1
|
Caterino M, Costanzo M, Castaldo A, Iacotucci P, Carnovale V, Ruoppolo M, Gelzo M, Castaldo G. Metabolomic profiling of saliva from cystic fibrosis patients. Sci Rep 2025; 15:479. [PMID: 39747338 PMCID: PMC11696459 DOI: 10.1038/s41598-024-84191-9] [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: 09/12/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
The development of targeted therapies that correct the effect of mutations in patients with cystic fibrosis (CF) and the relevant heterogeneity of the clinical expression of the disease require biomarkers correlated to the severity of the disease useful for monitoring the therapeutic effects. We applied a targeted metabolomic approach by LC-MS/MS on saliva samples from 70 adult CF patients and 63 age/sex-matched controls to investigate alterations in metabolic pathways related to pancreatic insufficiency (PI), Pseudomonas aeruginosa (PA) colonization, CF liver disease (CFLD), and CF related diabetes (CFRD). Sixty salivary metabolites were differentially expressed, with 11 being less abundant and 49 more abundant in CF patients. Among these, the most relevant alterations involved salivary ADMA, N-acetylornithine, methionine and methionine sulfoxide levels. Furthermore, methionine was significantly lower in CF patients with PI and salivary histamine levels were significantly lower in patients colonized by PA. Moreover, ADMA as well as N-acetylornithine and methionine were significantly lower in CF patients with CFRD than in patients without CFRD. Finally, the levels of DOPA resulted significantly lower in saliva from patients with liver disease. Our study revealed an imbalance in arginine methylation and tryptophan pathway related to CFRD and PI as well as alterations in dopaminergic pathway and Krebs cycle related to CFLD. This study also highlights different salivary metabolites as new potential biomarkers in a non-invasive sample that could represent a useful tool for the stratification and management of CF patients.
Collapse
Affiliation(s)
- M Caterino
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - M Costanzo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - A Castaldo
- SC di Pneumologia e UTSIR, AORN Santobono-Pausilipon, Naples, Italy
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, Naples, Italy
| | - P Iacotucci
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | - V Carnovale
- Dipartimento di Scienze Mediche Traslazionali, Università di Napoli Federico II, Naples, Italy
| | - M Ruoppolo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| | - M Gelzo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy.
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy.
| | - G Castaldo
- CEINGE-Biotecnologie avanzate Franco Salvatore, Via G. Salvatore 486, Naples, 80145, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Naples, Italy
| |
Collapse
|
2
|
Nunzi E, Pariano M, Costantini C, Garaci E, Puccetti P, Romani L. Host-microbe serotonin metabolism. Trends Endocrinol Metab 2025; 36:83-95. [PMID: 39142913 DOI: 10.1016/j.tem.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024]
Abstract
As a result of a long evolutionary history, serotonin plays a variety of physiological roles, including neurological, cardiovascular, gastrointestinal, and endocrine functions. While many of these activities can be accommodated within the serotoninergic activity, recent findings have revealed an unsuspected role of serotonin in orchestrating host and microbial dialogue at the tryptophan dining table, to the benefit of local and systemic homeostasis. Herein we review the dual role of serotonin at the host-microbe interface and discuss how unraveling the interconnections among the host and microbial pathways of tryptophan degradation may help to accommodate the versatility of serotonin in physiology and pathology.
Collapse
Affiliation(s)
- Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy; Casa di cura San Raffaele, Sulmona, L'Aquila, Italy.
| |
Collapse
|
3
|
Garaci E, Pariano M, Nunzi E, Costantini C, Bellet MM, Antognelli C, Russo MA, Romani L. Bacteria and fungi of the lung: allies or enemies? Front Pharmacol 2024; 15:1497173. [PMID: 39584143 PMCID: PMC11584946 DOI: 10.3389/fphar.2024.1497173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024] Open
Abstract
Moving from the earlier periods in which the lungs were believed to represent sterile environments, our knowledge on the lung microbiota has dramatically increased, from the first descriptions of the microbial communities inhabiting the healthy lungs and the definition of the ecological rules that regulate its composition, to the identification of the changes that occur in pathological conditions. Despite the limitations of lung as a microbiome reservoir due to the low microbial biomass and abundance, defining its microbial composition and function in the upper and lower airways may help understanding the impact on local homeostasis and its disruption in lung diseases. In particular, the understanding of the metabolic and immune significance of microbes, their presence or lack thereof, in health and disease states could be valuable in development of novel druggable targets in disease treatments. Next-generation sequencing has identified intricate inter-microbe association networks that comprise true mutualistic or antagonistic direct or indirect relationships in the respiratory tract. In this review, the tripartite interaction of bacteria, fungi and the mammalian host is addressed to provide an integrated view of the microbial-host cross-talk in lung health and diseases from an immune and metabolic perspective.
Collapse
Affiliation(s)
- Enrico Garaci
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Cinzia Antognelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Luigina Romani
- San Raffaele Research Center, Sulmona, L’Aquila, Italy
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
4
|
Yuasa HJ. Biochemical and kinetic properties of three indoleamine 2,3-dioxygenases of Aspergillus fumigatus: mechanism of increase in the apparent K m by ascorbate. FEBS J 2024; 291:5037-5050. [PMID: 39375930 DOI: 10.1111/febs.17290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/10/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is a monomeric heme enzyme that catalyzes the oxidative cleavage of tryptophan (L-Trp) to form N-formyl-kynurenine. Similar to other heme proteins, IDO only binds to O2 when the heme iron is ferrous (FeII), thereby rendering the enzyme active. Thus, ascorbate (Asc, a reducing agent) and methylene blue (MB, an electron carrier) are commonly added to in vitro IDO assay systems. However, Asc and MB have been recently reported to significantly impact the measurement of the enzymatic parameters of vertebrate IDO. Aspergillus fumigatus is a filamentous fungus and the most common cause of invasive aspergillosis; it has three IDO genes (IDOα, IDOβ, and IDOγ). The FeII-O2 IDOs of A. fumigatus, particularly FeII-O2 IDOγ, have relatively long half-lives in their autoxidation; however, the autoxidation was accelerated by Asc. Similar to vertebrate IDOs, Asc acted as a competitive (or mixed-competitive) inhibitor of the IDOs of A. fumigatus. A positive correlation (in the order of IDOγ > IDOβ > IDOα) was observed between the inhibitory sensitivity of the IDOs to Asc and the facilitation of their autoxidation by Asc. The FeII-O2 IDO can repeat the dioxygenase reaction as long as it reacts with L-Trp; however, substrate-free FeII-O2 IDO is converted into inactive FeIII-IDO by autoxidation. Thus, L-Trp (which keeps the IDO active) competes with Asc (which inactivates IDO by accelerating autoxidation). This is probably why Asc, which is structurally quite different from L-Trp, appears to function as a competitive (or mixed-competitive) inhibitor of IDOs.
