1
|
Liu Y, Wang D, Liu X, Yuan H, Liu D, Hu Y, Ning S. Biological and pharmacological roles of pyroptosis in pulmonary inflammation and fibrosis: recent advances and future directions. Cell Commun Signal 2024; 22:586. [PMID: 39639365 PMCID: PMC11619304 DOI: 10.1186/s12964-024-01966-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Pyroptosis, an inflammatory regulated cell death (RCD) mechanism, is characterized by cellular swelling, membrane rupture, and subsequent discharge of cellular contents, exerting robust proinflammatory effects. Recent studies have significantly advanced our understanding of pyroptosis, revealing that it can be triggered through inflammasome- and caspase-independent pathways, and interacts intricately with other RCD pathways (e.g., pyroptosis, necroptosis, ferroptosis, and cuproptosis). The pathogenesis of pulmonary fibrosis (PF), including idiopathic pulmonary fibrosis (IPF) and other interstitial lung diseases, involves a multifaceted interplay of factors such as pathogen infections, environmental pollutants, genetic variations, and immune dysfunction. This chronic and progressive interstitial lung disease is characterized by persistent inflammation, extracellular matrix (ECM) accumulation, and fibrotic alveolar wall thickening, which potentially contribute to deteriorated lung function. Despite recent advances in understanding pyroptosis, the mechanisms by which it regulates PF are not entirely elucidated, and effective strategies to improve clinical outcomes remain unclear. This review strives to deliver a comprehensive overview of the biological functions and molecular mechanisms of pyroptosis, exploring its roles in the pathogenesis of PF. Furthermore, it examines potential biomarkers and therapeutic agents for anti-fibrotic treatments.
Collapse
Affiliation(s)
- Ya Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, 411100, China
| | - Danxia Wang
- Department of Pharmacy, People's Hospital of Ningxiang City, Hunan University of Chinese Medicine, Changsha, 410600, China
| | - Xiang Liu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, 411100, China
| | - Haibin Yuan
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, School of Biomedical Sciences, Hunan University, Changsha, 410082, China
| | - Dan Liu
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
| | - Yixiang Hu
- Department of Clinical Pharmacy, Xiangtan Central Hospital (The Affiliated Hospital of Hunan University), Xiangtan, 411100, China.
| | - Shipeng Ning
- Department of Breast Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China.
| |
Collapse
|
2
|
Li C, Liu Q, Han L, Zhang H, Immler R, Rathkolb B, Secklehner J, de Angelis MH, Yildirim AÖ, Zeuschner D, Nicke A, Carlin LM, Sperandio M, Stoeger T, Rehberg M. The eATP/P2×7R Axis Drives Quantum Dot-Nanoparticle Induced Neutrophil Recruitment in the Pulmonary Microcirculation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404661. [PMID: 39364760 PMCID: PMC11615809 DOI: 10.1002/advs.202404661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/30/2024] [Indexed: 10/05/2024]
Abstract
Exposure to nanoparticles (NPs) is frequently associated with adverse cardiovascular effects. In contrast, NPs in nanomedicine hold great promise for precise lung-specific drug delivery, especially considering the extensive pulmonary capillary network that facilitates interactions with bloodstream-suspended particles. Therefore, exact knowledge about effects of engineered NPs within the pulmonary microcirculation are instrumental for future application of this technology in patients. To unravel the real-time dynamics of intravenously delivered NPs and their effects in the pulmonary microvasculature, we employed intravital microscopy of the mouse lung. Only PEG-amine-QDs, but not carboxyl-QDs triggered rapid neutrophil recruitment in microvessels and their subsequent recruitment to the alveolar space and was linked to cellular degranulation, TNF-α, and DAMP release into the circulation, particularly eATP. Stimulation of the ATP-gated receptor P2X7R induced expression of E-selectin on microvascular endothelium thereby mediating the neutrophilic immune response. Leukocyte integrins LFA-1 and MAC-1 facilitated adhesion and decelerated neutrophil crawling on the vascular surface. In summary, this study unravels the complex cascade of neutrophil recruitment during NP-induced sterile inflammation. Thereby we demonstrate novel adverse effects for NPs in the pulmonary microcirculation and provide critical insights for optimizing NP-based drug delivery and therapeutic intervention strategies, to ensure their efficacy and safety in clinical applications.
Collapse
Affiliation(s)
- Chenxi Li
- Institute of Lung Health and Immunity (LHI)Comprehensive Pneumology Center (CPC)Helmholtz Center MunichMember of the German Center for Lung Research (DZL)85764MunichGermany
- Department of Pulmonary and Critical CareShandong Provincial Hospital Affiliated toShandong First Medical UniversityJinanShandong250021China
| | - Qiongliang Liu
- Institute of Lung Health and Immunity (LHI)Comprehensive Pneumology Center (CPC)Helmholtz Center MunichMember of the German Center for Lung Research (DZL)85764MunichGermany
- Department of Thoracic SurgeryShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Lianyong Han
- Institute of Lung Health and Immunity (LHI)Comprehensive Pneumology Center (CPC)Helmholtz Center MunichMember of the German Center for Lung Research (DZL)85764MunichGermany
| | - Haiyun Zhang
- Institute of Lung Health and Immunity (LHI)Comprehensive Pneumology Center (CPC)Helmholtz Center MunichMember of the German Center for Lung Research (DZL)85764MunichGermany
| | - Roland Immler
- Walter Brendel Centre of Experimental MedicineBiomedical CenterInstitute of Cardiovascular Physiology and PathophysiologyLudwig‐Maximilians‐Universität München82152Planegg‐MartinsriedGermany
| | - Birgit Rathkolb
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum München85764NeuherbergGermany
- Institute of Experimental Animal Breeding and BiotechnologyLudwig‐Maximilians‐Universität München81377MunichGermany
| | - Judith Secklehner
- Cancer Research UK Scotland InstituteGlasgowG61 1BDUK
- School of Cancer SciencesUniversity of GlasgowGlasgowG12 8QQUK
| | - Martin Hrabe de Angelis
- Institute of Experimental Genetics and German Mouse ClinicHelmholtz Zentrum München85764NeuherbergGermany
- Chair of Experimental GeneticsTUM School of Life SciencesTechnische Universität München85354FreisingGermany
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI)Comprehensive Pneumology Center (CPC)Helmholtz Center MunichMember of the German Center for Lung Research (DZL)85764MunichGermany
- Institute of Experimental PneumologyLMU80539MunichGermany
| | - Dagmar Zeuschner
- Electron Microscopy FacilityMax Planck Institute for Molecular Biomedicine48149MuensterGermany
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and ToxicologyFaculty of MedicineLudwig‐Maximilians‐Universität München80336MunichGermany
| | - Leo M. Carlin
- Cancer Research UK Scotland InstituteGlasgowG61 1BDUK
- School of Cancer SciencesUniversity of GlasgowGlasgowG12 8QQUK
| | - Markus Sperandio
- Walter Brendel Centre of Experimental MedicineBiomedical CenterInstitute of Cardiovascular Physiology and PathophysiologyLudwig‐Maximilians‐Universität München82152Planegg‐MartinsriedGermany
| | - Tobias Stoeger
- Institute of Lung Health and Immunity (LHI)Comprehensive Pneumology Center (CPC)Helmholtz Center MunichMember of the German Center for Lung Research (DZL)85764MunichGermany
| | - Markus Rehberg
- Institute of Lung Health and Immunity (LHI)Comprehensive Pneumology Center (CPC)Helmholtz Center MunichMember of the German Center for Lung Research (DZL)85764MunichGermany
| |
Collapse
|
3
|
Yan X, Deqing Q, Yu F, Wang T, Xu D, Wen F, Chen J. Effects of cystic fibrosis transmembrane conductance regulator potentiators on clinical outcomes of chronic obstructive pulmonary disease: a systematic review and meta-analysis. Expert Rev Respir Med 2024; 18:893-902. [PMID: 39450920 DOI: 10.1080/17476348.2024.2421843] [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: 06/09/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 10/26/2024]
Abstract
INTRODUCTION Excessive mucus secretion is pivotal in chronic obstructive pulmonary disease (COPD) pathophysiology, particularly in chronic bronchitis phenotypes. Cystic fibrosis transmembrane conductance regulator (CFTR) has been implicated in COPD-related hypersecretion with acquired dysfunction, and emerged as a therapeutic target. However, the clinical efficacy of CFTR-potentiators in COPD remains controversial. METHODS We searched PubMed, Embase, Cochrane Library, China National Knowledge Infrastructure (CNKI), China Science and Technology Journal (CSTJ), and Wanfang Database to retrieve eligible studies published before 28 May 2024. RESULTS A total of 1172 COPD patients were included, meta-analysis showed that CFTR-potentiators significantly increased forced expiratory volume in 1 s (FEV1) and decreased sweat chloride and fibrinogen levels, with moderate-to-high quality evidence. However, no significant effects were observed on the percentage of detected FEV1 to predicted FEV1 (FEV1% predicted), forced vital capacity (FVC), COPD assessment test (CAT) score, St. George's Respiratory Questionnaire (SGRQ) score, or acute exacerbation times, with low-to-moderate quality evidence. CONCLUSION Our meta-analysis demonstrated CFTR-potentiators' potential efficacy in increasing FEV1, decreasing sweat chloride and fibrinogen levels, despite limited impacts on FEV1% predicted, FVC, CAT score, SGRQ score, and acute exacerbations, underscoring the necessity for future research to evaluate its effects on mucus hypersecretion, acute exacerbations, hospitalizations, and mortality in COPD management. Review registration PROSPERO Identifier: CRD42024538708.
