1
|
Berkel C. Potential Impact of Climate Change-Induced Alterations on Pyroptotic Cell Death in Animal Cells: A Review. Mol Biotechnol 2025; 67:1784-1799. [PMID: 38748072 DOI: 10.1007/s12033-024-01182-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/16/2024] [Indexed: 04/10/2025]
Abstract
Climate change-induced alterations in temperature variation, ozone exposure, water salinity and acidification, and hypoxia might influence immunity and thus survival in diverse groups of animals from fish to mammals. Pyroptosis is a type of lytic pro-inflammatory programmed cell death, which participates in the innate immune response, and is involved in multiple diseases characterized by inflammation and cell death, mostly studied in human cells. Diverse extrinsic factors can induce pyroptosis, leading to the extracellular release of pro-inflammatory molecules such as IL-18. Climate change-related factors, either directly or indirectly, can also promote animal cell death via different regulated mechanisms, impacting organismal fitness. However, pyroptosis has been relatively less studied in this context compared to another cell death process, apoptosis. This review covers previous research pointing to the potential impact of climate change, through various abiotic stressors, on pyroptotic cell death in different animal cells in various contexts. It was proposed that temperature, ozone exposure, water salinity, water acidification and hypoxia have the potential to induce pyroptotic cell death in animal cells and promote inflammation, and that these pyroptotic events should be better understood to be able to mitigate the adverse effects of climate change on animal physiology and health. This is of high importance considering the increasing frequency, intensity and duration of climate-based changes in these environmental parameters, and the critical function of pyroptosis in immune responses of animals and in their predisposition to multiple diseases including cancer. Furthermore, the need for further mechanistic studies showing the more direct impact of climate change-induced environmental alterations on pyroptotic cell death in animals at the organismal level was highlighted. A complete picture of the association between climate change and pyroptosis in animals will be also highly valuable in terms of ecological and clinical applications, and it requires an interdisciplinary approach. SIGNIFICANCE: Climate change-induced alterations might influence animal physiology. Pyroptosis is a form of cell death with pro-inflammatory characteristics. Previous research suggests that temperature variation, ozone exposure, water salinity and acidification, and hypoxia might have the potential to contribute to pyroptotic cell death in certain cell types and contexts. Climate change-induced pyroptotic cell death should be better understood to be able to mitigate the adverse effects of climate change on animal health.
Collapse
Affiliation(s)
- Caglar Berkel
- Deparment of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Türkiye.
| |
Collapse
|
2
|
Hynes J, Taggart CC, Tirouvanziam R, Coppinger JA. Innate Immunity in Cystic Fibrosis: Varied Effects of CFTR Modulator Therapy on Cell-to-Cell Communication. Int J Mol Sci 2025; 26:2636. [PMID: 40141278 PMCID: PMC11942055 DOI: 10.3390/ijms26062636] [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: 02/06/2025] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Cystic Fibrosis (CF) is a life-shortening, multi-organ disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene. Prominent clinical features of CF take place in the lung, hallmarked by cycles of bacterial infection and a dysfunctional inflammatory airway response, leading to eventual respiratory failure. Bidirectional crosstalk between epithelial cells, leukocytes (e.g., neutrophils, macrophages) and bacteria via release of intra-cellular mediators is key to driving inflammation in CF airways. In recent years, a highly effective combination of therapeutics targeting the CFTR defect have revolutionized treatment in CF. Despite these advancements and due to the complexity of the immune response in the CF airway, the full impact of highly effective modulator therapy (HEMT) on airway inflammation is not fully determined. This review provides the evidence to date on crosstalk mechanisms between host epithelium, leukocytes and bacteria and examines the effect of HEMT on both soluble and membrane-derived immune mediators in clinical samples. The varied effects of HEMT on expression of key proteases, cytokines and extracellular vesicles (EVs) in relation to clinical parameters is assessed. Advances in treatment with HEMT have shown potential in dampening the chronic inflammatory response in CF airways. However, to fully quell inflammation and maximize lung tissue resilience, further interventions may be necessary. Exploring the effects of HEMT on key immune mediators paves the way for identifying new anti-inflammatory approaches targeting host immune cell interactions, such as EV-directed lung therapies.
Collapse
Affiliation(s)
- Jennifer Hynes
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- Children’s Health Ireland Translational Research Centre, Children’s Health Ireland at Crumlin, D12 N512 Dublin, Ireland
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK;
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA;
- Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Judith A. Coppinger
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, D02 YN77 Dublin, Ireland;
- Children’s Health Ireland Translational Research Centre, Children’s Health Ireland at Crumlin, D12 N512 Dublin, Ireland
| |
Collapse
|
3
|
Zhao Y, Li K, Wang L, Kuang G, Xie K, Lin S. Dexmedetomidine Mitigates Acute Lung Injury by Enhancing M2 Macrophage Polarization and Inhibiting RAGE/Caspase-11-Mediated Pyroptosis. FRONT BIOSCI-LANDMRK 2024; 29:409. [PMID: 39735987 DOI: 10.31083/j.fbl2912409] [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: 07/04/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Acute lung injury (ALI) significantly impacts the survival rates in intensive care units (ICU). Releasing a lot of pro-inflammatory mediators during the progression of the disease is a core feature of ALI, which may lead to uncontrolled inflammation and further damages the tissues and organs of patients. This study explores the potential therapeutic mechanisms of Dexmedetomidine (Dex) in ALI. METHODS In present study, cecal ligation puncture (CLP)-established ALI model mice and lipopolysaccharide (LPS)-stimulated RAW264.7 cell line were established to discover the influence of Dex. The evaluation of lung injury in vivo using histopathology, TUNEL assay, and analysis of inflammatory factors in bronchoalveolar lavage fluid (BALF) and serum. The receptor for advanced glycation end products (RAGE)/Caspase-11-dependent pyroptosis-related proteins and macrophage polarization markers were analyzed using western blot, immunofluorescence, and flow cytometry. Finally, the mechanism of Dex in macrophages was further verified in vitro. RESULTS In vivo, Dex alleviated lung injury and decreased TUNEL-positive cell expression in CLP group. Dex decreased tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6 and IL-17A levels in BALF and serum, while increasing IL-10 expression. Dex treatment decreased the protein levels of RAGE, caspase-11, IL-1β and Gasdermin-D (GSDMD) in both in cells and in mice. Dex also down-regulated the synthesis of inducible nitric oxide synthase (iNOS) of classical activation phenotype (M1) markers, and up-regulated the synthesis of CD206 and Arg-1 of alternate activation phenotype (M2) markers. CONCLUSIONS Dex treatment can inhibit inflammation and reduce lung injury caused by CLP. It could be associated with mediating M1 and M2 polarization and suppressing RAGE/Caspase-11-depended pyroptosis.
Collapse
Affiliation(s)
- Yisi Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
- The Chongqing Key Laboratory of Translation Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Kefeng Li
- The Chongqing Key Laboratory of Translation Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
- Department of Critical Care Medicine, People's Hospital of Fengjie, 404600 Chongqing, China
| | - Liuyang Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Gang Kuang
- The Chongqing Key Laboratory of Translation Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
- Department of Critical Care Medicine, Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China
| | - Ke Xie
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| | - Shihui Lin
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, 400016 Chongqing, China
| |
Collapse
|
4
|
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
|
5
|
Yu C, Zhang Z, Xiao L, Ai M, Qing Y, Zhang Z, Xu L, Yu OY, Cao Y, Liu Y, Song K. IRE1α pathway: A potential bone metabolism mediator. Cell Prolif 2024; 57:e13654. [PMID: 38736291 PMCID: PMC11471397 DOI: 10.1111/cpr.13654] [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: 12/28/2023] [Revised: 04/07/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024] Open
Abstract
Osteoblasts and osteoclasts collaborate in bone metabolism, facilitating bone development, maintaining normal bone density and strength, and aiding in the repair of pathological damage. Endoplasmic reticulum stress (ERS) can disrupt the intracellular equilibrium between osteoclast and osteoblast, resulting in dysfunctional bone metabolism. The inositol-requiring enzyme-1α (IRE1α) pathway-the most conservative unfolded protein response pathway activated by ERS-is crucial in regulating cell metabolism. This involvement encompasses functions such as inflammation, autophagy, and apoptosis. Many studies have highlighted the potential roles of the IRE1α pathway in osteoblasts, chondrocytes, and osteoclasts and its implication in certain bone-related diseases. These findings suggest that it may serve as a mediator for bone metabolism. However, relevant reviews on the role of the IRE1α pathway in bone metabolism remain unavailable. Therefore, this review aims to explore recent research that elucidated the intricate roles of the IRE1α pathway in bone metabolism, specifically in osteogenesis, chondrogenesis, osteoclastogenesis, and osteo-immunology. The findings may provide novel insights into regulating bone metabolism and treating bone-related diseases.
Collapse
Affiliation(s)
- Chengbo Yu
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Zhixiang Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Li Xiao
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Mi Ai
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Ying Qing
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Zhixing Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Lianyi Xu
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Ollie Yiru Yu
- Faculty of DentistryThe University of Hong KongHong Kong SARChina
| | - Yingguang Cao
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and Metabolism, and the Institute for Advanced StudiesWuhan UniversityWuhanHubeiChina
| | - Ke Song
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| |
Collapse
|
6
|
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
|
7
|
Das Gupta K, Curson JEB, Tarique AA, Kapetanovic R, Schembri MA, Fantino E, Sly PD, Sweet MJ. CFTR is required for zinc-mediated antibacterial defense in human macrophages. Proc Natl Acad Sci U S A 2024; 121:e2315190121. [PMID: 38363865 PMCID: PMC10895263 DOI: 10.1073/pnas.2315190121] [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: 09/19/2023] [Accepted: 12/22/2023] [Indexed: 02/18/2024] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) is an anion transporter required for epithelial homeostasis in the lung and other organs, with CFTR mutations leading to the autosomal recessive genetic disease CF. Apart from excessive mucus accumulation and dysregulated inflammation in the airways, people with CF (pwCF) exhibit defective innate immune responses and are susceptible to bacterial respiratory pathogens such as Pseudomonas aeruginosa. Here, we investigated the role of CFTR in macrophage antimicrobial responses, including the zinc toxicity response that is used by these innate immune cells against intracellular bacteria. Using both pharmacological approaches, as well as cells derived from pwCF, we show that CFTR is required for uptake and clearance of pathogenic Escherichia coli by CSF-1-derived primary human macrophages. CFTR was also required for E. coli-induced zinc accumulation and zinc vesicle formation in these cells, and E. coli residing in macrophages exhibited reduced zinc stress in the absence of CFTR function. Accordingly, CFTR was essential for reducing the intramacrophage survival of a zinc-sensitive E. coli mutant compared to wild-type E. coli. Ectopic expression of the zinc transporter SLC30A1 or treatment with exogenous zinc was sufficient to restore antimicrobial responses against E. coli in human macrophages. Zinc supplementation also restored bacterial killing in GM-CSF-derived primary human macrophages responding to P. aeruginosa, used as an in vitro macrophage model relevant to CF. Thus, restoration of the zinc toxicity response could be pursued as a therapeutic strategy to restore innate immune function and effective host defense in pwCF.