Collapse
Affiliation(s)
- Hajime Julie Yuasa
- Laboratory of Biochemistry, Department of Chemistry and Biotechnology, Faculty of Science and Technology, National University Corporation Kochi University, Japan
| |
Collapse
|
5
|
Silva-Gomes R, Caldeira I, Fernandes R, Cunha C, Carvalho A. Metabolic regulation of the host-fungus interaction: from biological principles to therapeutic opportunities. J Leukoc Biol 2024; 116:469-486. [PMID: 38498599 DOI: 10.1093/jleuko/qiae045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/11/2024] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
Fungal infections present a significant global public health concern, impacting over 1 billion individuals worldwide and resulting in more than 3 million deaths annually. Despite considerable progress in recent years, the management of fungal infections remains challenging. The limited development of novel diagnostic and therapeutic approaches is largely attributed to our incomplete understanding of the pathogenetic mechanisms involved in these diseases. Recent research has highlighted the pivotal role of cellular metabolism in regulating the interaction between fungi and their hosts. In response to fungal infection, immune cells undergo complex metabolic adjustments to meet the energy demands necessary for an effective immune response. A comprehensive understanding of the metabolic circuits governing antifungal immunity, combined with the integration of individual host traits, holds the potential to inform novel medical interventions for fungal infections. This review explores recent insights into the immunometabolic regulation of host-fungal interactions and the infection outcome and discusses how the metabolic repurposing of immune cell function could be exploited in innovative and personalized therapeutic approaches.
Collapse
Affiliation(s)
- Rita Silva-Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Inês Caldeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Raquel Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
6
|
Pamart G, Gosset P, Le Rouzic O, Pichavant M, Poulain-Godefroy O. Kynurenine Pathway in Respiratory Diseases. Int J Tryptophan Res 2024; 17:11786469241232871. [PMID: 38495475 PMCID: PMC10943758 DOI: 10.1177/11786469241232871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/28/2024] [Indexed: 03/19/2024] Open
Abstract
The kynurenine pathway is the primary route for tryptophan catabolism and has received increasing attention as its association with inflammation and the immune system has become more apparent. This review provides a broad overview of the kynurenine pathway in respiratory diseases, from the initial observations to the characterization of the different cell types involved in the synthesis of kynurenine metabolites and the underlying immunoregulatory mechanisms. With a focus on respiratory infections, the various attempts to characterize the kynurenine/tryptophan (K/T) ratio as an inflammatory marker are reviewed. Its implication in chronic lung inflammation and its exacerbation by respiratory pathogens is also discussed. The emergence of preclinical interventional studies targeting the kynurenine pathway opens the way for the future development of new therapies.
Collapse
Affiliation(s)
- Guillaume Pamart
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Philippe Gosset
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Olivier Le Rouzic
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Muriel Pichavant
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Odile Poulain-Godefroy
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 -CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
7
|
Yang B, Rutkowski N, Elisseeff J. The foreign body response: emerging cell types and considerations for targeted therapeutics. Biomater Sci 2023; 11:7730-7747. [PMID: 37904536 DOI: 10.1039/d3bm00629h] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
The foreign body response (FBR) remains a clinical challenge in the field of biomaterials due to its ability to elicit a chronic and sustained immune response. Modulating the immune response to materials is a modern paradigm in tissue engineering to enhance repair while limiting fibrous encapsulation and implant isolation. Though the classical mediators of the FBR are well-characterized, recent studies highlight that our understanding of the cell types that shape the FBR may be incomplete. In this review, we discuss the emerging role of T cells, stromal-immune cell interactions, and senescent cells in the biomaterial response, particularly to synthetic materials. We emphasize future studies that will deepen the field's understanding of these cell types in the FBR, with the goal of identifying therapeutic targets that will improve implant integration. Finally, we briefly review several considerations that may influence our understanding of the FBR in humans, including rodent models, aging, gut microbiota, and sex differences. A better understanding of the heterogeneous host cell response during the FBR can enable the design and development of immunomodulatory materials that favor healing.
Collapse
Affiliation(s)
- Brenda Yang
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Natalie Rutkowski
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| | - Jennifer Elisseeff
- Translational Tissue Engineering Center, Wilmer Eye Institute and Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
Chadwick C, Lehman H, Luebbert S, Abdul-Aziz R, Borowitz D. Autoimmunity in people with cystic fibrosis. J Cyst Fibros 2023; 22:969-979. [PMID: 36966037 DOI: 10.1016/j.jcf.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 03/27/2023]
Abstract
Cystic fibrosis (CF) clinicians may see patients who have difficult-to-manage symptoms that do not have a clear CF-related etiology, such as unusual gastrointestinal (GI) complaints, vasculitis, or arthritis. Alterations in immunity, inflammation and intraluminal dysbiosis create a milieu that may lead to autoimmunity, and the CF transmembrane regulator protein may have a direct role as well. While autoantibodies and other autoimmune markers may develop, these may or may not lead to organ involvement, therefore they are helpful but not sufficient to establish an autoimmune diagnosis. Autoimmune involvement of the GI tract is the best-established association. Next steps to understand autoimmunity in CF should include a more in-depth assessment of the community perspective on its impact. In addition, bringing together specialists in various fields including, but not limited to, pulmonology, gastroenterology, immunology, and rheumatology, would lead to cross-dissemination and help define the path forward in basic science and clinical practice.
Collapse
Affiliation(s)
| | - Heather Lehman
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | | | - Rabheh Abdul-Aziz
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Drucy Borowitz
- Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
9
|
Guo B, Xue M, Zhang T, Gan H, Lin R, Liu M, Liao Y, Lyu J, Zheng P, Sun B. Correlation between immune-related Tryptophan-Kynurenine pathway and severity of severe pneumonia and inflammation-related polyunsaturated fatty acids. Immun Inflamm Dis 2023; 11:e1088. [PMID: 38018595 PMCID: PMC10659755 DOI: 10.1002/iid3.1088] [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: 05/29/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Immune dysfunction and oxidative stress caused by severe pneumonia can lead to multiple organ dysfunction and even death, causing a significant impact on health and the economy. Currently, great progress has been made in the diagnosis and treatment of this disease, but the mortality rate remains high (approximately 50%). Therefore, there is still potential for further exploration of the immune response mechanisms against severe pneumonia. OBJECTIVE This study analyzed the difference in serum metabolic profiles between patients with severe pneumonia and health individuals through metabolomics, aiming to uncover the correlation between the Tryptophan-Kynurenine pathway and the severity of severe pneumonia, as well as N-3/N-6 polyunsaturated fatty acids (PUFAs). METHODS In this study, 44 patients with severe pneumonia and 37 health controls were selected. According to the changes in the disease symptoms within the 7 days of admission, the patients were divided into aggravation (n = 22) and remission (n = 22) groups. Targeted metabolomics techniques were performed to quantify serum metabolites and analyze changes between groups. RESULTS Metabolomics analysis showed that serum kynurenine and kynurenine/tryptophan (K/T) were significantly increased and tryptophan was significantly decreased in patients with severe pneumonia; HETE and HEPE in lipids increased significantly, while eicosapentaenoic acid (EPA), docosapentaenoic acid (DPA), docosahexaenoic acid (DHA), α-linolenic acid (linolenic acid, α-LNA), arachidonic acid (ARA), Dihomo-γ-linolenic acid (DGLA), and 13(s)-hydroperoxylinoleic acid (HPODE) decreased significantly. Additionally, the longitudinal comparison revealed that Linolenic acid, DPA, and Tryptophan increased significantly in the remission group, while and kynurenine and K/T decreased significantly. In the aggravation group, Kynurenine and K/T increased significantly, while ARA, 8(S)-hydroxyeicosatetraenoic acid (HETE), 11(S)-HETE, and Tryptophan decreased significantly. The correlation analysis matrix demonstrated that Tryptophan was positively correlated with DGLA, 12(S)-hydroxyeicosapentaenoic acid (HEPE), ARA, EPA, α-LNA, DHA, and DPA. Kynurenine was positively correlated with 8(S)-HETE and negatively correlated with DHA. Additionally, K/T was negatively correlated with DGLA, ARA, EPA, α-LNA, DHA, and DPA. CONCLUSION This study revealed that during severe pneumonia, the Tryptophan-Kynurenine pathway was activated and was positively correlated with the disease progression. On the other hand, the activation of the Tryptophan-Kynurenine pathway was negatively correlated with N-3/N-6 PUFAs.