Collapse
Affiliation(s)
- Xi Yan
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, and Department of Respiratory and Critical Care Medicine, and Center for High Altitude Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Quzhen Deqing
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, and Department of Respiratory and Critical Care Medicine, and Center for High Altitude Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Feng Yu
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, and Department of Respiratory and Critical Care Medicine, and Center for High Altitude Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Tao Wang
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, and Department of Respiratory and Critical Care Medicine, and Center for High Altitude Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Dan Xu
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, and Department of Respiratory and Critical Care Medicine, and Center for High Altitude Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Fuqiang Wen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, and Department of Respiratory and Critical Care Medicine, and Center for High Altitude Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jun Chen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, and Department of Respiratory and Critical Care Medicine, and Center for High Altitude Medicine, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Bani Melhim S, Douglas LE, Reihill JA, Downey DG, Martin SL. The effect of triple CFTR modulator therapy and azithromycin on ion channels and inflammation in cystic fibrosis. ERJ Open Res 2024; 10:00502-2024. [PMID: 39687397 PMCID: PMC11647873 DOI: 10.1183/23120541.00502-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/07/2024] [Indexed: 12/18/2024] Open
Abstract
Background Inflammation in cystic fibrosis (CF) airways is difficult to treat with well-established regimens often including azithromycin (AZ) as an immunomodulatory drug. As AZ has been reported to require CF transmembrane conductance regulator (CFTR) to be able to reduce interleukin (IL)-8 and given the emergence of highly effective CFTR "triple" modulator therapy (elexacaftor/tezacaftor/ivacaftor; ETI), the aim of this study was to investigate the effect of AZ and ETI, singly and in combination, on ion channel activity and to assess the potential anti-inflammatory effects. Methods Electrophysiological assessment of ETI and AZ was performed on three-dimensional cultures of primary CF human bronchial epithelial (HBE) cells using a Multi Trans-Epithelial Current Clamp. IL-8 from NuLi-1 (non-CF) and CuFi-1 (CF) cells treated with AZ was measured by ELISA. Inflammatory mediators from primary CF HBE cells exposed to tumour necrosis factor-α in the presence of AZ, ETI and their combination, were screened using the Proteome Profiler™ Human Cytokine Array Kit, with selected targets validated by ELISA. Results AZ did not alter CFTR chloride efflux, nor did it have any synergistic/antagonistic effect in combination with ETI. AZ reduced IL-8 in NuLi-1 but not CuFi-1 cells. The Proteome Profiler™ screen identified several disease-relevant cytokines that were modulated by treatment. Subsequent analysis by ELISA showed IL-8, IL-6, CXCL1 and granulocyte-macrophage colony-stimulating factor to be significantly reduced by treatment with ETI, but not by AZ. Conclusions Incorporating ETI into the standard of CF care provides an opportunity to re-evaluate therapeutic regimens to reduce treatment burden and safely discontinue chronic treatments such as AZ, without loss of clinical benefit. Identification of redundant treatments in the era of CFTR modulation may improve medication adherence and overcome potential adverse effects associated with the chronic use AZ and other drugs.
Collapse
Affiliation(s)
- Suhad Bani Melhim
- School of Pharmacy, Queen's University Belfast, Belfast, UK
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa, Jordan
| | | | | | - Damian G. Downey
- Wellcome-Wolfson Institute of Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | | |
Collapse
|
5
|
Beucher L, Gabillard-Lefort C, Baris OR, Mialet-Perez J. Monoamine oxidases: A missing link between mitochondria and inflammation in chronic diseases ? Redox Biol 2024; 77:103393. [PMID: 39405979 PMCID: PMC11525629 DOI: 10.1016/j.redox.2024.103393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024] Open
Abstract
The role of mitochondria spans from the regulation of the oxidative phosphorylation, cell metabolism and survival/death pathways to a more recently identified function in chronic inflammation. In stress situations, mitochondria release some pro-inflammatory mediators such as ATP, cardiolipin, reactive oxygen species (ROS) or mitochondrial DNA, that are believed to participate in chronic diseases and aging. These mitochondrial Damage-Associated Molecular Patterns (mito-DAMPs) can modulate specific receptors among which TLR9, NLRP3 and cGAS-STING, triggering immune cells activation and sterile inflammation. In order to counter the development of chronic diseases, a better understanding of the underlying mechanisms of low grade inflammation induced by mito-DAMPs is needed. In this context, monoamine oxidases (MAO), the mitochondrial enzymes that degrade catecholamines and serotonin, have recently emerged as potent regulators of chronic inflammation in obesity-related disorders, cardiac diseases, cancer, rheumatoid arthritis and pulmonary diseases. The role of these enzymes in inflammation embraces their action in both immune and non-immune cells, where they regulate monoamines levels and generate toxic ROS and aldehydes, as by-products of enzymatic reaction. Here, we discuss the more recent advances on the role and mechanisms of action of MAOs in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Lise Beucher
- Univ Angers, Inserm, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, F-49000, France
| | | | - Olivier R Baris
- Univ Angers, Inserm, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, F-49000, France
| | - Jeanne Mialet-Perez
- Univ Angers, Inserm, CNRS, MITOVASC, Equipe MitoLab, SFR ICAT, Angers, F-49000, France.
| |
Collapse
|
6
|
Jarosz-Griffiths H, Caley L, Lara-Reyna S, Savic S, Clifton I, McDermott M, Peckham D. Heightened mitochondrial respiration in CF cells is normalised by triple CFTR modulator therapy through mechanisms involving calcium. Heliyon 2024; 10:e39244. [PMID: 39498005 PMCID: PMC11532250 DOI: 10.1016/j.heliyon.2024.e39244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024] Open
Abstract
Background Cystic fibrosis (CF) is associated with increased resting energy expenditure. However, the introduction of elexacaftor/tezacaftor/ivacaftor (ETI) has resulted in a paradigm shift in nutritional status for many people with CF, with increase body mass index and reduction in the need for nutritional support. While these changes are likely to reflect improved clinical status and an associated downregulation of energy expenditure, they may also reflect drug-induced alterations in metabolic perturbations within CF cells. We hypothesise that some of these changes relate to normalisation of mitochondrial respiration in CF. Methods Using wild-type (WT) and F508del/F508del CFTR human bronchial epithelial cell lines (HBE cell lines) and baby hamster kidney (BHK) cells we examined the impact of ETI on cellular metabolism. We monitored mitochondrial respiration, using Seahorse extracellular flux assays and monitored mitochondrial reactive oxygen species (mROS) and intracellular calcium levels by flow cytometry. Results Increased mitochondrial respiration was found in HBE cell lines and BHK cells expressing CFTR F508del/F508del when assessing basal, maximal, spare respiratory capacities and ATP production, as well as increased mitochondrial ROS generated via forward electron transport. ETI significantly decreased basal, maximal, spare respiratory capacity and ATP production to WT levels or below. Calcium blocker, BAPTA-AM normalised mitochondrial respiration, suggesting a calcium-mediated mechanism. ETI decreased intracellular calcium levels in CF cells to the same extent as BAPTA-AM, highlighting the importance of calcium and chloride in mitochondrial respiration in CF. Conclusions CF cell lines exhibit increased mitochondrial respiration, which can be downregulated by ETI therapy through mechanisms involving calcium.
Collapse
Affiliation(s)
| | - L.R. Caley
- Leeds Institute of Medical Research, University of Leeds, United Kingdom
| | - S. Lara-Reyna
- Institute of Microbiology and Infection, University of Birmingham, United Kingdom
| | - S. Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, United Kingdom
- Department of Clinical Immunology and Allergy, St James's University Hospital, United Kingdom
| | - I.J. Clifton
- Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - M.F. McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, United Kingdom
| | - D.G. Peckham
- Leeds Institute of Medical Research, University of Leeds, United Kingdom
- Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| |
Collapse
|
7
|
Liu Y, Pan R, Ouyang Y, Gu W, Xiao T, Yang H, Tang L, Wang H, Xiang B, Chen P. Pyroptosis in health and disease: mechanisms, regulation and clinical perspective. Signal Transduct Target Ther 2024; 9:245. [PMID: 39300122 DOI: 10.1038/s41392-024-01958-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
Pyroptosis is a type of programmed cell death characterized by cell swelling and osmotic lysis, resulting in cytomembrane rupture and release of immunostimulatory components, which play a role in several pathological processes. Significant cellular responses to various stimuli involve the formation of inflammasomes, maturation of inflammatory caspases, and caspase-mediated cleavage of gasdermin. The function of pyroptosis in disease is complex but not a simple angelic or demonic role. While inflammatory diseases such as sepsis are associated with uncontrollable pyroptosis, the potent immune response induced by pyroptosis can be exploited as a therapeutic target for anti-tumor therapy. Thus, a comprehensive review of the role of pyroptosis in disease is crucial for further research and clinical translation from bench to bedside. In this review, we summarize the recent advancements in understanding the role of pyroptosis in disease, covering the related development history, molecular mechanisms including canonical, non-canonical, caspase 3/8, and granzyme-mediated pathways, and its regulatory function in health and multiple diseases. Moreover, this review also provides updates on promising therapeutic strategies by applying novel small molecule inhibitors and traditional medicines to regulate pyroptosis. The present dilemmas and future directions in the landscape of pyroptosis are also discussed from a clinical perspective, providing clues for scientists to develop novel drugs targeting pyroptosis.
