1
|
Najm M, Martignetti L, Cornet M, Kelly-Aubert M, Sermet I, Calzone L, Stoven V. From CFTR to a CF signalling network: a systems biology approach to study Cystic Fibrosis. BMC Genomics 2024; 25:892. [PMID: 39342081 PMCID: PMC11438383 DOI: 10.1186/s12864-024-10752-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 08/30/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Cystic Fibrosis (CF) is a monogenic disease caused by mutations in the gene coding the Cystic Fibrosis Transmembrane Regulator (CFTR) protein, but its overall physio-pathology cannot be solely explained by the loss of the CFTR chloride channel function. Indeed, CFTR belongs to a yet not fully deciphered network of proteins participating in various signalling pathways. METHODS We propose a systems biology approach to study how the absence of the CFTR protein at the membrane leads to perturbation of these pathways, resulting in a panel of deleterious CF cellular phenotypes. RESULTS Based on publicly available transcriptomic datasets, we built and analyzed a CF network that recapitulates signalling dysregulations. The CF network topology and its resulting phenotypes were found to be consistent with CF pathology. CONCLUSION Analysis of the network topology highlighted a few proteins that may initiate the propagation of dysregulations, those that trigger CF cellular phenotypes, and suggested several candidate therapeutic targets. Although our research is focused on CF, the global approach proposed in the present paper could also be followed to study other rare monogenic diseases.
Collapse
Affiliation(s)
- Matthieu Najm
- Center for Computational Biology (CBIO), Mines Paris-PSL, 75006, Paris, France.
- Institut Curie, Université PSL, 75005, Paris, France.
- INSERM U900, 75005, Paris, France.
| | - Loredana Martignetti
- Center for Computational Biology (CBIO), Mines Paris-PSL, 75006, Paris, France
- Institut Curie, Université PSL, 75005, Paris, France
- INSERM U900, 75005, Paris, France
| | - Matthieu Cornet
- Center for Computational Biology (CBIO), Mines Paris-PSL, 75006, Paris, France
- Institut Curie, Université PSL, 75005, Paris, France
- INSERM U900, 75005, Paris, France
- Institut Necker Enfants Malades, INSERM U1151, 75015, Paris, France
| | - Mairead Kelly-Aubert
- Institut Necker Enfants Malades, INSERM U1151, 75015, Paris, France
- Université Paris Cité, 75015, Paris, France
| | - Isabelle Sermet
- Institut Necker Enfants Malades, INSERM U1151, 75015, Paris, France
- Université Paris Cité, 75015, Paris, France
- Centre de Référence Maladies Rares, Mucoviscidose et Maladies Apparentées, Hôpital Necker Enfants Malades AP-HP Centre Paris Cité, 75015, Paris, France
| | - Laurence Calzone
- Center for Computational Biology (CBIO), Mines Paris-PSL, 75006, Paris, France.
- Institut Curie, Université PSL, 75005, Paris, France.
- INSERM U900, 75005, Paris, France.
| | - Véronique Stoven
- Center for Computational Biology (CBIO), Mines Paris-PSL, 75006, Paris, France.
- Institut Curie, Université PSL, 75005, Paris, France.
- INSERM U900, 75005, Paris, France.
| |
Collapse
|
2
|
Wang J, Chen YL, Li YK, Chen DK, He JF, Yao N. Functions of Sphingolipids in Pathogenesis During Host-Pathogen Interactions. Front Microbiol 2021; 12:701041. [PMID: 34408731 PMCID: PMC8366399 DOI: 10.3389/fmicb.2021.701041] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/21/2021] [Indexed: 12/23/2022] Open
Abstract
Sphingolipids are a class of membrane lipids that serve as vital structural and signaling bioactive molecules in organisms ranging from yeast to animals. Recent studies have emphasized the importance of sphingolipids as signaling molecules in the development and pathogenicity of microbial pathogens including bacteria, fungi, and viruses. In particular, sphingolipids play key roles in regulating the delicate balance between microbes and hosts during microbial pathogenesis. Some pathogens, such as bacteria and viruses, harness host sphingolipids to promote development and infection, whereas sphingolipids from both the host and pathogen are involved in fungus-host interactions. Moreover, a regulatory role for sphingolipids has been described, but their effects on host physiology and metabolism remain to be elucidated. Here, we summarize the current state of knowledge about the roles of sphingolipids in pathogenesis and interactions with host factors, including how sphingolipids modify pathogen and host metabolism with a focus on pathogenesis regulators and relevant metabolic enzymes. In addition, we discuss emerging perspectives on targeting sphingolipids that function in host-microbe interactions as new therapeutic strategies for infectious diseases.
Collapse
Affiliation(s)
- Jian Wang
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resource, School of Agriculture, Sun Yat-sen University, Guangzhou, China
| | - Yi-Li Chen
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resource, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yong-Kang Li
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resource, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ding-Kang Chen
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resource, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jia-Fan He
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resource, School of Agriculture, Sun Yat-sen University, Guangzhou, China
| | - Nan Yao
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resource, School of Agriculture, Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Biocontrol, Guangdong Provincial Key Laboratory of Plant Resource, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Pathophysiology of Lung Disease and Wound Repair in Cystic Fibrosis. PATHOPHYSIOLOGY 2021; 28:155-188. [PMID: 35366275 PMCID: PMC8830450 DOI: 10.3390/pathophysiology28010011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/08/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive, life-threatening condition affecting many organs and tissues, the lung disease being the chief cause of morbidity and mortality. Mutations affecting the CF Transmembrane Conductance Regulator (CFTR) gene determine the expression of a dysfunctional protein that, in turn, triggers a pathophysiological cascade, leading to airway epithelium injury and remodeling. In vitro and in vivo studies point to a dysregulated regeneration and wound repair in CF airways, to be traced back to epithelial CFTR lack/dysfunction. Subsequent altered ion/fluid fluxes and/or signaling result in reduced cell migration and proliferation. Furthermore, the epithelial-mesenchymal transition appears to be partially triggered in CF, contributing to wound closure alteration. Finally, we pose our attention to diverse approaches to tackle this defect, discussing the therapeutic role of protease inhibitors, CFTR modulators and mesenchymal stem cells. Although the pathophysiology of wound repair in CF has been disclosed in some mechanisms, further studies are warranted to understand the cellular and molecular events in more details and to better address therapeutic interventions.
Collapse
|
4
|
Ling KM, Garratt LW, Gill EE, Lee AHY, Agudelo-Romero P, Sutanto EN, Iosifidis T, Rosenow T, Turvey SE, Lassmann T, Hancock REW, Kicic A, Stick SM. Rhinovirus Infection Drives Complex Host Airway Molecular Responses in Children With Cystic Fibrosis. Front Immunol 2020; 11:1327. [PMID: 32765492 PMCID: PMC7378398 DOI: 10.3389/fimmu.2020.01327] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 05/26/2020] [Indexed: 01/22/2023] Open
Abstract
Early-life viral infections are responsible for pulmonary exacerbations that can contribute to disease progression in young children with cystic fibrosis (CF). The most common respiratory viruses detected in the CF airway are human rhinoviruses (RV), and augmented airway inflammation in CF has been attributed to dysregulated airway epithelial responses although evidence has been conflicting. Here, we exposed airway epithelial cells from children with and without CF to RV in vitro. Using RNA-Seq, we profiled the transcriptomic differences of CF and non-CF airway epithelial cells at baseline and in response to RV. There were only modest differences between CF and non-CF cells at baseline. In response to RV, there were 1,442 and 896 differentially expressed genes in CF and non-CF airway epithelial cells, respectively. The core antiviral responses in CF and non-CF airway epithelial cells were mediated through interferon signaling although type 1 and 3 interferon signaling, when measured, were reduced in CF airway epithelial cells following viral challenge consistent with previous reports. The transcriptional responses in CF airway epithelial cells were more complex than in non-CF airway epithelial cells with diverse over-represented biological pathways, such as cytokine signaling and metabolic and biosynthetic pathways. Network analysis highlighted that the differentially expressed genes of CF airway epithelial cells' transcriptional responses were highly interconnected and formed a more complex network than observed in non-CF airway epithelial cells. We corroborate observations in fully differentiated air–liquid interface (ALI) cultures, identifying genes involved in IL-1 signaling and mucin glycosylation that are only dysregulated in the CF airway epithelial response to RV infection. These data provide novel insights into the CF airway epithelial cells' responses to RV infection and highlight potential pathways that could be targeted to improve antiviral and anti-inflammatory responses in CF.