Collapse
Affiliation(s)
- Baojun Guo
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
- School of MedicineHenan UniversityKaifengHenanChina
| | - Mingshan Xue
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
| | - Teng Zhang
- China Institute for Radiation ProtectionTaiyuanChina
| | - Hui Gan
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
| | - Runpei Lin
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
| | - Mingtao Liu
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
| | - Yuhong Liao
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
| | - Jiali Lyu
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
| | - Peiyan Zheng
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
| | - Baoqing Sun
- Department of Clinical LaboratoryNational Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University (The Key Laboratory of Advanced Interdisciplinary Studies Center, Advanced Interdisciplinary Studies Center)GuangzhouChina
| |
Collapse
|
10
|
Renga G, D'Onofrio F, Pariano M, Galarini R, Barola C, Stincardini C, Bellet MM, Ellemunter H, Lass-Flörl C, Costantini C, Napolioni V, Ehrlich AK, Antognelli C, Fini M, Garaci E, Nunzi E, Romani L. Bridging of host-microbiota tryptophan partitioning by the serotonin pathway in fungal pneumonia. Nat Commun 2023; 14:5753. [PMID: 37717018 PMCID: PMC10505232 DOI: 10.1038/s41467-023-41536-8] [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: 02/13/2023] [Accepted: 09/06/2023] [Indexed: 09/18/2023] Open
Abstract
The aromatic amino acid L-tryptophan (Trp) is essentially metabolized along the host and microbial pathways. While much is known about the role played by downstream metabolites of each pathways in intestinal homeostasis, their role in lung immune homeostasis is underappreciated. Here we have examined the role played by the Trp hydroxylase/5-hydroxytryptamine (5-HT) pathway in calibrating host and microbial Trp metabolism during Aspergillus fumigatus pneumonia. We found that 5-HT produced by mast cells essentially contributed to pathogen clearance and immune homeostasis in infection by promoting the host protective indoleamine-2,3-dioxygenase 1/kynurenine pathway and limiting the microbial activation of the indole/aryl hydrocarbon receptor pathway. This occurred via regulation of lung and intestinal microbiota and signaling pathways. 5-HT was deficient in the sputa of patients with Cystic fibrosis, while 5-HT supplementation restored the dysregulated Trp partitioning in murine disease. These findings suggest that 5-HT, by bridging host-microbiota Trp partitioning, may have clinical effects beyond its mood regulatory function in respiratory pathologies with an inflammatory component.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Fiorella D'Onofrio
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberta Galarini
- Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche "Togo Rosati,", Perugia, Italy
| | - Carolina Barola
- Istituto Zooprofilattico Sperimentale dell'Umbria e delle Marche "Togo Rosati,", Perugia, Italy
| | | | - Marina M Bellet
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Cornelia Lass-Flörl
- Division of Hygiene and Medical Microbiology, Innsbruck Medical University, Innsbruck, Austria
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Valerio Napolioni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Allison K Ehrlich
- Department of Environmental Toxicology, University of California, Davis, CA, USA
| | - Cinzia Antognelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimo Fini
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, Rome, Italy
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, Rome, Italy
| | - Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, Rome, Italy.
| |
Collapse
|
11
|
Lashgari NA, Roudsari NM, Shayan M, Niazi Shahraki F, Hosseini Y, Momtaz S, Abdolghaffari AH. IDO/Kynurenine; novel insight for treatment of inflammatory diseases. Cytokine 2023; 166:156206. [PMID: 37120946 DOI: 10.1016/j.cyto.2023.156206] [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: 01/07/2023] [Revised: 03/26/2023] [Accepted: 04/04/2023] [Indexed: 05/02/2023]
Abstract
Inflammation and oxidative stress play pivotal roles in pathogenesis of many diseases including cancer, type 2 diabetes, cardiovascular disease, atherosclerosis, neurological diseases, and inflammatory diseases such as inflammatory bowel disease (IBD). Inflammatory mediators such as interleukins (ILs), interferons (INF-s), and tumor necrosis factor (TNF)-α are related to an extended chance of inflammatory diseases initiation or progression due to the over expression of the nuclear factor Kappa B (NF-κB), signal transducer of activators of transcription (STAT), nod-like receptor family protein 3 (NLRP), toll-like receptors (TLR), mitogen-activated protein kinase (MAPK), and mammalian target of rapamycin (mTOR) pathways. These pathways are completely interconnected. Theindoleamine 2,3 dioxygenase (IDO) subset of the kynurenine (KYN) (IDO/KYN), is a metabolic inflammatory pathway involved in production of nicotinamide adenine dinucleotide (NAD + ). It has been shown that IDO/KYN actively participates in inflammatory processes and can increase the secretion of cytokines that provoke inflammatory diseases. Data were extracted from clinical and animal studies published in English between 1990-April 2022, which were collected from PubMed, Google Scholar, Scopus, and Cochrane library. IDO/KYN is completely associated with inflammatory-related pathways, thus leading to the production of cytokines such as TNF-α, IL-1β, and IL-6, and ultimately development and progression of various inflammatory disorders. Inhibition of the IDO/KYN pathway might be a novel therapeutic option for inflammatory diseases. Herein, we gathered data on probable interactions of the IDO/KYN pathway with induction of some inflammatory diseases.
Collapse
Affiliation(s)
- Naser-Aldin Lashgari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Momeni Roudsari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Shayan
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Niazi Shahraki
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Yasamin Hosseini
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Karaj, Iran; Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amir Hossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
12
|
Ribeiro CMP, Higgs MG, Muhlebach MS, Wolfgang MC, Borgatti M, Lampronti I, Cabrini G. Revisiting Host-Pathogen Interactions in Cystic Fibrosis Lungs in the Era of CFTR Modulators. Int J Mol Sci 2023; 24:ijms24055010. [PMID: 36902441 PMCID: PMC10003689 DOI: 10.3390/ijms24055010] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) modulators, a new series of therapeutics that correct and potentiate some classes of mutations of the CFTR, have provided a great therapeutic advantage to people with cystic fibrosis (pwCF). The main hindrances of the present CFTR modulators are related to their limitations in reducing chronic lung bacterial infection and inflammation, the main causes of pulmonary tissue damage and progressive respiratory insufficiency, particularly in adults with CF. Here, the most debated issues of the pulmonary bacterial infection and inflammatory processes in pwCF are revisited. Special attention is given to the mechanisms favoring the bacterial infection of pwCF, the progressive adaptation of Pseudomonas aeruginosa and its interplay with Staphylococcus aureus, the cross-talk among bacteria, the bronchial epithelial cells and the phagocytes of the host immune defenses. The most recent findings of the effect of CFTR modulators on bacterial infection and the inflammatory process are also presented to provide critical hints towards the identification of relevant therapeutic targets to overcome the respiratory pathology of pwCF.