Collapse
Affiliation(s)
- Yifan Liu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
- Department of Oncology, Xiangya Hospital, Central South University, 87th Xiangya road, Changsha, 410008, Hunan province, China
| | - Renjie Pan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Yuzhen Ouyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
- Department of Neurology, Xiangya Hospital, Central South University, 87th Xiangya road, Changsha, 410008, Hunan province, China
| | - Wangning Gu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Tengfei Xiao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Hongmin Yang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Ling Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| | - Bo Xiang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| | - Pan Chen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| |
Collapse
|
8
|
Chatterjee P, Moss CT, Omar S, Dhillon E, Hernandez Borges CD, Tang AC, Stevens DA, Hsu JL. Allergic Bronchopulmonary Aspergillosis (ABPA) in the Era of Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) Modulators. J Fungi (Basel) 2024; 10:656. [PMID: 39330416 PMCID: PMC11433030 DOI: 10.3390/jof10090656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Allergic bronchopulmonary aspergillosis (ABPA) is a hypersensitivity disease caused by Aspergillus fumigatus (Af), prevalent in persons with cystic fibrosis (CF) or asthma. In ABPA, Af proteases drive a T-helper cell-2 (Th2)-mediated allergic immune response leading to inflammation that contributes to permanent lung damage. Corticosteroids and antifungals are the mainstays of therapies for ABPA. However, their long-term use has negative sequelae. The treatment of patients with CF (pwCF) has been revolutionized by the efficacy of cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy. Pharmacological improvement in CFTR function with highly effective elexacaftor/tezacaftor/ivacaftor (ETI) provides unprecedented improvements in lung function and other clinical outcomes of pwCF. The mechanism behind the improvement in patient outcomes is a continued topic of investigation as our understanding of the role of CFTR function evolves. As ETI therapy gains traction in CF management, understanding its potential impact on ABPA, especially on the allergic immune response pathways and Af infection becomes increasingly crucial for optimizing patient outcomes. This literature review aims to examine the extent of these findings and expand our understanding of the already published research focusing on the intersection between ABPA therapeutic approaches in CF and the rapid impact of the evolving CFTR modulator landscape. While our literature search yielded limited reports specifically focusing on the role of CFTR modulator therapy on CF-ABPA, findings from epidemiologic and retrospective studies suggest the potential for CFTR modulator therapies to positively influence pulmonary outcomes by addressing the underlying pathophysiology of CF-ABPA, especially by decreasing inflammatory response and Af colonization. Thus, this review highlights the promising scope of CFTR modulator therapy in decreasing the overall prevalence and incidence of CF-ABPA.
Collapse
Affiliation(s)
- Paulami Chatterjee
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | - Carson Tyler Moss
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Sarah Omar
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | - Ekroop Dhillon
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| | | | - Alan C. Tang
- Department of Medicine, Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - David A. Stevens
- Division of Infectious Diseases and Geographic Medicine, Stanford University Medical School, Stanford, CA 94305, USA;
| | - Joe L. Hsu
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.C.); (S.O.); (E.D.)
| |
Collapse
|
9
|
Gogoi D, Yu H, Casey M, Baird R, Yusuf A, Forde L, O' Brien ME, West JR, Flagg T, McElvaney NG, Eden E, Mueller C, Brantly ML, Geraghty P, Reeves EP. Monocyte NLRP3 inflammasome and interleukin-1β activation modulated by alpha-1 antitrypsin therapy in deficient individuals. Thorax 2024; 79:822-833. [PMID: 38418195 PMCID: PMC11347198 DOI: 10.1136/thorax-2023-221071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/02/2024] [Indexed: 03/01/2024]
Abstract
INTRODUCTION Altered complement component 3 (C3) activation in patients with alpha-1 antitrypsin (AAT) deficiency (AATD) has been reported. To understand the potential impact on course of inflammation, the aim of this study was to investigate whether C3d, a cleavage-product of C3, triggers interleukin (IL)-1β secretion via activation of NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome. The objective was to explore the effect of AAT augmentation therapy in patients with AATD on the C3d/complement receptor 3 (CR3) signalling axis of monocytes and on circulating pro-inflammatory markers. METHODS Inflammatory mediators were detected in blood from patients with AATD (n=28) and patients with AATD receiving augmentation therapy (n=19). Inflammasome activation and IL-1β secretion were measured in monocytes of patients with AATD, and following C3d stimulation in the presence or absence of CR3 or NLRP3 inhibitors. RESULTS C3d acting via CR3 induces NLRP3 and pro-IL-1β production, and through induction of endoplasmic reticulum (ER) stress and calcium flux, triggers caspase-1 activation and IL-1β secretion. Treatment of individuals with AATD with AAT therapy results in decreased plasma levels of C3d (3.0±1.2 µg/mL vs 1.3±0.5 µg/mL respectively, p<0.0001) and IL-1β (115.4±30 pg/mL vs 73.3±20 pg/mL, respectively, p<0.0001), with a 2.0-fold decrease in monocyte NLRP3 protein expression (p=0.0303), despite continued ER stress activation. DISCUSSION These results provide strong insight into the mechanism of complement-driven inflammation associated with AATD. Although the described variance in C3d and NLRP3 activation decreased post AAT augmentation therapy, results demonstrate persistent C3d and monocyte ER stress, with implications for new therapeutics and clinical practice.
Collapse
Affiliation(s)
- Debananda Gogoi
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Howard Yu
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Michelle Casey
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rory Baird
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Azeez Yusuf
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Luke Forde
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michael E O' Brien
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jesse R West
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Tammy Flagg
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Edward Eden
- Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Christian Mueller
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, Worcester, MA, USA
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mark L Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Emer P Reeves
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
10
|
Li D, Donnelley M, Parsons D, Habgood MD, Schneider-Futschik EK. Extent of foetal exposure to maternal elexacaftor/tezacaftor/ivacaftor during pregnancy. Br J Pharmacol 2024; 181:2413-2428. [PMID: 38770951 DOI: 10.1111/bph.16417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Cystic fibrosis (CF) patients are living longer and healthier due to improved treatments, e.g. cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy elexacaftor/tezacaftor/ivacaftor (ETI), with treatment possibly occurring in pregnancy. The risk of ETI to foetuses remain unknown. Thus the effect of maternally administered ETI on foetal genetic and structural development was investigated. EXPERIMENTAL APPROACH Pregnant Sprague Dawley rats were orally treated with ETI (6.7 mg·kg-1·day-1 elexacaftor + 3.5 mg·kg-1·day-1 tezacaftor + 25 mg·kg-1·day-1 ivacaftor) for 7 days from E12 to E19. Tissue samples collected at E19 were analysed using histology and RNA sequencing. Histological changes and differentially expressed genes (DEG) were assessed. KEY RESULTS No overt structural abnormalities were found in foetal pancreas, liver, lung and small intestine after 7-day ETI exposure. Very few non-functionally associated DEG in foetal liver, lung and small intestine were identified using RNA-seq. 29 DEG were identified in thymus (27 up-regulated and two down-regulated) and most were functionally linked to each other. Gene ontology enrichment analysis revealed that multiple muscle-related terms were significantly enriched. Many more DEG were identified in cortex (44 up-regulated and four down-regulated) and a group of these were involved in central nervous system and brain development. CONCLUSION AND IMPLICATION Sub-chronic ETI treatment in late pregnancy does not appear to pose a significant risk to the genetic and structural development of many foetal tissues. However, significant gene changes in foetal thymic myoid cells and cortical neuronal development requires future follow-up studies to assess the risk to these organs.
Collapse
Affiliation(s)
- Danni Li
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Mark D Habgood
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| | - Elena K Schneider-Futschik
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
11
|
Perea L, Bottier M, Cant E, Richardson H, Dicker AJ, Shuttleworth M, Giam YH, Abo-Leyah H, Finch S, Huang JTJ, Shteinberg M, Goeminne PC, Polverino E, Altenburg J, Blasi F, Welte T, Aliberti S, Sibila O, Chalmers JD, Shoemark A. Airway IL-1β is related to disease severity and mucociliary function in bronchiectasis. Eur Respir J 2024; 64:2301966. [PMID: 38811046 DOI: 10.1183/13993003.01966-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/08/2024] [Indexed: 05/31/2024]
Abstract
RATIONALE The inflammasome is a key regulatory complex of the inflammatory response leading to interleukin-1β (IL-1β) release and activation. IL-1β amplifies inflammatory responses and induces mucus secretion and hyperconcentration in other diseases. The role of IL-1β in bronchiectasis has not been investigated. OBJECTIVES To characterise the role of airway IL-1β in bronchiectasis, including the association with mucus properties, ciliary function, airway inflammation, microbiome and disease severity. METHODS Stable bronchiectasis patients were enrolled in an international cohort study (n=269). IL-1β was measured in sputum supernatant. A validation cohort also had sputum rheology and hydration measured (n=53). For analysis, patients were stratified according to the median value of IL-1β in the population (high versus low) to compare disease severity, airway infection, microbiome (16S rRNA sequencing), inflammation and caspase-1 activity. Primary human nasal epithelial cells grown in air-liquid interface culture were used to study the effect of IL-1β on cilia function. RESULTS Patients with high sputum IL-1β had more severe disease, increased caspase-1 activity and an increased T-helper type 1, T-helper type 2 and neutrophil inflammatory response compared with patients with low IL-1β. The active-dominant form of IL-1β was associated with increased disease severity. High IL-1β was related to higher relative abundance of Proteobacteria in the microbiome and increased mucus solid content and viscoelastic properties. Chronic IL-1β treatment reduced the functionality of cilia and tight junctions of epithelial cells in vitro. CONCLUSIONS A subset of stable bronchiectasis patients show increased airway IL-1β, suggesting pulmonary inflammasome activation is linked with more severe disease, airway infection, mucus dehydration and epithelial dysfunction.