Collapse
Affiliation(s)
- Kak-Ming Ling
- Paediatrics, Medical School, Faculty of Healthy and Medical Science, The University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Luke W Garratt
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Erin E Gill
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Amy H Y Lee
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Patricia Agudelo-Romero
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Erika N Sutanto
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Thomas Iosifidis
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Tim Rosenow
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Stuart E Turvey
- Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Timo Lassmann
- Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia
| | - Robert E W Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Anthony Kicic
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| | - Stephen M Stick
- Telethon Kids Institute, Respiratory Research Centre, Nedlands, WA, Australia.,Telethon Kids Institute, Centre for Health Research, The University of Western Australia, Nedlands, WA, Australia.,School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory and Sleep Medicine, Perth Children's Hospital, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
5
|
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
|
6
|
Valdivieso ÁG, Mori C, Clauzure M, Massip-Copiz M, Santa-Coloma TA. CFTR modulates RPS27 gene expression using chloride anion as signaling effector. Arch Biochem Biophys 2017; 633:103-109. [DOI: 10.1016/j.abb.2017.09.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 09/13/2017] [Accepted: 09/20/2017] [Indexed: 12/13/2022]
|
7
|
Kormann MSD, Dewerth A, Eichner F, Baskaran P, Hector A, Regamey N, Hartl D, Handgretinger R, Antony JS. Transcriptomic profile of cystic fibrosis patients identifies type I interferon response and ribosomal stalk proteins as potential modifiers of disease severity. PLoS One 2017; 12:e0183526. [PMID: 28846703 PMCID: PMC5573219 DOI: 10.1371/journal.pone.0183526] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/04/2017] [Indexed: 12/25/2022] Open
Abstract
Cystic Fibrosis (CF) is the most common monogenic disease among people of Western European descent and caused by mutations in the CFTR gene. However, the disease severity is immensely variable even among patients with similar CFTR mutations due to the possible effect of 'modifier genes'. To identify genetic modifiers, we applied RNA-seq based transcriptomic analyses in CF patients with a mild and severe lung phenotype. Global gene expression and enrichment analyses revealed that genes of the type I interferon response and ribosomal stalk proteins are potential modifiers of CF related lung dysfunction. The results provide a new set of CF modifier genes with possible implications as new therapeutic targets for the treatment of CF.
Collapse
Affiliation(s)
- Michael S. D. Kormann
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - Alexander Dewerth
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - Felizitas Eichner
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
| | - Praveen Baskaran
- Center for Quantitative Biology, University of Tuebingen, Tuebingen, Germany
| | - Andreas Hector
- University Children's Clinic Department of Paediatrics I, Paediatric Infectiology & Immunology, University of Tuebingen, Tuebingen, Germany
| | - Nicolas Regamey
- Division of Paediatric Respiratory Medicine and Department of Clinical Research, University of Bern, Bern, Switzerland
- Children’s Hopsital of Lucerne, Paediatric Pulmonology, Lucerne, Switzerland
| | - Dominik Hartl
- University Children's Clinic Department of Paediatrics I, Paediatric Infectiology & Immunology, University of Tuebingen, Tuebingen, Germany
| | - Rupert Handgretinger
- University Children's Clinic Department of Paediatrics I, Hematology and Oncology, University of Tuebingen, Tuebingen, Germany
| | - Justin S. Antony
- Department of Pediatrics I, Pediatric Infectiology and Immunology, Translational Genomics and Gene Therapy in Pediatrics, University of Tuebingen, Tuebingen, Germany
- * E-mail:
| |
Collapse
|
8
|
Saint-Criq V, Villeret B, Bastaert F, Kheir S, Hatton A, Cazes A, Xing Z, Sermet-Gaudelus I, Garcia-Verdugo I, Edelman A, Sallenave JM. Pseudomonas aeruginosa LasB protease impairs innate immunity in mice and humans by targeting a lung epithelial cystic fibrosis transmembrane regulator-IL-6-antimicrobial-repair pathway. Thorax 2017; 73:49-61. [PMID: 28790180 PMCID: PMC5738602 DOI: 10.1136/thoraxjnl-2017-210298] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/11/2017] [Accepted: 07/17/2017] [Indexed: 11/16/2022]
Abstract
Background Pseudomonas aeruginosa lung infections are a huge problem in ventilator-associated pneumonia, cystic fibrosis (CF) and in chronic obstructive pulmonary disease (COPD) exacerbations. This bacterium secretes virulence factors that may subvert host innate immunity. Objective We evaluated the effect of P. aeruginosa elastase LasB, an important virulence factor secreted by the type II secretion system, on ion transport, innate immune responses and epithelial repair, both in vitro and in vivo. Methods Wild-type (WT) or cystic fibrosis transmembrane conductance regulator (CFTR)-mutated epithelial cells (cell lines and primary cells from patients) were treated with WT or ΔLasB pseudomonas aeruginosa O1 (PAO1) secretomes. The effect of LasB and PAO1 infection was also assessed in vivo in murine models. Results We showed that LasB was the most abundant protein in WT PAO1 secretomes and that it decreased epithelial CFTR expression and activity. In airway epithelial cell lines and primary bronchial epithelial cells, LasB degraded the immune mediators interleukin (IL)-6 and trappin-2, an important epithelial-derived antimicrobial molecule. We further showed that an IL-6/STAT3 signalling pathway was downregulated by LasB, resulting in inhibition of epithelial cell repair. In mice, intranasally instillated LasB induced significant weight loss, inflammation, injury and death. By contrast, we showed that overexpression of IL-6 and trappin-2 protected mice against WT-PAO1-induced death, by upregulating IL-17/IL-22 antimicrobial and repair pathways. Conclusions Our data demonstrate that PAO1 LasB is a major P. aeruginosa secreted factor that modulates ion transport, immune response and tissue repair. Targeting this virulence factor or upregulating protective factors such as IL-6 or antimicrobial molecules such as trappin-2 could be beneficial in P. aeruginosa-infected individuals.
Collapse
Affiliation(s)
- Vinciane Saint-Criq
- INSERM U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universtaire FIRE (Fibrosis, Inflammation and Remodeling), Université Paris Diderot, Sorbonne Paris Cité, Hopital Bichat - Claude-Bernard, Paris, France
| | - Bérengère Villeret
- INSERM U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universtaire FIRE (Fibrosis, Inflammation and Remodeling), Université Paris Diderot, Sorbonne Paris Cité, Hopital Bichat - Claude-Bernard, Paris, France
| | - Fabien Bastaert
- INSERM U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universtaire FIRE (Fibrosis, Inflammation and Remodeling), Université Paris Diderot, Sorbonne Paris Cité, Hopital Bichat - Claude-Bernard, Paris, France
| | - Saadé Kheir
- INSERM U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universtaire FIRE (Fibrosis, Inflammation and Remodeling), Université Paris Diderot, Sorbonne Paris Cité, Hopital Bichat - Claude-Bernard, Paris, France
| | - Aurélie Hatton
- INSERM U1151, Faculté de Médecine, site Necker, Université Paris Descartes, Paris, France
| | - Aurélie Cazes
- INSERM U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universtaire FIRE (Fibrosis, Inflammation and Remodeling), Université Paris Diderot, Sorbonne Paris Cité, Hopital Bichat - Claude-Bernard, Paris, France
| | - Zhou Xing
- McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | | | - Ignacio Garcia-Verdugo
- INSERM U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universtaire FIRE (Fibrosis, Inflammation and Remodeling), Université Paris Diderot, Sorbonne Paris Cité, Hopital Bichat - Claude-Bernard, Paris, France
| | - Aleksander Edelman
- INSERM U1151, Faculté de Médecine, site Necker, Université Paris Descartes, Paris, France
| | - Jean-Michel Sallenave
- INSERM U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universtaire FIRE (Fibrosis, Inflammation and Remodeling), Université Paris Diderot, Sorbonne Paris Cité, Hopital Bichat - Claude-Bernard, Paris, France
| |
Collapse
|
9
|
Farinha CM, Matos P. Rab GTPases regulate the trafficking of channels and transporters - a focus on cystic fibrosis. Small GTPases 2017; 9:136-144. [PMID: 28463591 DOI: 10.1080/21541248.2017.1317700] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The amount of ion channels and transporters present at the plasma membrane is a crucial component of the overall regulation of ion transport. The number of channels present result from an intricate network of proteins that controls the late events of channel trafficking, such as endocytosis, recycling and targeting to lysosomal degradation. Small GTPases of the Rab family are key players in these processes thus contributing to regulation of fluid secretion and ion homeostasis. In epithelia, this involves mainly the balance between the chloride channel CFTR and the sodium channel ENaC, whose misfunction is a hallmark of cystic fibrosis - the commonest recessive disorder in Caucasians. Here, we review the role of GTPases in regulating trafficking of ion channels and transporters, comparing what is known for CFTR and ENaC with other types of channels. We also discuss how feasible would be to target the Rab machinery to handle a disorder such as CF.