Collapse
Affiliation(s)
- Carla M. P. Ribeiro
- Marsico Lung Institute/Cystic Fibrosis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Correspondence: (C.M.P.R.); (G.C.)
| | - Matthew G. Higgs
- Marsico Lung Institute/Cystic Fibrosis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marianne S. Muhlebach
- Marsico Lung Institute/Cystic Fibrosis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew C. Wolfgang
- Marsico Lung Institute/Cystic Fibrosis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Monica Borgatti
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
- Innthera4CF, Center on Innovative Therapies for Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
| | - Ilaria Lampronti
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
- Innthera4CF, Center on Innovative Therapies for Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
| | - Giulio Cabrini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
- Innthera4CF, Center on Innovative Therapies for Cystic Fibrosis, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (C.M.P.R.); (G.C.)
| |
Collapse
|
13
|
Westhölter D, Raspe J, Uebner H, Pipping J, Schmitz M, Straßburg S, Sutharsan S, Welsner M, Taube C, Reuter S. Regulatory T cell enhancement in adults with cystic fibrosis receiving Elexacaftor/Tezacaftor/Ivacaftor therapy. Front Immunol 2023; 14:1107437. [PMID: 36875141 PMCID: PMC9978140 DOI: 10.3389/fimmu.2023.1107437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Cystic fibrosis (CF), especially CF lung disease, is characterized by chronic infection, immune dysfunction including impairment of regulatory T cells (Tregs) and an exaggerated inflammatory response. CF transmembrane conductance regulator (CFTR) modulators have shown to improve clinical outcomes in people with CF (PwCF) with a wide range of CFTR mutations. However, it remains unclear whether CFTR modulator therapy also affects CF-associated inflammation. We aimed to examine the effect of elexacaftor/tezacaftor/ivacaftor therapy on lymphocyte subsets and systemic cytokines in PwCF. Methods Peripheral blood mononuclear cells and plasma were collected before and at three and six months after the initiation of elexacaftor/tezacaftor/ivacaftor therapy; lymphocyte subsets and systemic cytokines were determined using flow cytometry. Results Elexacaftor/tezacaftor/ivacaftor treatment was initiated in 77 PwCF and improved percent predicted FEV1 by 12.5 points (p<0.001) at 3 months. During elexacaftor/tezacaftor/ivacaftor therapy, percentages of Tregs were enhanced (+18.7%, p<0.001), with an increased proportion of Tregs expressing CD39 as a marker of stability (+14.4%, p<0.001). Treg enhancement was more pronounced in PwCF clearing Pseudomonas aeruginosa infection. Only minor, non-significant shifts were observed among Th1-, Th2- and Th17-expressing effector T helper cells. These results were stable at 3- and 6-month follow-up. Cytokine measurements showed a significant decrease in interleukin-6 levels during treatment with elexacaftor/tezacaftor/ivacaftor (-50.2%, p<0.001). Conclusion Treatment with elexacaftor/tezacaftor/ivacaftor was associated with an increased percentage of Tregs, especially in PwCF clearing Pseudomonas aeruginosa infection. Targeting Treg homeostasis is a therapeutic option for PwCF with persistent Treg impairment.
Collapse
Affiliation(s)
- Dirk Westhölter
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Jonas Raspe
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Hendrik Uebner
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Johannes Pipping
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Mona Schmitz
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Svenja Straßburg
- Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Sivagurunathan Sutharsan
- Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Matthias Welsner
- Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| |
Collapse
|
14
|
Abstract
Cystic fibrosis (CF) pathophysiology is hallmarked by excessive inflammation and the inability to resolve lung infections, contributing to morbidity and eventually mortality. Paradoxically, despite a robust inflammatory response, CF lungs fail to clear bacteria and are susceptible to chronic infections. Impaired mucociliary transport plays a critical role in chronic infection but the immune mechanisms contributing to the adaptation of bacteria to the lung microenvironment is not clear. CFTR modulator therapy has advanced CF life expectancy opening up the need to understand changes in immunity as CF patients age. Here, we have summarized the current understanding of immune dysregulation in CF.
Collapse
Affiliation(s)
- Emanuela M Bruscia
- Department of Pediatrics, Section of Pulmonology, Allergy, Immunology and Sleep Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Tracey L Bonfield
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
15
|
Xu Y, Xu Z, Gu X, Xie Y, He R, Xu J, Jing B, Peng X, Yang G. Immunomodulatory effects of two recombinant arginine kinases in Sarcoptes Scabiei on host peripheral blood mononuclear cells. Front Immunol 2022; 13:1035729. [DOI: 10.3389/fimmu.2022.1035729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022] Open
Abstract
BackgroundAs an important zoonotic parasitic disease with global distribution, scabies causes serious public health and economic problems. Arginine kinase (AK) is involved in cell signal transduction, inflammation, and apoptosis. Two AKs were identified in Sarcoptes scabiei, but their functions in the host immune response remain unclear.MethodsrSsAK-1 and rSsAK-2 were expressed, purified, and immunolocalized. The effects of rSsAK-1 and rSsAK-2 on rabbit PBMC proliferation, apoptosis, and migration; Bcl-2, Bcl-xl, Fas, Bax, and NF-κB transcription levels; and IL-2, IFN-γ, IL-4, IL-10, TGF-β1, and IL-17 secretion were detected.ResultsrSsAK-1 and rSsAK-2 were cloned and expressed successfully. Both enzymes were ~57 kDa and contained 17-kDa tagged proteins, and had good catalytic activity and immunoreactivity. The proteins were located in the S. scabiei exoskeleton, chewing mouthparts, legs, stomach, and intestine. SsAK-1 and SsAK-2 were secreted in the pool and epidermis of the skin lesions, which may be involved in S. scabiei–host interaction. rSsAK-1 and rSsAK-2 significantly promoted cell proliferation, induced cell migration, inhibited apoptosis, and increased Bcl-2, Bcl-xl and NF-κB (p65) transcription levels concentration-dependently, and inhibited IL-2, IFN-γ, and IL-10 secretion and promoted IL-4 and IL-17 secretion.ConclusionrSsAK-1 and rSsAK-2 might increase Bcl-2 and Bcl-xl expression by activating the NF-κB signaling pathway to promote cell proliferation and inhibit apoptosis, which induced PBMC survival. By inducing PBMC migration to the infection site, rSsAK-1 and rSsAK-2 shifted the Th1/Th2 balance toward Th2 and changed the Th17/Treg balance, which indicated their immune role in S. scabiei allergic inflammation.
Collapse
|
16
|
Gonçalves SM, Ferreira AV, Cunha C, Carvalho A. Targeting immunometabolism in host-directed therapies to fungal disease. Clin Exp Immunol 2022; 208:158-166. [PMID: 35641161 PMCID: PMC9188340 DOI: 10.1093/cei/uxab014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/03/2021] [Accepted: 10/29/2021] [Indexed: 01/21/2024] Open
Abstract
Fungal infections affect over a billion people and are responsible for more than 1.5 million deaths each year. Despite progress in diagnostic and therapeutic approaches, the management of severe fungal infections remains a challenge. Recently, the reprogramming of cellular metabolism has emerged as a central mechanism through which the effector functions of immune cells are supported to promote antifungal activity. An improved understanding of the immunometabolic signatures that orchestrate antifungal immunity, together with the dissection of the mechanisms that underlie heterogeneity in individual immune responses, may therefore unveil new targets amenable to adjunctive host-directed therapies. In this review, we highlight recent advances in the metabolic regulation of host-fungus interactions and antifungal immune responses, and outline targetable pathways and mechanisms with promising therapeutic potential.