Collapse
Affiliation(s)
- Kaustav Das Gupta
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
| | - James E. B. Curson
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
| | - Abdullah A. Tarique
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD4101, Australia
| | - Ronan Kapetanovic
- Friedrich Miescher Institute for Biomedical Research, Basel, BS4058, Switzerland
- Institut national de recherche pour l’agriculture, l’alimentation et l’environnement (INRAE), Université de Tours, Infectiologie et Santé Publique (ISP), Nouzilly37380, France
| | - Mark A. Schembri
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD4072, Australia
| | - Emmanuelle Fantino
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD4101, Australia
| | - Peter D. Sly
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, QLD4101, Australia
| | - Matthew J. Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD4072, Australia
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Sposito F, Northey S, Charras A, McNamara PS, Hedrich CM. Hypertonic saline induces inflammation in human macrophages through the NLRP1 inflammasome. Genes Immun 2023; 24:263-269. [PMID: 37573430 PMCID: PMC10575766 DOI: 10.1038/s41435-023-00218-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Nebulized hypertonic saline (3-7%) is commonly used to increase mucociliary clearance in patients with chronic airway disease and/or virus infections. However, altered salt concentrations may contribute to inflammatory responses. The aim of this study was to investigate whether 500 mM NaCl (3%) triggers inflammation in human macrophages and identify the molecular mechanisms involved. NaCl-induced pyroptosis, IL-1β, IL-18 and ASC speck release were measured in primary human monocyte-derived macrophages. Treatment with the recombinant IL-1 receptor antagonist anakinra or the NLRP3 inhibitor MCC950 did not affect NaCl-mediated inflammasome assembly. Knock-down of NLRP1 expression, but not of NLRP3 and NLRC4, reduced NaCl-induced pyroptosis, pro-inflammatory cytokine and ASC speck release from human THP-1-derived macrophages. Data from this study suggest that 3% NaCl-induced inflammatory responses in human macrophages depend on NLRP1 and inflammasome assembly. Targeting inflammation in addition to inhalation with hypertonic saline may benefit patients with inflammatory airway disease.
Collapse
Affiliation(s)
- Francesca Sposito
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Sarah Northey
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Amandine Charras
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Paul S McNamara
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Respiratory Medicine, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| | - Christian M Hedrich
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
- Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK.
| |
Collapse
|
11
|
Lv J, Gao H, Ma J, Liu J, Tian Y, Yang C, Li M, Zhao Y, Li Z, Zhang X, Zhu Y, Zhang J, Wu L. Dynamic atlas of immune cells reveals multiple functional features of macrophages associated with progression of pulmonary fibrosis. Front Immunol 2023; 14:1230266. [PMID: 37771586 PMCID: PMC10525351 DOI: 10.3389/fimmu.2023.1230266] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/24/2023] [Indexed: 09/30/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease with a high mortality rate and unclarified aetiology. Immune response is elaborately regulated during the progression of IPF, but immune cells subsets are complicated which has not been detailed described during IPF progression. Therefore, in the current study, we sought to investigate the role of immune regulation by elaborately characterize the heterogeneous of immune cells during the progression of IPF. To this end, we performed single-cell profiling of lung immune cells isolated from four stages of bleomycin-induced pulmonary fibrosis-a classical mouse model that mimics human IPF. The results revealed distinct components of immune cells in different phases of pulmonary fibrosis and close communication between macrophages and other immune cells along with pulmonary fibrosis progression. Enriched signals of SPP1, CCL5 and CXCL2 were found between macrophages and other immune cells. The more detailed definition of the subpopulations of macrophages defined alveolar macrophages (AMs) and monocyte-derived macrophages (mo-Macs)-the two major types of primary lung macrophages-exhibited the highest heterogeneity and dynamic changes in expression of profibrotic genes during disease progression. Our analysis suggested that Gpnmb and Trem2 were both upregulated in macrophages and may play important roles in pulmonary fibrosis progression. Additionally, the metabolic status of AMs and mo-Macs varied with disease progression. In line with the published data on human IPF, macrophages in the mouse model shared some features regarding gene expression and metabolic status with that of macrophages in IPF patients. Our study provides new insights into the pathological features of profibrotic macrophages in the lung that will facilitate the identification of new targets for disease intervention and treatment of IPF.
Collapse
Affiliation(s)
- Jiaoyan Lv
- Institute for Immunology, Tsinghua-Peking Joint Centre for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Haoxiang Gao
- Department of Automation, Ministry of Education (MOE) Key Laboratory of Bioinformatics, Bioinformatics Division and Centre for Synthetic & Systems Biology, BNRist, Tsinghua University, Beijing, China
| | - Jie Ma
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Jiachen Liu
- Institute for Immunology, Tsinghua-Peking Joint Centre for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Yujie Tian
- Institute for Immunology, Tsinghua-Peking Joint Centre for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Chunyuan Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Mansheng Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yue Zhao
- Annoroad Gene Technology (Beijing) Co., Ltd., Beijing, China
| | - Zhimin Li
- Annoroad Gene Technology (Beijing) Co., Ltd., Beijing, China
| | - Xuegong Zhang
- Department of Automation, Ministry of Education (MOE) Key Laboratory of Bioinformatics, Bioinformatics Division and Centre for Synthetic & Systems Biology, BNRist, Tsinghua University, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yunping Zhu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Jianhong Zhang
- Institute for Immunology, Tsinghua-Peking Joint Centre for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Li Wu
- Institute for Immunology, Tsinghua-Peking Joint Centre for Life Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| |
Collapse
|
12
|
Iovino M, Colonval M, Wilkin C, L’homme L, Lassence C, Campas M, Peulen O, de Tullio P, Piette J, Legrand-Poels S. Novel XBP1s-independent function of IRE1 RNase in HIF-1α-mediated glycolysis upregulation in human macrophages upon stimulation with LPS or saturated fatty acid. Front Immunol 2023; 14:1204126. [PMID: 37711626 PMCID: PMC10498766 DOI: 10.3389/fimmu.2023.1204126] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/01/2023] [Indexed: 09/16/2023] Open
Abstract
In obesity, adipose tissue infiltrating macrophages acquire a unique pro-inflammatory polarization, thereby playing a key role in the development of chronic inflammation and Type 2 diabetes. Increased saturated fatty acids (SFAs) levels have been proposed to drive this specific polarization. Accordingly, we investigated the immunometabolic reprogramming in SFA-treated human macrophages. As expected, RNA sequencing highlighted a pro-inflammatory profile but also metabolic signatures including glycolysis and hypoxia as well as a strong unfolded protein response. Glycolysis upregulation was confirmed in SFA-treated macrophages by measuring glycolytic gene expression, glucose uptake, lactate production and extracellular acidification rate. Like in LPS-stimulated macrophages, glycolysis activation in SFA-treated macrophages was dependent on HIF-1α activation and fueled the production of pro-inflammatory cytokines. SFAs and LPS both induced IRE1α endoribonuclease activity, as demonstrated by XBP1 mRNA splicing, but with different kinetics matching HIF-1α activation and the glycolytic gene expression. Interestingly, the knockdown of IRE1α and/or the pharmacological inhibition of its RNase activity prevented HIF-1α activation and significantly decreased glycolysis upregulation. Surprisingly, XBP1s appeared to be dispensable, as demonstrated by the lack of inhibiting effect of XBP1s knockdown on glycolytic genes expression, glucose uptake, lactate production and HIF-1α activation. These experiments demonstrate for the first time a key role of IRE1α in HIF-1α-mediated glycolysis upregulation in macrophages stimulated with pro-inflammatory triggers like LPS or SFAs through XBP1s-independent mechanism. IRE1 could mediate this novel function by targeting other transcripts (mRNA or pre-miRNA) through a mechanism called regulated IRE1-dependent decay or RIDD. Deciphering the underlying mechanisms of this novel IRE1 function might lead to novel therapeutic targets to curtail sterile obesity- or infection-linked inflammation.
