme known to hydrolyze proapoptotic ceramide, generates sphingosine, which is then phosphorylated by sphingosine kinase to produce the prosurvival molecule sphingosine-1-phosphate. We hypothesized that Acid Ceramidase (AC) overexpression would counteract the negative effects of elevated ceramide and promote cell survival, thereby providing cardioprotection after MI. METHODS We performed transcriptomic, sphingolipid, and protein analyses to evaluate sphingolipid metabolism and signaling post-MI. We investigated the effect of altering ceramide metabolism through a loss (chemical inhibitors) or gain (modified mRNA [modRNA]) of AC function post hypoxia or MI. RESULTS We found that several genes involved in de novo ceramide synthesis were upregulated and that ceramide (C16, C20, C20:1, and C24) levels had significantly increased 24 hours after MI. AC inhibition after hypoxia or MI resulted in reduced AC activity and increased cell death. By contrast, enhancing AC activity via AC modRNA treatment increased cell survival after hypoxia or MI. AC modRNA-treated mice had significantly better heart function, longer survival, and smaller scar size than control mice 28 days post-MI. We attributed the improvement in heart function post-MI after AC modRNA delivery to decreased ceramide levels, lower cell death rates, and changes in the composition of the immune cell population in the left ventricle manifested by lowered abundance of proinflammatory detrimental neutrophils. CONCLUSIONS Our findings suggest that transiently altering sphingolipid metabolism through AC overexpression is sufficient and necessary to induce cardioprotection post-MI, thereby highlighting the therapeutic potential of AC modRNA in ischemic heart disease.
Collapse
Affiliation(s)
- Yoav Hadas
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam S. Vincek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elias Youssef
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Magdalena M. Żak
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elena Chepurko
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nishat Sultana
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mohammad Tofael Kabir Sharkar
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ningning Guo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rinat Komargodski
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ann Anu Kurian
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keerat Kaur
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ajit Magadum
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anthony Fargnoli
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michael G. Katz
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nadia Hossain
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ephraim Kenigsberg
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicole C. Dubois
- Department of Developmental and Regenerative Biology and The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Multiscale Biology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roger Hajjar
- Phospholamban Foundation, Amsterdam, Netherlands
| | - Efrat Eliyahu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Multiscale Biology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lior Zangi
- Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
107
|
Plasma Levels of the Bioactive Sphingolipid Metabolite S1P in Adult Cystic Fibrosis Patients: Potential Target for Immunonutrition? Nutrients 2020; 12:nu12030765. [PMID: 32183316 PMCID: PMC7146441 DOI: 10.3390/nu12030765] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 01/03/2023] Open
Abstract
Recent research has linked sphingolipid (SL) metabolism with cystic fibrosis transmembrane conductance regulator (CFTR) activity, affecting bioactive lipid mediator sphingosine-1-phosphate (S1P). We hypothesize that loss of CFTR function in cystic fibrosis (CF) patients influenced plasma S1P levels. Total and unbound plasma S1P levels were measured in 20 lung-transplanted adult CF patients and 20 healthy controls by mass spectrometry and enzyme-linked immunosorbent assay (ELISA). S1P levels were correlated with CFTR genotype, routine laboratory parameters, lung function and pathogen colonization, and clinical symptoms. Compared to controls, CF patients showed lower unbound plasma S1P, whereas total S1P levels did not differ. A positive correlation of total and unbound S1P levels was found in healthy controls, but not in CF patients. Higher unbound S1P levels were measured in ΔF508-homozygous compared to ΔF508-heterozygous CF patients (p = 0.038), accompanied by higher levels of HDL in ΔF508-heterozygous patients. Gastrointestinal symptoms were more common in ΔF508 heterozygotes compared to ΔF508 homozygotes. This is the first clinical study linking plasma S1P levels with CFTR function and clinical presentation in adult CF patients. Given the emerging role of immunonutrition in CF, our study might pave the way for using S1P as a novel biomarker and nutritional target in CF.
Collapse
|
108
|
Bal Topçu D, Tugcu G, Ozcan F, Aslan M, Yalcinkaya A, Polat SE, Hizal M, Yalcin EE, Ersoz DD, Ozcelik U, Kiper N, Lay I, Oztas Y. Plasma Ceramides and Sphingomyelins of Pediatric Patients Increase in Primary Ciliary Dyskinesia but Decrease in Cystic Fibrosis. Lipids 2020; 55:213-223. [PMID: 32120452 DOI: 10.1002/lipd.12230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/09/2020] [Accepted: 02/18/2020] [Indexed: 11/06/2022]
Abstract
We investigated plasma sphingomyelin (CerPCho) and ceramide (Cer) levels in pediatric patients with cystic fibrosis (CF) and primary ciliary dyskinesia (PCD). Plasma samples were obtained from CF (n = 19) and PCD (n = 7) patients at exacerbation, discharge, and stable periods. Healthy children (n = 17) of similar age served as control. Levels of 16-24 CerPCho and 16-24 Cer were measured by LC-MS/MS. Concentrations of all CerPCho and Cer species measured at exacerbation were significantly lower in patients with CF than PCD. 16, 18, 24 CerPCho, and 22, 24 Cer in exacerbation; 18, 24 CerPCho, and 18, 20, 22, 24 Cer at discharge; 18, 24 CerPCho and 24 Cer at stable period were significantly lower in CF patients than healthy children (p < 0.001 and p < 0.05). All CerPCho and Cer levels of PCD patients were significantly higher except 24 CerPCho and 24 Cer during exacerbation, 24 CerPCho at discharge, and 18, 22 CerPCho levels at stable period (p < 0.001 and p < 0.05) compared with healthy children. There was no significant difference among exacerbation, discharge, and stable periods in each group for Cer and CerPCho levels. This is the first study measuring plasma Cer and CerPCho levels in PCD and third study in CF patients. The dramatic difference in plasma levels of most CerPCho and Cer species found between two diseases suggest that cilia pathology in PCD and CFTR mutation in CF seem to alter sphingolipid metabolism possibly in opposite directions.
Collapse
Affiliation(s)
- Dilara Bal Topçu
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Gokcen Tugcu
- Department of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Filiz Ozcan
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya, Konyaaltı, 07070, Turkey
| | - Mutay Aslan
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya, Konyaaltı, 07070, Turkey
| | - Ahmet Yalcinkaya
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Sanem Eryilmaz Polat
- Department of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Mina Hizal
- Department of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Ebru Elmas Yalcin
- Department of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Deniz Dogru Ersoz
- Department of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Ugur Ozcelik
- Department of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Nural Kiper
- Department of Pediatric Pulmonology, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Incilay Lay
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| | - Yesim Oztas
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara, Sıhhıye, 06100, Turkey
| |
Collapse
|
109
|
Gupta G, Baumlin N, Poon J, Ahmed B, Chiang YP, Railwah C, Kim MD, Rivas M, Goldenberg H, Elgamal Z, Salathe M, Panwala AA, Dabo A, Huan C, Foronjy R, Jiang XC, Wadgaonkar R, Geraghty P. Airway Resistance Caused by Sphingomyelin Synthase 2 Insufficiency in Response to Cigarette Smoke. Am J Respir Cell Mol Biol 2020; 62:342-353. [PMID: 31517509 PMCID: PMC7055695 DOI: 10.1165/rcmb.2019-0133oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022] Open
Abstract
Sphingomyelin synthase is responsible for the production of sphingomyelin (SGM), the second most abundant phospholipid in mammalian plasma, from ceramide, a major sphingolipid. Knowledge of the effects of cigarette smoke on SGM production is limited. In the present study, we examined the effect of chronic cigarette smoke on sphingomyelin synthase (SGMS) activity and evaluated how the deficiency of Sgms2, one of the two isoforms of mammalian SGMS, impacts pulmonary function. Sgms2-knockout and wild-type control mice were exposed to cigarette smoke for 6 months, and pulmonary function testing was performed. SGMS2-dependent signaling was investigated in these mice and in human monocyte-derived macrophages of nonsmokers and human bronchial epithelial (HBE) cells isolated from healthy nonsmokers and subjects with chronic obstructive pulmonary disease (COPD). Chronic cigarette smoke reduces SGMS activity and Sgms2 gene expression in mouse lungs. Sgms2-deficient mice exhibited enhanced airway and tissue resistance after chronic cigarette smoke exposure, but had similar degrees of emphysema, compared with smoke-exposed wild-type mice. Sgms2-/- mice had greater AKT phosphorylation, peribronchial collagen deposition, and protease activity in their lungs after smoke inhalation. Similarly, we identified reduced SGMS2 expression and enhanced phosphorylation of AKT and protease production in HBE cells isolated from subjects with COPD. Selective inhibition of AKT activity or overexpression of SGMS2 reduced the production of several matrix metalloproteinases in HBE cells and monocyte-derived macrophages. Our study demonstrates that smoke-regulated Sgms2 gene expression influences key COPD features in mice, including airway resistance, AKT signaling, and protease production.
Collapse
Affiliation(s)
- Gayatri Gupta
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Nathalie Baumlin
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Justin Poon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Begum Ahmed
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | | | - Michael D. Kim
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Melissa Rivas
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Hannah Goldenberg
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Ziyad Elgamal
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Matthias Salathe
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Apurav A. Panwala
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | - Abdoulaye Dabo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell Biology, and
| | - Chongmin Huan
- Department of Cell Biology, and
- Department of Surgery, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Robert Foronjy
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell Biology, and
| | - Xian-Cheng Jiang
- Department of Cell Biology, and
- VA Medical Center, Brooklyn, New York
| | - Raj Wadgaonkar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell Biology, and
- VA Medical Center, Brooklyn, New York
| | - Patrick Geraghty
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Department of Cell Biology, and
| |
Collapse
|
110
|
Horati H, Janssens HM, Margaroli C, Veltman M, Stolarczyk M, Kilgore MB, Chou J, Peng L, Tiddens HAMW, Chandler JD, Tirouvanziam R, Scholte BJ. Airway profile of bioactive lipids predicts early progression of lung disease in cystic fibrosis. J Cyst Fibros 2020; 19:902-909. [PMID: 32057679 DOI: 10.1016/j.jcf.2020.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/20/2020] [Accepted: 01/26/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Previously, we showed that abnormal levels of bioactive lipids in bronchoalveolar lavage fluid (BALF) from infants with cystic fibrosis (CF) correlated with early structural lung damage. METHOD To extend these studies, BALF bioactive lipid measurement by mass spectrometry and chest computed tomography (CT, combined with the sensitive PRAGMA-CF scoring method) were performed longitudinally at 2-year intervals in a new cohort of CF children (n = 21, aged 1-5 yrs). RESULTS PRAGMA-CF, neutrophil elastase activity, and myeloperoxidase correlated with BALF lysolipids and isoprostanes, markers of oxidative stress, as well as prostaglandin E2 and combined ceramide precursors (Spearman's Rho > 0.5; P < 0.01 for all). Multiple protein agonists of inflammation and tissue remodeling, measured by Olink protein array, correlated positively (r = 0.44-0.79, p < 0.05) with PRAGMA-CF scores and bioactive lipid levels. Notably, levels of lysolipids, prostaglandin E2 and isoprostanes at first BALF predicted the evolution of PRAGMA-CF scores 2 years later. In wild-type differentiated primary bronchial epithelial cells, and in CFTR-inducible iCFBE cells, treatment with a lysolipid receptor agonist (VPC3114) enhanced shedding of pro-inflammatory and pro-fibrotic proteins. CONCLUSIONS Together, our findings suggest that bioactive lipids in BALF correlate with and possibly predict structural lung disease in CF children, which supports their use as biomarkers of disease progression and treatment efficacy. Furthermore, our data suggest a causative role of airway lysolipids and oxidative stress in the progression of early CF lung disease, unveiling potential therapeutic targets.