Collapse
Affiliation(s)
- Samuel M Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Anaísa V Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| |
Collapse
|
17
|
Aryl hydrocarbon receptor-targeted therapy for CD4+ T cell-mediated idiopathic pneumonia syndrome in mice. Blood 2022; 139:3325-3339. [PMID: 35226727 DOI: 10.1182/blood.2021013849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/10/2022] [Indexed: 12/15/2022] Open
Abstract
We previously demonstrated that interferon γ (IFN-γ) derived from donor T cells co-opts the indoleamine 2,3-dioxygenase 1 (IDO1) → aryl hydrocarbon receptor (AHR) axis to suppress idiopathic pneumonia syndrome (IPS). Here we report that the dysregulated expression of AP-1 family genes in Ahr-/- lung epithelial cells exacerbated IPS in allogeneic bone marrow transplantation settings. AHR repressed transcription of Jund by preventing STAT1 from binding to its promoter. As a consequence, decreased interleukin-6 impaired the differentiation of CD4+ T cells toward Th17 cells. IFN-γ- and IDO1-independent induction of Ahr expression indicated that the AHR agonist might be a better therapeutic target for IPS than the IDO1 activator. We developed a novel synthetic AHR agonist (referred to here as PB502) that potently inhibits Jund expression. PB502 was highly effective at inducing AHR activation and ameliorating IPS. Notably, PB502 was by far superior to the endogenous AHR ligand, L-kynurenine, in promoting the differentiation of both mouse and human FoxP3+ regulatory CD4+ T cells. Our results suggest that the IDO1-AHR axis in lung epithelial cells is associated with IPS repression. A specific AHR agonist may exhibit therapeutic activity against inflammatory and autoimmune diseases by promoting regulatory T-cell differentiation.
Collapse
|
18
|
Cabrini G, Rimessi A, Borgatti M, Pinton P, Gambari R. Overview of CF lung pathophysiology. Curr Opin Pharmacol 2022; 64:102214. [PMID: 35453033 DOI: 10.1016/j.coph.2022.102214] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 03/02/2022] [Accepted: 03/16/2022] [Indexed: 12/21/2022]
Abstract
Defects of the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) protein affect the homeostasis of chloride, bicarbonate, sodium, and water in the airway surface liquid, influencing the mucus composition and viscosity, which induces a severe condition of infection and inflammation along the whole life of CF patients. The introduction of CFTR modulators, novel drugs directly intervening to rescue the function of CFTR protein, opens a new era of experimental research. The review summarizes the most recent advancements to understand the characteristics of the infective and inflammatory pathology of CF lungs.
Collapse
Affiliation(s)
- Giulio Cabrini
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Center of Innovative Therapies for Cystic Fibrosis, University of Ferrara, Ferrara, Italy.
| | - Alessandro Rimessi
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy; Center of Innovative Therapies for Cystic Fibrosis, University of Ferrara, Ferrara, Italy
| | - Monica Borgatti
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Center of Innovative Therapies for Cystic Fibrosis, University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy; Center of Innovative Therapies for Cystic Fibrosis, University of Ferrara, Ferrara, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Center of Innovative Therapies for Cystic Fibrosis, University of Ferrara, Ferrara, Italy
| |
Collapse
|
19
|
A High-Risk Profile for Invasive Fungal Infections Is Associated with Altered Nasal Microbiota and Niche Determinants. Infect Immun 2022; 90:e0004822. [PMID: 35311544 DOI: 10.1128/iai.00048-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is becoming increasingly clear that the communities of microorganisms that populate the surfaces exposed to the external environment, termed microbiota, are key players in the regulation of pathogen-host cross talk affecting the onset as well as the outcome of infectious diseases. We have performed a multicenter, prospective, observational study in which nasal and oropharyngeal swabs were collected for microbiota predicting the risk of invasive fungal infections (IFIs) in patients with hematological malignancies. Here, we demonstrate that the nasal and oropharyngeal microbiota are different, although similar characteristics differentiate high-risk from low-risk samples at both sites. Indeed, similar to previously published results on the oropharyngeal microbiota, high-risk samples in the nose were characterized by low diversity, a loss of beneficial bacteria, and an expansion of potentially pathogenic taxa, in the presence of reduced levels of tryptophan (Trp). At variance with oropharyngeal samples, however, low Trp levels were associated with defective host-derived kynurenine production, suggesting reduced tolerance mechanisms at the nasal mucosal surface. This was accompanied by reduced levels of the chemokine interleukin-8 (IL-8), likely associated with a reduced recruitment of neutrophils and impaired fungal clearance. Thus, the nasal and pharyngeal microbiomes of hematological patients provide complementary information that could improve predictive tools for the risk of IFI in hematological patients.
Collapse
|
20
|
Mirzaei R, Sabokroo N, Ahmadyousefi Y, Motamedi H, Karampoor S. Immunometabolism in biofilm infection: lessons from cancer. Mol Med 2022; 28:10. [PMID: 35093033 PMCID: PMC8800364 DOI: 10.1186/s10020-022-00435-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Biofilm is a community of bacteria embedded in an extracellular matrix, which can colonize different human cells and tissues and subvert the host immune reactions by preventing immune detection and polarizing the immune reactions towards an anti-inflammatory state, promoting the persistence of biofilm-embedded bacteria in the host. MAIN BODY OF THE MANUSCRIPT It is now well established that the function of immune cells is ultimately mediated by cellular metabolism. The immune cells are stimulated to regulate their immune functions upon sensing danger signals. Recent studies have determined that immune cells often display distinct metabolic alterations that impair their immune responses when triggered. Such metabolic reprogramming and its physiological implications are well established in cancer situations. In bacterial infections, immuno-metabolic evaluations have primarily focused on macrophages and neutrophils in the planktonic growth mode. CONCLUSION Based on differences in inflammatory reactions of macrophages and neutrophils in planktonic- versus biofilm-associated bacterial infections, studies must also consider the metabolic functions of immune cells against biofilm infections. The profound characterization of the metabolic and immune cell reactions could offer exciting novel targets for antibiofilm therapy.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Niloofar Sabokroo
- Department of Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Motamedi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Averna M, Melotti P, Sorio C. Revisiting the Role of Leukocytes in Cystic Fibrosis. Cells 2021; 10:cells10123380. [PMID: 34943888 PMCID: PMC8699441 DOI: 10.3390/cells10123380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022] Open
Abstract
Cystic fibrosis in characterized by pulmonary bacterial colonization and hyperinflammation. Lymphocytes, monocytes/macrophages, neutrophils, and dendritic cells of patients with CF express functional CFTR and are directly affected by altered CFTR expression/function, impairing their ability to resolve infections and inflammation. However, the mechanism behind and the contribution of leukocytes in the pathogenesis of CF are still poorly characterized. The recent clinical introduction of specific CFTR modulators added an important tool not only for the clinical management of the disease but also to the investigation of the pathophysiological mechanisms related to CFTR dysfunction and dysregulated immunity. These drugs treat the basic defect in cystic fibrosis (CF) by increasing CFTR function with improvement of lung function and quality of life, and may improve clinical outcomes also by correcting the dysregulated immune function that characterizes CF. Measure of CFTR function, protein expression profiling and several omics methods were used to identify molecular changes in freshly isolated leukocytes of CF patients, highlighting two roles of leukocytes in CF: one more generally related to the mechanism(s) causing immune dysregulation in CF and unresolved inflammation, and another more applicative role, which identifies in myeloid cells, an important tool predictive of the therapeutic response of CF patients. In this review we will summarize available data on CFTR expression and function in leukocyte populations and will discuss potential clinical applications based on available data.