Collapse
Affiliation(s)
- Margaud Iovino
- Laboratory of Immunometabolism and Nutrition, GIGA, ULiège, Liège, Belgium
| | - Megan Colonval
- Laboratory of Immunometabolism and Nutrition, GIGA, ULiège, Liège, Belgium
| | - Chloé Wilkin
- Laboratory of Immunometabolism and Nutrition, GIGA, ULiège, Liège, Belgium
| | - Laurent L’homme
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Cédric Lassence
- Laboratory of Virology and Immunology, GIGA, ULiège, Liège, Belgium
| | - Manon Campas
- Clinical Metabolomics Group, CIRM, ULiège, Liege, Belgium
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA, ULiège, Liège, Belgium
| | | | - Jacques Piette
- Laboratory of Virology and Immunology, GIGA, ULiège, Liège, Belgium
| | | |
Collapse
|
13
|
Fehsel K. Why Is Iron Deficiency/Anemia Linked to Alzheimer's Disease and Its Comorbidities, and How Is It Prevented? Biomedicines 2023; 11:2421. [PMID: 37760862 PMCID: PMC10526115 DOI: 10.3390/biomedicines11092421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Impaired iron metabolism has been increasingly observed in many diseases, but a deeper, mechanistic understanding of the cellular impact of altered iron metabolism is still lacking. In addition, deficits in neuronal energy metabolism due to reduced glucose import were described for Alzheimer's disease (AD) and its comorbidities like obesity, depression, cardiovascular disease, and type 2 diabetes mellitus. The aim of this review is to present the molecular link between both observations. Insufficient cellular glucose uptake triggers increased ferritin expression, leading to depletion of the cellular free iron pool and stabilization of the hypoxia-induced factor (HIF) 1α. This transcription factor induces the expression of the glucose transporters (Glut) 1 and 3 and shifts the cellular metabolism towards glycolysis. If this first line of defense is not adequate for sufficient glucose supply, further reduction of the intracellular iron pool affects the enzymes of the mitochondrial electron transport chain and activates the AMP-activated kinase (AMPK). This enzyme triggers the translocation of Glut4 to the plasma membrane as well as the autophagic recycling of cell components in order to mobilize energy resources. Moreover, AMPK activates the autophagic process of ferritinophagy, which provides free iron urgently needed as a cofactor for the synthesis of heme- and iron-sulfur proteins. Excessive activation of this pathway ends in ferroptosis, a special iron-dependent form of cell death, while hampered AMPK activation steadily reduces the iron pools, leading to hypoferremia with iron sequestration in the spleen and liver. Long-lasting iron depletion affects erythropoiesis and results in anemia of chronic disease, a common condition in patients with AD and its comorbidities. Instead of iron supplementation, drugs, diet, or phytochemicals that improve energy supply and cellular glucose uptake should be administered to counteract hypoferremia and anemia of chronic disease.
Collapse
Affiliation(s)
- Karin Fehsel
- Neurobiochemical Research Unit, Department of Psychiatry, Medical Faculty, Heinrich-Heine-University, 240629 Düsseldorf, Germany
| |
Collapse
|
14
|
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
|
15
|
D'Urso A, Oltolina F, Borsotti C, Prat M, Colangelo D, Follenzi A. Macrophage Reprogramming via the Modulation of Unfolded Protein Response with siRNA-Loaded Magnetic Nanoparticles in a TAM-like Experimental Model. Pharmaceutics 2023; 15:1711. [PMID: 37376159 DOI: 10.3390/pharmaceutics15061711] [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: 05/02/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
New therapeutic strategies are required in cancer therapy. Considering the prominent role of tumor-associated macrophages (TAMs) in the development and progression of cancer, the re-education of TAMs in the tumor microenvironment (TME) could represent a potential approach for cancer immunotherapy. TAMs display an irregular unfolded protein response (UPR) in their endoplasmic reticulum (ER) to endure environmental stress and ensure anti-cancer immunity. Therefore, nanotechnology could be an attractive tool to modulate the UPR in TAMs, providing an alternative strategy for TAM-targeted repolarization therapy. Herein, we developed and tested polydopamine-coupled magnetite nanoparticles (PDA-MNPs) functionalized with small interfering RNAs (siRNA) to downregulate the protein kinase R (PKR)-like ER kinase (PERK) expression in TAM-like macrophages derived from murine peritoneal exudate (PEMs). After the evaluation of the cytocompatibility, the cellular uptake, and the gene silencing efficiency of PDA-MNPs/siPERK in PEMs, we analyzed their ability to re-polarize in vitro these macrophages from M2 to the M1 inflammatory anti-tumor phenotype. Our results indicate that PDA-MNPs, with their magnetic and immunomodulator features, are cytocompatible and able to re-educate TAMs toward the M1 phenotype by PERK inhibition, a UPR effector contributing to TAM metabolic adaptation. These findings can provide a novel strategy for the development of new tumor immunotherapies in vivo.
Collapse
Affiliation(s)
- Annarita D'Urso
- Department of Health Sciences, School Medicine, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy
| | - Francesca Oltolina
- Department of Health Sciences, School Medicine, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy
| | - Chiara Borsotti
- Department of Health Sciences, School Medicine, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy
| | - Maria Prat
- Department of Health Sciences, School Medicine, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy
| | - Donato Colangelo
- Department of Health Sciences, School Medicine, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy
| | - Antonia Follenzi
- Department of Health Sciences, School Medicine, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy
| |
Collapse
|
16
|
Harris VK, Bishop D, Wollowitz J, Carling G, Carlson AL, Daviaud N, Sadiq SA. Mesenchymal stem cell-derived neural progenitors attenuate proinflammatory microglial activation via paracrine mechanisms. Regen Med 2023; 18:259-273. [PMID: 36852422 DOI: 10.2217/rme-2023-0005] [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] [Indexed: 03/01/2023] Open
Abstract
Background: Mesenchymal stem cell-derived neural progenitor cell (MSC-NP) therapy is an experimental approach to treat multiple sclerosis. The influence of MSC-NPs on microglial activation was investigated. Methods: Microglia were stimulated in the presence of MSC-NP-conditioned media, and proinflammatory or proregenerative marker expression was assessed by quantitative PCR and ELISA. Results: Microglia stimulated in the presence of MSC-NP-conditioned media displayed reduced expression of proinflammatory markers including CCL2, increased expression of proregenerative markers and reduced phagocytic activity. The paracrine effects of MSC-NPs from multiple donors correlated with TGF-β3 gene expression and was reversed by TGF-β signaling inhibition. Conclusion: MSC-NPs promote beneficial microglial polarization through secreted factors. This study suggests that microglia are a potential therapeutic target of MSC-NP cell therapy.
Collapse
Affiliation(s)
| | - Derek Bishop
- Tisch Multiple Sclerosis Research Center of New York, NY 10019, USA
| | - Jaina Wollowitz
- Tisch Multiple Sclerosis Research Center of New York, NY 10019, USA
| | - Gillian Carling
- Tisch Multiple Sclerosis Research Center of New York, NY 10019, USA
| | - Alyssa L Carlson
- Tisch Multiple Sclerosis Research Center of New York, NY 10019, USA
| | - Nicolas Daviaud
- Tisch Multiple Sclerosis Research Center of New York, NY 10019, USA
| | - Saud A Sadiq
- Tisch Multiple Sclerosis Research Center of New York, NY 10019, USA
| |
Collapse
|
17
|
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
|
18
|
Proulx J, Stacy S, Park IW, Borgmann K. A Non-Canonical Role for IRE1α Links ER and Mitochondria as Key Regulators of Astrocyte Dysfunction: Implications in Methamphetamine use and HIV-Associated Neurocognitive Disorders. Front Neurosci 2022; 16:906651. [PMID: 35784841 PMCID: PMC9247407 DOI: 10.3389/fnins.2022.906651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Astrocytes are one of the most numerous glial cells in the central nervous system (CNS) and provide essential support to neurons to ensure CNS health and function. During a neuropathological challenge, such as during human immunodeficiency virus (HIV)-1 infection or (METH)amphetamine exposure, astrocytes shift their neuroprotective functions and can become neurotoxic. Identifying cellular and molecular mechanisms underlying astrocyte dysfunction are of heightened importance to optimize the coupling between astrocytes and neurons and ensure neuronal fitness against CNS pathology, including HIV-1-associated neurocognitive disorders (HAND) and METH use disorder. Mitochondria are essential organelles for regulating metabolic, antioxidant, and inflammatory profiles. Moreover, endoplasmic reticulum (ER)-associated signaling pathways, such as calcium and the unfolded protein response (UPR), are important messengers for cellular fate and function, including inflammation and mitochondrial homeostasis. Increasing evidence supports that the three arms of the UPR are involved in the direct contact and communication between ER and mitochondria through mitochondria-associated ER membranes (MAMs). The current study investigated the effects of HIV-1 infection and chronic METH exposure on astrocyte ER and mitochondrial homeostasis and then examined the three UPR messengers as potential regulators of astrocyte mitochondrial dysfunction. Using primary human astrocytes infected with pseudotyped HIV-1 or exposed to low doses of METH for 7 days, astrocytes had increased mitochondrial oxygen consumption rate (OCR), cytosolic calcium flux and protein expression of UPR mediators. Notably, inositol-requiring protein 1α (IRE1α) was most prominently upregulated following both HIV-1 infection and chronic METH exposure. Moreover, pharmacological inhibition of the three UPR arms highlighted IRE1α as a key regulator of astrocyte metabolic function. To further explore the regulatory role of astrocyte IRE1α, astrocytes were transfected with an IRE1α overexpression vector followed by activation with the proinflammatory cytokine interleukin 1β. Overall, our findings confirm IRE1α modulates astrocyte mitochondrial respiration, glycolytic function, morphological activation, inflammation, and glutamate uptake, highlighting a novel potential target for regulating astrocyte dysfunction. Finally, these findings suggest both canonical and non-canonical UPR mechanisms of astrocyte IRE1α. Thus, additional studies are needed to determine how to best balance astrocyte IRE1α functions to both promote astrocyte neuroprotective properties while preventing neurotoxic properties during CNS pathologies.
Collapse
|
19
|
Fehsel K, Christl J. Comorbidity of osteoporosis and Alzheimer's disease: Is `AKT `-ing on cellular glucose uptake the missing link? Ageing Res Rev 2022; 76:101592. [PMID: 35192961 DOI: 10.1016/j.arr.2022.101592] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2023]
Abstract
Osteoporosis and Alzheimer's disease (AD) are both degenerative diseases. Osteoporosis often proceeds cognitive deficits, and multiple studies have revealed common triggers that lead to energy deficits in brain and bone. Risk factors for osteoporosis and AD, such as obesity, type 2 diabetes, aging, chemotherapy, vitamin deficiency, alcohol abuse, and apolipoprotein Eε4 and/or Il-6 gene variants, reduce cellular glucose uptake, and protective factors, such as estrogen, insulin, exercise, mammalian target of rapamycin inhibitors, hydrogen sulfide, and most phytochemicals, increase uptake. Glucose uptake is a fine-tuned process that depends on an abundance of glucose transporters (Gluts) on the cell surface. Gluts are stored in vesicles under the plasma membrane, and protective factors cause these vesicles to fuse with the membrane, resulting in presentation of Gluts on the cell surface. This translocation depends mainly on AKT kinase signaling and can be affected by a range of factors. Reduced AKT kinase signaling results in intracellular glucose deprivation, which causes endoplasmic reticulum stress and iron depletion, leading to activation of HIF-1α, the transcription factor necessary for higher Glut expression. The link between diseases and aging is a topic of growing interest. Here, we show that diseases that affect the same biochemical pathways tend to co-occur, which may explain why osteoporosis and/or diabetes are often associated with AD.