Collapse
Affiliation(s)
- Hamed Horati
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, the Netherlands
| | - Hettie M Janssens
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, the Netherlands
| | - Camilla Margaroli
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Mieke Veltman
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, the Netherlands; Department of Cell Biology, Erasmus MC, Rotterdam, the Netherlands
| | - Marta Stolarczyk
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Matthew B Kilgore
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Jeffrey Chou
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Limin Peng
- Department of Biostatistics and Bioinformatics, Emory University School of public Health, Atlanta, GA, USA
| | - Harm A M W Tiddens
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, the Netherlands
| | - Joshua D Chandler
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | - Bob J Scholte
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC-Sophia Children's Hospital, University Hospital Rotterdam, the Netherlands; Department of Cell Biology, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
111
|
Arterial Medial Calcification through Enhanced small Extracellular Vesicle Release in Smooth Muscle-Specific Asah1 Gene Knockout Mice. Sci Rep 2020; 10:1645. [PMID: 32015399 PMCID: PMC6997457 DOI: 10.1038/s41598-020-58568-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 01/17/2020] [Indexed: 11/09/2022] Open
Abstract
Arterial medial calcification (AMC) involves an increased small extracellular vesicle (sEV) secretion and apatite calcium precipitation in the arterial wall. The mechanisms mediating AMC remain poorly understood. In the present study, smooth muscle-specific acid ceramidase (Ac) gene knockout mice (Asah1fl/fl/SMCre) were used to demonstrate the role of lysosomal ceramide signaling pathway in AMC. Asah1fl/fl/SMCre mice were found to have more severe AMC in both aorta and coronary arteries compared to their littermates (Asah1fl/fl/SMwt and WT/WT mice) after receiving a high dose vitamin D. These mice also had pronounced upregulation of osteopontin and RUNX2 (osteogenic markers), CD63, AnX2 (sEV markers) and ALP expression (mineralization marker) in the arterial media. In cultured coronary arterial smooth muscle cells (CASMCs) from Asah1fl/fl/SMCre mice, high dose of Pi led to a significantly increased calcium deposition, phenotypic change and sEV secretion compared to WT CASMCs, which was associated with reduced lysosome-multivesicular body (MVB) interaction. Also, GW4869, sEV release inhibitor decreased sEV secretion and calcification in these cells. Lysosomal transient receptor potential mucolipin 1 (TRPML1) channels regulating lysosome interaction with MVBs were found remarkably inhibited in Asah1fl/fl/SMCre CASMCs as shown by GCaMP3 Ca2+ imaging and Port-a-Patch patch clamping of lysosomes. Lysosomal Ac in SMCs controls sEV release by regulating lysosomal TRPML1 channel activity and lysosome-MVB interaction, which importantly contributes to phenotypic transition and AMC.
Collapse
|
112
|
Lu Z, Li Y, Syn WK, Wang Z, Lopes-Virella MF, Lyons TJ, Huang Y. Amitriptyline inhibits nonalcoholic steatohepatitis and atherosclerosis induced by high-fat diet and LPS through modulation of sphingolipid metabolism. Am J Physiol Endocrinol Metab 2020; 318:E131-E144. [PMID: 31821039 PMCID: PMC7052581 DOI: 10.1152/ajpendo.00181.2019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We reported previously that increased acid sphingomyelinase (ASMase)-catalyzed hydrolysis of sphingomyelin, which leads to increases in ceramide and sphingosine 1 phosphate (S1P), played a key role in the synergistic upregulation of proinflammatory cytokines by palmitic acid (PA), a major saturated fatty acid, and lipopolysaccharide (LPS) in macrophages. Since macrophages are vital players in nonalcoholic steatohepatitis (NASH) and atherosclerosis, we assessed the effect of ASMase inhibition on NASH and atherosclerosis cooperatively induced by high-PA-containing high-fat diet (HP-HFD) and LPS in LDL receptor-deficient (LDLR-/-) mice. LDLR-/- mice were fed HP-HFD, injected with low dose of LPS and treated with or without the ASMase inhibitor amitriptyline. The neutral sphingomyelinase inhibitor GW4869 was used as control. Metabolic study showed that both amitriptyline and GW4869 reduced glucose, lipids, and insulin resistance. Histological analysis and Oil Red O staining showed that amitriptyline robustly reduced hepatic steatosis while GW4869 had modest effects. Interestingly, immunohistochemical study showed that amitriptyline, but not GW4869, strongly reduced hepatic inflammation. Furthermore, results showed that both amitriptyline and GW4869 attenuated atherosclerosis. To elucidate the underlying mechanisms whereby amitriptyline inhibited both NASH and atherosclerosis, but GW4869 only inhibited atherosclerosis, we found that amitriptyline, but not GW4869, downregulated proinflammatory cytokines in macrophages. Finally, we found that inhibition of sphingosine 1 phosphate production is a potential mechanism whereby amitriptyline inhibited proinflammatory cytokines. Collectively, this study showed that amitriptyline inhibited NASH and atherosclerosis through modulation of sphingolipid metabolism in LDLR-/- mice, indicating that sphingolipid metabolism in macrophages plays a crucial role in the linkage of NASH and atherosclerosis.
Collapse
Affiliation(s)
- Zhongyang Lu
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Yanchun Li
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Wing-Kin Syn
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
- Division of Gastroenterology and Hepatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, Euskal Herriko Unibertsitatea/Universidad del País Vasco, Leioa, Spain
| | - Zhewu Wang
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Maria F Lopes-Virella
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Timothy J Lyons
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Yan Huang
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
- Division of Endocrinology, Diabetes and Medical Genetics, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
113
|
Guzman G, Niekamp P, Tafesse FG. The Squeaky Yeast Gets Greased: The Roles of Host Lipids in the Clearance of Pathogenic Fungi. J Fungi (Basel) 2020; 6:E19. [PMID: 32024011 PMCID: PMC7151219 DOI: 10.3390/jof6010019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/21/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
Fungal infections remain a global health threat with high morbidity and mortality. The human immune system must, therefore, perpetually defend against invasive fungal infections. Phagocytosis is critical for the clearance of fungal pathogens, as this cellular process allows select immune cells to internalize and destroy invading fungal cells. While much is known about the protein players that enable phagocytosis, the various roles that lipids play during this fundamental innate immune process are still being illuminated. In this review, we describe recent discoveries that shed new light on the mechanisms by which host lipids enable the phagocytic uptake and clearance of fungal pathogens.
Collapse
Affiliation(s)
- Gaelen Guzman
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA; (G.G.); (P.N.)
| | - Patrick Niekamp
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA; (G.G.); (P.N.)
- Biology & Chemistry Department, University of Osnabrück, Fachbereich Biologie/Chemie, Barbarastrasse 13, 49076 Osnabrück, Germany
| | - Fikadu Geta Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA; (G.G.); (P.N.)
| |
Collapse
|
114
|
Infection of Human Tracheal Epithelial Cells by H5 Avian Influenza Virus Is Regulated by the Acid Stability of Hemagglutinin and the pH of Target Cell Endosomes. Viruses 2020; 12:v12010082. [PMID: 31936692 PMCID: PMC7019350 DOI: 10.3390/v12010082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/04/2020] [Accepted: 01/05/2020] [Indexed: 01/04/2023] Open
Abstract
Despite the possible relationships between tracheal infection and concomitant infection of the terminal part of the lower respiratory tract (bronchioles/alveoli), the behavior of avian influenza viruses (AIVs), such as H5N1, in the conducting airways is unclear. To examine the tropism of AIVs for cells lining the conducting airways of humans, we established human tracheal epithelial cell clones (HTEpC-Ts) and examined their susceptibility to infection by AIVs. The HTEpC-Ts showed differing susceptibility to H5N1 and non-zoonotic AIVs. Viral receptors expressed by HTEpC-Ts bound all viruses; however, the endosomal pH was associated with the overall susceptibility to infection by AIVs. Moreover, H5N1 hemagglutinin broadened viral tropism to include HTEpC-Ts, because it had a higher pH threshold for viral-cell membrane fusion. Thus, H5N1 viruses infect human tracheal epithelial cells as a result of their higher pH threshold for membrane fusion which may be one mechanism underlying H5N1 pathogenesis in human airway epithelia. Efficient replication of H5N1 in the conducting airways of humans may facilitate infection of the lower respiratory tract.
Collapse
|
115
|
Yeganeh B, Lee J, Bilodeau C, Lok I, Ermini L, Ackerley C, Caniggia I, Tibboel J, Kroon A, Post M. Acid Sphingomyelinase Inhibition Attenuates Cell Death in Mechanically Ventilated Newborn Rat Lung. Am J Respir Crit Care Med 2020; 199:760-772. [PMID: 30326731 DOI: 10.1164/rccm.201803-0583oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
RATIONALE Premature infants subjected to mechanical ventilation (MV) are prone to lung injury that may result in bronchopulmonary dysplasia. MV causes epithelial cell death and halts alveolar development. The exact mechanism of MV-induced epithelial cell death is unknown. OBJECTIVES To determine the contribution of autophagy to MV-induced epithelial cell death in newborn rat lungs. METHODS Newborn rat lungs and fetal rat lung epithelial (FRLE) cells were exposed to MV and cyclic stretch, respectively, and were then analyzed by immunoblotting and mass spectrometry for autophagy, apoptosis, and bioactive sphingolipids. MEASUREMENTS AND MAIN RESULTS Both MV and stretch first induce autophagy (ATG 5-12 [autophagy related 5-12] and LC3B-II [microtubule-associated proteins 1A/1B light chain 3B-II] formation) followed by extrinsic apoptosis (cleaved CASP8/3 [caspase-8/3] and PARP [poly(ADP-ribose) polymerase] formation). Stretch-induced apoptosis was attenuated by inhibiting autophagy. Coimmunoprecipitation revealed that stretch promoted an interaction between LC3B and the FAS (first apoptosis signal) cell death receptor in FRLE cells. Ceramide levels, in particular C16 ceramide, were rapidly elevated in response to ventilation and stretch, and C16 ceramide treatment of FRLE cells induced autophagy and apoptosis in a temporal pattern similar to that seen with MV and stretch. SMPD1 (sphingomyelin phosphodiesterase 1) was activated by ventilation and stretch, and its inhibition prevented ceramide production, LC3B-II formation, LC3B/first apoptosis signal interaction, caspase-3 activation, and, ultimately, FLRE cell death. SMPD1 inhibition also attenuated ventilation-induced autophagy and apoptosis in newborn rats. CONCLUSIONS Ventilation-induced ceramides promote autophagy-mediated cell death, and identifies SMPD1 as a potential therapeutic target for the treatment of ventilation-induced lung injury in newborns.