Collapse
Affiliation(s)
- Monica Averna
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy;
| | - Paola Melotti
- Cystic Fibrosis Centre, Azienda Ospedaliera Universitaria Integrata Verona, 37126 Verona, Italy;
| | - Claudio Sorio
- Department of Medicine, General Pathology Division, University of Verona, 37134 Verona, Italy
- Correspondence: ; Tel.: +39-045-802-7688
| |
Collapse
|
22
|
Al Shakirchi M, Sorjonen K, Klingspor L, Bergman P, Hjelte L, de Monestrol I. The Effects of Aspergillus fumigatus Colonization on Lung Function in Patients with Cystic Fibrosis. J Fungi (Basel) 2021; 7:jof7110944. [PMID: 34829231 PMCID: PMC8618016 DOI: 10.3390/jof7110944] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 11/28/2022] Open
Abstract
Aspergillus fumigatus is commonly isolated from CF airways. However, the impact on CF lung progression is not completely understood. In this study, using a 16-year retrospective observational cohort study (2000–2015) that included 132 patients, we determined the annual lung function, measured as percent predicted forced expiratory volume in the first second (ppFEV1), decline before and after the first colonization with A. fumigatus. Further, in the same individual, the ratios of lung function when patients were colonized with A. fumigatus and when they were not were calculated. The impact of eradication, with antifungal treatment or spontaneously, was assessed. The annual ppFEV1 was significantly lower after the first colonization with A. fumigatus. Furthermore, within the same individual, colonization with A. fumigatus for two and three years in a row was associated with 4.3% and 7.9% lower ppFEV1, respectively, compared to when not colonized. Finally, patients who eradicated A. fumigatus the following two years after colonization exhibited 9.9% and 14.5% higher ppFEV1 compared to patients who continued to produce cultures with A. fumigatus for two and three years. Our study demonstrated that A. fumigatus colonization was associated with a negative impact on lung function in the long term and eradication, spontaneously or with treatment, was associated with a better pulmonary outcome.
Collapse
Affiliation(s)
- Mahasin Al Shakirchi
- Stockholm Cystic Fibrosis Centre, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden; (L.H.); (I.d.M.)
- Department of Clinical Science, Intervention and Technology, Division of Pediatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
- Correspondence:
| | - Kimmo Sorjonen
- Department of Clinical Neuroscience, Division of Psychology, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Lena Klingspor
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, 171 77 Stockholm, Sweden; (L.K.); (P.B.)
| | - Peter Bergman
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, 171 77 Stockholm, Sweden; (L.K.); (P.B.)
- Department of Infectious Diseases, The Immunodeficiency Unit, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden
| | - Lena Hjelte
- Stockholm Cystic Fibrosis Centre, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden; (L.H.); (I.d.M.)
- Department of Clinical Science, Intervention and Technology, Division of Pediatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Isabelle de Monestrol
- Stockholm Cystic Fibrosis Centre, Karolinska University Hospital Huddinge, 141 86 Stockholm, Sweden; (L.H.); (I.d.M.)
- Department of Clinical Science, Intervention and Technology, Division of Pediatrics, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
23
|
Mishra R, Nawas AF, Mendelson CR. Role of NRF2 in immune modulator expression in developing lung. FASEB J 2021; 35:e21758. [PMID: 34245611 DOI: 10.1096/fj.202100129rr] [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: 01/20/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 11/11/2022]
Abstract
After birth, the alveolar epithelium is exposed to environmental pathogens and high O2 tensions. The alveolar type II cells may protect this epithelium through surfactant production. Surfactant protein, SP-A, an immune modulator, is developmentally upregulated in fetal lung with surfactant phospholipid synthesis. Herein, we observed that the redox-regulated transcription factor, NRF2, and co-regulated C/EBPβ and PPARγ, were markedly induced during cAMP-mediated differentiation of cultured human fetal lung (HFL) epithelial cells. This occurred with enhanced expression of immune modulators, SP-A, TDO2, AhR, and NQO1. Like SP-A, cAMP induction of NRF2 was prevented when cells were exposed to hypoxia. NRF2 knockdown inhibited induction of C/EBPβ, PPARγ, and immune modulators. Binding of endogenous NRF2 to promoters of SP-A and other immune modulator genes increased during HFL cell differentiation. In mouse fetal lung (MFL), a developmental increase in Nrf2, SP-A, Tdo2, Ahr, and Nqo1 and decrease in Keap1 occurred from 14.5 to 18.5 dpc. Developmental induction of Nrf2 in MFL was associated with increased nuclear localization of NF-κB p65, a decline in p38 MAPK phosphorylation, increase in the MAPK phosphatase, DUSP1, induction of the histone acetylase, CBP, and decline in the histone deacetylase, HDAC4. Thus, together with surfactant production, type II cells protect the alveolar epithelium through increased expression of NRF2 and immune modulators to prevent inflammation and oxidative stress. Our findings further suggest that lung cancer cells have usurped this developmental pathway to promote immune tolerance and enhance survival.
Collapse
Affiliation(s)
- Ritu Mishra
- Department of Biochemistry, North Texas March of Dimes Birth Defects Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Afshan Fathima Nawas
- Department of Biochemistry, North Texas March of Dimes Birth Defects Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carole R Mendelson
- Department of Biochemistry, North Texas March of Dimes Birth Defects Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Obstetrics & Gynecology, North Texas March of Dimes Birth Defects Center, The University of Texas Southwestern Med Center, Dallas, TX, USA
| |
Collapse
|
24
|
Hérivaux A, Willis JR, Mercier T, Lagrou K, Gonçalves SM, Gonçales RA, Maertens J, Carvalho A, Gabaldón T, Cunha C. Lung microbiota predict invasive pulmonary aspergillosis and its outcome in immunocompromised patients. Thorax 2021; 77:283-291. [PMID: 34172558 PMCID: PMC8867272 DOI: 10.1136/thoraxjnl-2020-216179] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 05/26/2021] [Indexed: 11/17/2022]
Abstract
Rationale Recent studies have revealed that the lung microbiota of critically ill patients is altered and predicts clinical outcomes. The incidence of invasive fungal infections, namely, invasive pulmonary aspergillosis (IPA), in immunocompromised patients is increasing, but the clinical significance of variations in lung bacterial communities is unknown. Objectives To define the contribution of the lung microbiota to the development and course of IPA. Methods and measurements We performed an observational cohort study to characterise the lung microbiota in 104 immunocompromised patients using bacterial 16S ribosomal RNA gene sequencing on bronchoalveolar lavage samples sampled on clinical suspicion of infection. Associations between lung dysbiosis in IPA and pulmonary immunity were evaluated by quantifying alveolar cytokines and chemokines and immune cells. The contribution of microbial signatures to patient outcome was assessed by estimating overall survival. Main results Patients diagnosed with IPA displayed a decreased alpha diversity, driven by a markedly increased abundance of the Staphylococcus, Escherichia, Paraclostridium and Finegoldia genera and a decreased proportion of the Prevotella and Veillonella genera. The overall composition of the lung microbiome was influenced by the neutrophil counts and associated with differential levels of alveolar cytokines. Importantly, the degree of bacterial diversity at the onset of IPA predicted the survival of infected patients. Conclusions Our results reveal the lung microbiota as an understudied source of clinical variation in patients at risk of IPA and highlight its potential as a diagnostic and therapeutic target in the context of respiratory fungal diseases.