Collapse
|
20
|
Catellani C, Cirillo F, Graziano S, Montanini L, Marmiroli N, Gullì M, Street ME. MicroRNA global profiling in cystic fibrosis cell lines reveals dysregulated pathways related with inflammation, cancer, growth, glucose and lipid metabolism, and fertility: an exploratory study. ACTA BIO-MEDICA : ATENEI PARMENSIS 2022; 93:e2022133. [PMID: 35775757 PMCID: PMC9335447 DOI: 10.23750/abm.v93i3.12842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/10/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND AIM Cystic fibrosis (CF), is due to CF transmembrane conductance regulator (CFTR) loss of function, and is associated with comorbidities. The increasing longevity of CF patients has been associated with increased cancer risk besides the other known comorbidities. The significant heterogeneity among patients, suggests potential epigenetic regulation. Little attention has been given to how CFTR influences microRNA (miRNA) expression and how this may impact on biological processes and pathways. METHODS We assessed the changes in miRNAs and subsequently identified the affected molecular pathways using CFBE41o-, and IB3 human immortalized cell lines since they reflect the most common genetic mutations in CF patients, and 16HBE14o- cells were used as controls. RESULTS In the CF cell lines, 41 miRNAs showed significant changes (FC (log2) ≥ +2 or FC (log2) ≤ -2 and p-value≤0.05). Gene target analysis evidenced 511 validated miRNA target genes. Gene Ontology analysis evidenced cancer, inflammation, body growth, glucose, and lipid metabolism as the biological processes most impacted by these miRNAs. Protein-protein interaction and pathway analysis highlighted 50 significantly enriched pathways among which RAS, TGF beta, JAK/STAT and insulin signaling. CONCLUSIONS CFTR loss of function is associated with changes in the miRNA network, which regulates genes involved in the major comorbidities that affect CF patients suggesting that further research is warranted.
Collapse
Affiliation(s)
- Cecilia Catellani
- Department of Mother and Child, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy, PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy, These authors contributed equally to this work
| | - Francesca Cirillo
- Department of Mother and Child, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy, These authors contributed equally to this work
| | - Sara Graziano
- Interdepartmental Center SITEIA.PARMA, University of Parma, Parma, Italy
| | - Luisa Montanini
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Nelson Marmiroli
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Mariolina Gullì
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Maria E. Street
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
21
|
Sharma P, Alizadeh J, Juarez M, Samali A, Halayko AJ, Kenyon NJ, Ghavami S, Zeki AA. Autophagy, Apoptosis, the Unfolded Protein Response, and Lung Function in Idiopathic Pulmonary Fibrosis. Cells 2021; 10:1642. [PMID: 34209019 PMCID: PMC8307368 DOI: 10.3390/cells10071642] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 01/18/2023] Open
Abstract
Autophagy, apoptosis, and the unfolded protein response (UPR) are fundamental biological processes essential for manifold cellular functions in health and disease. Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal pulmonary disorder associated with aging that has limited therapies, reflecting our incomplete understanding. We conducted an observational study linking molecular markers of cell stress response pathways (UPR: BiP, XBP1; apoptosis: cleaved caspase-3; autophagy: LC3β) in lung tissues from IPF patients and correlated the expression of these protein markers to each subject's lung function measures. We hypothesized that changes in lung tissue expression of apoptosis, autophagy, and UPR markers correlate with lung function deficits in IPF. The cell stress markers BiP, XBP1, LC3β puncta, and cleaved caspase-3 were found to be elevated in IPF lungs compared to non-IPF lungs, and, further, BiP and cleaved caspase-3 co-localized in IPF lungs. Considering lung function independently, we observed that increased XBP1, BiP, and cleaved caspase-3 were each associated with reduced lung function (FEV1, FVC, TLC, RV). However, increased lung tissue expression of LC3β puncta was significantly associated with increased diffusion capacity (DLCO), an indicator of alveolar-capillary membrane function. Similarly, the co-localization of UPR (XBP1, BiP) and autophagy (LC3β puncta) markers was positively correlated with increased lung function (FEV1, FVC, TLC, DLCO). However, the presence of LC3β puncta can indicate either autophagy flux inhibition or activation. While the nature of our observational cross-sectional study design does not allow conclusions regarding causal links between increased expression of these cell stress markers, lung fibrosis, and lung function decline, it does provide some insights that are hypothesis-generating and suggests that within the milieu of active UPR, changes in autophagy flux may play an important role in determining lung function. Further research is necessary to investigate the mechanisms linking UPR and autophagy in IPF and how an imbalance in these cell stress pathways can lead to progressive fibrosis and loss of lung function. We conclude by presenting five testable hypotheses that build on the research presented here. Such an understanding could eventually lead to the development of much-needed therapies for IPF.
Collapse
Affiliation(s)
- Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Javad Alizadeh
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada;
| | - Maya Juarez
- Davis Lung Center, School of Medicine; Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, Davis, CA 95616, USA; (M.J.); (N.J.K.)
| | - Afshin Samali
- Apoptosis Research Centre, School of Natural Sciences, National University of Ireland, H91 W2TY Galway, Ireland;
| | - Andrew J. Halayko
- Department of Physiology and Pathophysiology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada;
| | - Nicholas J. Kenyon
- Davis Lung Center, School of Medicine; Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, Davis, CA 95616, USA; (M.J.); (N.J.K.)
- Veterans Affairs Medical Center, Mather, CA 95655, USA
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 3P4, Canada;
- Research Institute of Hematology and Oncology, Cancer Care Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine, Katowice School of Technology, 40-555 Katowice, Poland
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Amir A. Zeki
- Davis Lung Center, School of Medicine; Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, Davis, CA 95616, USA; (M.J.); (N.J.K.)
- Veterans Affairs Medical Center, Mather, CA 95655, USA
| |
Collapse
|
22
|
Nakada EM, Sun R, Fujii U, Martin JG. The Impact of Endoplasmic Reticulum-Associated Protein Modifications, Folding and Degradation on Lung Structure and Function. Front Physiol 2021; 12:665622. [PMID: 34122136 PMCID: PMC8188853 DOI: 10.3389/fphys.2021.665622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
The accumulation of unfolded/misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and induces the unfolded protein response (UPR) and other mechanisms to restore ER homeostasis, including translational shutdown, increased targeting of mRNAs for degradation by the IRE1-dependent decay pathway, selective translation of proteins that contribute to the protein folding capacity of the ER, and activation of the ER-associated degradation machinery. When ER stress is excessive or prolonged and these mechanisms fail to restore proteostasis, the UPR triggers the cell to undergo apoptosis. This review also examines the overlooked role of post-translational modifications and their roles in protein processing and effects on ER stress and the UPR. Finally, these effects are examined in the context of lung structure, function, and disease.
Collapse
Affiliation(s)
- Emily M. Nakada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Rui Sun
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Utako Fujii
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - James G. Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| |
Collapse
|
23
|
Hyperinflammation and airway surface liquid dehydration in cystic fibrosis: purinergic system as therapeutic target. Inflamm Res 2021; 70:633-649. [PMID: 33904934 DOI: 10.1007/s00011-021-01464-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/06/2021] [Accepted: 04/19/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE AND DESIGN The exacerbate inflammatory response contributes to the progressive loss of lung function in cystic fibrosis (CF), a genetic disease that affects the osmotic balance of mucus and mucociliary clearance, resulting in a microenvironment that favors infection and inflammation. The purinergic system, an extracellular signaling pathway characterized by nucleotides, enzymes and receptors, may have a protective role in the disease, through its action in airway surface liquid (ASL) and anti-inflammatory response. MATERIALS AND METHODS To make up this review, studies covering topics of CF, inflammation, ASL and purinergic system were selected from the main medical databases, such as Pubmed and ScienceDirect. CONCLUSION We propose several ways to modulate the purinergic system as a potential therapy for CF, like inhibition of P2X7, activation of P2Y2, A2A and A2B receptors and blocking of adenosine deaminase. Among them, we postulate that the most suitable strategy is to block the action of adenosine deaminase, which culminates in the increase of Ado levels that presents anti-inflammatory actions and improves mucociliary clearance. Furthermore, it is possible to maintain the physiological levels of ATP to control the hydration of ASL. These therapies could correct the main mechanisms that contribute to the progression of CF.
Collapse
|
24
|
Abed S, Turner R, Serniuck N, Tat V, Naiel S, Hayat A, Mekhael O, Vierhout M, Ask K, Rullo AF. Cell-specific drug targeting in the lung. Biochem Pharmacol 2021; 190:114577. [PMID: 33887259 DOI: 10.1016/j.bcp.2021.114577] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 11/26/2022]
Abstract
Non-targeted drug delivery systems have several limitations including the decreased bioavailability of the drug, poor stability and rapid clearance in addition to off-target distribution. Cell-specific targeted delivery approaches promise to overcome some of these limitations and enhance therapeutic selectivity. In this review, we aim to discuss cell-specific targeted approachesin the lung at the biochemical and molecular levels. These approaches include;a) directly administered small molecule drugs with intracellular action; b) targeted biologics and synthetic hybrids with extracellular action; c) site activateddrugs; and d) delivery systems.We discuss the pharmaceutical and biochemical parameters that govern the fate of drug molecules at delivery sites while presenting an overview of relevant literature surrounding this area of research and current advancements.