Collapse
Affiliation(s)
- Behzad Yeganeh
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joyce Lee
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,2 Institute of Medical Science and
| | - Claudia Bilodeau
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,3 Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Irene Lok
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Leonardo Ermini
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Cameron Ackerley
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Isabella Caniggia
- 4 Mount Sinai Hospital, the Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada; and
| | - Jeroen Tibboel
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,5 Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
| | - Andre Kroon
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,5 Department of Pediatrics, Erasmus MC-Sophia, Rotterdam, the Netherlands
| | - Martin Post
- 1 Translational Medicine Program, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada.,2 Institute of Medical Science and
| |
Collapse
|
116
|
Iron and Sphingolipids as Common Players of (Mal)Adaptation to Hypoxia in Pulmonary Diseases. Int J Mol Sci 2020; 21:ijms21010307. [PMID: 31906427 PMCID: PMC6981703 DOI: 10.3390/ijms21010307] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/24/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022] Open
Abstract
Hypoxia, or lack of oxygen, can occur in both physiological (high altitude) and pathological conditions (respiratory diseases). In this narrative review, we introduce high altitude pulmonary edema (HAPE), acute respiratory distress syndrome (ARDS), Chronic Obstructive Pulmonary Disease (COPD), and Cystic Fibrosis (CF) as examples of maladaptation to hypoxia, and highlight some of the potential mechanisms influencing the prognosis of the affected patients. Among the specific pathways modulated in response to hypoxia, iron metabolism has been widely explored in recent years. Recent evidence emphasizes hepcidin as highly involved in the compensatory response to hypoxia in healthy subjects. A less investigated field in the adaptation to hypoxia is the sphingolipid (SPL) metabolism, especially through Ceramide and sphingosine 1 phosphate. Both individually and in concert, iron and SPL are active players of the (mal)adaptation to physiological hypoxia, which can result in the pathological HAPE. Our aim is to identify some pathways and/or markers involved in the physiological adaptation to low atmospheric pressures (high altitudes) that could be involved in pathological adaptation to hypoxia as it occurs in pulmonary inflammatory diseases. Hepcidin, Cer, S1P, and their interplay in hypoxia are raising growing interest both as prognostic factors and therapeutical targets.
Collapse
|
117
|
Hou W, Chen Q, Wang H, Qiu P, Lyu X, Chen W, Chua MLK, Chinn YE, Deng CX, Wang R. The metabolic footprint during adipocyte commitment highlights ceramide modulation as an adequate approach for obesity treatment. EBioMedicine 2020; 51:102605. [PMID: 31901865 PMCID: PMC6940659 DOI: 10.1016/j.ebiom.2019.102605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 12/12/2019] [Accepted: 12/12/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Metabolic modulation is capable of maintaining cell potency, regulating niche homeostasis, or determining cell fate. However, little is known regarding the metabolic landscape during early adipogenesis or whether metabolic modulation could be a potential approach for obesity treatment. METHODS The metabolic footprint during adipocyte commitment was evaluated by metabolomics analysis in mouse embryonic fibroblasts (MEFs). The role of apoptosis induced by ceramide and how ceramide is regulated were evaluated by omics analysis in vitro, human database and the adipocyte-specific Sirt1 knockout mouse. FINDINGS The metabolic footprint showed that a complicated diversity of metabolism was enriched as early as 3 h and tended to fluctuate throughout differentiation. Subsequently, the scale of these perturbed metabolic patterns was reduced to reach a balanced state. Of high relevance is the presence of apoptosis induced by ceramide accumulation, which is associated with metabolic dynamics. Interestingly, apoptotic cells were not merely a byproduct of adipogenesis but rather promoted the release of lipid components to facilitate adipogenesis. Mechanistically, ceramide accumulation stemming from hydrolysis and the de novo pathway during early adipogenesis is regulated by Sirt1 upon epigenetic alterations of constitutive Histone H3K4 methylation and H3K9 acetylation. INTERPRETATION The metabolic footprint during adipocyte commitment highlights that apoptosis induced by ceramide is essential for adipogenesis, which is reversed by suppression of Sirt1. Therefore, Sirt1 may constitute a target to treat obesity or other ceramide-associated metabolic syndromes. FUNDING This project was supported by grants from the University of Macau (SRG2015-00008-FHS, MYRG2016-00054-FHS and MYRG2017-00096-FHS to RHW; CPG2019-00019-FHS to CXD) and from the National Natural Science Foundation of China (81672603 and 81401978) to QC.
Collapse
Affiliation(s)
- Weilong Hou
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Qiang Chen
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Haitao Wang
- Faculty of Health Sciences, University of Macau, Macau SAR, China; Division of Radiation Oncology, National Cancer Centre Singapore, Singapore; Division of Medical Sciences, National Cancer Centre Singapore, Singapore; Oncology Academic Programme, Duke-NUS Medical School, Singapore
| | - Pengxiang Qiu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Xueying Lyu
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Weiping Chen
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| | - Melvin L K Chua
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore; Division of Medical Sciences, National Cancer Centre Singapore, Singapore; Oncology Academic Programme, Duke-NUS Medical School, Singapore
| | - Y Eugene Chinn
- Institute of Biology and Medical Sciences, Soochow University School of Medicine, 199# Ren'ai Road, Suzhou Jiangsu 215123, China
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China.
| | - Ruihong Wang
- Faculty of Health Sciences, University of Macau, Macau SAR, China; Center for Cancer Research, Nation Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
118
|
Lights and Shadows in the Use of Mesenchymal Stem Cells in Lung Inflammation, a Poorly Investigated Topic in Cystic Fibrosis. Cells 2019; 9:cells9010020. [PMID: 31861724 PMCID: PMC7016730 DOI: 10.3390/cells9010020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent non-hematopoietic stem cells residing in many tissues, including the lung. MSCs have long been regarded as a promising tool for cell-based therapy because of their ability to replace damaged tissue by differentiating into the resident cell and repopulating the injured area. Their ability to release soluble factors and extracellular vesicles has emerged as crucial in the resolution of inflammation and injury. There is a growing literature on the use of MSCs and MSC secretome to hamper inflammation in different lung pathologies, including: asthma, pneumonia, acute lung injury (ALI), pulmonary hypertension, and chronic obstructive pulmonary disease (COPD). However, their potential therapeutic role in the context of Cystic Fibrosis (CF) lung inflammation is still not fully characterized. CF morbidity and mortality are mainly due to progressive lung dysfunction. Lung inflammation is a chronic and unresolved condition that triggers progressive tissue damage. Thus, it becomes even more important to develop innovative immunomodulatory therapies aside from classic anti-inflammatory agents. Here, we address the main features of CF and the implications in lung inflammation. We then review how MSCs and MSC secretome participate in attenuating inflammation in pulmonary pathologies, emphasizing the significant potential of MSCs as new therapeutic approach in CF.
Collapse
|
119
|
Lee SB, Han YR, Jeon HJ, Jun CH, Kim SK, Chin J, Lee SJ, Jeong M, Lee JE, Lee CH, Cho SJ, Kim DS, Jeon YH. Medical fluorophore 1 (MF1), a benzoquinolizinium-based fluorescent dye, as an inflammation imaging agent. J Mater Chem B 2019; 7:7326-7331. [PMID: 31681930 DOI: 10.1039/c9tb01266d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Structure-based targeting of fluorescent dyes is essential for their use as imaging agents for disease diagnosis. Here, we describe the development of the benzoquinolizinium compound Medical fluorophore 1 (MF1) as a novel biomedical imaging agent that allows the visualization of inflammation by virtue of its unique chemical structure. Lipopolysaccharide treatment stimulated the uptake of MF1 by bone marrow-derived macrophages, with no adverse effects on cell proliferation. In vivo fluorescence lifetime imaging revealed the accumulation of MF1 in carrageenan-induced acute inflammatory lesions in mice, which peaked at 6 h. MF1-based imaging also allowed monitoring of the response to the anti-inflammatory drugs dexamethasone and sulfasalazine. Thus, MF1 can be used to diagnose diseases characterized by inflammation as well as treatment efficacy.
Collapse
Affiliation(s)
- Sang Bong Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Ye Ri Han
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Hui-Jeon Jeon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Chul-Ho Jun
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea and Center for NanoMedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea and Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Sang-Kyoon Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Su-Jeong Lee
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Minseon Jeong
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Jae-Eon Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea. and Department of Biomaterials Science, College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Pusan, Republic of Korea
| | - Chang-Hee Lee
- Department of Chemistry, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea and Center for NanoMedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea and Yonsei-IBS Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Sung Jin Cho
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Dong-Su Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| | - Yong Hyun Jeon
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation, 80 Chembok-ro Dong-gu Daegu, Republic of Korea.
| |
Collapse
|
120
|
Beckmann N, Becker KA, Kadow S, Schumacher F, Kramer M, Kühn C, Schulz-Schaeffer WJ, Edwards MJ, Kleuser B, Gulbins E, Carpinteiro A. Acid Sphingomyelinase Deficiency Ameliorates Farber Disease. Int J Mol Sci 2019; 20:ijms20246253. [PMID: 31835809 PMCID: PMC6941101 DOI: 10.3390/ijms20246253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/03/2019] [Accepted: 12/07/2019] [Indexed: 12/20/2022] Open
Abstract
Farber disease is a rare lysosomal storage disorder resulting from acid ceramidase deficiency and subsequent ceramide accumulation. No treatments for Farber disease are clinically available, and affected patients have a severely shortened lifespan. We have recently reported a novel acid ceramidase deficiency model that mirrors the human disease closely. Acid sphingomyelinase is the enzyme that generates ceramide upstream of acid ceramidase in the lysosomes. Using our acid ceramidase deficiency model, we tested if acid sphingomyelinase could be a potential novel therapeutic target for the treatment of Farber disease. A number of functional acid sphingomyelinase inhibitors are clinically available and have been used for decades to treat major depression. Using these as a therapeutic for Farber disease, thus, has the potential to improve central nervous symptoms of the disease as well, something all other treatment options for Farber disease can’t achieve so far. As a proof-of-concept study, we first cross-bred acid ceramidase deficient mice with acid sphingomyelinase deficient mice in order to prevent ceramide accumulation. Double-deficient mice had reduced ceramide accumulation, fewer disease manifestations, and prolonged survival. We next targeted acid sphingomyelinase pharmacologically, to test if these findings would translate to a setting with clinical applicability. Surprisingly, the treatment of acid ceramidase deficient mice with the acid sphingomyelinase inhibitor amitriptyline was toxic to acid ceramidase deficient mice and killed them within a few days of treatment. In conclusion, our study provides the first proof-of-concept that acid sphingomyelinase could be a potential new therapeutic target for Farber disease to reduce disease manifestations and prolong survival. However, we also identified previously unknown toxicity of the functional acid sphingomyelinase inhibitor amitriptyline in the context of Farber disease, strongly cautioning against the use of this substance class for Farber disease patients.