Collapse
Affiliation(s)
- Carlos M Farinha
- a University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute , Campo Grande, Lisboa , Portugal
| | - Paulo Matos
- a University of Lisboa, Faculty of Sciences, BioISI - Biosystems & Integrative Sciences Institute , Campo Grande, Lisboa , Portugal.,b Department of Human Genetics , National Health Institute 'Dr. Ricardo Jorge' , Av. Padre Cruz, Lisboa , Portugal
| |
Collapse
|
10
|
In silico search for modifier genes associated with pancreatic and liver disease in Cystic Fibrosis. PLoS One 2017; 12:e0173822. [PMID: 28339466 PMCID: PMC5365109 DOI: 10.1371/journal.pone.0173822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/27/2017] [Indexed: 12/15/2022] Open
Abstract
Cystic Fibrosis is the most common lethal autosomal recessive disorder in the white population, affecting among other organs, the lung, the pancreas and the liver. Whereas Cystic Fibrosis is a monogenic disease, many studies reveal a very complex relationship between genotype and clinical phenotype. Indeed, the broad phenotypic spectrum observed in Cystic Fibrosis is far from being explained by obvious genotype-phenotype correlations and it is admitted that Cystic Fibrosis disease is the result of multiple factors, including effects of the environment as well as modifier genes. Our objective was to highlight new modifier genes with potential implications in the lung, pancreatic and liver outcomes of the disease. For this purpose we performed a system biology approach which combined, database mining, literature mining, gene expression study and network analysis as well as pathway enrichment analysis and protein-protein interactions. We found that IFI16, CCNE2 and IGFBP2 are potential modifiers in the altered lung function in Cystic Fibrosis. We also found that EPHX1, HLA-DQA1, HLA-DQB1, DSP and SLC33A1, GPNMB, NCF2, RASGRP1, LGALS3 and PTPN13, are potential modifiers in pancreas and liver, respectively. Associated pathways indicate that immune system is likely involved and that Ubiquitin C is probably a central node, linking Cystic Fibrosis to liver and pancreatic disease. We highlight here new modifier genes with potential implications in Cystic Fibrosis. Nevertheless, our in silico analysis requires functional analysis to give our results a physiological relevance.
Collapse
|
11
|
Balloy V, Varet H, Dillies MA, Proux C, Jagla B, Coppée JY, Tabary O, Corvol H, Chignard M, Guillot L. Normal and Cystic Fibrosis Human Bronchial Epithelial Cells Infected with Pseudomonas aeruginosa Exhibit Distinct Gene Activation Patterns. PLoS One 2015; 10:e0140979. [PMID: 26485688 PMCID: PMC4618526 DOI: 10.1371/journal.pone.0140979] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/25/2015] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND AIMS In cystic fibrosis (CF), Pseudomonas aeruginosa is not eradicated from the lower respiratory tract and is associated with epithelial inflammation that eventually causes tissue damage. To identify the molecular determinants of an effective response to P. aeruginosa infection, we performed a transcriptomic analysis of primary human bronchial epithelial cells from healthy donors (CTRL) 2, 4, and 6 h after induced P. aeruginosa infection. Compared to noninfected cells, infected cells showed changes in gene activity, which were most marked 6 h postinfection and usually consisted in upregulation. RESULTS By comparing for each time point of infection, the transcriptomic response of epithelial cells from CF patients and healthy donors, we identified 851, 638, 667, and 980 differentially expressed genes 0, 2, 4, and 6 h postinfection, respectively. Gene selection followed by bioinformatic analysis showed that most of the differentially expressed genes, either up- or downregulated, were in the protein-binding and catalytic gene-ontology categories. Finally, we established that the protein products of the genes exhibiting the greatest differential upregulation (CSF2, CCL2, TNF, CSF3, MMP1, and MMP10) between CF patients and CTRL were produced in higher amounts by infected cells from CF patients versus CTRL. CONCLUSIONS The differentially expressed genes in CF patients may constitute a signature for a detrimental inflammatory response and for an inefficient P. aeruginosa host-cell response.
Collapse
Affiliation(s)
- Viviane Balloy
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France
| | - Hugo Varet
- Institut Pasteur, Plate-forme Transcriptome Epigenome, Centre Innovation et Recherche Technologiques, Paris, France
| | - Marie-Agnès Dillies
- Institut Pasteur, Plate-forme Transcriptome Epigenome, Centre Innovation et Recherche Technologiques, Paris, France
| | - Caroline Proux
- Institut Pasteur, Plate-forme Transcriptome Epigenome, Centre Innovation et Recherche Technologiques, Paris, France
| | - Bernd Jagla
- Institut Pasteur, Plate-forme Transcriptome Epigenome, Centre Innovation et Recherche Technologiques, Paris, France
| | - Jean-Yves Coppée
- Institut Pasteur, Plate-forme Transcriptome Epigenome, Centre Innovation et Recherche Technologiques, Paris, France
| | - Olivier Tabary
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France
| | - Harriet Corvol
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France
- Pneumologie pédiatrique, AP-HP, Hôpital Trousseau, Paris, France
| | - Michel Chignard
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France
| | - Loïc Guillot
- INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, Paris, France
- * E-mail:
| |
Collapse
|
12
|
Venkatakrishnan V, Packer NH, Thaysen-Andersen M. Host mucin glycosylation plays a role in bacterial adhesion in lungs of individuals with cystic fibrosis. Expert Rev Respir Med 2014; 7:553-76. [DOI: 10.1586/17476348.2013.837752] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
13
|
Clarke LA, Sousa L, Barreto C, Amaral MD. Changes in transcriptome of native nasal epithelium expressing F508del-CFTR and intersecting data from comparable studies. Respir Res 2013; 14:38. [PMID: 23537407 PMCID: PMC3637641 DOI: 10.1186/1465-9921-14-38] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 03/07/2013] [Indexed: 01/06/2023] Open
Abstract
Background Microarray studies related to cystic fibrosis (CF) airway gene expression have gone some way in clarifying the complex molecular background of CF lung diseases, but have made little progress in defining a robust “molecular signature” associated with mutant CFTR expression. Disparate methodological and statistical analyses complicate comparisons between independent studies of the CF transcriptome, and although each study may be valid in isolation, the conclusions reached differ widely. Methods We carried out a small-scale whole genome microarray study of gene expression in human native nasal epithelial cells from F508del-CFTR homozygotes in comparison to non-CF controls. We performed superficial comparisons with other microarray datasets in an attempt to identify a subset of regulated genes that could act as a signature of F508del-CFTR expression in native airway tissue samples. Results Among the alterations detected in CF, up-regulation of genes involved in cell proliferation, and down-regulation of cilia genes were the most notable. Other changes involved gene expression changes in calcium and membrane pathways, inflammation, defence response, wound healing and the involvement of estrogen signalling. Comparison of our data set with previously published studies allowed us to assess the consistency of independent microarray data sets, and shed light on the limitations of such snapshot studies in measuring a system as subtle and dynamic as the transcriptome. Comparison of in-vivo studies nevertheless yielded a small molecular CF signature worthy of future investigation. Conclusions Despite the variability among the independent studies, the current CF transcriptome meta-analysis identified subsets of differentially expressed genes in native airway tissues which provide both interesting clues to CF pathogenesis and a possible CF biomarker.
Collapse
Affiliation(s)
- Luka A Clarke
- BioFIG-Centre for Biodiversity, Functional and Integrative Genomics, FCUL-Faculty of Sciences, University of Lisboa, Lisboa 1749-016, Portugal.
| | | | | | | |
Collapse
|
14
|
Tsuchiya M, Kumar P, Bhattacharyya S, Chattoraj S, Srivastava M, Pollard HB, Biswas R. Differential Regulation of Inflammation by Inflammatory Mediators in Cystic Fibrosis Lung Epithelial Cells. J Interferon Cytokine Res 2013; 33:121-9. [DOI: 10.1089/jir.2012.0074] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Motohiro Tsuchiya
- Molecular and Cell Biology Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Parameet Kumar
- Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sharmistha Bhattacharyya
- Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Sangbrita Chattoraj
- Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Meera Srivastava
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Harvey B. Pollard
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Roopa Biswas
- Graduate School of Nursing, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
15
|
Eiserich JP, Yang J, Morrissey BM, Hammock BD, Cross CE. Omics approaches in cystic fibrosis research: a focus on oxylipin profiling in airway secretions. Ann N Y Acad Sci 2012; 1259:1-9. [PMID: 22758630 DOI: 10.1111/j.1749-6632.2012.06580.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cystic fibrosis (CF) is associated with abnormal lipid metabolism, intense respiratory tract (RT) infection, and inflammation, eventually resulting in lung tissue destruction and respiratory failure. The CF RT inflammatory milieu, as reflected by airway secretions, includes a complex array of inflammatory mediators, bacterial products, and host secretions. It is dominated by neutrophils and their proteolytic and oxidative products and includes a wide spectrum of bioactive lipids produced by both host and presumably microbial metabolic pathways. The fairly recent advent of "omics" technologies has greatly increased capabilities of further interrogating this easily obtainable RT compartment that represents the apical culture media of the underlying RT epithelial cells. This paper discusses issues related to the study of CF omics with a focus on the profiling of CF RT oxylipins. Challenges in their identification/quantitation in RT fluids, their pathways of origin, and their potential utility for understanding CF RT inflammatory and oxidative processes are highlighted. Finally, the utility of oxylipin metabolic profiling in directing optimal therapeutic approaches and determining the efficacy of various interventions is discussed.