Collapse
Affiliation(s)
- Soumeya Abed
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Rebecca Turner
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Nickolas Serniuck
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Victor Tat
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | - Safaa Naiel
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Aaron Hayat
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Olivia Mekhael
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Megan Vierhout
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada.
| | - Anthony F Rullo
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada; Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
25
|
García R, Falduti C, Clauzure M, Jara R, Massip-Copiz MM, de Los Ángeles Aguilar M, Santa-Coloma TA, Valdivieso ÁG. CFTR chloride channel activity modulates the mitochondrial morphology in cultured epithelial cells. Int J Biochem Cell Biol 2021; 135:105976. [PMID: 33845203 DOI: 10.1016/j.biocel.2021.105976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 01/10/2023]
Abstract
The impairment of the CFTR channel activity, a cAMP-activated chloride (Cl-) channel responsible for cystic fibrosis (CF), has been associated with a variety of mitochondrial alterations such as modified gene expression, impairment in oxidative phosphorylation, increased reactive oxygen species (ROS), and a disbalance in calcium homeostasis. The mechanisms by which these processes occur in CF are not fully understood. Previously, we demonstrated a reduced MTND4 expression and a failure in the mitochondrial complex I (mCx-I) activity in CF cells. Here we hypothesized that the activity of CFTR might modulate the mitochondrial fission/fusion balance, explaining the decreased mCx-I. The mitochondrial morphology and the levels of mitochondrial dynamic proteins MFN1 and DRP1 were analysed in IB3-1 CF cells, and S9 (IB3-1 expressing wt-CFTR), and C38 (IB3-1 expressing a truncated functional CFTR) cells. The mitochondrial morphology of IB3-1 cells compared to S9 and C38 cells showed that the impaired CFTR activity induced a fragmented mitochondrial network with increased rounded mitochondria and shorter branches. Similar results were obtained by using the CFTR pharmacological inhibitors CFTR(inh)-172 and GlyH101 on C38 cells. These morphological changes were accompanied by modifications in the levels of the mitochondrial dynamic proteins MFN1, DRP1, and p(616)-DRP1. IB3-1 CF cells treated with Mdivi-1, an inhibitor of mitochondrial fission, restored the mCx-I activity to values similar to those seen in S9 and C38 cells. These results suggest that the mitochondrial fission/fusion balance is regulated by the CFTR activity and might be a potential target to treat the impaired mCx-I activity in CF.
Collapse
Affiliation(s)
- Rocío García
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), the National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Camila Falduti
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), the National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Mariángeles Clauzure
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), the National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Raquel Jara
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), the National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - María M Massip-Copiz
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), the National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - María de Los Ángeles Aguilar
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), the National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Tomás A Santa-Coloma
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), the National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Ángel G Valdivieso
- Laboratory of Cellular and Molecular Biology, Institute for Biomedical Research (BIOMED), School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), the National Scientific and Technical Research Council of Argentina (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
26
|
Bradley KL, Stokes CA, Marciniak SJ, Parker LC, Condliffe AM. Role of unfolded proteins in lung disease. Thorax 2021; 76:92-99. [PMID: 33077618 PMCID: PMC7803888 DOI: 10.1136/thoraxjnl-2019-213738] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 01/01/2023]
Abstract
The lungs are exposed to a range of environmental toxins (including cigarette smoke, air pollution, asbestos) and pathogens (bacterial, viral and fungal), and most respiratory diseases are associated with local or systemic hypoxia. All of these adverse factors can trigger endoplasmic reticulum (ER) stress. The ER is a key intracellular site for synthesis of secretory and membrane proteins, regulating their folding, assembly into complexes, transport and degradation. Accumulation of misfolded proteins within the lumen results in ER stress, which activates the unfolded protein response (UPR). Effectors of the UPR temporarily reduce protein synthesis, while enhancing degradation of misfolded proteins and increasing the folding capacity of the ER. If successful, homeostasis is restored and protein synthesis resumes, but if ER stress persists, cell death pathways are activated. ER stress and the resulting UPR occur in a range of pulmonary insults and the outcome plays an important role in many respiratory diseases. The UPR is triggered in the airway of patients with several respiratory diseases and in corresponding experimental models. ER stress has been implicated in the initiation and progression of pulmonary fibrosis, and evidence is accumulating suggesting that ER stress occurs in obstructive lung diseases (particularly in asthma), in pulmonary infections (some viral infections and in the setting of the cystic fibrosis airway) and in lung cancer. While a number of small molecule inhibitors have been used to interrogate the role of the UPR in disease models, many of these tools have complex and off-target effects, hence additional evidence (eg, from genetic manipulation) may be required to support conclusions based on the impact of such pharmacological agents. Aberrant activation of the UPR may be linked to disease pathogenesis and progression, but at present, our understanding of the context-specific and disease-specific mechanisms linking these processes is incomplete. Despite this, the ability of the UPR to defend against ER stress and influence a range of respiratory diseases is becoming increasingly evident, and the UPR is therefore attracting attention as a prospective target for therapeutic intervention strategies.
Collapse
Affiliation(s)
- Kirsty L Bradley
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | - Clare A Stokes
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | | | - Lisa C Parker
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| |
Collapse
|
27
|
Coderre L, Debieche L, Plourde J, Rabasa-Lhoret R, Lesage S. The Potential Causes of Cystic Fibrosis-Related Diabetes. Front Endocrinol (Lausanne) 2021; 12:702823. [PMID: 34394004 PMCID: PMC8361832 DOI: 10.3389/fendo.2021.702823] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/06/2021] [Indexed: 12/16/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator gene (CFTR). Cystic fibrosis-related diabetes (CFRD) is the most common comorbidity, affecting more than 50% of adult CF patients. Despite this high prevalence, the etiology of CFRD remains incompletely understood. Studies in young CF children show pancreatic islet disorganization, abnormal glucose tolerance, and delayed first-phase insulin secretion suggesting that islet dysfunction is an early feature of CF. Since insulin-producing pancreatic β-cells express very low levels of CFTR, CFRD likely results from β-cell extrinsic factors. In the vicinity of β-cells, CFTR is expressed in both the exocrine pancreas and the immune system. In the exocrine pancreas, CFTR mutations lead to the obstruction of the pancreatic ductal canal, inflammation, and immune cell infiltration, ultimately causing the destruction of the exocrine pancreas and remodeling of islets. Both inflammation and ductal cells have a direct effect on insulin secretion and could participate in CFRD development. CFTR mutations are also associated with inflammatory responses and excessive cytokine production by various immune cells, which infiltrate the pancreas and exert a negative impact on insulin secretion, causing dysregulation of glucose homeostasis in CF adults. In addition, the function of macrophages in shaping pancreatic islet development may be impaired by CFTR mutations, further contributing to the pancreatic islet structural defects as well as impaired first-phase insulin secretion observed in very young children. This review discusses the different factors that may contribute to CFRD.
Collapse
Affiliation(s)
- Lise Coderre
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
| | - Lyna Debieche
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
- Département de médecine, Université de Montréal, Montréal, QC, Canada
| | - Joëlle Plourde
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
- Département de médecine, Université de Montréal, Montréal, QC, Canada
| | - Rémi Rabasa-Lhoret
- Division of Cardiovascular and Metabolic Diseases, Institut de recherche clinique de Montréal, Montréal, QC, Canada
- Département de nutrition, Université de Montréal, Montréal, QC, Canada
- Cystic Fibrosis Clinic, Centre Hospitalier de l’Université de Montréal (CHUM), Montréal, QC, Canada
| | - Sylvie Lesage
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
- *Correspondence: Sylvie Lesage,
| |
Collapse
|
28
|
Macrophage metabolic reprogramming during chronic lung disease. Mucosal Immunol 2021; 14:282-295. [PMID: 33184475 PMCID: PMC7658438 DOI: 10.1038/s41385-020-00356-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/13/2020] [Accepted: 10/24/2020] [Indexed: 02/04/2023]
Abstract
Airway macrophages (AMs) play key roles in the maintenance of lung immune tolerance. Tissue tailored, highly specialised and strategically positioned, AMs are critical sentinels of lung homoeostasis. In the last decade, there has been a revolution in our understanding of how metabolism underlies key macrophage functions. While these initial observations were made during steady state or using in vitro polarised macrophages, recent studies have indicated that during many chronic lung diseases (CLDs), AMs adapt their metabolic profile to fit their local niche. By generating reactive oxygen species (ROS) for pathogen defence, utilising aerobic glycolysis to rapidly generate cytokines, and employing mitochondrial respiration to fuel inflammatory responses, AMs utilise metabolic reprogramming for host defence, although these changes may also support chronic pathology. This review focuses on how metabolic alterations underlie AM phenotype and function during CLDs. Particular emphasis is given to how our new understanding of AM metabolic plasticity may be exploited to develop AM-focused therapies.
Collapse
|
29
|
UPR modulation of host immunity by Pseudomonas aeruginosa in cystic fibrosis. Clin Sci (Lond) 2020; 134:1911-1934. [PMID: 32537652 DOI: 10.1042/cs20200066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
Cystic fibrosis (CF) is a progressive multiorgan autosomal recessive disease with devastating impact on the lungs caused by derangements of the CF transmembrane conductance regulator (CFTR) gene. Morbidity and mortality are caused by the triad of impaired mucociliary clearance, microbial infections and chronic inflammation. Pseudomonas aeruginosa is the main respiratory pathogen in individuals with CF infecting most patients in later stages. Despite its recognized clinical impact, molecular mechanisms that underlie P. aeruginosa pathogenesis and the host response to P. aeruginosa infection remain incompletely understood. The nuclear hormone receptor peroxisome proliferator-activated receptor (PPAR) γ (PPARγ), has shown to be reduced in CF airways. In the present study, we sought to investigate the upstream mechanisms repressing PPARγ expression and its impact on airway epithelial host defense. Endoplasmic reticulum-stress (ER-stress) triggered unfolded protein response (UPR) activated by misfolded CFTR and P. aeruginosa infection contributed to attenuated expression of PPARγ. Specifically, the protein kinase RNA (PKR)-like ER kinase (PERK) signaling pathway led to the enhanced expression of the CCAAT-enhancer-binding-protein homologous protein (CHOP). CHOP induction led to the repression of PPARγ expression. Mechanistically, we showed that CHOP induction mediated PPARγ attenuation, impacted the innate immune function of normal and ∆F508 primary airway epithelial cells by reducing expression of antimicrobial peptide (AMP) and paraoxanse-2 (PON-2), as well as enhancing IL-8 expression. Furthermore, mitochondrial reactive oxygen species production (mt-ROS) and ER-stress positive feedforward loop also dysregulated mitochondrial bioenergetics. Additionally, our findings implicate that PPARγ agonist pioglitazone (PIO) has beneficial effect on the host at the multicellular level ranging from host defense to mitochondrial re-energization.