Collapse
Affiliation(s)
- Nadine Beckmann
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | - Stephanie Kadow
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | - Fabian Schumacher
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany;
| | - Melanie Kramer
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | - Claudine Kühn
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | | | - Michael J. Edwards
- Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45229, USA;
| | - Burkhard Kleuser
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany;
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
- Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45229, USA;
| | - Alexander Carpinteiro
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
- Department of Hematology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
- Correspondence: ; Tel.: +49-201-723-84579; Fax: +49-201-723-5974
| |
Collapse
|
121
|
O'Brien TJ, Welch M. Recapitulation of polymicrobial communities associated with cystic fibrosis airway infections: a perspective. Future Microbiol 2019; 14:1437-1450. [PMID: 31778075 DOI: 10.2217/fmb-2019-0200] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The airways of persons with cystic fibrosis are prone to infection by a diverse and dynamic polymicrobial consortium. Currently, no models exist that permit recapitulation of this consortium within the laboratory. Such microbial ecosystems likely have a network of interspecies interactions, serving to modulate metabolic pathways and impact upon disease severity. The contribution of less abundant/fastidious microbial species on this cross-talk has often been neglected due to lack of experimental tractability. Here, we critically assess the existing models for studying polymicrobial infections. Particular attention is paid to 3Rs-compliant in vitro and in silico infection models, offering significant advantages over mammalian infection models. We outline why these models will likely become the 'go to' approaches when recapitulating polymicrobial cystic fibrosis infection.
Collapse
Affiliation(s)
- Thomas J O'Brien
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| |
Collapse
|
122
|
The role of endothelial cells in cystic fibrosis. J Cyst Fibros 2019; 18:752-761. [DOI: 10.1016/j.jcf.2019.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/18/2019] [Accepted: 07/23/2019] [Indexed: 12/22/2022]
|
123
|
Spengler D, Rintz N, Krause MF. An Unsettled Promise: The Newborn Piglet Model of Neonatal Acute Respiratory Distress Syndrome (NARDS). Physiologic Data and Systematic Review. Front Physiol 2019; 10:1345. [PMID: 31736777 PMCID: PMC6831728 DOI: 10.3389/fphys.2019.01345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022] Open
Abstract
Despite great advances in mechanical ventilation and surfactant administration for the newborn infant with life-threatening respiratory failure no specific therapies are currently established to tackle major pro-inflammatory pathways. The susceptibility of the newborn infant with neonatal acute respiratory distress syndrome (NARDS) to exogenous surfactant is linked with a suppression of most of the immunologic responses by the innate immune system, however, additional corticosteroids applied in any severe pediatric lung disease with inflammatory background do not reduce morbidity or mortality and may even cause harm. Thus, the neonatal piglet model of acute lung injury serves as an excellent model to study respiratory failure and is the preferred animal model for reasons of availability, body size, similarities of porcine and human lung, robustness, and costs. In addition, similarities to the human toll-like receptor 4, the existence of intraalveolar macrophages, the sensitivity to lipopolysaccharide, and the production of nitric oxide make the piglet indispensable in anti-inflammatory research. Here we present the physiologic and immunologic data of newborn piglets from three trials involving acute lung injury secondary to repeated airway lavage (and others), mechanical ventilation, and a specific anti-inflammatory intervention via the intratracheal route using surfactant as a carrier substance. The physiologic data from many organ systems of the newborn piglet—but with preference on the lung—are presented here differentiating between baseline data from the uninjured piglet, the impact of acute lung injury on various parameters (24 h), and the follow up data after 72 h of mechanical ventilation. Data from the control group and the intervention groups are listed separately or combined. A systematic review of the newborn piglet meconium aspiration model and the repeated airway lavage model is finally presented. While many studies assessed lung injury scores, leukocyte infiltration, and protein/cytokine concentrations in bronchoalveolar fluid, a systematic approach to tackle major upstream pro-inflammatory pathways of the innate immune system is still in the fledgling stages. For the sake of newborn infants with life-threatening NARDS the newborn piglet model still is an unsettled promise offering many options to conquer neonatal physiology/immunology and to establish potent treatment modalities.
Collapse
Affiliation(s)
- Dietmar Spengler
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Nele Rintz
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Martin F Krause
- Department of Pediatrics, Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
124
|
McElvaney OJ, Wade P, Murphy M, Reeves EP, McElvaney NG. Targeting airway inflammation in cystic fibrosis. Expert Rev Respir Med 2019; 13:1041-1055. [PMID: 31530195 DOI: 10.1080/17476348.2019.1666715] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: The major cause of morbidity and mortality in patients with cystic fibrosis (CF) is lung disease. Inflammation in the CF airways occurs from a young age and contributes significantly to disease progression and shortened life expectancy. Areas covered: In this review, we discuss the key immune cells involved in airway inflammation in CF, the contribution of the intrinsic genetic defect to the CF inflammatory phenotype, and anti-inflammatory strategies designed to overcome what is a critical factor in the pathogenesis of CF lung disease. Review of the literature was carried out using the MEDLINE (from 1975 to 2018), Google Scholar and The Cochrane Library databases. Expert opinion: Therapeutic interventions specifically targeting the defective CF transmembrane conductance regulator (CFTR) protein have changed the clinical landscape and significantly improved the outlook for CF. As survival estimates for people with CF increase, long-term management has become an important focus, with an increased need for therapies targeted at specific elements of inflammation, to complement CFTR modulator therapies.
Collapse
Affiliation(s)
- Oliver J McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Patricia Wade
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Mark Murphy
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| |
Collapse
|
125
|
Zulueta A, Peli V, Dei Cas M, Colombo M, Paroni R, Falleni M, Baisi A, Bollati V, Chiaramonte R, Del Favero E, Ghidoni R, Caretti A. Inflammatory role of extracellular sphingolipids in Cystic Fibrosis. Int J Biochem Cell Biol 2019; 116:105622. [PMID: 31563560 DOI: 10.1016/j.biocel.2019.105622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 09/24/2019] [Accepted: 09/25/2019] [Indexed: 12/13/2022]
Abstract
Ceramide is emerging as one of the players of inflammation in lung diseases. However, data on its inflammatory role in Cystic Fibrosis (CF) as part of the extracellular machinery driven by lung mesenchymal stem cells (MSCs)-derived extracellular vesicles (EVs) are missing. We obtained an in vitro model of CF-MSC by treating control human lung MSCs with a specific CFTR inhibitor. We characterized EVs populations derived from MSCs (ctr EVs) and CF-MSCs (CF-EVs) and analyzed their sphingolipid profile by LC-MS/MS. To evaluate their immunomodulatory function, we treated an in vitro human model of CF, with both EVs populations. Our data show that the two EVs populations differ for the average size, amount, and rate of uptake. CF-EVs display higher ceramide and dihydroceramide accumulation as compared to control EVs, suggesting the involvement of the de novo biosynthesis pathway in the parental CF-MSCs. Higher sphingomyelinase activity in CF-MSCs, driven by inflammation-induced ceramide accumulation, sustains the exocytosis of vesicles that export new formed pro-inflammatory ceramide. Our results suggest that CFTR dysfunction associates with an enhanced sphingolipid metabolism leading to the release of EVs that export the excess of pro-inflammatory Cer to the recipient cells, thus contributing to maintain the unresolved inflammatory status of CF.
Collapse
Affiliation(s)
- Aida Zulueta
- Biochemistry and Molecular Biology Lab., Health Sciences Department, University of Milan, Via A. di Rudinì, 8, Milan, Italy.
| | - Valeria Peli
- Biochemistry and Molecular Biology Lab., Health Sciences Department, University of Milan, Via A. di Rudinì, 8, Milan, Italy.
| | - Michele Dei Cas
- Biochemistry and Molecular Biology Lab., Health Sciences Department, University of Milan, Via A. di Rudinì, 8, Milan, Italy.
| | - Michela Colombo
- Laboratory of Experimental Medicine and Pathophysiology, Health Sciences Department, University of Milan, Via A. di Rudinì, 8, Milan, Italy; Haematopoietic Stem Cell Biology Laboratory, Medical Research Council(MRC) Weatherall Institute of Molecular Medicine (WIMM), University of Oxford, Oxford OX39DS, UK.
| | - Rita Paroni
- Biochemistry and Molecular Biology Lab., Health Sciences Department, University of Milan, Via A. di Rudinì, 8, Milan, Italy.
| | - Monica Falleni
- Pathology Division, Health Sciences Department, University of Milan, San Paolo Hospital Medical School, Via A. di Rudinì, 8, Milan, Italy.
| | - Alessandro Baisi
- Thoracic Surgery Unit, Health Sciences Department, University of Milan, San Paolo Hospital Medical School, Via A. di Rudinì, 8, Milan, Italy.
| | - Valentina Bollati
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Raffaella Chiaramonte
- Laboratory of Experimental Medicine and Pathophysiology, Health Sciences Department, University of Milan, Via A. di Rudinì, 8, Milan, Italy.
| | - Elena Del Favero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, Milan, Italy.
| | - Riccardo Ghidoni
- Biochemistry and Molecular Biology Lab., Health Sciences Department, University of Milan, Via A. di Rudinì, 8, Milan, Italy.
| | - Anna Caretti
- Biochemistry and Molecular Biology Lab., Health Sciences Department, University of Milan, Via A. di Rudinì, 8, Milan, Italy.
| |
Collapse
|
126
|
Ceramide Domains in Health and Disease: A Biophysical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1159:79-108. [DOI: 10.1007/978-3-030-21162-2_6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
127
|
Li G, Huang D, Hong J, Bhat OM, Yuan X, Li PL. Control of lysosomal TRPML1 channel activity and exosome release by acid ceramidase in mouse podocytes. Am J Physiol Cell Physiol 2019; 317:C481-C491. [PMID: 31268777 PMCID: PMC6766620 DOI: 10.1152/ajpcell.00150.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
The transient receptor potential mucolipin 1 (TRPML1) channel has been reported to mediate lysosomal Ca2+ release that is involved in Ca2+-dependent lysosome trafficking and autophagic flux. However, this regulatory mechanism of lysosomal TRPML1 channel activity in podocytes remains poorly understood. In the present study, we tested whether the TRPML1 channel in podocytes mediates lysosome trafficking, which is essential for multivesicular body (MVB) degradation by lysosomes. We first demonstrated the abundant expression of TRPML1 channel in podocytes. By GCaMP3 Ca2+ imaging, we characterized the lysosomal specificity of TRPML1 channel-mediated Ca2+ release in podocytes. Given the important role of acid ceramidase (AC) in lysosome function and podocyte injury, we tested whether AC regulates this TRPML1 channel-mediated Ca2+ release and consequent lysosome-dependent MVB degradation in podocytes. Pharmacologically, it was found that TRPML1 channel activity was remarkably attenuated by the AC inhibitor carmofur. Sphingosine, as an AC product, was demonstrated to induce TRPML1-mediated Ca2+ release, which was inhibited by a TRPML1 blocker, verapamil. Using a Port-a-Patch planar patch-clamp system, we found that AC-associated sphingolipids, sphingomyelin, ceramide, and sphingosine had different effects on TRPML1 channel activity in podocytes. Functionally, the inhibition of AC or blockade of TRPML1 channels was found to suppress the interaction of lysosomes and MVBs, leading to increased exosome release from podocytes. These results suggest that AC is critical for TRPML1 channel-mediated Ca2+ release, which controls lysosome-MVB interaction and exosome release in podocytes.