Collapse
Affiliation(s)
- Anaïs Hérivaux
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Jesse R Willis
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Toine Mercier
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Katrien Lagrou
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.,Clinical Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Samuel M Gonçalves
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Relber A Gonçales
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Johan Maertens
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Agostinho Carvalho
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| | - Toni Gabaldón
- Barcelona Supercomputing Centre (BSC-CNS), Barcelona, Spain .,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Cristina Cunha
- Life and Health Sciences Research Institute (ICVS), University of Minho, Braga, Portugal .,ICVS/3B's - PT Government Associate Laboratory, Guimarães/Braga, Portugal
| |
Collapse
|
25
|
Westhölter D, Beckert H, Straßburg S, Welsner M, Sutharsan S, Taube C, Reuter S. Pseudomonas aeruginosa infection, but not mono or dual-combination CFTR modulator therapy affects circulating regulatory T cells in an adult population with cystic fibrosis. J Cyst Fibros 2021; 20:1072-1079. [PMID: 34030985 DOI: 10.1016/j.jcf.2021.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/20/2021] [Accepted: 05/01/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Chronic infection and an exaggerated inflammatory response are key drivers of the pathogenesis of cystic fibrosis (CF), especially CF lung disease. An imbalance of pro- and anti-inflammatory mediators, including dysregulated Th2/Th17 cells and impairment of regulatory T cells (Tregs), maintain CF inflammation. CF transmembrane conductance regulator (CFTR) modulator therapy might influence these immune cell abnormalities. METHODS Peripheral blood mononuclear cells and serum samples were collected from 108 patients with CF (PWCF) and 40 patients with non-CF bronchiectasis. Samples were analysed for peripheral blood lymphocytes subsets (Tregs; Th1-, Th1/17-, Th17- and Th2-effector cells) and systemic T helper cell-associated cytokines (interleukin [IL]-5, IL-13, IL-2, IL-6, IL-9, IL-10, IL-17A, IL-17F, IL-4, IL-22, interferon-γ, tumour necrosis factor-α) using flow cytometry. RESULTS 51% of PWCF received CFTR modulators (ivacaftor, ivacaftor/ lumacaftor or tezacaftor/ ivacaftor). There were no differences in proportions of analysed T cell subsets or cytokines between PWCF who were versus were not receiving CFTR modulators. Additional analysis revealed lower percentages of Tregs in PWCF and chronic pulmonary Pseudomonas aeruginosa infection; this difference was also present in PWCF treated with CFTR modulators. Patients with non-CF bronchiectasis tended to have higher percentages of Th2- and Th17-cells and higher levels of peripheral cytokines versus PWCF. CONCLUSIONS Chronic P. aeruginosa lung infection appears to impair Tregs in PWCF (independent of CFTR modulator therapy) but not those with non-CF bronchiectasis. Moreover, our data showed no statistically significant differences in major subsets of peripheral lymphocytes and cytokines among PWCF who were versus were not receiving CFTR modulators.
Collapse
Affiliation(s)
- Dirk Westhölter
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany.
| | - Hendrik Beckert
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Svenja Straßburg
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany; Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Essen, Germany
| | - Matthias Welsner
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany; Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Essen, Germany
| | - Sivagurunathan Sutharsan
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany; Adult Cystic Fibrosis Center, Department of Pulmonary Medicine, University Hospital Essen - Ruhrlandklinik, Essen, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen- Ruhrlandklinik, Essen, Germany
| |
Collapse
|
26
|
CFTR regulates embryonic T lymphopoiesis via Wnt signaling in zebrafish. Immunol Lett 2021; 234:47-53. [PMID: 33951474 DOI: 10.1016/j.imlet.2021.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/06/2021] [Accepted: 04/25/2021] [Indexed: 11/23/2022]
Abstract
The number and function of T cells are abnormal as observed in cystic fibrosis (CF) patients and CF mouse models, and our previous work shows that the CFTR mutant leads to deficiency of primitive and definitive hematopoietic in zebrafish. However, the functions and underlying mechanisms of CFTR in T cell development during early embryogenesis have not been explored. Here, we report that the genetic ablation of CFTR in zebrafish resulted in abrogated embryonic T lymphopoiesis, which was ascribed to impaired thymic homing and expansion of hematopoietic stem cells (HSCs). Transcriptome analysis of isolated HSCs in zebrafish embryos at 48 hpf showed a significant alteration of key factors essential for T cell development and Wnt signaling, consistent with our previous work on CFTR regulating hematopoiesis. In brief, we uncovered the function of CFTR in embryonic T cell development and suggest that the immune deficiency of CF patients may originate from an early embryonic stage.
Collapse
|
27
|
Small Molecule CCR4 Antagonists Protect Mice from Aspergillus Infection and Allergy. Biomolecules 2021; 11:biom11030351. [PMID: 33669094 PMCID: PMC7996545 DOI: 10.3390/biom11030351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/18/2021] [Accepted: 02/23/2021] [Indexed: 12/29/2022] Open
Abstract
The ability to regulate the recruitment of immune cells makes chemokines and their receptors attractive drug targets in many inflammatory diseases. Based on its preferential expression on T helper type 2 (Th2) cells, C-C chemokine receptor type 4 (CCR4) has been widely studied in the context of allergic diseases, but recent evidence on the expression of CCR4 in other cell types has considerably expanded the potential applications of CCR4 antagonism. However, the current number of approved indications, as well as the portfolio of CCR4-targeting drugs, are still limited. In the present study, we have assessed the potential therapeutic efficacy of a CCR4 small molecule antagonist, SP50, discovered via an in silico-based approach, against a variety of pre-clinical settings of infection with the fungus Aspergillus fumigatus. We show that SP50 efficiently worked as prophylactic vaccine adjuvant in immunocompetent mice, protected against invasive aspergillosis in immunosuppressed mice. Further, the CCR4 antagonist prevented allergic bronchopulmonary aspergillosis in susceptible mice, and in a murine model of cystic fibrosis, a genetic disorder characterized by chronic pulmonary inflammation and recurrent infections. In conclusion, our results extend the potential applications of CCR4 antagonism and prompt for the development of novel compounds with the potential to progress to clinical trials.
Collapse
|
28
|
Correlation of Indoleamine-2,3-Dioxygenase and Chronic Kidney Disease: A Pilot Study. J Immunol Res 2021; 2021:8132569. [PMID: 33506062 PMCID: PMC7806403 DOI: 10.1155/2021/8132569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 11/28/2020] [Accepted: 12/19/2020] [Indexed: 12/13/2022] Open
Abstract
Objective To explore the correlation of indoleamine-2,3-dioxygenase (IDO) and chronic kidney disease (CKD). Methods A total of 154 CKD patients and 42 non-CKD patients were recruited. Patients were grouped into ACR1~ACR3 (<30 mg/g, 30-300 mg/g, and >300 mg/g). Biomarkers in different groups were compared by ANOVA. Correlation was calculated by Pearson or Spearman analysis and binary logistic regression. The ROC curve was also performed. Results The levels of albumin, serum creatinine (sCr), and IDO in non-CKD patients were significantly different from those in CKD3-CKD5 stages (p < 0.05). IDO was correlated with age, proteinuria, ACR, and eGFR (p < 0.01). After adjusting for CKD-related indices, ln(IDO) was an independent risk factor for CKD (3.48, p < 0.05). The analysis of ROC curve revealed a best cutoff for IDO was 0.0466 and yielded a sensitivity of 83.8% and a specificity of 75%. Hemoglobin, total protein, and albumin in the ACR1 group were significantly higher than those in the ACR2 and ACR3 groups (p < 0.01), while sCr and IDO levels were significantly lower than those in the ACR2 and ACR3 groups (p < 0.01 or p < 0.05). After adjusting for CKD-related indices, ln(IDO) was still an independent risk factor for ACR (OR = 2.7, p < 0.05). The analysis of ROC curve revealed a best cutoff for IDO was 0.075 and yielded a sensitivity of 71.9% and a specificity of 72.2%. Conclusion IDO may be a promising biomarker to predict CKD and assess kidney function.