Collapse
Affiliation(s)
- Jason P Eiserich
- Department of Internal Medicine, University of California, Davis, California, USA
| | | | | | | | | |
Collapse
|
16
|
Hampton TH, Ballok AE, Bomberger JM, Rutkowski MR, Barnaby R, Coutermarsh B, Conejo-Garcia JR, O'Toole GA, Stanton BA. Does the F508-CFTR mutation induce a proinflammatory response in human airway epithelial cells? Am J Physiol Lung Cell Mol Physiol 2012; 303:L509-18. [PMID: 22821996 DOI: 10.1152/ajplung.00226.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In the clinical setting, mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene enhance the inflammatory response in the lung to Pseudomonas aeruginosa (P. aeruginosa) infection. However, studies on human airway epithelial cells in vitro have produced conflicting results regarding the effect of mutations in CFTR on the inflammatory response to P. aeruginosa, and there are no comprehensive studies evaluating the effect of P. aeruginosa on the inflammatory response in airway epithelial cells with the ΔF508/ΔF508 genotype and their matched CF cell line rescued with wild-type (wt)-CFTR. CFBE41o- cells (ΔF508/ΔF508) and CFBE41o- cells complemented with wt-CFTR (CFBE-wt-CFTR) have been used extensively as an experimental model to study CF. Thus the goal of this study was to examine the effect of P. aeruginosa on gene expression and cytokine/chemokine production in this pair of cells. P. aeruginosa elicited a more robust increase in cytokine and chemokine expression (e.g., IL-8, CXCL1, CXCL2 and TNF-α) in CFBE-wt-CFTR cells compared with CFBE-ΔF508-CFTR cells. These results demonstrate that CFBE41o- cells complemented with wt-CFTR mount a more robust inflammatory response to P. aeruginosa than CFBE41o-ΔF508/ΔF508-CFTR cells. Taken together with other published studies, our data demonstrate that there is no compelling evidence to support the view that mutations in CFTR induce a hyperinflammatory response in human airway epithelial cells in vivo. Although the lungs of patients with CF have abundant levels of proinflammatory cytokines and chemokines, because the lung is populated by immune cells and epithelial cells there is no way to know, a priori, whether airway epithelial cells in the CF lung in vivo are hyperinflammatory in response to P. aeruginosa compared with non-CF lung epithelial cells. Thus studies on human airway epithelial cell lines and primary cells in vitro that propose to examine the effect of mutations in CFTR on the inflammatory response to P. aeruginosa have uncertain clinical significance with regard to CF.
Collapse
Affiliation(s)
- Thomas H Hampton
- Dept. of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
|
18
|
Abstract
To understand the links between CFTR mutations and the development of cystic fibrosis (CF) phenotypes, it is imperative to study the transcriptome in affected cell types. Microarray expression profiling provides a platform to study global gene expression in detail. This approach may provide the necessary information to segregate phenotypic characteristics of CF, differentiate between genetic or environmental factors, and assess the advent and progression of disease phenotypes. Moreover, if a "CF signature" of genes with altered expression is defined, this can be used to monitor effectiveness of treatment. We provide here detailed protocols and tips for collecting and preserving tissues and cells, and preparing total RNA. We also outline novel strategies for experimental design and data analysis, and describe some powerful gene and pathway discovery tools.
Collapse
|
19
|
Martino AT, Mueller C, Braag S, Cruz PE, Campbell-Thompson M, Jin S, Flotte TR. N-glycosylation augmentation of the cystic fibrosis epithelium improves Pseudomonas aeruginosa clearance. Am J Respir Cell Mol Biol 2010; 44:824-30. [PMID: 20693405 DOI: 10.1165/rcmb.2009-0285oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chronic lung colonization with Pseudomonas aeruginosa is anticipated in cystic fibrosis (CF). Abnormal terminal glycosylation has been implicated as a candidate for this condition. We previously reported a down-regulation of mannose-6-phosphate isomerase (MPI) for core N-glycan production in the CFTR-defective human cell line (IB3). We found a 40% decrease in N-glycosylation of IB3 cells compared with CFTR-corrected human cell line (S9), along with a threefold-lower surface attachment of P. aeruginosa strain, PAO1. There was a twofold increase in intracellular bacteria in S9 cells compared with IB3 cells. After a 4-hour clearance period, intracellular bacteria in IB3 cells increased twofold. Comparatively, a twofold decrease in intracellular bacteria occurred in S9 cells. Gene augmentation in IB3 cells with hMPI or hCFTR reversed these IB3 deficiencies. Mannose-6-phosphate can be produced from external mannose independent of MPI, and correction in the IB3 clearance deficiencies was observed when cultured in mannose-rich medium. An in vivo model for P. aeruginosa colonization in the upper airways revealed an increased bacterial burden in the trachea and oropharynx of nontherapeutic CF mice compared with mice treated either with an intratracheal delivery adeno-associated viral vector 5 expressing murine MPI, or a hypermannose water diet. Finally, a modest lung inflammatory response was observed in CF mice, and was partially corrected by both treatments. Augmenting N-glycosylation to attenuate colonization of P. aeruginosa in CF airways reveals a new therapeutic avenue for a hallmark disease condition in CF.
Collapse
Affiliation(s)
- Ashley T Martino
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Farberman MM, Ibricevic A, Joseph TD, Akers KT, Garcia-Medina R, Crosby S, Clarke LL, Brody SL, Ferkol TW. Effect of polarized release of CXC-chemokines from wild-type and cystic fibrosis murine airway epithelial cells. Am J Respir Cell Mol Biol 2010; 45:221-8. [PMID: 20639462 DOI: 10.1165/rcmb.2009-0249oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The respiratory epithelium lining the airway relies on mucociliary clearance and a complex network of inflammatory mediators to protect the lung. Alterations in the composition and volume of the periciliary liquid layer, as occur in cystic fibrosis (CF), lead to impaired mucociliary clearance and persistent airway infection. Moreover, the respiratory epithelium releases chemoattractants after infection, inciting airway inflammation. However, characterizing the inflammatory response of primary human airway epithelial cells to infection can be challenging because of genetic heterogeneity. Using well-characterized, differentiated, primary murine tracheal cells grown at an air-liquid interface, which provides an in vitro polarized epithelial model, we compared inflammatory gene expression and secretion in wild-type and ΔF508 CF airway cells after infection with Pseudomonas aeruginosa. The expression of several CXC-chemokines, including macrophage inflammatory protein-2, small inducible cytokine subfamily member 2, lipopolysaccharide-induced chemokine, and interferon-inducible cytokine-10, was markedly increased after infection, and these proinflammatory mediators were asymmetrically released from the airway epithelium, predominantly from the basolateral surface. Equal amounts of CXC-chemokines were released from wild-type and CF cells. Secreted mediators were concentrated in the thin, periciliary fluid layer, and the dehydrated apical microenvironment of CF airway epithelial cells amplified the inflammatory signal, potentially resulting in high chemokine concentration gradients across the epithelium. Consistent with this observation, the enhanced chemotaxis of wild-type neutrophils was detected in CF airway epithelial cultures, compared with wild-type cells. These data suggest that P. aeruginosa infection of the airway epithelium induces the expression and polarized secretion of CXC-chemokines, and the increased concentration gradient across the CF airway leads to an exaggerated inflammatory response.