Collapse
Affiliation(s)
- Guangbi Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Dandan Huang
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jinni Hong
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Xinxu Yuan
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
128
|
Rolando M, Buchrieser C. A Comprehensive Review on the Manipulation of the Sphingolipid Pathway by Pathogenic Bacteria. Front Cell Dev Biol 2019; 7:168. [PMID: 31497599 PMCID: PMC6712060 DOI: 10.3389/fcell.2019.00168] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 08/06/2019] [Indexed: 12/31/2022] Open
Abstract
Bacterial pathogens have developed many different strategies to hijack host cell responses to promote their own survival. The manipulation of lipid biogenesis and cell membrane stability is emerging as a key player in bacterial host cell control. Indeed, many bacterial pathogens such as Legionella, Pseudomonas, Neisseria, Staphylococci, Mycobacteria, Helicobacter, or Clostridia are able to manipulate and use host sphingolipids during multiple steps of the infectious process. Sphingolipids have long been considered only as structural components of cell membranes, however, it is now well known that they are also intracellular and intercellular signaling molecules that play important roles in many eukaryotic cell functions as well as in orchestrating immune responses. Furthermore, they are important to eliminate invading pathogens and play a crucial role in infectious diseases. In this review, we focus on the different strategies employed by pathogenic bacteria to hijack the sphingolipid balance in the host cell to promote cellular colonization.
Collapse
Affiliation(s)
- Monica Rolando
- Biologie des Bactéries Intracellulaires, CNRS UMR 3525, Institut Pasteur, Paris, France
| | - Carmen Buchrieser
- Biologie des Bactéries Intracellulaires, CNRS UMR 3525, Institut Pasteur, Paris, France
| |
Collapse
|
129
|
Garić D, De Sanctis JB, Shah J, Dumut DC, Radzioch D. Biochemistry of very-long-chain and long-chain ceramides in cystic fibrosis and other diseases: The importance of side chain. Prog Lipid Res 2019:100998. [PMID: 31445070 DOI: 10.1016/j.plipres.2019.100998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/01/2019] [Accepted: 03/10/2019] [Indexed: 12/18/2022]
Abstract
Ceramides, the principal building blocks of all sphingolipids, have attracted the attention of many scientists around the world interested in developing treatments for cystic fibrosis, the most common genetic disease of Caucasians. Many years of fruitful research in this field have produced some fundamentally important, yet controversial results. Here, we aimed to summarize the current knowledge on the role of long- and very-long- chain ceramides, the most abundant species of ceramides in animal cells, in cystic fibrosis and other diseases. We also aim to explain the importance of the length of their side chain in the context of stability of transmembrane proteins through a concise synthesis of their biophysical chemistry, cell biology, and physiology. This review also addresses several remaining riddles in this field. Finally, we discuss the technical challenges associated with the analysis and quantification of ceramides. We provide the evaluation of the antibodies used for ceramide quantification and we demonstrate their lack of specificity. Results and discussion presented here will be of interest to anyone studying these enigmatic lipids.
Collapse
Affiliation(s)
- Dušan Garić
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Juan B De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Juhi Shah
- Department of Pharmacology and Experimental Therapeutics, McGill University, Montreal, QC, Canada
| | - Daciana Catalina Dumut
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Danuta Radzioch
- Department of Human Genetics, McGill University, Montreal, QC, Canada; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic; Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
130
|
Otopathogenic Staphylococcus aureus Invades Human Middle Ear Epithelial Cells Primarily through Cholesterol Dependent Pathway. Sci Rep 2019; 9:10777. [PMID: 31346200 PMCID: PMC6658548 DOI: 10.1038/s41598-019-47079-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 06/26/2019] [Indexed: 01/20/2023] Open
Abstract
Chronic suppurative otitis media (CSOM) is one of the most common infectious diseases of the middle ear especially affecting children, leading to delay in language development and communication. Although Staphylococcus aureus is the most common pathogen associated with CSOM, its interaction with middle ear epithelial cells is not well known. In the present study, we observed that otopathogenic S. aureus has the ability to invade human middle ear epithelial cells (HMEECs) in a dose and time dependent manner. Scanning electron microscopy demonstrated time dependent increase in the number of S. aureus on the surface of HMEECs. We observed that otopathogenic S. aureus primarily employs a cholesterol dependent pathway to colonize HMEECs. In agreement with these findings, confocal microscopy showed that S. aureus colocalized with lipid rafts in HMEECs. The results of the present study provide new insights into the pathogenesis of S. aureus induced CSOM. The availability of in vitro cell culture model will pave the way to develop novel effective treatment modalities for CSOM beyond antibiotic therapy.
Collapse
|
131
|
Neisseria meningitidis Type IV Pili Trigger Ca 2+-Dependent Lysosomal Trafficking of the Acid Sphingomyelinase To Enhance Surface Ceramide Levels. Infect Immun 2019; 87:IAI.00410-19. [PMID: 31160362 DOI: 10.1128/iai.00410-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 01/26/2023] Open
Abstract
Acid sphingomyelinase (ASM) is a lipid hydrolase that converts sphingomyelin to ceramide and that can be activated by various cellular stress mechanisms, including bacterial pathogens. Vesicle transportation or trafficking of ASM from the lysosomal compartment to the cell membrane is a prerequisite for its activation in response to bacterial infections; however, the effectors and mechanisms of ASM translocation and activation are poorly defined. Our recent work documented the key importance of ASM for Neisseria meningitidis uptake into human brain microvascular endothelial cells (HBMEC). We clearly identified OpcA to be one bacterial effector promoting ASM translocation and activity, though it became clear that additional bacterial components were involved, as up to 80% of ASM activity and ceramide generation was retained in cells infected with an opcA-deficient mutant. We hypothesized that N. meningitidis might use pilus components to promote the translocation of ASM into HBMEC. Indeed, we found that both live, piliated N. meningitidis and pilus-enriched fractions trigger transient ASM surface display, followed by the formation of ceramide-rich platforms (CRPs). By using indirect immunocytochemistry and direct stochastic optical reconstruction microscopy, we show that the overall number of CRPs with a size of ∼80 nm in the plasma membrane is significantly increased after exposure to pilus-enriched fractions. Infection with live bacteria as well as exposure to pilus-enriched fractions transiently increased cytosolic Ca2+ levels in HBMEC, and this was found to be important for ASM surface display mediated by lysosomal exocytosis, as depletion of cytosolic Ca2+ resulted in a significant decrease in ASM surface levels, ASM activity, and CRP formation.
Collapse
|
132
|
Wang J, Zhao H, Xu F, Zhang P, Zheng Y, Jia N. Human epididymis protein 4 (HE4) protects against cystic pulmonary fibrosis associated-inflammation through inhibition of NF-κB and MAPK singnaling. Genes Genomics 2019; 41:1045-1053. [PMID: 31165362 DOI: 10.1007/s13258-019-00836-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 05/27/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cystic pulmonary fibrosis (CF) affects mostly the lung of the newborns. Chronic infection and inflammation become the major causes of morbidity and mortality in CF. However, the underlying molecular mechanisms causing CF still remain unclear. METHODS ELISA assay was used to examine the expression of HE4 and pro-inflammatory cytokines in W126VA4 cells supernatant fluid. qRT-PCR was applicable to determine the mRNA level of HE4, α-SMA, collagen 1, MMP2, MMP9 and various interleukins. Immunofluorescent assay was used to test the expression of HE4 in WI-26 VA4 cells. Major elements of MAPK and NF-κB signals pathways were examined by western blot. RESULTS We found higher expression of HE4 in CF patients serum and lung biopsy. Interestingly, HE4 expression was positively correlated with fibrosis markers expression. In addition,HE4 overexpression increased inflammatory cytokines secretion and fibrosis markers expression in WI-26 VA4 cells. And NF-κB pathways were responsible for elevated inflammation. In addition, HE4/MAPK/MMPs signaling cascades destroyed the normal extracellular matrix (ECM) and promoted fibrosis. CONCLUSIONS Overall, we first identified that HE4 promoted CF-associated inflammation. Additionally, NF-κB and MAPK signalings were further validated to be responsible for CF-associated inflammation and ECM destruction. Characterization of lumacaftor/ivacaftor in CF-associated inflammation may provide a novel insight into clinical CF treatment.
Collapse
Affiliation(s)
- Jinli Wang
- Department of Pediatrics, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Hongyang Zhao
- Department of Pediatrics, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Fenfen Xu
- Department of Pediatrics, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Piaopiao Zhang
- Graduate School of Taishan Medical University, Tai'an, China
| | - Yuan Zheng
- Department of Pediatrics, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Nan Jia
- Department of Neonatal, The Second Affiliated Hospital of Xi'an Medical College, No. 167, Textile City East Street, Baqiao District, Xi'an, 710038, Shanxi, China.
| |
Collapse
|
133
|
Abu-Arish A, Pandžić E, Kim D, Tseng HW, Wiseman PW, Hanrahan JW. Agonists that stimulate secretion promote the recruitment of CFTR into membrane lipid microdomains. J Gen Physiol 2019; 151:834-849. [PMID: 31048413 PMCID: PMC6572005 DOI: 10.1085/jgp.201812143] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 04/05/2019] [Indexed: 01/20/2023] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a tightly regulated anion channel that mediates secretion by epithelia and is mutated in the disease cystic fibrosis. CFTR forms macromolecular complexes with many proteins; however, little is known regarding its associations with membrane lipids or the regulation of its distribution and mobility at the cell surface. We report here that secretagogues (agonists that stimulate secretion) such as the peptide hormone vasoactive intestinal peptide (VIP) and muscarinic agonist carbachol increase CFTR aggregation into cholesterol-dependent clusters, reduce CFTR lateral mobility within and between membrane microdomains, and trigger the fusion of clusters into large (3.0 µm2) ceramide-rich platforms. CFTR clusters are closely associated with motile cilia and with the enzyme acid sphingomyelinase (ASMase) that is constitutively bound on the cell surface. Platform induction is prevented by pretreating cells with cholesterol oxidase to disrupt lipid rafts or by exposure to the ASMase functional inhibitor amitriptyline or the membrane-impermeant reducing agent 2-mercaptoethanesulfonate. Platforms are reversible, and their induction does not lead to an increase in apoptosis; however, blocking platform formation does prevent the increase in CFTR surface expression that normally occurs during VIP stimulation. These results demonstrate that CFTR is colocalized with motile cilia and reveal surprisingly robust regulation of CFTR distribution and lateral mobility, most likely through autocrine redox activation of extracellular ASMase. Formation of ceramide-rich platforms containing CFTR enhances transepithelial secretion and likely has other functions related to inflammation and mucosal immunity.