Collapse
Affiliation(s)
- Lidia Perea
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Biomedical Research Institute August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mathieu Bottier
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Royal Brompton Hospital, Guy's and St Thomas' NHS Foundation Trust, National Heart and Lung Institute, Imperial College London, London, UK
| | - Erin Cant
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Hollian Richardson
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Alison J Dicker
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Morven Shuttleworth
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Yan Hui Giam
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Hani Abo-Leyah
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Simon Finch
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Jeffrey T-J Huang
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Michal Shteinberg
- Pulmonology Institute and CF Center, Carmel Medical Center, Haifa, Israel
| | | | | | | | - Francesco Blasi
- Department of Pathophysiology and Transplantation, Università Degli Studi Di Milano, Milan, Italy
- Department of Internal Medicine Department, Respiratory Unit and Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - Tobias Welte
- Department of Respiratory Medicine, Medizinische Hochschule Hannover, Hannover, Germany
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Milan, Italy
| | - Oriol Sibila
- Respiratory Department, Hospital Clinic, IDIBAPS, CIBERES, University of Barcelona, Barcelona, Spain
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Senior authors contributed equally to this manuscript
| | - Amelia Shoemark
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
- Senior authors contributed equally to this manuscript
| |
Collapse
|
12
|
Cohen A, Mass A, Reiter J, Zangen DH, Cohen-Cymberknoh M. Long-term therapy with CFTR modulators consistently improves glucose metabolism in adolescents and adults with cystic fibrosis. Respir Med 2024; 228:107664. [PMID: 38759874 DOI: 10.1016/j.rmed.2024.107664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Impaired glycemic control and the subsequent development of Cystic fibrosis Related Diabetes (CFRD) are prevalent complications, affecting up to 50 % of adults with cystic fibrosis (CF). CFTR modulator (CFTRm) therapies improve pulmonary functions, reduce exacerbation rates, increase survival in people with CF (pwCF) and appear to have a positive effect on extrapulmonary manifestations, such as nutritional state, improvements in upper respiratory symptoms, and quality of life. Initial findings indicate that CFTRm may have a positive impact on short-term glycemic control; however, long-term effects remain uncertain at present. METHODS In this retrospective study, data were collected and analyzed on 15 pwCF, ages 13-37 years, started on CFTRm therapy. Oral Glucose Tolerance Test (OGTT) results were compared pre- and post-CFTRm therapy. RESULTS The 120-min OGTT value decreased from 159.7 mg/dL to 130.4 mg/dL post-CFTRm (p = 0.047). The average time elapsed between the two OGTTs was 49.87 months (ranging 9-157 months, median 38 months). Glycemic status improved in six pwCF (two CFRD to normal (NGT)/indeterminate (INDET) glucose tolerance; two impaired glucose tolerance (IGT) to INDET; two INDET to NGT) and worsened in one (IGT to CFRD). Six pwCF and NGT remained stable with no changes in glycemic status throughout the follow-up period. CONCLUSIONS CFTRm therapy may decelerate the glycemic control deterioration in pwCF over an extended period. These findings indicate the need for periodic OGTTs following the initiation of CFTRm therapy to appropriately adjust insulin requirements and prevent hypoglycemia. Further larger cohorts are required to authenticate and substantiate these findings.
Collapse
Affiliation(s)
- Amitay Cohen
- Division of Pediatric Endocrinology, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Alon Mass
- Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Joel Reiter
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; Pediatric Pulmonary Unit and Cystic fibrosis Center, Hadassah Medical Center, Jerusalem, Israel
| | - David Haim Zangen
- Division of Pediatric Endocrinology, Hadassah Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Israel
| | - Malena Cohen-Cymberknoh
- Faculty of Medicine, Hebrew University of Jerusalem, Israel; Pediatric Pulmonary Unit and Cystic fibrosis Center, Hadassah Medical Center, Jerusalem, Israel.
| |
Collapse
|
13
|
Jarosz-Griffiths HH, Gillgrass L, Caley LR, Spoletini G, Clifton IJ, Etherington C, Savic S, McDermott MF, Peckham D. Anti-inflammatory effects of elexacaftor/tezacaftor/ivacaftor in adults with cystic fibrosis heterozygous for F508del. PLoS One 2024; 19:e0304555. [PMID: 38820269 PMCID: PMC11142445 DOI: 10.1371/journal.pone.0304555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
Inflammation is a key driver in the pathogenesis of cystic fibrosis (CF). We assessed the effectiveness of elexacaftor/tezacaftor/ivacaftor (ETI) therapy on downregulating systemic and immune cell-derived inflammatory cytokines. We also monitored the impact of ETI therapy on clinical outcome. Adults with CF, heterozygous for F508del (n = 19), were assessed at baseline, one month and three months following ETI therapy, and clinical outcomes were measured, including sweat chloride, lung function, weight, neutrophil count and C-reactive protein (CRP). Cytokine quantifications were measured in serum and following stimulation of peripheral blood mononuclear cells (PBMCs) with lipopolysaccharide (LPS) and adenosine triphosphate and analysed using LEGEND plex™ Human Inflammation Panel 1 by flow cytometry (n = 19). ASC specks were measured in serum and caspase-1 activity and mRNA levels determined from stimulated PBMCs were determined. Patients remained stable over the study period. ETI therapy resulted in decreased sweat chloride concentrations (p < 0.0001), CRP (p = 0.0112) and neutrophil count (p = 0.0216) and increased percent predicted forced expiratory volume (ppFEV1) (p = 0.0399) from baseline to three months, alongside a trend increase in weight. Three months of ETI significantly decreased IL-18 (p< 0.0011, p < 0.0001), IL-1β (p<0.0013, p = 0.0476), IL-6 (p = 0.0109, p = 0.0216) and TNF (p = 0.0028, p = 0.0033) levels in CF serum and following PBMCs stimulation respectively. The corresponding mRNA levels were also found to be reduced in stimulated PBMCs, as well as reduced ASC specks and caspase-1 levels, indicative of NLRP3-mediated production of pro-inflammatory cytokines, IL-1β and IL-18. While ETI therapy is highly effective at reducing sweat chloride and improving lung function, it also displays potent anti-inflammatory properties, which are likely to contribute to improved long-term clinical outcomes.
Collapse
Affiliation(s)
| | - Lindsey Gillgrass
- Adult Cystic Fibrosis Unit, St James’s University Hospital, Leeds, United Kingdom
| | - Laura R. Caley
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Giulia Spoletini
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
- Adult Cystic Fibrosis Unit, St James’s University Hospital, Leeds, United Kingdom
| | - Ian J. Clifton
- Adult Cystic Fibrosis Unit, St James’s University Hospital, Leeds, United Kingdom
| | | | - Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Michael F. McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Daniel Peckham
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
- Adult Cystic Fibrosis Unit, St James’s University Hospital, Leeds, United Kingdom
| |
Collapse
|
14
|
Wei C, Fu M, Zhang H, Yao B. How is the P2X7 receptor signaling pathway involved in epileptogenesis? Neurochem Int 2024; 173:105675. [PMID: 38211839 DOI: 10.1016/j.neuint.2024.105675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
Epilepsy, a condition characterized by spontaneous recurrent epileptic seizures, is among the most prevalent neurological disorders. This disorder is estimated to affect approximately 70 million people worldwide. Although antiseizure medications are considered the first-line treatments for epilepsy, most of the available antiepileptic drugs are not effective in nearly one-third of patients. This calls for the development of more effective drugs. Evidence from animal models and epilepsy patients suggests that strategies that interfere with the P2X7 receptor by binding to adenosine triphosphate (ATP) are potential treatments for this patient population. This review describes the role of the P2X7 receptor signaling pathways in epileptogenesis. We highlight the genes, purinergic signaling, Pannexin1, glutamatergic signaling, adenosine kinase, calcium signaling, and inflammatory response factors involved in the process, and conclude with a synopsis of these key connections. By unraveling the intricate interplay between P2X7 receptors and epileptogenesis, this review provides ideas for designing potent clinical therapies that will revolutionize both prevention and treatment for epileptic patients.