Collapse
|
30
|
Hamilton K, Krause K, Badr A, Daily K, Estfanous S, Eltobgy M, Khweek AA, Anne MNK, Carafice C, Baetzhold D, Tonniges JR, Zhang X, Gavrilin MA, Parinandi NL, Amer AO. Defective immunometabolism pathways in cystic fibrosis macrophages. J Cyst Fibros 2020; 20:664-672. [PMID: 33208300 DOI: 10.1016/j.jcf.2020.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Mitochondria play a key role in immune defense pathways, particularly for macrophages. We and others have previously demonstrated that cystic fibrosis (CF) macrophages exhibit weak autophagy activity and exacerbated inflammatory responses. Previous studies have revealed that mitochondria are defective in CF epithelial cells, but to date, the connection between defective mitochondrial function and CF macrophage immune dysregulation has not been fully elucidated. Here, we present a characterization of mitochondrial dysfunction in CF macrophages. METHODS Mitochondrial function in wild-type (WT) and CF F508del/F508del murine macrophages was measured using the Seahorse Extracellular Flux analyzer. Mitochondrial morphology was investigated using transmission electron and confocal microscopy. Mitochondrial membrane potential (MMP) as well as mitochondrial reactive oxygen species (mROS) were measured using TMRM and MitoSOX Red fluorescent dyes, respectively. All assays were performed at baseline and following infection by Burkholderia cenocepacia, a multi-drug resistant bacterium that causes detrimental infections in CF patients. RESULTS We have identified impaired oxygen consumption in CF macrophages without and with B. cenocepacia infection. We also observed increased mitochondrial fragmentation in CF macrophages following infection. Lastly, we observed increased MMP and impaired mROS production in CF macrophages following infection with B. cenocepacia. CONCLUSIONS The mitochondrial defects identified are key components of the macrophage response to infection. Their presence suggests that mitochondrial dysfunction contributes to impaired bacterial killing in CF macrophages. Our current study will enhance our understanding of the pathobiology of CF and lead to the identification of novel mitochondrial therapeutic targets for CF.
Collapse
Affiliation(s)
- Kaitlin Hamilton
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kathrin Krause
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Asmaa Badr
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Kylene Daily
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Shady Estfanous
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Arwa Abu Khweek
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; Department of Biology and Biochemistry, Birzeit University, Birzeit, West Bank, Palestine
| | - Midhun N K Anne
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Cierra Carafice
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Baetzhold
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jeffrey R Tonniges
- Campus Microscopy and Imaging Facility, The Ohio State University, Columbus, OH 43210, USA
| | - Xiaoli Zhang
- Center for Biostatistics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mikhail A Gavrilin
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Narasimham L Parinandi
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Amal O Amer
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
31
|
Lara-Reyna S, Holbrook J, Jarosz-Griffiths HH, Peckham D, McDermott MF. Dysregulated signalling pathways in innate immune cells with cystic fibrosis mutations. Cell Mol Life Sci 2020; 77:4485-4503. [PMID: 32367193 PMCID: PMC7599191 DOI: 10.1007/s00018-020-03540-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 12/12/2022]
Abstract
Cystic fibrosis (CF) is one of the most common life-limiting recessive genetic disorders in Caucasians, caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR). CF is a multi-organ disease that involves the lungs, pancreas, sweat glands, digestive and reproductive systems and several other tissues. This debilitating condition is associated with recurrent lower respiratory tract bacterial and viral infections, as well as inflammatory complications that may eventually lead to pulmonary failure. Immune cells play a crucial role in protecting the organs against opportunistic infections and also in the regulation of tissue homeostasis. Innate immune cells are generally affected by CFTR mutations in patients with CF, leading to dysregulation of several cellular signalling pathways that are in continuous use by these cells to elicit a proper immune response. There is substantial evidence to show that airway epithelial cells, neutrophils, monocytes and macrophages all contribute to the pathogenesis of CF, underlying the importance of the CFTR in innate immune responses. The goal of this review is to put into context the important role of the CFTR in different innate immune cells and how CFTR dysfunction contributes to the pathogenesis of CF, highlighting several signalling pathways that may be dysregulated in cells with CFTR mutations.
Collapse
Affiliation(s)
- Samuel Lara-Reyna
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, LS9 7TF, UK.
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, LS9 7TF, UK.
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, LS9 7TF, UK.
| | - Jonathan Holbrook
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, LS9 7TF, UK
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, LS9 7TF, UK
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, LS9 7TF, UK
| | - Heledd H Jarosz-Griffiths
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, LS9 7TF, UK
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, LS9 7TF, UK
| | - Daniel Peckham
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, LS9 7TF, UK
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, LS9 7TF, UK
- Adult Cystic Fibrosis Unit, St James's University Hospital, Leeds, LS9 7TF, UK
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, LS9 7TF, UK.
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, LS9 7TF, UK.
| |
Collapse
|
32
|
Airway Inflammation and Host Responses in the Era of CFTR Modulators. Int J Mol Sci 2020; 21:ijms21176379. [PMID: 32887484 PMCID: PMC7504341 DOI: 10.3390/ijms21176379] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
The arrival of cystic fibrosis transmembrane conductance regulator (CFTR) modulators as a new class of treatment for cystic fibrosis (CF) in 2012 represented a pivotal advance in disease management, as these small molecules directly target the upstream underlying protein defect. Further advancements in the development and scope of these genotype-specific therapies have been transformative for an increasing number of people with CF (PWCF). Despite clear improvements in CFTR function and clinical endpoints such as lung function, body mass index (BMI), and frequency of pulmonary exacerbations, current evidence suggests that CFTR modulators do not prevent continued decline in lung function, halt disease progression, or ameliorate pathogenic organisms in those with established lung disease. Furthermore, it remains unknown whether their restorative effects extend to dysfunctional CFTR expressed in phagocytes and other immune cells, which could modulate airway inflammation. In this review, we explore the effects of CFTR modulators on airway inflammation, infection, and their influence on the impaired pulmonary host defences associated with CF lung disease. We also consider the role of inflammation-directed therapies in light of the widespread clinical use of CFTR modulators and identify key areas for future research.
Collapse
|
33
|
Briottet M, Shum M, Urbach V. The Role of Specialized Pro-Resolving Mediators in Cystic Fibrosis Airways Disease. Front Pharmacol 2020; 11:1290. [PMID: 32982730 PMCID: PMC7493015 DOI: 10.3389/fphar.2020.01290] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/04/2020] [Indexed: 12/26/2022] Open
Abstract
Cystic Fibrosis (CF) is a recessive genetic disease due to mutations of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene encoding the CFTR chloride channel. The ion transport abnormalities related to CFTR mutation generate a dehydrated airway surface liquid (ASL) layer, which is responsible for an altered mucociliary clearance, favors infections and persistent inflammation that lead to progressive lung destruction and respiratory failure. The inflammatory response is normally followed by an active resolution phase to return to tissue homeostasis, which involves specialized pro-resolving mediators (SPMs). SPMs promote resolution of inflammation, clearance of microbes, tissue regeneration and reduce pain, but do not evoke unwanted immunosuppression. The airways of CF patients showed a decreased production of SPMs even in the absence of pathogens. SPMs levels in the airway correlated with CF patients' lung function. The prognosis for CF has greatly improved but there remains a critical need for more effective treatments that prevent excessive inflammation, lung damage, and declining pulmonary function for all CF patients. This review aims to highlight the recent understanding of CF airway inflammation and the possible impact of SPMs on functions that are altered in CF airways.
Collapse
Affiliation(s)
| | | | - Valerie Urbach
- Institut national de la santé et de la recherche médicale (Inserm) U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
| |
Collapse
|
34
|
Pehote G, Vij N. Autophagy Augmentation to Alleviate Immune Response Dysfunction, and Resolve Respiratory and COVID-19 Exacerbations. Cells 2020; 9:cells9091952. [PMID: 32847034 PMCID: PMC7565665 DOI: 10.3390/cells9091952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
The preservation of cellular homeostasis requires the synthesis of new proteins (proteostasis) and organelles, and the effective removal of misfolded or impaired proteins and cellular debris. This cellular homeostasis involves two key proteostasis mechanisms, the ubiquitin proteasome system and the autophagy–lysosome pathway. These catabolic pathways have been known to be involved in respiratory exacerbations and the pathogenesis of various lung diseases, such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and coronavirus disease-2019 (COVID-19). Briefly, proteostasis and autophagy processes are known to decline over time with age, cigarette or biomass smoke exposure, and/or influenced by underlying genetic factors, resulting in the accumulation of misfolded proteins and cellular debris, elevating apoptosis and cellular senescence, and initiating the pathogenesis of acute or chronic lung disease. Moreover, autophagic dysfunction results in an impaired microbial clearance, post-bacterial and/or viral infection(s) which contribute to the initiation of acute and recurrent respiratory exacerbations as well as the progression of chronic obstructive and restrictive lung diseases. In addition, the autophagic dysfunction-mediated cystic fibrosis transmembrane conductance regulator (CFTR) immune response impairment further exacerbates the lung disease. Recent studies demonstrate the therapeutic potential of novel autophagy augmentation strategies, in alleviating the pathogenesis of chronic obstructive or restrictive lung diseases and exacerbations such as those commonly seen in COPD, CF, ALI/ARDS and COVID-19.