Collapse
Affiliation(s)
- Asmahan Abu-Arish
- Department of Physiology, McGill University, Montréal, Canada
- Department of Physics, McGill University, Montréal, Canada
- Cystic Fibrosis Translational Research Centre, McGill University, Montréal, Canada
| | - Elvis Pandžić
- Department of Physics, McGill University, Montréal, Canada
| | - Dusik Kim
- Department of Physiology, McGill University, Montréal, Canada
- Cystic Fibrosis Translational Research Centre, McGill University, Montréal, Canada
| | - Hsin Wei Tseng
- Department of Physiology, McGill University, Montréal, Canada
| | - Paul W Wiseman
- Department of Physics, McGill University, Montréal, Canada
- Department of Chemistry, McGill University, Montréal, Canada
- Cystic Fibrosis Translational Research Centre, McGill University, Montréal, Canada
| | - John W Hanrahan
- Department of Physiology, McGill University, Montréal, Canada
- Cystic Fibrosis Translational Research Centre, McGill University, Montréal, Canada
- McGill University Health Centre Research Institute, Montréal, Canada
| |
Collapse
|
134
|
Simonis A, Schubert-Unkmeir A. The role of acid sphingomyelinase and modulation of sphingolipid metabolism in bacterial infection. Biol Chem 2019; 399:1135-1146. [PMID: 29924727 DOI: 10.1515/hsz-2018-0200] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/14/2018] [Indexed: 01/01/2023]
Abstract
Acid sphingomyelinase (ASM) is a key enzyme in sphingolipid metabolism that converts sphingomyelin to ceramide, thereby modulating membrane structures and signal transduction. Bacterial pathogens can manipulate ASM activity and function, and use host sphingolipids during multiple steps of their infection process. An increase in ceramides upon infection results in the formation of ceramide-enriched membrane platforms that serve to cluster receptor molecules and organize intracellular signaling molecules, thus facilitating bacterial uptake. In this review, we focus on how extracellular bacterial pathogens target ASM and modulate membrane properties and signaling pathways to gain entry into eukaryotic cells or induce cell death. We describe how intracellular pathogens interfere with the intralysosomal functions of ASM to favor replication and survival. In addition, bacteria utilize their own sphingomyelinases as virulence factors to modulate sphingolipid metabolism. The potential of ASM as a target for treating bacterial infections is also discussed.
Collapse
Affiliation(s)
- Alexander Simonis
- Division of Hematology, University Hospital Zurich, Rämistrasse 100, 8091 Zürich, Switzerland
| | - Alexandra Schubert-Unkmeir
- Institute of Hygiene and Microbiology, University of Würzburg, Josef-Schneider-Straße 2, D-97080 Würzburg, Germany
| |
Collapse
|
135
|
Keitsch S, Riethmüller J, Soddemann M, Sehl C, Wilker B, Edwards MJ, Caldwell CC, Fraunholz M, Gulbins E, Becker KA. Pulmonary infection of cystic fibrosis mice with Staphylococcus aureus requires expression of α-toxin. Biol Chem 2019; 399:1203-1213. [PMID: 29613852 DOI: 10.1515/hsz-2018-0161] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 03/28/2018] [Indexed: 01/25/2023]
Abstract
Pulmonary infections of cystic fibrosis (CF) patients with Staphylococcus aureus (S. aureus) occur very early in the disease. The molecular details that cause infection-susceptibility of CF patients to and mediate infection with S. aureus are poorly characterized. Therefore, we aimed to identify the role of α-toxin, a major S. aureus toxin, for pulmonary infection of CF mice. Infection with S. aureus JE2 resulted in severe pneumonia in CF mice, while wildtype mice were almost unaffected. Deficiency of α-toxin in JE2-Δhla reduced the pathogenicity of S. aureus in CF mice. However, CF mice were still more susceptible to the mutant S. aureus strain than wildtype mice. The S. aureus JE2 induced a marked increase of ceramide and a downregulation of sphingosine and acid ceramidase expression in bronchi of CF mice. Deletion of α-toxin reduced these changes after infection of CF mice. Similar changes were observed in wildtype mice, but at much lower levels. Our data indicate that expression of α-toxin is a major factor causing S. aureus infections in CF mice. Wildtype S. aureus induces a marked increase of ceramide and a reduction of sphingosine and acid ceramidase expression in bronchial epithelial cells of wildtype and CF mice, changes that determine infection susceptibility.
Collapse
Affiliation(s)
- Simone Keitsch
- Department of Molecular Biology, Medical School, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| | - Joachim Riethmüller
- Center for Pediatric Clinical Studies, Children's Clinic, University of Tübingen, Hoppe-Seyler-Str. 1, D-72076 Tübingen, Germany
| | - Matthias Soddemann
- Department of Molecular Biology, Medical School, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| | - Carolin Sehl
- Department of Molecular Biology, Medical School, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| | - Barbara Wilker
- Department of Molecular Biology, Medical School, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| | - Michael J Edwards
- Department of Surgery, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA
| | - Charles C Caldwell
- Department of Surgery, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA
| | - Martin Fraunholz
- Chair of Microbiology, University of Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | - Erich Gulbins
- Department of Molecular Biology, Medical School, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany.,Department of Surgery, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0558, USA
| | - Katrin Anne Becker
- Department of Molecular Biology, Medical School, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| |
Collapse
|
136
|
Beckmann N, Kadow S, Schumacher F, Göthert JR, Kesper S, Draeger A, Schulz-Schaeffer WJ, Wang J, Becker JU, Kramer M, Kühn C, Kleuser B, Becker KA, Gulbins E, Carpinteiro A. Pathological manifestations of Farber disease in a new mouse model. Biol Chem 2019; 399:1183-1202. [PMID: 29908121 DOI: 10.1515/hsz-2018-0170] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/07/2018] [Indexed: 11/15/2022]
Abstract
Farber disease (FD) is a rare lysosomal storage disorder resulting from acid ceramidase deficiency and subsequent ceramide accumulation. No treatments are clinically available and affected patients have a severely shortened lifespan. Due to the low incidence, the pathogenesis of FD is still poorly understood. Here, we report a novel acid ceramidase mutant mouse model that enables the study of pathogenic mechanisms of FD and ceramide accumulation. Asah1tmEx1 mice were generated by deletion of the acid ceramidase signal peptide sequence. The effects on lysosomal targeting and activity of the enzyme were assessed. Ceramide and sphingomyelin levels were quantified by liquid chromatography tandem-mass spectrometry (LC-MS/MS) and disease manifestations in several organ systems were analyzed by histology and biochemistry. We show that deletion of the signal peptide sequence disrupts lysosomal targeting and enzyme activity, resulting in ceramide and sphingomyelin accumulation. The affected mice fail to thrive and die early. Histiocytic infiltrations were observed in many tissues, as well as lung inflammation, liver fibrosis, muscular disease manifestations and mild kidney injury. Our new mouse model mirrors human FD and thus offers further insights into the pathogenesis of this disease. In the future, it may also facilitate the development of urgently needed therapies.
Collapse
Affiliation(s)
- Nadine Beckmann
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Stephanie Kadow
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Fabian Schumacher
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany.,Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | - Joachim R Göthert
- Department of Hematology, University Hospital Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Stefanie Kesper
- Department of Hematology, University Hospital Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Annette Draeger
- Institute of Anatomy, University of Bern, Baltzerstr. 2, CH-3012 Bern, Switzerland
| | - Walter J Schulz-Schaeffer
- Insitute of Neuropathology, University of the Saarland, Kirrberger Str. 100, D-66421 Homburg, Germany
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, UC Health University Hospital, 234 Goodman Street, Cincinnati, OH 45219, USA
| | - Jan U Becker
- Institute of Pathology, University Hospital Cologne, Kerpener Straße 62, D-50937 Cologne, Germany
| | - Melanie Kramer
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Claudine Kühn
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Burkhard Kleuser
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany.,Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45229, USA
| | - Alexander Carpinteiro
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany.,Department of Hematology, University Hospital Essen, Hufelandstraße 55, D-45147 Essen, Germany
| |
Collapse
|
137
|
Wu Y, Gulbins E, Grassmé H. The function of sphingomyelinases in mycobacterial infections. Biol Chem 2019; 399:1125-1133. [PMID: 29924725 DOI: 10.1515/hsz-2018-0179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/06/2018] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, is one of the deadliest and most important infectious diseases worldwide. The sphingomyelinase/ceramide system, which has been shown several times to be a crucial factor in the internalization, processing and killing of diverse pathogens, also modulates the pro-inflammatory response and the state of mycobacteria in macrophages. Both acid and neutral sphingomyelinases are important in this activity. However, studies of the role of sphingomyelinases in TB are still at an early stage.
Collapse
Affiliation(s)
- Yuqing Wu
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany.,Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH 45267, USA
| | - Heike Grassmé
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstrasse 55, D-45122 Essen, Germany
| |
Collapse
|
138
|
Scholte BJ, Horati H, Veltman M, Vreeken RJ, Garratt LW, Tiddens HAWM, Janssens HM, Stick SM. Oxidative stress and abnormal bioactive lipids in early cystic fibrosis lung disease. J Cyst Fibros 2019; 18:781-789. [PMID: 31031161 DOI: 10.1016/j.jcf.2019.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 10/26/2022]
Abstract
BACKGROUND Clinical data indicate that airway inflammation in children with cystic fibrosis (CF) arises early, is associated with structural lung damage, and predicts progression. In bronchoalveolar lavage fluid (BALF) from CFTR mutant mice, several aspects of lipid metabolism are abnormal that contributes to lung disease. We aimed to determine whether lipid pathway dysregulation is also observed in BALF from children with CF, to identify biomarkers of early lung disease and potential therapeutic targets. METHODS A comprehensive panel of lipids that included Sphingolipids, oxylipins, isoprostanes and lysolipids, all bioactive lipid species known to be involved in inflammation and tissue remodeling, were measured in BALF from children with CF (1-6 years, N = 33) and age-matched non-CF patients with unexplained inflammatory disease (N = 16) by HPLC-MS/MS. Lipid data were correlated with chest CT scores and BALF inflammation biomarkers. RESULTS The ratio of long chain to very long chain ceramide species (LCC/VLCC) and lysolipid levels were enhanced in CF compared to non-CF patients, despite comparable neutrophil counts and bacterial load. In CF patients both LCC/VLCC and lysolipid levels correlated with inflammation and chest CT scores. The ceramide precursors Sphingosine, Sphinganine, Sphingomyelin, correlated with inflammation, whilst the oxidative stress marker isoprostane correlated with inflammation and chest CT scores. No correlation between lipids and current bacterial infection in CF (N = 5) was observed. CONCLUSIONS Several lipid biomarkers of early CF lung disease were identified, which point toward potential disease monitoring and therapeutic approaches that can be used to complement CFTR modulators.