Collapse
Affiliation(s)
- Michelle M Farberman
- Division of Pediatric Allergy, Immunology, and Pulmonary Medicine, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Hampton TH, Stanton BA. A novel approach to analyze gene expression data demonstrates that the DeltaF508 mutation in CFTR downregulates the antigen presentation pathway. Am J Physiol Lung Cell Mol Physiol 2009; 298:L473-82. [PMID: 20044437 DOI: 10.1152/ajplung.00379.2009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Gene array studies comparing cystic fibrosis (CF) and non-CF genotypes should reveal factors that explain variability in CF lung disease progression, yielding insights that lead to improved CF care. To date, studies have reached conflicting conclusions, perhaps due to experimental differences and divergent statistical approaches. This review aims: 1) to summarize the findings of four recent gene studies comparing CF and non-CF genotypes, and 2) to reanalyze original data using a recently developed statistical approach, with the aim of identifying genes and paths consistently regulated by the CF genotype. We identified four studies evaluating the effect of the DeltaF508-CFTR mutation on human airway epithelial cell gene expression, restricting our investigation to human airway epithelial cell studies whose data were accessible in NCBI's Gene Expression Omnibus or the European Bioinformatic Institute's ArrayExpress. Gene expression patterns showed consistent repression of MHC class I antigen presentation genes in CF human airway epithelia, suggesting a novel mechanistic explanation for poor clearance of viral and bacterial infections by CF patients. We also examined proinflammatory and NF-kappaB genes, whose induction is widely accepted as a hallmark of the CF genotype, but found little evidence of induction, consistent with a recent review (Machen TE, Am J Physiol Cell Physiol 291: C218-C230, 2006.). In conclusion, our analysis suggests that the CF genotype may impair immune function in airway epithelial cells but may not increase inflammation. Additional studies are required to determine whether MHC class I gene repression in CF reduces antigen presentation at the protein level and whether repression impairs immune function.
Collapse
Affiliation(s)
- Thomas H Hampton
- Department of Physiology, Dartmouth Medical School, Hanover, New Hampshire 03755, USA
| | | |
Collapse
|
22
|
Stalvey MS, Flotte TR. Endocrine parameters of cystic fibrosis: Back to basics. J Cell Biochem 2009; 108:353-61. [DOI: 10.1002/jcb.22284] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
23
|
Rottner M, Freyssinet JM, Martínez MC. Mechanisms of the noxious inflammatory cycle in cystic fibrosis. Respir Res 2009; 10:23. [PMID: 19284656 PMCID: PMC2660284 DOI: 10.1186/1465-9921-10-23] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Accepted: 03/13/2009] [Indexed: 01/09/2023] Open
Abstract
Multiple evidences indicate that inflammation is an event occurring prior to infection in patients with cystic fibrosis. The self-perpetuating inflammatory cycle may play a pathogenic part in this disease. The role of the NF-κB pathway in enhanced production of inflammatory mediators is well documented. The pathophysiologic mechanisms through which the intrinsic inflammatory response develops remain unclear. The unfolded mutated protein cystic fibrosis transmembrane conductance regulator (CFTRΔF508), accounting for this pathology, is retained in the endoplasmic reticulum (ER), induces a stress, and modifies calcium homeostasis. Furthermore, CFTR is implicated in the transport of glutathione, the major antioxidant element in cells. CFTR mutations can alter redox homeostasis and induce an oxidative stress. The disturbance of the redox balance may evoke NF-κB activation and, in addition, promote apoptosis. In this review, we examine the hypotheses of the integrated pathogenic processes leading to the intrinsic inflammatory response in cystic fibrosis.
Collapse
Affiliation(s)
- Mathilde Rottner
- 1INSERM U 770; Université Paris-Sud 11, Faculté de Médecine, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France.
| | | | | |
Collapse
|
24
|
Pashuck TD, Franz SE, Altman MK, Wasserfall CH, Atkinson MA, Wronski TJ, Flotte TR, Stalvey MS. Murine model for cystic fibrosis bone disease demonstrates osteopenia and sex-related differences in bone formation. Pediatr Res 2009; 65:311-6. [PMID: 19047917 PMCID: PMC4201032 DOI: 10.1203/pdr.0b013e3181961e80] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
As the incidence of cystic fibrosis (CF) bone disease is increasing, we analyzed CF transmembrane conductance regulator (CFTR) deficient mice (CF mice) to gain pathogenic insights. In these studies comparing adult (14 wk) CF and C57BL/6J mice, both bone length and total area were decreased in CF mice. Metaphyseal trabecular and cortical density were also decreased, as well as diaphyseal cortical and total density. Trabecular bone volume was diminished in CF mice. Female CF mice revealed decreased trabecular width and number compared with C57BL/6J, whereas males demonstrated no difference in trabecular number. Female CF mice had reduced mineralizing surface and bone formation rates. Conversely, male CF mice had increased mineralizing surface, mineral apposition, and bone formation rates compared with C57BL/6J males. Bone formation rate was greater in males compared with female CF mice. Smaller bones with decreased density in CF, despite absent differences in osteoblast and osteoclast surfaces, suggest CF transmembrane conductance regulator influences bone cell activity rather than number. Differences in bone formation rate in CF mice are suggestive of inadequate bone formation in females but increased bone formation in males. This proanabolic observation in male CF mice is consistent with other clinical sex differences in CF.
Collapse
Affiliation(s)
- Troy D. Pashuck
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610
| | - Sarah E. Franz
- Department of Physiological Sciences, University of Florida, College of Veterinary Medicine, Gainesville, FL 32610
| | - Molly K. Altman
- Department of Physiological Sciences, University of Florida, College of Veterinary Medicine, Gainesville, FL 32610
| | - Clive H. Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610
| | - Mark A. Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610
| | - Thomas J. Wronski
- Department of Physiological Sciences, University of Florida, College of Veterinary Medicine, Gainesville, FL 32610
| | - Terence R. Flotte
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655
| | - Michael S. Stalvey
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA 01655; Department of Pediatrics, University of Florida, College of Medicine, Gainesville, FL 32610
| |
Collapse
|
25
|
Yu H, Zeidan YH, Wu BX, Jenkins RW, Flotte TR, Hannun YA, Virella-Lowell I. Defective acid sphingomyelinase pathway with Pseudomonas aeruginosa infection in cystic fibrosis. Am J Respir Cell Mol Biol 2009; 41:367-75. [PMID: 19168701 DOI: 10.1165/rcmb.2008-0295oc] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Acid sphingomyelinase (ASMase) is a key enzyme in sphingolipid metabolism, which can be activated by various cellular stress mechanisms including bacterial pathogens. Activation of ASMase generates ceramide, which is important for innate immune response to eliminate infected pathogens. The current study reveals a defective ASMase pathway after Pseudomonas aeruginosa infection in both a cystic fibrosis (CF) bronchial epithelial cell line (IB3-1 cell) and in the lungs of CF transmembrane conductance regulator (CFTR) knockout (KO) mice as compared with S9 cells and wild-type C57BL/6 mice. ASMase activity and total ceramide levels significantly increased in S9 cells and C57BL/6 mice with P. aeruginosa infection, but not in IB3-1 cells and CFTR KO mice. The silencing of CFTR by CFTR RNAi in S9 cells significantly decreased ASMase activity after bacterial infection as compared with controls. This study also demonstrates that induction of ASMase is responsible for modulating the immune response to bacterial infection. Blocking ASMase activity with specific ASMase RNAi, an ASMase inhibitor, or an ASMase antibody in S9 cells significantly increased IL-8 levels with P. aeruginosa infection compared with controls. Reciprocally, adding exogenous bacterial sphingomyelinase to IB3-1 cells significantly decreased IL-8 levels compared with untreated cells. In addition, silencing of ASMase in S9 cells also significantly decreased bacterial internalization. Adding exogenous bacterial sphingomyelinase to IB3-1 cells reconstituted the cell death response to P. aeruginosa infection. This study demonstrates that the defective ASMase pathway in CF is a key contributor to the unabated IL-8 response with P. aeruginosa infection and to the compromised host response failing to eradicate bacteria.