Collapse
Affiliation(s)
- Caichuan Wei
- Department of Pediatrics, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuchang District, Wuhan, Hubei Province 430060, China
| | - Miaoying Fu
- Department of Pediatrics, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuchang District, Wuhan, Hubei Province 430060, China
| | - Haiju Zhang
- Department of Pediatrics, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuchang District, Wuhan, Hubei Province 430060, China
| | - Baozhen Yao
- Department of Pediatrics, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuchang District, Wuhan, Hubei Province 430060, China.
| |
Collapse
|
15
|
Li D, Zhu Y, Donnelley M, Parsons D, Habgood MD, Schneider-Futschik EK. Fetal drug exposure after maternally administered CFTR modulators Elexacaftor/Tezacaftor/Ivacaftor in a rat model. Biomed Pharmacother 2024; 171:116155. [PMID: 38232663 DOI: 10.1016/j.biopha.2024.116155] [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: 11/20/2023] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND The potential effects of the very effective cystic fibrosis triple combination drug, Elexacaftor/Tezacaftor/Ivacaftor (ETI) in pregnancy on prenatal development of offspring remain largely unknown. RESEARCH QUESTION We aimed to investigate the fetal tissue distribution pattern of maternally administered ETI by placental transfer in the rat fetuses. STUDY DESIGN AND METHODS Sprague Dawley pregnant rats were administered ETI (6.7 mg/kg/d elexacaftor + 3.5 mg/kg/d tezacaftor + 25 mg/kg/d ivacaftor) traced with [3 H]-ivacaftor in single dose acute experiments (intraperitoneal injection) or treated orally with ETI (the same dose) for 7 days in sub-chronic experiments. Fetal tissue samples were collected at embryonic day (E) 19 and analyzed using liquid scintillation counting for acute experiments or liquid chromatography-mass spectrometry for sub-chronic experiments. RESULTS On day E19, after acute exposure, the entry of ivacaftor into fetal brain (brain/plasma concentration ratios <50%) was significantly lower than to other tissues (>100%). However, after sub-chronic exposure, the entry of all 3 components into the developing brain was comparably extensive as into other tissues (tissue/plasma ratios, 260 - 1000%). Each component of ETI accumulated in different fetal tissues to approximately equal extent. Inter-litter differences on fetal drug distribution were found in cortex for ivacaftor, muscle for tezacaftor and cortex and mid/hindbrain for elexacaftor. Fetal plasma concentrations of ETI (ng/mL) were variable between litters. The entry of ivacaftor and tezacaftor into adult brain appeared to be restricted (<100%). INTERPRETATION Fetal rats are exposed to maternally ingested ETI after sub-chronic exposure, potentially impacting fetal development. The brain entry data highlights the need for attention be paid to any long-term potential effects ETI exposure could have on normal brain development.
Collapse
Affiliation(s)
- Danni Li
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Yimin Zhu
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Martin Donnelley
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia 5006, Australia
| | - David Parsons
- Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia; Department of Respiratory and Sleep Medicine, Women's and Children's Hospital, 72 King William Rd, North Adelaide, South Australia 5006, Australia
| | - Mark D Habgood
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elena K Schneider-Futschik
- Department of Biochemistry & Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
16
|
Wellems D, Hu Y, Jennings S, Wang G. Loss of CFTR function in macrophages alters the cell transcriptional program and delays lung resolution of inflammation. Front Immunol 2023; 14:1242381. [PMID: 38035088 PMCID: PMC10687418 DOI: 10.3389/fimmu.2023.1242381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder caused by mutations in the CF Transmembrane-conductance Regulator (CFTR) gene. The most severe pathologies of CF occur in the lung, manifesting as chronic bacterial infection, persistent neutrophilic inflammation, and mucopurulent airway obstruction. Despite increasing knowledge of the CF primary defect and the resulting clinical sequelae, the relationship between the CFTR loss of function and the neutrophilic inflammation remains incompletely understood. Here, we report that loss of CFTR function in macrophages causes extended lung inflammation. After intratracheal inoculation with Pseudomonas aeruginosa, mice with a macrophage-specific Cftr-knockout (Mac-CF) were able to mount an effective host defense to clear the bacterial infection. However, three days post-inoculation, Mac-CF lungs demonstrated significantly more neutrophil infiltration and higher levels of inflammatory cytokines, suggesting that Mac-CF mice had a slower resolution of inflammation. Single-cell RNA sequencing revealed that absence of CFTR in the macrophages altered the cell transcriptional program, affecting the cell inflammatory and immune responses, antioxidant system, and mitochondrial respiration. Thus, loss of CFTR function in macrophages influences cell homeostasis, leading to a dysregulated cellular response to infection that may exacerbate CF lung disease.
Collapse
Affiliation(s)
| | | | | | - Guoshun Wang
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
17
|
Arslan M, Bahadir Z, Basiaga ML, Chalmers SJ, Demirel N. A pediatric cystic fibrosis arthropathy case who responded to Elexacaftor/Tezacaftor/Ivacaftor therapy. J Cyst Fibros 2023; 22:1120-1122. [PMID: 37709627 DOI: 10.1016/j.jcf.2023.08.012] [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: 02/28/2023] [Revised: 08/04/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023]
Abstract
Cystic fibrosis arthropathy (CFA) is a transient, intermittent form of arthritis that cannot be associated with any other disease other than CF thus making CFA a diagnosis of exclusion. NSAIDs, short-term intermittent splinting, glucocorticoids, and disease-modifying anti-rheumatic drugs are treatment options for CFA. Currently, there is no consensus on how to best treat CFA. Diagnosis and treatment of CFA remain a challenge for physicians and people with CF. The newest CFTR modulator therapy, elexacaftor/tezacaftor/ivacaftor (ETI), was approved by the FDA recently for children over the age of 6 with at least one Phe508del allele in the CFTR gene. Multiple clinical benefits of ETI in pulmonary functions and overall disease burden have been reported since its approval, however, the data on the musculoskeletal therapeutic benefits of ETI has been limited. In this report, we present a 7-year-old female with CF whose CFA symptoms resolved after starting ETI therapy.
Collapse
Affiliation(s)
| | - Zeynep Bahadir
- Istanbul Medipol University School of Medicine, Istanbul, Turkey
| | - Matthew L Basiaga
- Mayo Clinic, Department of Pediatric and Adolescent Medicine, Division of Pediatric Rheumatology, Rochester, MN, United States of America
| | - Sarah J Chalmers
- Mayo Clinic, Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Rochester, MN, United States of America
| | - Nadir Demirel
- Mayo Clinic, Department of Pediatric and Adolescent Medicine, Division of Pediatric Pulmonology, Rochester, MN, United States of America
| |
Collapse
|
18
|
Caley LR, Jarosz-Griffiths HH, Smith L, Gale L, Barrett J, Kinsey L, Davey V, Nash M, Jones AM, Whitehouse JL, Shimmin D, Floto RA, White H, Peckham DG. Body mass index and nutritional intake following Elexacaftor/Tezacaftor/Ivacaftor modulator therapy in adults with cystic fibrosis. J Cyst Fibros 2023; 22:1002-1009. [PMID: 37422432 DOI: 10.1016/j.jcf.2023.06.010] [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: 02/02/2023] [Revised: 06/01/2023] [Accepted: 06/25/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Elexacaftor/Tezacaftor/Ivacaftor (ETI) modulator therapy is often associated with increased body mass index (BMI) in people with cystic fibrosis (CF). This is thought to reflect improved clinical stability and increased appetite and nutritional intake. We explored the change in BMI and nutritional intake following ETI modulator therapy in adults with CF. METHODS Dietary intake, measured with myfood24®, and BMI were collected from adults with CF at baseline and follow-up as part of an observational study. Changes in BMI and nutritional intake in participants who commenced ETI therapy between time points were assessed. To contextualize findings, we also assessed changes in BMI and nutritional intake between study points in a group on no modulators. RESULTS In the pre and post ETI threapy group (n = 40), BMI significantly increased from 23.0 kg/m2 (IQR 21.4, 25.3) at baseline to 24.6 kg/m2 (IQR 23.0, 26.7) at follow-up (p<0.001), with a median of 68 weeks between time points (range 20-94 weeks) and median duration of ETI therapy was 23 weeks (range 7-72 weeks). There was a significant decrease in energy intake from 2551 kcal/day (IQR 2107, 3115) to 2153 kcal/day (IQR 1648, 2606), p<0.001. In the no modulator group (n = 10), BMI and energy intake did not significantly change between time points (p>0.05), a median of 28 weeks apart (range 20-76 weeks). CONCLUSIONS These findings tentatively suggest that the increase in BMI with ETI therapy may not simply be attributable to an increase in oral intake. Further exploration into the underlying aetiology of weight gain with ETI therapy is needed.