Collapse
Affiliation(s)
- Bob J Scholte
- Erasmus MC, Rotterdam, the Netherlands, Cell Biology; Erasmus MC, Sophia Children Hospital, Pediatric Pulmonology, the Netherlands.
| | - Hamed Horati
- Erasmus MC, Sophia Children Hospital, Pediatric Pulmonology, the Netherlands
| | - Mieke Veltman
- Erasmus MC, Rotterdam, the Netherlands, Cell Biology; Erasmus MC, Sophia Children Hospital, Pediatric Pulmonology, the Netherlands
| | - Rob J Vreeken
- Netherlands Metabolomics Centre, LACDR, Leiden, the Netherlands
| | - Luke W Garratt
- Telethon Kids Institute, University of Western Australia, Subiaco, 6008, Western Australia, Australia
| | - Harm A W M Tiddens
- Erasmus MC, Sophia Children Hospital, Pediatric Pulmonology, the Netherlands
| | - Hettie M Janssens
- Erasmus MC, Sophia Children Hospital, Pediatric Pulmonology, the Netherlands
| | - Stephen M Stick
- Telethon Kids Institute, University of Western Australia, Subiaco, 6008, Western Australia, Australia; Division of Paediatrics and Child Health, University of Western Australia, Nedlands, 6009, Western Australia, Australia; Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, 6009, Western Australia, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, University of Western Australia, Nedlands, 6009, Western Australia, Australia
| | | |
Collapse
|
139
|
Cockburn CL, Green RS, Damle SR, Martin RK, Ghahrai NN, Colonne PM, Fullerton MS, Conrad DH, Chalfant CE, Voth DE, Rucks EA, Gilk SD, Carlyon JA. Functional inhibition of acid sphingomyelinase disrupts infection by intracellular bacterial pathogens. Life Sci Alliance 2019; 2:e201800292. [PMID: 30902833 PMCID: PMC6431796 DOI: 10.26508/lsa.201800292] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022] Open
Abstract
Intracellular bacteria that live in host cell-derived vacuoles are significant causes of human disease. Parasitism of low-density lipoprotein (LDL) cholesterol is essential for many vacuole-adapted bacteria. Acid sphingomyelinase (ASM) influences LDL cholesterol egress from the lysosome. Using functional inhibitors of ASM (FIASMAs), we show that ASM activity is key for infection cycles of vacuole-adapted bacteria that target cholesterol trafficking-Anaplasma phagocytophilum, Coxiella burnetii, Chlamydia trachomatis, and Chlamydia pneumoniae. Vacuole maturation, replication, and infectious progeny generation by A. phagocytophilum, which exclusively hijacks LDL cholesterol, are halted and C. burnetii, for which lysosomal cholesterol accumulation is bactericidal, is killed by FIASMAs. Infection cycles of Chlamydiae, which hijack LDL cholesterol and other lipid sources, are suppressed but less so than A. phagocytophilum or C. burnetii A. phagocytophilum fails to productively infect ASM-/- or FIASMA-treated mice. These findings establish the importance of ASM for infection by intracellular bacteria and identify FIASMAs as potential host-directed therapies for diseases caused by pathogens that manipulate LDL cholesterol.
Collapse
Affiliation(s)
- Chelsea L Cockburn
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Ryan S Green
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Sheela R Damle
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Rebecca K Martin
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Naomi N Ghahrai
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Punsiri M Colonne
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Marissa S Fullerton
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Daniel H Conrad
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Daniel E Voth
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Elizabeth A Rucks
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Stacey D Gilk
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| |
Collapse
|
140
|
Warris A, Bercusson A, Armstrong-James D. Aspergillus colonization and antifungal immunity in cystic fibrosis patients. Med Mycol 2019; 57:S118-S126. [PMID: 30816976 DOI: 10.1093/mmy/myy074] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/20/2018] [Accepted: 07/26/2018] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF), caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, is the most common inherited life-limiting disease in North European people affecting 90,000 people worldwide. Progressive lung damage caused by recurrent infection and chronic airway inflammation is the major determinant of survival with a median age at death of 29 years. Approximately 60% of CF patients are infected with Aspergillus fumigatus, a ubiquitous environmental fungus, and its presence has been associated with accelerated lung function decline. Half of the patients infected with Aspergillus are <18 years of age. Yet time of acquisition of this fungus and determinants of CF-related Aspergillus disease severity and progression are not known. CFTR expression has been demonstrated in cells of the innate and adaptive immune system and has shown to be critical for normal function. Research delineating the role of CFTR-deficient phagocytes in Aspergillus persistence and infection in the CF lung, has only recently received attention. In this concise review we aim to present the current understanding with respect to when people with CF acquire infection with A. fumigatus and antifungal immune responses by CF immune cells.
Collapse
Affiliation(s)
- Adilia Warris
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, University of Aberdeen, United Kingdom
| | - Amelia Bercusson
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | | |
Collapse
|
141
|
Nambiar S, Bong How S, Gummer J, Trengove R, Moodley Y. Metabolomics in chronic lung diseases. Respirology 2019; 25:139-148. [PMID: 30907495 DOI: 10.1111/resp.13530] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/31/2019] [Accepted: 02/25/2019] [Indexed: 12/11/2022]
Abstract
Chronic lung diseases represent a significant global burden. Their increasing incidence and complexity render a comprehensive, multidisciplinary and personalized approach to each patient, critically important. Most recently, unique biochemical pathways and disease markers have been identified through large-scale metabolomic studies. Metabolomics is the study of metabolic pathways and the measurement of unique biomolecules in a living system. Analysing samples from different compartments such as bronchoalveolar lavage fluid (BALF) and plasma has proven useful for the characterization of a number of pathological conditions and offers promise as a clinical tool. For example, several studies using mass spectrometry (MS) have shown alterations in the sphingolipid metabolism of chronic obstructive pulmonary disease (COPD) sufferers. In this article, we present a practical review of the application of metabolomics to the study of chronic lung diseases (CLD): COPD, idiopathic pulmonary fibrosis (IPF) and asthma. The insights, which the analytical strategies employed in metabolomics, have provided to the dissection of the biochemistry of CLD and future clinical biomarkers are explored.
Collapse
Affiliation(s)
- Shabarinath Nambiar
- Separation Science and Metabolomics Laboratory, Murdoch University, Perth, WA, Australia
| | - Sze Bong How
- Separation Science and Metabolomics Laboratory, Murdoch University, Perth, WA, Australia.,Metabolomics Australia, Murdoch University, Perth, WA, Australia
| | - Joel Gummer
- Separation Science and Metabolomics Laboratory, Murdoch University, Perth, WA, Australia.,Metabolomics Australia, Murdoch University, Perth, WA, Australia
| | - Robert Trengove
- Separation Science and Metabolomics Laboratory, Murdoch University, Perth, WA, Australia.,Metabolomics Australia, Murdoch University, Perth, WA, Australia
| | - Yuben Moodley
- School of Medicine, University of Western Australia, Perth, WA, Australia.,Department of Respiratory Medicine, Fiona Stanley Hospital, Perth, WA, Australia.,Institute of Respiratory Health, Sir Charles Gairdner Hospital, Perth, WA, Australia
| |
Collapse
|
142
|
Garić D, De Sanctis JB, Shah J, Dumut DC, Radzioch D. Biochemistry of very-long-chain and long-chain ceramides in cystic fibrosis and other diseases: The importance of side chain. Prog Lipid Res 2019; 74:130-144. [PMID: 30876862 DOI: 10.1016/j.plipres.2019.03.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/01/2019] [Accepted: 03/10/2019] [Indexed: 12/19/2022]
Abstract
Ceramides, the principal building blocks of all sphingolipids, have attracted the attention of many scientists around the world interested in developing treatments for cystic fibrosis, the most common genetic disease of Caucasians. Many years of fruitful research in this field have produced some fundamentally important, yet controversial results. Here, we aimed to summarize the current knowledge on the role of long- and very-long- chain ceramides, the most abundant species of ceramides in animal cells, in cystic fibrosis and other diseases. We also aim to explain the importance of the length of their side chain in the context of stability of transmembrane proteins through a concise synthesis of their biophysical chemistry, cell biology, and physiology. This review also addresses several remaining riddles in this field. Finally, we discuss the technical challenges associated with the analysis and quantification of ceramides. We provide the evaluation of the antibodies used for ceramide quantification and we demonstrate their lack of specificity. Results and discussion presented here will be of interest to anyone studying these enigmatic lipids.
Collapse
Affiliation(s)
- Dušan Garić
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Juan B De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Juhi Shah
- Department of Pharmacology and Experimental Therapeutics, McGill University, Montreal, QC, Canada
| | - Daciana Catalina Dumut
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Danuta Radzioch
- Department of Human Genetics, McGill University, Montreal, QC, Canada; Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic; Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
143
|
Bezzerri V, Piacenza F, Caporelli N, Malavolta M, Provinciali M, Cipolli M. Is cellular senescence involved in cystic fibrosis? Respir Res 2019; 20:32. [PMID: 30764828 PMCID: PMC6376730 DOI: 10.1186/s12931-019-0993-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/31/2019] [Indexed: 02/06/2023] Open
Abstract
Pulmonary disease is the main cause of the morbidity and mortality of patients affected by cystic fibrosis (CF). The lung pathology is dominated by excessive recruitment of neutrophils followed by an exaggerated inflammatory process that has also been reported to occur in the absence of apparent pathogenic infections. Airway surface dehydration and mucus accumulation are the driving forces of this process. The continuous release of reactive oxygen species and proteases by neutrophils contributes to tissue damage, which eventually leads to respiratory insufficiency. CF has been considered a paediatric problem for several decades. Nevertheless, during the last 40 years, therapeutic options for CF have been greatly improved, turning CF into a chronic disease and extending the life expectancy of patients. Unfortunately, chronic inflammatory processes, which are characterized by a substantial release of cytokines and chemokines, along with ROS and proteases, can accelerate cellular senescence, leading to further complications in adulthood. The alterations and mechanisms downstream of CFTR functional defects that can stimulate cellular senescence remain unclear. However, while there are correlative data suggesting that cellular senescence may be implicated in CF, a causal or consequential relationship between cellular senescence and CF is still far from being established. Senescence can be both beneficial and detrimental. Senescence may suppress bacterial infections and cooperate with tissue repair. Additionally, it may act as an effective anticancer mechanism. However, it may also promote a pro-inflammatory environment, thereby damaging tissues and leading to chronic age-related diseases. In this review, we present the most current knowledge on cellular senescence and contextualize its possible involvement in CF.