Collapse
Affiliation(s)
- Hong Yu
- Department of Pediatrics, Medical University of South Carolina, 173 Ashley BSB 749, Charleston, SC 29403, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
CFTR mutations impart elevated immune reactivity in a murine model of cystic fibrosis related diabetes. Cytokine 2008; 44:154-9. [DOI: 10.1016/j.cyto.2008.07.468] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2007] [Revised: 04/11/2008] [Accepted: 07/17/2008] [Indexed: 11/21/2022]
|
27
|
Bartling TR, Drumm ML. Oxidative stress causes IL8 promoter hyperacetylation in cystic fibrosis airway cell models. Am J Respir Cell Mol Biol 2008; 40:58-65. [PMID: 18635816 DOI: 10.1165/rcmb.2007-0464oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Dysregulated inflammation has been implicated in cystic fibrosis (CF) airway pathophysiology. The expression of inflammatory genes, like interleukin 8 (IL8), involves chromatin remodeling through histone acetylation. Inflammatory gene hyperacetylation could explain inflammatory mediator dysregulation seen in CF airways. CF airways are exposed to high levels of oxidative stress, and oxidative stress increases histone acetylation and inflammatory gene transcription. Loss of cystic fibrosis transmembrane conductance regulator (CFTR) may even reduce protection against oxidative stress. Consequently, increasing oxidative stress would likely lead to an imbalance of histone acetyl-transferase (HAT) and deacetylase (HDAC) stoichiometry and contribute to the heightened inflammatory response seen in the CF airway. We hypothesize that oxidative stress in CF airways causes increased acetylation of inflammatory gene promoters, contributing to transcriptional activity of these loci. Messenger RNA levels of IL8, IL6, CXCL1, CXCL2, CXCL3, and IL1 are significantly elevated in CF epithelial cell models. Histone H4 acetylation is lower at the IL8 promoter of the non-CF cell lines than the CF models. The reducing agent N-acetyl-cysteine decreases IL8 message and promoter H4 acetylation to non-CF levels, suggesting that oxidative stress contributes to IL8 expression in these models. H(2)O(2) treatment causes increased IL-8 acetylation and mRNA in all cells, but less in the CF-model cells. Together these data suggest a model in which cells without functional CFTR are under increased oxidative stress. Our data suggest intrinsic alterations in the HAT/HDAC balance in CFTR-deficient cells, and that oxidative stress contributes to this alteration.
Collapse
Affiliation(s)
- Toni R Bartling
- Department of Genetics, Case Western Reserve University, Cleveland, OH 44106-4948, USA
| | | |
Collapse
|
28
|
Boncoeur E, Roque T, Bonvin E, Saint-Criq V, Bonora M, Clement A, Tabary O, Henrion-Caude A, Jacquot J. Cystic fibrosis transmembrane conductance regulator controls lung proteasomal degradation and nuclear factor-kappaB activity in conditions of oxidative stress. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:1184-94. [PMID: 18372427 DOI: 10.2353/ajpath.2008.070310] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cystic fibrosis is a lethal inherited disorder caused by mutations in a single gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR) protein, resulting in progressive oxidative lung damage. In this study, we evaluated the role of CFTR in the control of ubiquitin-proteasome activity and nuclear factor (NF)-kappaB/IkappaB-alpha signaling after lung oxidative stress. After a 64-hour exposure to hyperoxia-mediated oxidative stress, CFTR-deficient (cftr(-/-)) mice exhibited significantly elevated lung proteasomal activity compared with wild-type (cftr(+/+)) animals. This was accompanied by reduced lung caspase-3 activity and defective degradation of NF-kappaB inhibitor IkappaB-alpha. In vitro, human CFTR-deficient lung cells exposed to oxidative stress exhibited increased proteasomal activity and decreased NF-kappaB-dependent transcriptional activity compared with CFTR-sufficient lung cells. Inhibition of the CFTR Cl(-) channel by CFTR(inh-172) in the normal bronchial immortalized cell line 16HBE14o- increased proteasomal degradation after exposure to oxidative stress. Caspase-3 inhibition by Z-DQMD in CFTR-sufficient lung cells mimicked the response profile of increased proteasomal degradation and reduced NF-kappaB activity observed in CFTR-deficient lung cells exposed to oxidative stress. Taken together, these results suggest that functional CFTR Cl(-) channel activity is crucial for regulation of lung proteasomal degradation and NF-kappaB activity in conditions of oxidative stress.
Collapse
|
29
|
Boncoeur E, Criq VS, Bonvin E, Roque T, Henrion-Caude A, Gruenert DC, Clement A, Jacquot J, Tabary O. Oxidative stress induces extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase in cystic fibrosis lung epithelial cells: Potential mechanism for excessive IL-8 expression. Int J Biochem Cell Biol 2007; 40:432-46. [PMID: 17936667 DOI: 10.1016/j.biocel.2007.08.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 08/18/2007] [Accepted: 08/20/2007] [Indexed: 10/22/2022]
Abstract
Cystic fibrosis (CF) is a lethal disease caused by defective function of the cftr gene product, the CF transmembrane conductance regulator (CFTR) that leads to oxidative damage and excessive inflammatory response in lungs of CF patients. We here report the effects of oxidative stress (hyperoxia, 95% O(2)) on the expression of pro-inflammatory interleukin (IL)-8 and CXCR1/2 receptors in two human CF lung epithelial cell lines (IB3-1, with the heterozygous F508del/W1282X mutation and CFBE41o- with the homozygous F508del/F508del mutation) and two control non-CF lung epithelial cell lines (S9 cell line derived from IB3-1 after correction with wtCFTR and the normal bronchial cell line 16HBE14o-). Under oxidative stress, the expression of IL-8 and CXCR1/2 receptors was increased in CF, corrected and normal lung cell lines. The effects of oxidative stress were also investigated by measuring the transcription nuclear factor kappaB (NF-kappaB) and activator protein-1 (AP-1) activities. Under oxidative stress, no increase of NF-kappaB activation was observed in CF lung cells in contrast to that observed in normal and corrected CF lung cells. The signalling of mitogen-activated protein (MAP) kinases was further studied. We demonstrated that extracellular signal-regulated kinase (ERK1/2) and AP-1 activity was markedly enhanced in CF but not non-CF lung cells under oxidative stress. Consistently, inhibition of ERK1/2 in oxidative stress-exposed CF lung cells strongly decreased both the IL-8 production and CXCR1/2 expression. Therefore, targeting of ERK1/2 MAP kinase may be critical to reduce oxidative stress-mediated inflammation in lungs of CF patients.
Collapse
|
30
|
Vos JB, Datson NA, Rabe KF, Hiemstra PS. Exploring host-pathogen interactions at the epithelial surface: application of transcriptomics in lung biology. Am J Physiol Lung Cell Mol Physiol 2007; 292:L367-77. [PMID: 17041013 DOI: 10.1152/ajplung.00242.2006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The epithelial surface of the airways is the largest barrier-forming interface between the human body and the outside world. It is now well recognized that, at this strategic position, airway epithelial cells play an eminent role in host defense by recognizing and responding to microbial exposure. Conversely, inhaled microorganisms also respond to contact with epithelial cells. Our understanding of this cross talk is limited, requiring sophisticated experimental approaches to analyze these complex interactions. High-throughput technologies, such as DNA microarray analysis and serial analysis of gene expression (SAGE), have been developed to screen for gene expression levels at large scale within single experiments. Since their introduction, these hypothesis-generating technologies have been widely used in diverse areas such as oncology and brain research. Successful application of these genomics-based technologies has also revealed novel insights in host-pathogen interactions in both the host and pathogen. This review aims to provide an overview of the SAGE and microarray technology illustrated by their application in the analysis of host-pathogen interactions. In particular, the interactions between epithelial cells in the human lungs and clinically relevant microorganisms are the central focus of this review.
Collapse
Affiliation(s)
- Joost B Vos
- Department of Pulmonology, Leiden Amsterdam Center for Drug Research, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | |
Collapse
|
31
|
Emam A, Yu AR, Park HJ, Mahfoud R, Kus J, Burrows LL, Lingwood CA. Laboratory and clinical Pseudomonas aeruginosa strains do not bind glycosphingolipids in vitro or during type IV pili-mediated initial host cell attachment. MICROBIOLOGY-SGM 2006; 152:2789-2799. [PMID: 16946273 DOI: 10.1099/mic.0.28863-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The glycosphingolipids (GSLs) gangliotriaosylceramide (Gg(3)) and gangliotetraosylceramide (Gg(4)) have been implicated as receptors for type IV pili (T4P)-mediated Pseudomonas aeruginosa epithelial cell attachment. Since P. aeruginosa T4P are divided into five groups, the authors determined whether GSLs in general, and Gg(3) and Gg(4) in particular, are specifically bound and required for host epithelial cell attachment of clinical and laboratory strains within these groups. An enterohaemorrhagic Escherichia coli strain, CL56, known to bind to both Gg(3) and Gg(4), provided a positive control. TLC overlay showed no binding of more than 12 P. aeruginosa strains to either Gg(3) or Gg(4) (or other GSLs), while CL56 Gg(3)/Gg(4) binding was readily detectable. GSL ELISA similarly demonstrated no significant P. aeruginosa binding to Gg(3) or Gg(4), compared with CL56. Using a selective chemical inhibitor, epithelial cell GSL synthesis was abrogated, and Gg(3) and Gg(4) expression deleted, but P. aeruginosa attachment was not impaired. Target cell attachment was mediated by T4P, since non-piliated, but flagellated, mutants were unable to bind to the target cells. CFTR (cystic fibrosis transmembrane conductance regulator) has also been implicated as a receptor; however, in this work, overexpression of CFTR had no effect on P. aeruginosa binding. It is concluded that neither Gg(3) nor Gg(4) are specifically recognized by P. aeruginosa, and that endogenous GSLs do not have a role in the attachment of live intact P. aeruginosa to cultured lung epithelial cells. In contrast to whole piliated P. aeruginosa, T4P sheared from such bacteria showed significant Gg(3) and Gg(4) binding, which may explain the results of other studies.