Collapse
Affiliation(s)
- L R Caley
- Leeds Institute of Medical Research at St James's, University of Leeds, UK; Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| | | | - L Smith
- Leeds Institute of Health Sciences, University Hospital, University of Leeds, UK
| | - L Gale
- Cambridge Centre for Lung Infection, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - J Barrett
- West Midlands Regional Adult CF Centre, University Hospitals Birmingham NHS, Foundation Trust, Birmingham, UK
| | - L Kinsey
- Manchester Adult Cystic Fibrosis Centre, Wythenshawe Hospital, Manchester University Hospitals NHS Foundation Trust, UK
| | - V Davey
- Dietetic Department, Manchester University NHS Foundation Trust, Manchester Royal Infirmary, Oxford Road, Manchester, UK
| | - M Nash
- North East Essex Community Services, East Suffolk and North Essex NHS Foundation Trust, UK
| | - A M Jones
- Manchester Adult Cystic Fibrosis Centre, Wythenshawe Hospital, Manchester University Hospitals NHS Foundation Trust, UK
| | - J L Whitehouse
- West Midlands Regional Adult CF Centre, University Hospitals Birmingham NHS, Foundation Trust, Birmingham, UK
| | - D Shimmin
- Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - R A Floto
- Cambridge Centre for Lung Infection, Royal Papworth Hospital NHS Foundation Trust, Cambridge, UK; Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - H White
- Leeds Beckett University, Nutrition, Health & Environment, Leeds, UK
| | - D G Peckham
- Leeds Institute of Medical Research at St James's, University of Leeds, UK; Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| |
Collapse
|
19
|
Carbone A, Vitullo P, Di Gioia S, Conese M. Lung Inflammatory Genes in Cystic Fibrosis and Their Relevance to Cystic Fibrosis Transmembrane Conductance Regulator Modulator Therapies. Genes (Basel) 2023; 14:1966. [PMID: 37895314 PMCID: PMC10606852 DOI: 10.3390/genes14101966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Cystic fibrosis (CF) is a monogenic syndrome determined by over 2000 mutations in the CF Transmembrane Conductance Regulator (CFTR) gene harbored on chromosome 7. In people with CF (PWCF), lung disease is the major determinant of morbidity and mortality and is characterized by a clinical phenotype which differs in the presence of equal mutational assets, indicating that genetic and environmental modifiers play an important role in this variability. Airway inflammation determines the pathophysiology of CF lung disease (CFLD) both at its onset and progression. In this narrative review, we aim to depict the inflammatory process in CF lung, with a particular emphasis on those genetic polymorphisms that could modify the clinical outcome of the respiratory disease in PWCF. The natural history of CF has been changed since the introduction of CFTR modulator therapies in the clinical arena. However, also in this case, there is a patient-to-patient variable response. We provide an overview on inflammatory/immunity gene variants that affect CFLD severity and an appraisal of the effects of CFTR modulator therapies on the inflammatory process in lung disease and how this knowledge may advance the optimization of the management of PWCF.
Collapse
Affiliation(s)
- Annalucia Carbone
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Pamela Vitullo
- Cystic Fibrosis Support Center, Ospedale “G. Tatarella”, 71042 Cerignola, Italy;
| | - Sante Di Gioia
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| | - Massimo Conese
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy; (A.C.); (S.D.G.)
| |
Collapse
|
20
|
Aridgides DS, Mellinger DL, Gwilt LL, Hampton TH, Mould DL, Hogan DA, Ashare A. Comparative effects of CFTR modulators on phagocytic, metabolic and inflammatory profiles of CF and nonCF macrophages. Sci Rep 2023; 13:11995. [PMID: 37491532 PMCID: PMC10368712 DOI: 10.1038/s41598-023-38300-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/06/2023] [Indexed: 07/27/2023] Open
Abstract
Macrophage dysfunction has been well-described in Cystic Fibrosis (CF) and may contribute to bacterial persistence in the lung. Whether CF macrophage dysfunction is related directly to Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) in macrophages or an indirect consequence of chronic inflammation and mucostasis is a subject of ongoing debate. CFTR modulators that restore CFTR function in epithelial cells improve global CF monocyte inflammatory responses but their direct effects on macrophages are less well understood. To address this knowledge gap, we measured phagocytosis, metabolism, and cytokine expression in response to a classical CF pathogen, Pseudomonas aeruginosa in monocyte-derived macrophages (MDM) isolated from CF F508del homozygous subjects and nonCF controls. Unexpectedly, we found that CFTR modulators enhanced phagocytosis in both CF and nonCF cohorts. CFTR triple modulators also inhibited MDM mitochondrial function, consistent with MDM activation. In contrast to studies in humans where CFTR modulators decreased serum inflammatory cytokine levels, modulators did not alter cytokine secretion in our system. Our studies therefore suggest modulator induced metabolic effects may promote bacterial clearance in both CF and nonCF monocyte-derived macrophages.
Collapse
Affiliation(s)
- Daniel S Aridgides
- Section of Pulmonary and Critical Care Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
| | - Diane L Mellinger
- Section of Pulmonary and Critical Care Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Lorraine L Gwilt
- Section of Pulmonary and Critical Care Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Thomas H Hampton
- Department of Microbiology and Immunology, Dartmouth College, Geisel School of Medicine, Hanover, NH, USA
| | - Dallas L Mould
- Department of Microbiology and Immunology, Dartmouth College, Geisel School of Medicine, Hanover, NH, USA
| | - Deborah A Hogan
- Department of Microbiology and Immunology, Dartmouth College, Geisel School of Medicine, Hanover, NH, USA
| | - Alix Ashare
- Section of Pulmonary and Critical Care Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Department of Microbiology and Immunology, Dartmouth College, Geisel School of Medicine, Hanover, NH, USA
| |
Collapse
|
21
|
Schmidt H, Höpfer LM, Wohlgemuth L, Knapp CL, Mohamed AOK, Stukan L, Münnich F, Hüsken D, Koller AS, Stratmann AEP, Müller P, Braun CK, Fabricius D, Bode SFN, Huber-Lang M, Messerer DAC. Multimodal analysis of granulocytes, monocytes, and platelets in patients with cystic fibrosis before and after Elexacaftor-Tezacaftor-Ivacaftor treatment. Front Immunol 2023; 14:1180282. [PMID: 37457734 PMCID: PMC10347380 DOI: 10.3389/fimmu.2023.1180282] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/15/2023] [Indexed: 07/18/2023] Open
Abstract
Cystic fibrosis (CF) is a monogenetic disease caused by an impairment of the cystic fibrosis transmembrane conductance regulator (CFTR). CF affects multiple organs and is associated with acute and chronic inflammation. In 2020, Elexacaftor-Tezacaftor-Ivacaftor (ETI) was approved to enhance and restore the remaining CFTR functionality. This study investigates cellular innate immunity, with a focus on neutrophil activation and phenotype, comparing healthy volunteers with patients with CF before (T1, n = 13) and after six months (T2, n = 11) of ETI treatment. ETI treatment reduced sweat chloride (T1: 95 mmol/l (83|108) vs. T2: 32 mmol/l (25|62), p < 0.01, median, first|third quartile) and significantly improved pulmonal function (FEV1 T1: 2.66 l (1.92|3.04) vs. T2: 3.69 l (3.00|4.03), p < 0.01). Moreover, there was a significant decrease in the biomarker human epididymis protein 4 (T1: 6.2 ng/ml (4.6|6.3) vs. T2: 3.0 ng/ml (2.2|3.7), p < 0.01) and a small but significant decrease in matrix metallopeptidase 9 (T1: 45.5 ng/ml (32.5|140.1) vs. T2: 28.2 ng/ml (18.2|33.6), p < 0.05). Neutrophil phenotype (CD10, CD11b, CD62L, and CD66b) and function (radical oxygen species generation, chemotactic and phagocytic activity) remained largely unaffected by ETI treatment. Likewise, monocyte phenotype and markers of platelet activation were similar at T1 and T2. In summary, the present study confirmed a positive impact on patients with CF after ETI treatment. However, neither beneficial nor harmful effects of ETI treatment on cellular innate immunity could be detected, possibly due to the study population consisting of patients with well-controlled CF.
Collapse
Affiliation(s)
- Hanna Schmidt
- Department of Pediatric and Adolescent Medicine, University Hospital Ulm, Ulm, Germany
| | - Larissa Melina Höpfer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Christiane Leonie Knapp
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | | | - Laura Stukan
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Frederik Münnich
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Dominik Hüsken
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | | | | | - Paul Müller
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Christian Karl Braun
- Department of Pediatric and Adolescent Medicine, University Hospital Ulm, Ulm, Germany
- Institute of Transfusion Medicine, Ulm University, Ulm, Germany
- Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service and University Hospital Ulm, Ulm, Germany
| | - Dorit Fabricius
- Department of Pediatric and Adolescent Medicine, University Hospital Ulm, Ulm, Germany
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
- Department of Transfusion Medicine and Hemostaseology, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
22
|
Nichols DP, Morgan SJ, Skalland M, Vo AT, Van Dalfsen JM, Singh SB, Ni W, Hoffman LR, McGeer K, Heltshe SL, Clancy JP, Rowe SM, Jorth P, Singh PK. Pharmacologic improvement of CFTR function rapidly decreases sputum pathogen density, but lung infections generally persist. J Clin Invest 2023; 133:e167957. [PMID: 36976651 PMCID: PMC10178839 DOI: 10.1172/jci167957] [Citation(s) in RCA: 94] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
BackgroundLung infections are among the most consequential manifestations of cystic fibrosis (CF) and are associated with reduced lung function and shortened survival. Drugs called CF transmembrane conductance regulator (CFTR) modulators improve activity of dysfunctional CFTR channels, which is the physiological defect causing CF. However, it is unclear how improved CFTR activity affects CF lung infections.MethodsWe performed a prospective, multicenter, observational study to measure the effect of the newest and most effective CFTR modulator, elexacaftor/tezacaftor/ivacaftor (ETI), on CF lung infections. We studied sputum from 236 people with CF during their first 6 months of ETI using bacterial cultures, PCR, and sequencing.ResultsMean sputum densities of Staphylococcus aureus, Pseudomonas aeruginosa, Stenotrophomonas maltophilia, Achromobacter spp., and Burkholderia spp. decreased by 2-3 log10 CFU/mL after 1 month of ETI. However, most participants remained culture positive for the pathogens cultured from their sputum before starting ETI. In those becoming culture negative after ETI, the pathogens present before treatment were often still detectable by PCR months after sputum converted to culture negative. Sequence-based analyses confirmed large reductions in CF pathogen genera, but other bacteria detected in sputum were largely unchanged. ETI treatment increased average sputum bacterial diversity and produced consistent shifts in sputum bacterial composition. However, these changes were caused by ETI-mediated decreases in CF pathogen abundance rather than changes in other bacteria.ConclusionsTreatment with the most effective CFTR modulator currently available produced large and rapid reductions in traditional CF pathogens in sputum, but most participants remain infected with the pathogens present before modulator treatment.Trial RegistrationClinicalTrials.gov NCT04038047.FundingThe Cystic Fibrosis Foundation and the NIH.