Collapse
Affiliation(s)
- Garrett Pehote
- Michigan State University College of Osteopathic Medicine, East Lansing, MI 48823, USA;
| | - Neeraj Vij
- Department of Pediatrics and Pulmonary Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- PRECISION THERANOSTICS INC, Baltimore, MD 21202, USA
- VIJ BIOTECH, Baltimore, MD 21202, USA
- Correspondence: or ; Tel.: +1-240-623-0757
| |
Collapse
|
35
|
Zhang S, Shrestha CL, Wisniewski BL, Pham H, Hou X, Li W, Dong Y, Kopp BT. Consequences of CRISPR-Cas9-Mediated CFTR Knockout in Human Macrophages. Front Immunol 2020; 11:1871. [PMID: 32973772 PMCID: PMC7461958 DOI: 10.3389/fimmu.2020.01871] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/13/2020] [Indexed: 01/12/2023] Open
Abstract
Macrophage dysfunction is fundamentally related to altered immunity in cystic fibrosis (CF). How genetic deficits in the cystic fibrosis transmembrane conductance regulator (CFTR) lead to these defects remains unknown. Rapid advances in genomic editing such as the clustered regularly interspaced short palindromic repeats associated protein 9 (CRISPR/Cas9) system provide new tools for scientific study. We aimed to create a stable CFTR knockout (KO) in human macrophages in order to study how CFTR regulates macrophage function. Peripheral blood monocytes were isolated from non-CF healthy volunteers and differentiated into monocyte-derived macrophages (MDMs). MDMs were transfected with a CRISPR Cas9 CFTR KO plasmid. CFTR KO efficiency was verified and macrophage halide efflux, phagocytosis, oxidative burst, apoptosis, and cytokine functional assays were performed. CFTR KO in human MDMs was efficient and stable after puromycin selection. CFTR KO was confirmed by CFTR mRNA and protein expression. CFTR function was abolished in CFTR KO MDMs. CFTR KO recapitulated known defects in human CF MDM (CFTR class I/II variants) dysfunction including (1) increased apoptosis, (2) decreased phagocytosis, (3) reduced oxidative burst, and (4) increased bacterial load. Activation of the oxidative burst via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase assembly was diminished in CFTR KO MDMs (decreased phosphorylated p47phox). Cytokine production was unchanged or decreased in response to infection in CFTR KO MDMs. In conclusion, we developed a primary human macrophage CFTR KO system. CFTR KO mimics most pathology observed in macrophages obtained from persons with CF, which suggests that many aspects of CF macrophage dysfunction are CFTR-dependent and not just reflective of the CF inflammatory milieu.
Collapse
Affiliation(s)
- Shuzhong Zhang
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Chandra L Shrestha
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Benjamin L Wisniewski
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Hanh Pham
- Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| | - Xucheng Hou
- Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH, United States
| | - Wenqing Li
- Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH, United States
| | - Yizhou Dong
- Pharmaceutics and Pharmacology, The Ohio State University, Columbus, OH, United States
| | - Benjamin T Kopp
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States.,Division of Pulmonary Medicine, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
36
|
Turton KB, Ingram RJ, Valvano MA. Macrophage dysfunction in cystic fibrosis: Nature or nurture? J Leukoc Biol 2020; 109:573-582. [PMID: 32678926 DOI: 10.1002/jlb.4ru0620-245r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) affect the homeostasis of chloride flux by epithelial cells. This has deleterious consequences, especially in respiratory epithelia, where the defect results in mucus accumulation distinctive of cystic fibrosis. CFTR is, however, also expressed in phagocytic cells, like macrophages. Immune cells are highly sensitive to conditioning by their environment; thus, CFTR dysfunction in epithelia influences macrophages by affecting the lung milieu, but the mutations also appear to be directly consequential for intrinsic macrophage functions. Particular mutations can alter CFTR's folding, traffic of the protein to the membrane and function. As such, understanding the intrinsic effects of CFTR mutation requires distinguishing the secondary effects of misfolded CFTR on cell stress pathways from the primary defect of CFTR dysfunction/absence. Investigations into CFTR's role in macrophages have exploited various models, each with their own advantages and limitations. This review summarizes these methodologic approaches, discussing their physiological correspondence and highlighting key findings. The controversy surrounding CFTR-dependent acidification is used as a case study to highlight difficulties in commensurability across model systems. Recent work in macrophage biology, including polarization and host-pathogen interaction studies, brought into the context of CFTR research, offers potential explanations for observed discrepancies between studies. Moreover, the rapid advancement of novel gene editing technologies and new macrophage model systems makes this assessment of the field's models and methodologies timely.
Collapse
Affiliation(s)
- Keren B Turton
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Rebecca J Ingram
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| | - Miguel A Valvano
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
37
|
Aoe T. Pathological Aspects of COVID-19 as a Conformational Disease and the Use of Pharmacological Chaperones as a Potential Therapeutic Strategy. Front Pharmacol 2020; 11:1095. [PMID: 32754041 PMCID: PMC7366900 DOI: 10.3389/fphar.2020.01095] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/09/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), the seventh human coronavirus infectious disease, was first reported in Wuhan, China, in December 2019, followed by its rapid spread globally (251,059 deaths, on May 5, 2020, by Johns Hopkins University). An early clinical report showed that fever, cough, fatigue, sputum production, and myalgia were initial symptoms, with the development of pneumonia as the disease progressed. Increases in the level of serum liver enzymes, D-dimer, cardiac troponin I, and creatinine have been observed in severely ill patients, indicating that multiple organ failure had occurred in these cases. Lymphopenia and an increase in interleukin-6 (IL-6) were also observed. Although COVID-19 patients are administered glucocorticoid therapy to treat the excessive immune response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection, the efficacy of this form of therapy is unclear. Viremia is observed in severe cases, suggesting that in addition to type II alveolar epithelial cells, many cell types, such as vascular endothelial cells, cardiomyocytes, renal tubular cells, neuronal cells, and lymphocytes, may be damaged. The improvement of survival rates requires elucidation of the mechanism by which cellular damage occurs during viral infection. Cellular therapy, along with organ support systems such as oxygen therapy, artificial ventilation, extra corporeal membrane oxygenation and dialysis, as well as antiviral therapy, are required. Viral replication in infected host cells may perturb protein folding in the endoplasmic reticulum (ER), causing ER stress. Although an adaptive cellular response, i.e. the unfolded protein response, can compensate for the misfolded protein burden to some extent, continued viral proliferation may induce inflammation and cell death. Therefore, we propose that proteostasis dysfunction may cause conformational disorders in COVID-19. The application of pharmacological chaperone therapy to treat COVID-19 patients is additionally discussed.
Collapse
Affiliation(s)
- Tomohiko Aoe
- Pain Center, Teikyo University Chiba Medical Center, Ichihara, Japan.,Department of Medicine, Teikyo University, Tokyo, Japan
| |
Collapse
|
38
|
Altered iron metabolism in cystic fibrosis macrophages: the impact of CFTR modulators and implications for Pseudomonas aeruginosa survival. Sci Rep 2020; 10:10935. [PMID: 32616918 PMCID: PMC7331733 DOI: 10.1038/s41598-020-67729-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
Cystic fibrosis (CF) is a genetic disease caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, resulting in chronic bacterial lung infections and tissue damage. CF macrophages exhibit reduced bacterial killing and increased inflammatory signaling. Iron is elevated in the CF lung and is a critical nutrient for bacteria, including the common CF pathogen Pseudomonas aeruginosa (Pa). While macrophages are a key regulatory component of extracellular iron, iron metabolism has yet to be characterized in human CF macrophages. Secreted and total protein levels were analyzed in non-CF and F508del/F508del CF monocyte derived macrophages (MDMs) with and without clinically approved CFTR modulators ivacaftor/lumacaftor. CF macrophage transferrin receptor 1 (TfR1) was reduced with ivacaftor/lumacaftor treatment. When activated with LPS, CF macrophage expressed reduced ferroportin (Fpn). After the addition of exogenous iron, total iron was elevated in conditioned media from CF MDMs and reduced in conditioned media from ivacaftor/lumacaftor treated CF MDMs. Pa biofilm formation and viability were elevated in conditioned media from CF MDMs and biofilm formation was reduced in the presence of conditioned media from ivacaftor/lumacaftor treated CF MDMs. Defects in iron metabolism observed in this study may inform host–pathogen interactions between CF macrophages and Pa.
Collapse
|
39
|
Batista A, Rodvold JJ, Xian S, Searles SC, Lew A, Iwawaki T, Almanza G, Waller TC, Lin J, Jepsen K, Carter H, Zanetti M. IRE1α regulates macrophage polarization, PD-L1 expression, and tumor survival. PLoS Biol 2020; 18:e3000687. [PMID: 32520957 PMCID: PMC7307794 DOI: 10.1371/journal.pbio.3000687] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/22/2020] [Accepted: 05/20/2020] [Indexed: 12/14/2022] Open
Abstract
In the tumor microenvironment, local immune dysregulation is driven in part by macrophages and dendritic cells that are polarized to a mixed proinflammatory/immune-suppressive phenotype. The unfolded protein response (UPR) is emerging as the possible origin of these events. Here we report that the inositol-requiring enzyme 1 (IRE1α) branch of the UPR is directly involved in the polarization of macrophages in vitro and in vivo, including the up-regulation of interleukin 6 (IL-6), IL-23, Arginase1, as well as surface expression of CD86 and programmed death ligand 1 (PD-L1). Macrophages in which the IRE1α/X-box binding protein 1 (Xbp1) axis is blocked pharmacologically or deleted genetically have significantly reduced polarization and CD86 and PD-L1 expression, which was induced independent of IFNγ signaling, suggesting a novel mechanism in PD-L1 regulation in macrophages. Mice with IRE1α- but not Xbp1-deficient macrophages showed greater survival than controls when implanted with B16.F10 melanoma cells. Remarkably, we found a significant association between the IRE1α gene signature and CD274 gene expression in tumor-infiltrating macrophages in humans. RNA sequencing (RNASeq) analysis showed that bone marrow-derived macrophages with IRE1α deletion lose the integrity of the gene connectivity characteristic of regulated IRE1α-dependent decay (RIDD) and the ability to activate CD274 gene expression. Thus, the IRE1α/Xbp1 axis drives the polarization of macrophages in the tumor microenvironment initiating a complex immune dysregulation leading to failure of local immune surveillance.