Collapse
Affiliation(s)
- Aufaugh Emam
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Analyn R Yu
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Hyun-Joo Park
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Radhia Mahfoud
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Julianne Kus
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Lori L Burrows
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Clifford A Lingwood
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
32
|
Stalvey MS, Muller C, Schatz DA, Wasserfall CH, Campbell-Thompson ML, Theriaque DW, Flotte TR, Atkinson MA. Cystic fibrosis transmembrane conductance regulator deficiency exacerbates islet cell dysfunction after beta-cell injury. Diabetes 2006; 55:1939-45. [PMID: 16804061 DOI: 10.2337/db05-1647] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The cause of cystic fibrosis-related diabetes (CFRD) remains unknown, but cystic fibrosis transmembrane conductance regulator (CFTR) mutations contribute directly to multiple aspects of the cystic fibrosis phenotype. We hypothesized that susceptibility to islet dysfunction in cystic fibrosis is determined by the lack of functional CFTR. To address this, glycemia was assessed in CFTR null (CFTR(-/-)), C57BL/6J, and FVB/NJ mice after streptozotocin (STZ)-induced beta-cell injury. Fasting blood glucose levels were similar among age-matched non-STZ-administered animals, but they were significantly higher in CFTR(-/-) mice 4 weeks after STZ administration (288.4 +/- 97.4, 168.4 +/- 35.9, and 188.0 +/- 42.3 mg/dl for CFTR(-/-), C57BL/6J, and FVB/NJ, respectively; P < 0.05). After intraperitoneal glucose administration, elevated blood glucose levels were also observed in STZ-administered CFTR(-/-) mice. STZ reduced islets among all strains; however, only CFTR(-/-) mice demonstrated a negative correlation between islet number and fasting blood glucose (P = 0.02). To determine whether a second alteration associated with cystic fibrosis (i.e., airway inflammation) could impact glucose control, animals were challenged with Aspergillus fumigatus. The A. fumigatus-sensitized CFTR(-/-) mice demonstrated similar fasting and stimulated glucose responses in comparison to nonsensitized animals. These studies suggest metabolic derangements in CFRD originate from an islet dysfunction inherent to the CFTR(-/-) state.
Collapse
Affiliation(s)
- Michael S Stalvey
- Department of Pathology, University of Florida, College of Medicine, PO Box 100275, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Guilbault C, Novak JP, Martin P, Boghdady ML, Saeed Z, Guiot MC, Hudson TJ, Radzioch D. Distinct pattern of lung gene expression in theCftr-KO mice developing spontaneous lung disease compared with their littermate controls. Physiol Genomics 2006; 25:179-93. [PMID: 16418321 DOI: 10.1152/physiolgenomics.00206.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cystic fibrosis (CF) is caused by a defect in the CF transmembrane conductance regulator (CFTR) protein that functions as a chloride channel. Dysfunction of the CFTR protein results in salty sweat, pancreatic insufficiency, intestinal obstruction, male infertility, and severe pulmonary disease. Most of the morbidity and mortality of CF patients results from pulmonary complications. Differences in susceptibility to bacterial infection and variable degree of CF lung disease among CF patients remain unexplained. Many phenotypic expressions of the disease do not directly correlate with the type of mutation in the Cftr gene. Using a unique CF mouse model that mimics aspects of human CF lung disease, we analyzed the differential gene expression pattern between the normal lungs of wild-type mice (WT) and the affected lungs of CFTR knockout mice (KO). Using microarray analysis followed by quantitation of candidate gene mRNA and protein expression, we identified many interesting genes involved in the development of CF lung disease in mice. These findings point to distinct mechanisms of gene expression regulation between mice with CF and control mice.
Collapse
|
34
|
Büscher R, Hoerning A, Patel HH, Zhang S, Arthur DB, Grasemann H, Ratjen F, Insel PA. P2Y2 receptor polymorphisms and haplotypes in cystic fibrosis and their impact on Ca2+ influx. Pharmacogenet Genomics 2006; 16:199-205. [PMID: 16495779 DOI: 10.1097/01.fpc.0000189798.11468.6a] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Activation of P2Y2 receptors in airway epithelia by ATP and UTP stimulates a Ca2+-regulated Cl- channel, which regulates Cl- secretion in cystic fibrosis (CF). We hypothesized that genetic alterations in the P2Y2 receptor may act as disease modifiers in CF and thus analyzed the coding region of this gene for polymorphisms in 146 CF patients and 64 healthy controls. We also assessed the impact of the genetic variants on Ca2+-influx of P2Y2-null cells transfected with several P2Y2 receptor haplotypes. RESULTS We identified three frequent nonsynonymous P2Y2 receptor polymorphisms: Leu46Pro; Arg312Ser and Arg334Cys, of which only Arg312Ser was significantly more common in CF: Arg = 0.80, Ser = 0.20 (CF) vs. Arg = 0.72, Ser = 0.28 (controls), P < 0.05; for Leu46Pro, Leu = 0.92, Pro = 0.08 (CF) vs. Leu = 0.96, Pro = 0.04 (controls), P = 0.65 and for Arg334Cys, Arg = 0.79, Cys = 0.21 (CF) vs. Arg = 0.84, Cys = 0.16 (controls), P = 0.79. The most frequent haplotype was Leu46Leu/Arg312Arg/Arg334Arg (28% in CF, 31% in controls) but 6% of CF patients and none of the controls had Leu46Leu/Ser312Ser/Arg334Cys or Leu46Leu/Arg312Arg/Cys334Cys. To assess function of the receptor haplotypes, we stably transfected 1321N1 (P2Y-null) cells to similar levels of mRNA expression with Leu46Leu/Arg312Arg/Arg334Arg (wild-type), Leu46Leu/Ser312Ser/Arg334Arg and Leu46Leu/Arg312Arg/Cys334Cys and measured ATP-stimulated transient Ca2+-influx. Cells expressing the homozygous Cys334 variant had significantly increased Ca2+-influx compared to wild-type (P<0.01). The increase in Ca2+-influx was more pronounced in cells carrying the homozygous Ser312 variant than in cells with the other two genotypes (P<0.01). CONCLUSIONS These data indicate that P2Y2 receptor gene haplotypes influence intracellular Ca2+-release. Such genetic variants might therefore represent modifiers of Cl- secretion or of response to P2Y2 agonist therapy in CF.
Collapse
Affiliation(s)
- Rainer Büscher
- Department of Pediatrics, University Children's Hospital, Hufelandstr. 55, 45122 Essen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Boncoeur E, Tabary O, Bonvin E, Muselet C, Fritah A, Lefait E, Redeuilh G, Clement A, Jacquot J, Henrion-Caude A. Oxidative stress response results in increased p21WAF1/CIP1 degradation in cystic fibrosis lung epithelial cells. Free Radic Biol Med 2006; 40:75-86. [PMID: 16337881 DOI: 10.1016/j.freeradbiomed.2005.08.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2004] [Revised: 07/27/2005] [Accepted: 08/15/2005] [Indexed: 12/31/2022]
Abstract
Lung epithelium in cystic fibrosis (CF) patients is characterized by structural damage and altered repair due to oxidative stress. To gain insight into the oxidative stress-related damage in CF, we studied the effects of hyperoxia in CF and normal lung epithelial cell lines. In response to a 95% O2 exposure, both cell lines exhibited increased reactive oxygen species. Unexpectedly, the cyclin-dependent kinase inhibitor p21WAF1/CIP1 protein was undetectable in CF cells under hyperoxia, contrasting with increased levels of p21WAF1/CIP1 in normal cells. In both cell lines, exposure to hyperoxia led to S-phase arrest. Apoptotic features including nuclear condensation, DNA laddering, Annexin V incorporation, and elevated caspase-3 activity were not readily observed in CF cells in contrast to normal cells. Interestingly, treatment of hyperoxia-exposed CF cells with two proteasome inhibitors, MG132 and lactacystin, restored p21WAF1/CIP1 protein and was associated with an increase of caspase-3 activity. Moreover, transfection of p21WAF1/CIP1 protein in CF cells led to increased caspase-3 activity and was associated with increased apoptotic cell death, specifically under hyperoxia. Taken together, our data suggest that modulating p21WAF1/CIP1 degradation may have the therapeutic potential of reducing lung epithelial damage related to oxidative stress in CF patients.