Collapse
Affiliation(s)
- Valentino Bezzerri
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Nicole Caporelli
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy
| | - Marco Malavolta
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Mauro Provinciali
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, 60121, Ancona, Italy
| | - Marco Cipolli
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Ospedali Riuniti, 60121, Ancona, Italy.
| |
Collapse
|
144
|
Bodas M, Vij N. Adapting Proteostasis and Autophagy for Controlling the Pathogenesis of Cystic Fibrosis Lung Disease. Front Pharmacol 2019; 10:20. [PMID: 30774592 PMCID: PMC6367269 DOI: 10.3389/fphar.2019.00020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/09/2019] [Indexed: 12/20/2022] Open
Abstract
Cystic fibrosis (CF), a fatal genetic disorder predominant in the Caucasian population, is caused by mutations in the cystic fibrosis transmembrane conductance regulator (Cftr) gene. The most common mutation is the deletion of phenylalanine from the position-508 (F508del-CFTR), resulting in a misfolded-CFTR protein, which is unable to fold, traffic and retain its plasma membrane (PM) localization. The resulting CFTR dysfunction, dysregulates variety of key cellular mechanisms such as chloride ion transport, airway surface liquid (ASL) homeostasis, mucociliary-clearance, inflammatory-oxidative signaling, and proteostasis that includes ubiquitin-proteasome system (UPS) and autophagy. A collective dysregulation of these key homoeostatic mechanisms contributes to the development of chronic obstructive cystic fibrosis lung disease, instead of the classical belief focused exclusively on ion-transport defect. Hence, therapeutic intervention(s) aimed at rescuing chronic CF lung disease needs to correct underlying defect that mediates homeostatic dysfunctions and not just chloride ion transport. Since targeting all the myriad defects individually could be quite challenging, it will be prudent to identify a process which controls almost all disease-promoting processes in the CF airways including underlying CFTR dysfunction. There is emerging experimental and clinical evidence that supports the notion that impaired cellular proteostasis and autophagy plays a central role in regulating pathogenesis of chronic CF lung disease. Thus, correcting the underlying proteostasis and autophagy defect in controlling CF pulmonary disease, primarily via correcting the protein processing defect of F508del-CFTR protein has emerged as a novel intervention strategy. Hence, we discuss here both the rationale and significant therapeutic utility of emerging proteostasis and autophagy modulating drugs/compounds in controlling chronic CF lung disease, where targeted delivery is a critical factor-influencing efficacy.
Collapse
Affiliation(s)
- Manish Bodas
- Department of Medicine, University of Oklahoma, Oklahoma City, OK, United States
| | - Neeraj Vij
- Department of Pediatric Pulmonary Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- 4Dx Limited, Los Angeles, CA, United States
- VIJ Biotech LLC, Baltimore, MD, United States
| |
Collapse
|
145
|
Bodas M, Pehote G, Silverberg D, Gulbins E, Vij N. Autophagy augmentation alleviates cigarette smoke-induced CFTR-dysfunction, ceramide-accumulation and COPD-emphysema pathogenesis. Free Radic Biol Med 2019; 131:81-97. [PMID: 30500419 DOI: 10.1016/j.freeradbiomed.2018.11.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 01/27/2023]
Abstract
In this study, we aimed to investigate precise mechanism(s) of sphingolipid-imbalance and resulting ceramide-accumulation in COPD-emphysema. Where, human and murine emphysema lung tissues or human bronchial epithelial cells (Beas2b) were used for experimental analysis. We found that lungs of smokers and COPD-subjects with increasing emphysema severity demonstrate sphingolipid-imbalance, resulting in significant ceramide-accumulation and increased ceramide/sphingosine ratio, as compared to non-emphysema/non-smoker controls. Next, we found a substantial increase in emphysema chronicity-related ceramide-accumulation in murine (C57BL/6) lungs, while sphingosine levels only slightly increased. In accordance, the expression of the acid ceramidase decreased after CS-exposure. Moreover, CS-induced (sub-chronic) ceramide-accumulation was significantly (p < 0.05) reduced by treatment with TFEB/autophagy-inducing drug, gemfibrozil (GEM), suggesting that autophagy regulates CS-induced ceramide-accumulation. Next, we validated experimentally that autophagy/lipophagy-induction using an anti-oxidant, cysteamine, significantly (p < 0.05) reduces CS-extract (CSE)-mediated intracellular-ceramide-accumulation in p62 + aggresome-bodies. In addition to intracellular-accumulation, we found that CSE also induces membrane-ceramide-accumulation by ROS-dependent acid-sphingomyelinase (ASM) activation and plasma-membrane translocation, which was significantly controlled (p < 0.05) by cysteamine (an anti-oxidant) and amitriptyline (AMT, an inhibitor of ASM). Cysteamine-mediated and CSE-induced membrane-ceramide regulation was nullified by CFTR-inhibitor-172, demonstrating that CFTR controls redox impaired-autophagy dependent membrane-ceramide accumulation. In summary, our data shows that CS-mediated autophagy/lipophagy-dysfunction results in intracellular-ceramide-accumulation, while acquired CFTR-dysfunction-induced ASM causes membrane ceramide-accumulation. Thus, CS-exposure alters the sphingolipid-rheostat leading to the increased membrane- and intracellular- ceramide-accumulation inducing COPD-emphysema pathogenesis that is alleviated by treatment with cysteamine, a potent anti-oxidant with CFTR/autophagy-augmenting properties.
Collapse
Affiliation(s)
- Manish Bodas
- College of Medicine, Central Michigan University, Mt Pleasant, MI, USA
| | - Garrett Pehote
- College of Medicine, Central Michigan University, Mt Pleasant, MI, USA
| | - David Silverberg
- College of Medicine, Central Michigan University, Mt Pleasant, MI, USA
| | - Erich Gulbins
- Dept. of Molecular Biology, University of Duisburg-Essen, Germany and Dept. of Surgery, University of Cincinnati, OH, USA
| | - Neeraj Vij
- College of Medicine, Central Michigan University, Mt Pleasant, MI, USA; The Johns Hopkins University SOM University, Baltimore, MD, USA; VIJ Biotech LLC, Baltimore, MD, USA and 4Dx Ltd, Los Angeles, CA, USA.
| |
Collapse
|
146
|
Acid ceramidase, an emerging target for anti-cancer and anti-angiogenesis. Arch Pharm Res 2019; 42:232-243. [DOI: 10.1007/s12272-019-01114-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/10/2019] [Indexed: 02/07/2023]
|
147
|
Yagci ZB, Esvap E, Ozkara HA, Ulgen KO, Olmez EO. Inflammatory response and its relation to sphingolipid metabolism proteins: Chaperones as potential indirect anti-inflammatory agents. MOLECULAR CHAPERONES IN HUMAN DISORDERS 2019; 114:153-219. [DOI: 10.1016/bs.apcsb.2018.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
148
|
Baker JE, Boudreau RM, Seitz AP, Caldwell CC, Gulbins E, Edwards MJ. Sphingolipids and Innate Immunity: A New Approach to Infection in the Post-Antibiotic Era? Surg Infect (Larchmt) 2018; 19:792-803. [DOI: 10.1089/sur.2018.187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Jennifer E. Baker
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Ryan M. Boudreau
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Aaron P. Seitz
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Charles C. Caldwell
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
- Division of Research, Shriners Hospital for Children, Cincinnati, Ohio
| | - Erich Gulbins
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael J. Edwards
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
149
|
Castellani S, Di Gioia S, di Toma L, Conese M. Human Cellular Models for the Investigation of Lung Inflammation and Mucus Production in Cystic Fibrosis. Anal Cell Pathol (Amst) 2018; 2018:3839803. [PMID: 30581723 PMCID: PMC6276497 DOI: 10.1155/2018/3839803] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/04/2018] [Accepted: 09/23/2018] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation, oxidative stress, mucus plugging, airway remodeling, and respiratory infections are the hallmarks of the cystic fibrosis (CF) lung disease. The airway epithelium is central in the innate immune responses to pathogens colonizing the airways, since it is involved in mucociliary clearance, senses the presence of pathogens, elicits an inflammatory response, orchestrates adaptive immunity, and activates mesenchymal cells. In this review, we focus on cellular models of the human CF airway epithelium that have been used for studying mucus production, inflammatory response, and airway remodeling, with particular reference to two- and three-dimensional cultures that better recapitulate the native airway epithelium. Cocultures of airway epithelial cells, macrophages, dendritic cells, and fibroblasts are instrumental in disease modeling, drug discovery, and identification of novel therapeutic targets. Nevertheless, they have to be implemented in the CF field yet. Finally, novel systems hijacking on tissue engineering, including three-dimensional cocultures, decellularized lungs, microfluidic devices, and lung organoids formed in bioreactors, will lead the generation of relevant human preclinical respiratory models a step forward.
Collapse
Affiliation(s)
- Stefano Castellani
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Lorena di Toma
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Laboratory of Regenerative and Experimental Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
150
|
Haque AKMA, Dewerth A, Antony JS, Riethmüller J, Schweizer GR, Weinmann P, Latifi N, Yasar H, Pedemonte N, Sondo E, Weidensee B, Ralhan A, Laval J, Schlegel P, Seitz C, Loretz B, Lehr CM, Handgretinger R, Kormann MSD. Chemically modified hCFTR mRNAs recuperate lung function in a mouse model of cystic fibrosis. Sci Rep 2018; 8:16776. [PMID: 30425265 PMCID: PMC6233194 DOI: 10.1038/s41598-018-34960-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022] Open
Abstract
Gene therapy has always been a promising therapeutic approach for Cystic Fibrosis (CF). However, numerous trials using DNA or viral vectors encoding the correct protein resulted in a general low efficacy. In the last years, chemically modified messenger RNA (cmRNA) has been proven to be a highly potent, pulmonary drug. Consequently, we first explored the expression, function and immunogenicity of human (h)CFTR encoded by cmRNAhCFTR in vitro and ex vivo, quantified the expression by flow cytometry, determined its function using a YFP based assay and checked the immune response in human whole blood. Similarly, we examined the function of cmRNAhCFTR in vivo after intratracheal (i.t.) or intravenous (i.v.) injection of the assembled cmRNAhCFTR together with Chitosan-coated PLGA (poly-D, L-lactide-co-glycolide 75:25 (Resomer RG 752 H)) nanoparticles (NPs) by FlexiVent. The amount of expression of human hCFTR encoded by cmRNAhCFTR was quantified by hCFTR ELISA, and cmRNAhCFTR values were assessed by RT-qPCR. Thereby, we observed a significant improvement of lung function, especially in regards to FEV0.1, suggesting NP-cmRNAhCFTR as promising therapeutic option for CF patients independent of their CFTR genotype.
Collapse
Affiliation(s)
- A K M Ashiqul Haque
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Alexander Dewerth
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Justin S Antony
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany.,Department of Hematology, Oncology, Clinical Immunology, University of Tuebingen, Tuebingen, Germany
| | | | - Georg R Schweizer
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Petra Weinmann
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Ngadhnjim Latifi
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Hanzey Yasar
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbruecken, Germany
| | | | - Elvira Sondo
- U.O.C. Genetica Medica, Istituto Giannina Gaslini, Genova, Italy
| | - Brian Weidensee
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Anjali Ralhan
- Department of Pediatrics I - Immunology and Pneumology/Cystic fibrosis, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Julie Laval
- Department of Pediatrics I - Immunology and Pneumology/Cystic fibrosis, Department of Pediatrics I, University of Tuebingen, Tuebingen, Germany
| | - Patrick Schlegel
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Christian Seitz
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbruecken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Center for Infection Research (HZI), Saarbruecken, Germany.,Department of Pharmacy, Saarland University, Saarbruecken, Germany
| | - Rupert Handgretinger
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany.,Department of Hematology, Oncology, Clinical Immunology, University of Tuebingen, Tuebingen, Germany
| | - Michael S D Kormann
- Department of Pediatrics I - Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|