Collapse
Affiliation(s)
- Alyssa Batista
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Jeffrey J. Rodvold
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Su Xian
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Stephen C. Searles
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Alyssa Lew
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - Takao Iwawaki
- Laboratory for Cell Recovery Mechanisms, Brain Science Institute, RIKEN, Hirosawa, Japan
| | - Gonzalo Almanza
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| | - T. Cameron Waller
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Jonathan Lin
- Department of Pathology, Stanford University, Palo Alto, California, United States of America
| | - Kristen Jepsen
- IGM Genomics Center, University of California, San Diego, La Jolla, California, United States of America
| | - Hannah Carter
- Division of Medical Genetics; Department of Medicine, and Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, California, United States of America
| | - Maurizio Zanetti
- The Laboratory of Immunology, Department of Medicine and Moores Cancer Center, University of California, San Diego, La Jolla, California, United States of America
| |
Collapse
|
40
|
Giacalone VD, Dobosh BS, Gaggar A, Tirouvanziam R, Margaroli C. Immunomodulation in Cystic Fibrosis: Why and How? Int J Mol Sci 2020; 21:ijms21093331. [PMID: 32397175 PMCID: PMC7247557 DOI: 10.3390/ijms21093331] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 01/09/2023] Open
Abstract
Cystic fibrosis (CF) lung disease is characterized by unconventional mechanisms of inflammation, implicating a chronic immune response dominated by innate immune cells. Historically, therapeutic development has focused on the mutated cystic fibrosis transmembrane conductance regulator (CFTR), leading to the discovery of small molecules aiming at modulating and potentiating the presence and activity of CFTR at the plasma membrane. However, treatment burden sustained by CF patients, side effects of current medications, and recent advances in other therapeutic areas have highlighted the need to develop novel disease targeting of the inflammatory component driving CF lung damage. Furthermore, current issues with standard treatment emphasize the need for directed lung therapies that could minimize systemic side effects. Here, we summarize current treatment used to target immune cells in the lungs, and highlight potential benefits and caveats of novel therapeutic strategies.
Collapse
Affiliation(s)
- Vincent D. Giacalone
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (V.D.G.); (B.S.D.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Brian S. Dobosh
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (V.D.G.); (B.S.D.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.G.); (C.M.)
- Pulmonary Section, Birmingham VA Medical Center, Birmingham, AL 35233, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (V.D.G.); (B.S.D.)
- Center for CF & Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Correspondence:
| | - Camilla Margaroli
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA; (A.G.); (C.M.)
| |
Collapse
|
41
|
Savic S, Caseley EA, McDermott MF. Moving towards a systems-based classification of innate immune-mediated diseases. NATURE REVIEWS. RHEUMATOLOGY 2020. [PMID: 32107482 DOI: 10.1038/s41584-020-0377-5)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Autoinflammation as a distinct disease category was first reported in 1999 as a group of monogenic disorders characterized by recurrent episodes of systemic and organ-specific inflammation, known as periodic fever syndromes. Since this original description, the focus has shifted considerably to the inclusion of complex multifactorial conditions with an autoinflammatory basis. Furthermore, the boundaries of what are considered to be autoinflammatory disorders are constantly evolving and currently encompass elements of immunodeficiency and autoimmunity. Notable developments in the intervening 20 years include substantial progress in understanding how the different inflammasomes are activated, how infection is sensed by the innate immune system and how intracellular signalling systems are consequently activated and integrated with many different cellular functions in the autoinflammatory process. With these developments, the field of autoinflammation is moving from a gene-centric view of innate immune-mediated disease towards a systems-based concept, which describes how various convergent pathways, including pyrin and the actin cytoskeleton, protein misfolding and cellular stress, NF-κB dysregulation and interferon activation, contribute to the autoinflammatory process. The development and adoption of a systems-based concept of systemic autoinflammatory diseases is anticipated to have implications for the development of treatments that target specific components of the innate immune system.
Collapse
Affiliation(s)
- Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital, Leeds, UK. .,National Institute for Health Research-Leeds Biomedical Research Centre, Chapel Allerton Hospital, Leeds, UK. .,Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, UK.
| | - Emily A Caseley
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital, Leeds, UK
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, St James's University Hospital, Leeds, UK.
| |
Collapse
|
42
|
Moving towards a systems-based classification of innate immune-mediated diseases. Nat Rev Rheumatol 2020; 16:222-237. [DOI: 10.1038/s41584-020-0377-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2020] [Indexed: 02/07/2023]
|
43
|
Li A, Song NJ, Riesenberg BP, Li Z. The Emerging Roles of Endoplasmic Reticulum Stress in Balancing Immunity and Tolerance in Health and Diseases: Mechanisms and Opportunities. Front Immunol 2020; 10:3154. [PMID: 32117210 PMCID: PMC7026265 DOI: 10.3389/fimmu.2019.03154] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/30/2019] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum (ER) is an organelle equipped with mechanisms for proper protein folding, trafficking, and degradation to maintain protein homeostasis in the secretory pathway. As a defense mechanism, perturbation of ER proteostasis by ER stress agents activates a cascade of signaling pathways from the ER to the nucleus known as unfolded protein response (UPR). The primary goal of UPR is to induce transcriptional and translational programs to restore ER homeostasis for cell survival. As such, defects in UPR signaling have been implicated as a key contributor to multiple diseases including metabolic diseases, degenerative diseases, inflammatory disorders, and cancer. Growing evidence support the critical role of ER stress in regulating the fate as well as the magnitude of the immune response. Moreover, the availability of multiple UPR pharmacological inhibitors raises the hope that targeting UPR can be a new strategy for immune modulation and immunotherapy of diseases. This paper reviews the principal mechanisms by which ER stress affects immune cell biology and function, with a focus of discussion on UPR-associated immunopathology and the development of potential ER stress-targeted therapeutics.
Collapse
Affiliation(s)
- Anqi Li
- College of Medicine, The Ohio State University, Columbus, OH, United States.,The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
| | - No-Joon Song
- The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
| | - Brian P Riesenberg
- The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States
| | - Zihai Li
- College of Medicine, The Ohio State University, Columbus, OH, United States.,The James Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, United States.,Division of Medical Oncology, Department of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
44
|
Choi JA, Song CH. Insights Into the Role of Endoplasmic Reticulum Stress in Infectious Diseases. Front Immunol 2020; 10:3147. [PMID: 32082307 PMCID: PMC7005066 DOI: 10.3389/fimmu.2019.03147] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 12/27/2019] [Indexed: 11/13/2022] Open
Abstract
The endoplasmic reticulum (ER) is the major organelle in the cell for protein folding and plays an important role in cellular functions. The unfolded protein response (UPR) is activated in response to misfolded or unfolded protein accumulation in the ER. However, the UPR successfully alleviates the ER stress. If UPR fails to restore ER homeostasis, apoptosis is induced. ER stress plays an important role in innate immune signaling in response to microorganisms. Dysregulation of UPR signaling contributes to the pathogenesis of a variety of infectious diseases. In this review, we summarize the contribution of ER stress to the innate immune response to invading microorganisms and its role in the pathogenesis of infectious diseases.
Collapse
Affiliation(s)
- Ji-Ae Choi
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Chang-Hwa Song
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, South Korea.,Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, South Korea.,Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, South Korea
| |
Collapse
|
45
|
Abstract
Cystic fibrosis (CF) is an autosomal-recessive multi-organ disease characterized by airways obstruction, recurrent infections, and systemic inflammation. Vasculitis is a severe complication of CF that affects 2-3% of CF patients and is generally associated with poor prognosis. Various pathogenic mechanisms may be involved in the development of CF-related vasculitis. Bacterial colonization leads to persistent activation of neutrophilic granulocytes, inflammation and damage, contributing to the production of antineutrophil cytoplasmic autoantibodies (ANCAs). The presence of ANCA may on the other hand predispose to bacterial colonization and infection, likely entertaining a vicious circle amplifying inflammation and damage. As a result, in CF-associated vasculitis, ongoing inflammation, immune cell activation, the presence of pathogens, and the use of numerous medications may lead to immune complex formation and deposition, subsequently causing leukocytoclastic vasculitis. Published individual case reports and small case series suggest that patients with CF-associated vasculitis require immune modulating treatment, including non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids, hydroxychloroquine, and/or disease-modifying anti-rheumatic drugs (DMARDs). As immunosuppression increases the risk of infection and/or malignancy, which are both already increased in people with CF, possible alternative medications may involve the blockade of individual cytokine or inflammatory pathways, or the use of novel CFTR modulators. This review summarizes molecular alterations involved in CF-associated vasculitis, clinical presentation, and complications, as well as currently available and future treatment options.
Collapse
Affiliation(s)
- Francesca Sposito
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Paul S McNamara
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Christian M Hedrich
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.,Department of Paediatric Rheumatology, Alder Hey Children's National Health Service Foundation Trust Hospital, Liverpool, United Kingdom
| |
Collapse
|
46
|
Scambler T, Jarosz-Griffiths HH, Lara-Reyna S, Pathak S, Wong C, Holbrook J, Martinon F, Savic S, Peckham D, McDermott MF. ENaC-mediated sodium influx exacerbates NLRP3-dependent inflammation in cystic fibrosis. eLife 2019; 8:49248. [PMID: 31532390 PMCID: PMC6764826 DOI: 10.7554/elife.49248] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022] Open
Abstract
Cystic Fibrosis (CF) is a monogenic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, resulting in defective CFTR-mediated chloride and bicarbonate transport, with dysregulation of epithelial sodium channels (ENaC). These changes alter fluid and electrolyte homeostasis and result in an exaggerated proinflammatory response driven, in part, by infection. We tested the hypothesis that NLRP3 inflammasome activation and ENaC upregulation drives exaggerated innate-immune responses in this multisystem disease. We identify an enhanced proinflammatory signature, as evidenced by increased levels of IL-18, IL-1β, caspase-1 activity and ASC-speck release in monocytes, epithelia and serum with CF-associated mutations; these differences were reversed by pretreatment with NLRP3 inflammasome inhibitors and notably, inhibition of amiloride-sensitive sodium (Na+) channels. Overexpression of β-ENaC, in the absence of CFTR dysfunction, increased NLRP3-mediated inflammation, indicating that dysregulated, ENaC-dependent signalling may drive exaggerated inflammatory responses in CF. These data support a role for sodium in modulating NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Thomas Scambler
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Heledd H Jarosz-Griffiths
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom.,Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom
| | - Samuel Lara-Reyna
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Shelly Pathak
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Chi Wong
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom.,Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom
| | - Jonathan Holbrook
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom.,Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom
| | - Fabio Martinon
- Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom.,Department of Biochemistry, University of Lausanne, Lausanne, Switzerland
| | - Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom.,Department of Clinical Immunology and Allergy, St James's University Hospital, Leeds, United Kingdom
| | - Daniel Peckham
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom.,Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom.,Adult Cystic Fibrosis Unit, St James' University Hospital, Leeds, United Kingdom
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom.,Leeds Cystic Fibrosis Trust Strategic Research Centre, University of Leeds, Leeds, United Kingdom
| |
Collapse
|