Collapse
Affiliation(s)
| | - Sarah J. Morgan
- Departments of Microbiology and Medicine, University of Washington, Seattle, Washington, USA
| | - Michelle Skalland
- Therapeutics Development Network Coordinating Center, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Anh T. Vo
- Departments of Microbiology and Medicine, University of Washington, Seattle, Washington, USA
| | - Jill M. Van Dalfsen
- Therapeutics Development Network Coordinating Center, Seattle Children’s Research Institute, Seattle, Washington, USA
| | | | - Wendy Ni
- Departments of Microbiology and Medicine, University of Washington, Seattle, Washington, USA
| | | | - Kailee McGeer
- Departments of Microbiology and Medicine, University of Washington, Seattle, Washington, USA
| | - Sonya L. Heltshe
- Department of Pediatrics and
- Therapeutics Development Network Coordinating Center, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - John P. Clancy
- Department of Medicine, University of Alabama, Birmingham, Alabama, USA
| | - Steven M. Rowe
- Department of Medicine, University of Alabama, Birmingham, Alabama, USA
| | - Peter Jorth
- Departments of Pathology and Laboratory Medicine, Medicine, and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Pradeep K. Singh
- Departments of Microbiology and Medicine, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
23
|
Bruscia EM. The effects of elexafactor/tezafactor/ivacaftor beyond the epithelium: spurring macrophages to fight infections. Eur Respir J 2023; 61:61/4/2300216. [PMID: 37003613 DOI: 10.1183/13993003.00216-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/25/2023] [Indexed: 04/03/2023]
|
24
|
Cavinato L, Luly FR, Pastore V, Chiappetta D, Sangiorgi G, Ferrara E, Baiocchi P, Mandarello G, Cimino G, Del Porto P, Ascenzioni F. Elexacaftor/tezacaftor/ivacaftor corrects monocyte microbicidal deficiency in cystic fibrosis. Eur Respir J 2023; 61:2200725. [PMID: 36455959 PMCID: PMC10066567 DOI: 10.1183/13993003.00725-2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 11/12/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND Cystic fibrosis (CF), which is caused by mutations in the CF transmembrane conductance regulator (CFTR), is characterised by chronic bacterial lung infection and inflammation. In CF, monocytes and monocyte-derived macrophages have been shown to display defective phagocytosis and antimicrobial activity against relevant lung pathogens, including Pseudomonas aeruginosa. Thus, we addressed the effect of CFTR triple modulator therapy (elexacaftor/tezacaftor/ivacaftor (ETI)) on the activity of CF monocytes against P. aeruginosa. METHODS Monocytes from people with CF (PWCF) before and after 1 and 6 months of ETI therapy were isolated from blood and infected with P. aeruginosa to assess phagocytic activity and intracellular bacterial killing. The oxidative burst and interleukin-6 secretion were also determined. Monocytes from healthy controls were also included. RESULTS Longitudinal analysis of the clinical parameters confirmed an improvement of lung function and lung microbiology by ETI. Both the phagocytic and microbicidal deficiencies of CF monocytes also improved significantly, although not completely. Furthermore, we measured an exuberant oxidative burst in CF monocytes before therapy, which was reduced considerably by ETI. This led to an improvement of reactive oxygen species-dependent bactericidal activity. Inflammatory response to bacterial stimuli was also lowered compared with pre-therapy. CONCLUSIONS PWCF on ETI therapy, in a real-life setting, in addition to clinical recovery, showed significant improvement in monocyte activity against P. aeruginosa, which may have contributed to the overall effect of ETI on pulmonary disease. This also suggests that CF monocyte dysfunctions may be specifically targeted to ameliorate lung function in CF.
Collapse
Affiliation(s)
- Luca Cavinato
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Francesco R Luly
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Valentina Pastore
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Daniele Chiappetta
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Gloria Sangiorgi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Eva Ferrara
- Cystic Fibrosis Reference Center of Lazio Region, Policlinico Umberto I, Rome, Italy
| | - Pia Baiocchi
- Department of Public Health and Infectious Disease, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Mandarello
- Department of Onco-Hematology, Immunotransfusion Service, ASL Viterbo, Rome, Italy
| | - Giuseppe Cimino
- Cystic Fibrosis Reference Center of Lazio Region, Policlinico Umberto I, Rome, Italy
| | - Paola Del Porto
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Fiorentina Ascenzioni
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Tümmler B. Post-approval studies with the CFTR modulators Elexacaftor-Tezacaftor-Ivacaftor. Front Pharmacol 2023; 14:1158207. [PMID: 37025483 PMCID: PMC10072268 DOI: 10.3389/fphar.2023.1158207] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/07/2023] [Indexed: 03/31/2023] Open
Abstract
Triple combination therapy with the CFTR modulators elexacaftor (ELX), tezacaftor (TEZ) and ivacaftor (IVA) has been qualified as a game changer in cystic fibrosis (CF). We provide an overview of the body of literature on ELX/TEZ/IVA published between November 2019 and February 2023 after approval by the regulators. Recombinant ELX/TEZ/IVA-bound Phe508del CFTR exhibits a wild type conformation in vitro, but in patient's tissue a CFTR glyoisoform is synthesized that is distinct from the wild type and Phe508del isoforms. ELX/TEZ/IVA therapy improved the quality of life of people with CF in the real-life setting irrespective of their anthropometry and lung function at baseline. ELX/TEZ/IVA improved sinonasal and abdominal disease, lung function and morphology, airway microbiology and the basic defect of impaired epithelial chloride and bicarbonate transport. Pregnancy rates were increasing in women with CF. Side effects of mental status changes deserve particular attention in the future.
Collapse
Affiliation(s)
- Burkhard Tümmler
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research, Hannover Medical School, Hannover, Germany
| |
Collapse
|
26
|
Wang Q, Peng D, Huang B, Men L, Jiang T, Huo S, Wang M, Guo J, Lv J, Lin L. Notopterol Ameliorates Hyperuricemia-Induced Cardiac Dysfunction in Mice. Pharmaceuticals (Basel) 2023; 16:ph16030361. [PMID: 36986461 PMCID: PMC10052463 DOI: 10.3390/ph16030361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/02/2023] Open
Abstract
Notopterol is a naturally occurring furanocoumarin compound found in the root of Notopterygium incisum. Hyperuricemia involves the activation of chronic inflammation and leads to cardiac damage. Whether notopterol has cardioprotective potential in hyperuricemia mice remains elusive. The hyperuricemic mouse model was constructed by administration of potassium oxonate and adenine every other day for six weeks. Notopterol (20 mg/kg) and allopurinol (10 mg/kg) were given daily as treatment, respectively. The results showed that hyperuricemia dampened heart function and reduced exercise capacity. Notopterol treatment improved exercise capacity and alleviated cardiac dysfunction in hyperuricemic mice. P2X7R and pyroptosis signals were activated both in hyperuricemic mice and in uric acid-stimulated H9c2 cells. Additionally, it was verified that inhibition of P2X7R alleviated pyroptosis and inflammatory signals in uric acid-treated H9c2 cells. Notopterol administration significantly suppressed expression levels of pyroptosis associated proteins and P2X7R in vivo and in vitro. P2X7R overexpression abolished the inhibition effect of notopterol on pyroptosis. Collectively, our findings suggested that P2X7R played a critical role in uric acid-induced NLRP3 inflammatory signals. Notopterol inhibited pyroptosis via inhibiting the P2X7R/NLRP3 signaling pathway under uric acid stimulation. Notopterol might represent a potential therapeutic strategy against pyroptosis and improve cardiac function in hyperuricemic mice.