Collapse
Affiliation(s)
- Emilie Boncoeur
- Inserm U719, Université Pierre et Marie Curie, Hôpital Saint-Antoine, 184 rue du Fg St Antoine, Bâtiment Kourilsky, 75571 Paris Cedex 12, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Reiniger N, Ichikawa JK, Pier GB. Influence of cystic fibrosis transmembrane conductance regulator on gene expression in response to Pseudomonas aeruginosa infection of human bronchial epithelial cells. Infect Immun 2005; 73:6822-30. [PMID: 16177360 PMCID: PMC1230967 DOI: 10.1128/iai.73.10.6822-6830.2005] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chronic lung infection by Pseudomonas aeruginosa causes significant morbidity in cystic fibrosis patients initiated by the failure of innate immune responses. We used microarray analysis and real-time PCR to detect transcriptional changes associated with cytokine production in isogenic bronchial epithelial cell lines with either wild-type (WT) or mutant cystic fibrosis transmembrane conductance regulator (CFTR) in response to P. aeruginosa infection. The transcription of four NF-kappaB-regulated cytokine genes was maximal in the presence of WT CFTR: the interleukin-8 (IL-8), IL-6, CXCL1, and intracellular adhesion molecule 1 (ICAM-1) genes. Analysis of protein expression in two cell lines paired for wild-type and mutant CFTR with three P. aeruginosa strains showed IL-6 and IL-8 expressions were consistently enhanced by the presence of WT CFTR in both cell lines with all three strains of P. aeruginosa, although some strains gave small IL-8 increases in cells with mutant CFTR. CXCL1 production showed consistent enhancement in cells with WT CFTR using all three bacterial strains in one cell line, whereas in the other cell line, CXCL1 showed a significant increase in cells with either WT or mutant CFTR. ICAM-1 was unchanged at the protein level in one of the cell lines but did show mild enhancement with WT CFTR in the other cell pair. Inhibitions of NF-kappaB prior to infection indicated differing degrees of dependence on NF-kappaB for production of the cytokines, contingent on the cell line. Cytokine effectors of innate immunity to P. aeruginosa were found to be positively influenced by the presence of WT CFTR, indicating a role in resistance to P. aeruginosa infection.
Collapse
Affiliation(s)
- Nina Reiniger
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
37
|
Tabary O, Corvol H, Boncoeur E, Chadelat K, Fitting C, Cavaillon JM, Clément A, Jacquot J. Adherence of airway neutrophils and inflammatory response are increased in CF airway epithelial cell-neutrophil interactions. Am J Physiol Lung Cell Mol Physiol 2005; 290:L588-96. [PMID: 16272177 DOI: 10.1152/ajplung.00013.2005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Persistent presence of PMN in airways is the hallmark of CF. Our aim was to assess PMN adherence, percentage of apoptotic airway PMN (aPMN), and IL-6 and IL-8 production when aPMN are in contact with airway epithelial cells. Before coculture, freshly isolated CF aPMN have greater spontaneous and TNF-alpha-induced apoptosis compared with blood PMN from the same CF patients and from aPMN of non-CF patients. We then examined cocultures of PMN isolated from CF and non-CF airways with bronchial epithelial cells bearing mutated cftr compared with cftr-corrected bronchial epithelial cells. After 18-h coculture, the number of CF aPMN adhered on cftr-deficient bronchial epithelial cells was 2.3-fold higher compared with the coculture of non-CF aPMN adhered on cftr-corrected bronchial epithelial cells. The percentage of CF apoptotic aPMN (9.5 +/- 0.2%) adhered on cftr-deficient bronchial epithelial cells was similar to the percentage of non-CF apoptotic aPMN adhered on cftr-corrected bronchial epithelial cells (10.3 +/- 0.7%). IL-6 and IL-8 levels were enhanced 6.5- and 2.9-fold, respectively, in coculture of CF aPMN adhered on cftr-deficient bronchial epithelial cells compared with coculture of non-CF aPMN adhered on cftr-corrected bronchial epithelial cells. Moreover, blocking surface adhesion molecules ICAM-1, VCAM-1, and E-selectin on cftr-deficient bronchial epithelial cells with specific MAbs inhibited the adherence of CF aPMN by 64, 51, and 50%, respectively. Our data suggest that in CF patients a high number of nonapoptotic PMN adhered on airway epithelium associated with elevated IL-6 and IL-8 levels may contribute to sustained and exaggerated inflammatory response in CF airways.
Collapse
Affiliation(s)
- Olivier Tabary
- Inserm U719, Hôpital St-Antoine, Bâtiment Kourilsky, 184, rue du Fg St-Antoine, F-75571 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Swiatecka-Urban A, Brown A, Moreau-Marquis S, Renuka J, Coutermarsh B, Barnaby R, Karlson KH, Flotte TR, Fukuda M, Langford GM, Stanton BA. The Short Apical Membrane Half-life of Rescued ΔF508-Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Results from Accelerated Endocytosis of ΔF508-CFTR in Polarized Human Airway Epithelial Cells. J Biol Chem 2005; 280:36762-72. [PMID: 16131493 DOI: 10.1074/jbc.m508944200] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The most common mutation in the cystic fibrosis transmembrane conductance regulator (CFTR) gene in individuals with cystic fibrosis, DeltaF508, causes retention of DeltaF508-CFTR in the endoplasmic reticulum and leads to the absence of CFTR Cl(-) channels in the apical plasma membrane. Rescue of DeltaF508-CFTR by reduced temperature or chemical means reveals that the DeltaF508 mutation reduces the half-life of DeltaF508-CFTR in the apical plasma membrane. Because DeltaF508-CFTR retains some Cl(-) channel activity, increased expression of DeltaF508-CFTR in the apical membrane could serve as a potential therapeutic approach for cystic fibrosis. However, little is known about the mechanisms responsible for the short apical membrane half-life of DeltaF508-CFTR in polarized human airway epithelial cells. Accordingly, the goal of this study was to determine the cellular defects in the trafficking of rescued DeltaF508-CFTR that lead to the decreased apical membrane half-life of DeltaF508-CFTR in polarized human airway epithelial cells. We report that in polarized human airway epithelial cells (CFBE41o-) the DeltaF508 mutation increased endocytosis of CFTR from the apical membrane without causing a global endocytic defect or affecting the endocytic recycling of CFTR in the Rab11a-specific apical recycling compartment.
Collapse
|
39
|
Flotte TR, Schwiebert EM, Zeitlin PL, Carter BJ, Guggino WB. Correlation between DNA transfer and cystic fibrosis airway epithelial cell correction after recombinant adeno-associated virus serotype 2 gene therapy. Hum Gene Ther 2005; 16:921-8. [PMID: 16076250 DOI: 10.1089/hum.2005.16.921] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recombinant adeno-associated virus serotype 2 (rAAV2)-based human gene therapy for cystic fibrosis has progressed through a series of preclinical studies and phase I and II clinical trials. This agent has shown an encouraging safety profile, consistent levels of DNA transfer, and positive evidence of short-term clinical improvement in lung function in a prospective, placebo-controlled phase II trial of aerosol administration. Nonetheless, it has been difficult to assess the relationship between its molecular action and the observed clinical improvements, because of the lack of positive results from a highly specific assay for vector mRNA. This issue is further complicated by the fact that the clinical vector utilizes a small cryptic rAAV2 promoter sequence that is less robust for mRNA expression than typical viral promoters. In this paper, we report the results of more sensitive assays performed on primary nasal cells harvested from rAAV2-CFTR gene therapy recipients. These studies demonstrate a correlation between the presence of rAAV2-CFTR vector genomes, CFTR mRNA expression, and cAMP-activated chloride channel function in these cells. The observation of sizeable physiological correction in the face of low mRNA levels may reflect the regulatory role of low levels of CFTR protein as an activator of other chloride channels.
Collapse
Affiliation(s)
- Terence R Flotte
- Department of Pediatrics, University of Florida, Gainesville, 32610, USA.
| | | | | | | | | |
Collapse
|
40
|
Correlation Between DNA Transfer and Cystic Fibrosis Airway Epithelial Cell Correction After Recombinant Adeno-Associated Virus Serotype 2 Gene Therapy. Hum Gene Ther 2005. [DOI: 10.1089/hum.2005.16.ft-99] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|