Collapse
Affiliation(s)
- Qian Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dewei Peng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bingyu Huang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lintong Men
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tao Jiang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shengqi Huo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Moran Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Junyi Guo
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jiagao Lv
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Li Lin
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
- Correspondence: or
| |
Collapse
|
27
|
β-sitosterol ameliorates inflammation and Pseudomonas aeruginosa lung infection in a mouse model. J Cyst Fibros 2023; 22:156-160. [PMID: 35973900 DOI: 10.1016/j.jcf.2022.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/29/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022]
Abstract
We previously demonstrated that β-sitosterol (BSS) inhibits the expression of the chemokine IL-8 in CF bronchial epithelial cells exposed to P. aeruginosa. In the mouse model of lung chronic infection, herein shown, BSS significantly reduced leukocyte recruitment in the bronchoalveolar lavage fluid and decreased bacteria recovered in the airways. Treatment with BSS decreased the expression of key cytokines involved in immune response, mainly neutrophil chemotaxis, in the lung homogenate. This anti-inflammatory activity is accompanied by a beneficial protecting activity against infection and improvement of health status. Our data suggest that BSS has the potential to become a new drug to target the excessive neutrophil recruitment in lungs chronically infected by P. aeruginosa and encourage future investigations on mechanism of protection driven by BSS.
Collapse
|
28
|
Öz HH, Cheng EC, Di Pietro C, Tebaldi T, Biancon G, Zeiss C, Zhang PX, Huang PH, Esquibies SS, Britto CJ, Schupp JC, Murray TS, Halene S, Krause DS, Egan ME, Bruscia EM. Recruited monocytes/macrophages drive pulmonary neutrophilic inflammation and irreversible lung tissue remodeling in cystic fibrosis. Cell Rep 2022; 41:111797. [PMID: 36516754 PMCID: PMC9833830 DOI: 10.1016/j.celrep.2022.111797] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 09/30/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022] Open
Abstract
Persistent neutrophil-dominated lung inflammation contributes to lung damage in cystic fibrosis (CF). However, the mechanisms that drive persistent lung neutrophilia and tissue deterioration in CF are not well characterized. Starting from the observation that, in patients with CF, c-c motif chemokine receptor 2 (CCR2)+ monocytes/macrophages are abundant in the lungs, we investigate the interplay between monocytes/macrophages and neutrophils in perpetuating lung tissue damage in CF. Here we show that CCR2+ monocytes in murine CF lungs drive pathogenic transforming growth factor β (TGF-β) signaling and sustain a pro-inflammatory environment by facilitating neutrophil recruitment. Targeting CCR2 to lower the numbers of monocytes in CF lungs ameliorates neutrophil inflammation and pathogenic TGF-β signaling and prevents lung tissue damage. This study identifies CCR2+ monocytes as a neglected contributor to the pathogenesis of CF lung disease and as a therapeutic target for patients with CF, for whom lung hyperinflammation and tissue damage remain an issue despite recent advances in CF transmembrane conductance regulator (CFTR)-specific therapeutic agents.
Collapse
Affiliation(s)
- Hasan H Öz
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Ee-Chun Cheng
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | | | - Toma Tebaldi
- Department of Hematology, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA; Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Giulia Biancon
- Department of Hematology, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Caroline Zeiss
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ping-Xia Zhang
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA; Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Pamela H Huang
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Sofia S Esquibies
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Clemente J Britto
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jonas C Schupp
- Department of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease Hannover, German Lung Research Center (DZL), Hannover, Germany
| | - Thomas S Murray
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Department of Hematology, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA
| | - Diane S Krause
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA; Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Marie E Egan
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Emanuela M Bruscia
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Yale Stem Cell Center, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
29
|
Caverly LJ, Riquelme SA, Hisert KB. The Impact of Highly Effective Modulator Therapy on Cystic Fibrosis Microbiology and Inflammation. Clin Chest Med 2022; 43:647-665. [PMID: 36344072 PMCID: PMC10224747 DOI: 10.1016/j.ccm.2022.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Highly effective cystic fibrosis (CF) transmembrane conductance regulator (CFTR) modulator therapy (HEMT) corrects the underlying molecular defect causing CF disease. HEMT decreases symptom burden and improves clinical metrics and quality of life for most people with CF (PwCF) and eligible cftr mutations. Improvements in measures of pulmonary health suggest that restoration of function of defective CFTR anion channels by HEMT not only enhances airway mucociliary clearance, but also reduces chronic pulmonary infection and inflammation. This article reviews the evidence for how HEMT influences the dynamic and interdependent processes of infection and inflammation in the CF airway, and what questions remain unanswered.
Collapse
Affiliation(s)
- Lindsay J Caverly
- Department of Pediatrics, University of Michigan Medical School, L2221 UH South, 1500 East Medical Center Drive, Ann Arbor, MI 48109-5212, USA
| | - Sebastián A Riquelme
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, Columbia University Medical Center, 650West 168th Street, New York, NY 10032, USA
| | - Katherine B Hisert
- Department of Medicine, National Jewish Health, Smith A550, 1400 Jackson Street, Denver, CO 80205, USA.
| |
Collapse
|
30
|
Meoli A, Eickmeier O, Pisi G, Fainardi V, Zielen S, Esposito S. Impact of CFTR Modulators on the Impaired Function of Phagocytes in Cystic Fibrosis Lung Disease. Int J Mol Sci 2022; 23:12421. [PMID: 36293274 PMCID: PMC9604330 DOI: 10.3390/ijms232012421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Cystic fibrosis (CF), the most common genetically inherited disease in Caucasian populations, is a multi-systemic life-threatening autosomal recessive disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. In 2012, the arrival of CFTR modulators (potentiators, correctors, amplifiers, stabilizers, and read-through agents) revolutionized the therapeutic approach to CF. In this review, we examined the physiopathological mechanism of chronic dysregulated innate immune response in the lungs of CF patients with pulmonary involvement with particular reference to phagocytes, critically analyzing the role of CFTR modulators in influencing and eventually restoring their function. Our literature review highlighted that the role of CFTR in the lungs is crucial not only for the epithelial function but also for host defense, with particular reference to phagocytes. In macrophages and neutrophils, the CFTR dysfunction compromises both the intricate process of phagocytosis and the mechanisms of initiation and control of inflammation which then reverberates on the epithelial environment already burdened by the chronic colonization of pathogens leading to irreversible tissue damage. In this context, investigating the impact of CFTR modulators on phagocytic functions is therefore crucial not only for explaining the underlying mechanisms of pleiotropic effects of these molecules but also to better understand the physiopathological basis of this disease, still partly unexplored, and to develop new complementary or alternative therapeutic approaches.
Collapse
Affiliation(s)
- Aniello Meoli
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Olaf Eickmeier
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Giovanna Pisi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Valentina Fainardi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Stefan Zielen
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
31
|
Caley L, Peckham D. Time to change course and tackle CF related obesity. J Cyst Fibros 2022; 21:732-734. [PMID: 35970693 DOI: 10.1016/j.jcf.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 08/07/2022] [Indexed: 10/15/2022]
Affiliation(s)
- Laura Caley
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Daniel Peckham
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom; Leeds Adult Cystic Fibrosis Unit, St James's University Hospital, Leeds, United Kingdom.
| |
Collapse
|
32
|
Pyroptosis in inflammation-related respiratory disease. J Physiol Biochem 2022; 78:721-737. [PMID: 35819638 PMCID: PMC9684248 DOI: 10.1007/s13105-022-00909-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022]
Abstract
Pyroptosis is commonly induced by the gasdermin (GSDM) family and is accompanied by the release of inflammatory cytokines such as IL-1β and IL-18. Recently, increasing evidence suggests that pyroptosis plays a role in respiratory diseases. This review aimed to summarize the roles and mechanisms of pyroptosis in inflammation-related respiratory diseases. There are several pathways involved in pyroptosis, such as the canonical inflammasome-induced pathway, non-canonical inflammasome-induced pathway, caspase-1/3/6/7/GSDMB pathway, caspase-8/GSDMC pathway, caspase-8/GSDMD pathway, and caspase-3/GSEME pathway. Pyroptosis may be involved in asthma, chronic obstructive pulmonary disease (COPD), lung cancer, acute lung injury (ALI), silicosis, pulmonary hypertension (PH), and tuberculosis (TB), in which the NLRP3 inflammasome-induced pathway is mostly highlighted. Pyroptosis contributes to the deterioration of asthma, COPD, ALI, silicosis, and PH. In addition, pyroptosis has dual effects on lung cancer and TB. Additionally, whether pyroptosis participates in cystic fibrosis (CF) and sarcoidosis or not is largely unknown, though the activation of NLRP3 inflammasome is found in CF and sarcoidosis. In conclusion, pyroptosis may play a role in inflammation-related respiratory diseases, providing new therapeutic targets.
Collapse
|
33
|
Cantin AM. The P2X7 Receptor in Cystic Fibrosis Monocytes: Linking CFTR Deficiency to Inflammation. Am J Respir Crit Care Med 2022; 205:740-742. [PMID: 35139320 PMCID: PMC9836219 DOI: 10.1164/rccm.202201-0008ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- André M. Cantin
- Faculty of Medicine and Health SciencesUniversity of SherbrookeSherbrooke, Quebec, Canada,Centre de Recherche CentreHospitalier Universitaire de SherbrookeSherbrooke, Quebec, Canada
| |
Collapse
|