Collapse
|
29
|
Abstract
Cystic fibrosis (CF) is a hereditary, multisystemic disease caused by different mutations in the CFTR gene encoding CF transmembrane conductance regulator. CF is mainly characterized by pulmonary dysfunction as a result of deterioration in the mucociliary clearance and anion transport of airways. Mortality is mostly caused by bronchiectasis, bronchiole obstruction, and progressive respiratory dysfunction in the early years of life. Over the last decade, new therapeutic strategies rather than symptomatic treatment have been proposed, such as the small molecule approach, ion channel therapy, and pulmonary gene therapy. Due to considerable progress in the treatment options, CF has become an adult disease rather than a pediatric disease in recent years. Pulmonary gene therapy has gained special attention due to its mutation type independent aspect, therefore being applicable to all CF patients. On the other hand, the major obstacle for CF treatment is to predict the drug response of patients due to genetic complexity and heterogeneity. The advancement of 3D culture systems has made it possible to extrapolate the disease modeling and individual drug response in vitro by producing mini adult organs called "organoids" obtained from rectal cell biopsies. In this review, we summarize the advances in the novel therapeutic approaches, clinical interventions, and precision medicine concept for CF.
Collapse
|
30
|
Keown K, Reid A, Moore JE, Taggart CC, Downey DG. Coinfection with Pseudomonas aeruginosa and Aspergillus fumigatus in cystic fibrosis. Eur Respir Rev 2020; 29:29/158/200011. [PMID: 33208485 DOI: 10.1183/16000617.0011-2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/16/2020] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Cystic fibrosis (CF) lung disease is characterised by mucus stasis, chronic infection and inflammation, causing progressive structural lung disease and eventual respiratory failure. CF airways are inhabited by an ecologically diverse polymicrobial environment with vast potential for interspecies interactions, which may be a contributing factor to disease progression. Pseudomonas aeruginosa and Aspergillus fumigatus are the most common bacterial and fungal species present in CF airways respectively and coinfection results in a worse disease phenotype. METHODS In this review we examine existing expert knowledge of chronic co-infection with P. aeruginosa and A. fumigatus in CF patients. We summarise the mechanisms of interaction and evaluate the clinical and inflammatory impacts of this co-infection. RESULTS P. aeruginosa inhibits A. fumigatus through multiple mechanisms: phenazine secretion, iron competition, quorum sensing and through diffusible small molecules. A. fumigatus reciprocates inhibition through gliotoxin release and phenotypic adaptations enabling evasion of P. aeruginosa inhibition. Volatile organic compounds secreted by P. aeruginosa stimulate A. fumigatus growth, while A. fumigatus stimulates P. aeruginosa production of cytotoxic elastase. CONCLUSION A complex bi-directional relationship exists between P. aeruginosa and A. fumigatus, exhibiting both mutually antagonistic and cooperative facets. Cross-sectional data indicate a worsened disease state in coinfected patients; however, robust longitudinal studies are required to derive causality and to determine whether interspecies interaction contributes to disease progression.
Collapse
Affiliation(s)
- Karen Keown
- Royal Belfast Hospital for Sick Children, Belfast Health and Social Care Trust, Belfast, UK.,Wellcome Wolfson Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Alastair Reid
- Royal Belfast Hospital for Sick Children, Belfast Health and Social Care Trust, Belfast, UK
| | - John E Moore
- Northern Ireland Public Health Laboratory, Dept of Bacteriology, Belfast City Hospital, Belfast, UK
| | - Clifford C Taggart
- Wellcome Wolfson Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Damian G Downey
- Wellcome Wolfson Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| |
Collapse
|
31
|
Bellet MM, Borghi M, Pariano M, Renga G, Stincardini C, D'Onofrio F, Brancorsini S, Garaci E, Costantini C, Romani L. Thymosin alpha 1 exerts beneficial extrapulmonary effects in cystic fibrosis. Eur J Med Chem 2020; 209:112921. [PMID: 33071052 DOI: 10.1016/j.ejmech.2020.112921] [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: 03/25/2020] [Revised: 08/12/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the gene encoding for the ion channel Cystic Fibrosis Transmembrane conductance Regulator (CFTR). Long considered a lung disease for the devastating impact on the respiratory function, the recent diagnostic and therapeutic advances have shed the light on the extra-pulmonary manifestations of CF, including gastrointestinal, hepatobiliary and pancreatic symptoms. We have previously demonstrated that thymosin alpha1 (Tα1), a naturally occurring immunomodulatory peptide, displays multi-sided beneficial effects in CF that concur in ameliorating the lung inflammatory pathology. In the present study, by resorting to murine models of gut inflammation with clinical relevance for CF patients, we demonstrate that Tα1 can also have beneficial effects in extrapulmonary pathology. Specifically, Tα1 restored barrier integrity and immune homeostasis in the inflamed gut of CF mice as well as in mice with the metabolic syndrome, a disorder that may arise in CF patients with high caloric intake despite pancreatic sufficiency. The protective effects of Tα1 also extended to pancreas and liver, further emphasizing the beneficial effects of Tα1 in extra-pulmonary complications of CF. By performing wide-ranging multi-organ anti-inflammatory effects, Tα1 could potentially integrate current therapeutic approaches to tackle the complex symptomatology of CF disease.
Collapse
Affiliation(s)
- Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.
| | - Monica Borghi
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Claudia Stincardini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Fiorella D'Onofrio
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Stefano Brancorsini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, 00166, Rome, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.
| |
Collapse
|
32
|
Moss RB. Mucosal humoral immunity in cystic fibrosis - a tangled web of failed proteostasis, infection and adaptive immunity. EBioMedicine 2020; 60:103035. [PMID: 32971469 PMCID: PMC7516060 DOI: 10.1016/j.ebiom.2020.103035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Richard B Moss
- Center of Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University School of Medicine, 770 Welch Rd, Suite 350, Palo Alto, CA 94304, United States.
| |
Collapse
|
33
|
Costantini C, Puccetti M, Pariano M, Renga G, Stincardini C, D'Onofrio F, Bellet MM, Cellini B, Giovagnoli S, Romani L. Selectively targeting key inflammatory pathways in cystic fibrosis. Eur J Med Chem 2020; 206:112717. [PMID: 32823008 DOI: 10.1016/j.ejmech.2020.112717] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/10/2020] [Accepted: 07/21/2020] [Indexed: 01/04/2023]
Abstract
Cystic fibrosis (CF) is a rare genetic disorder caused by a defect in the ion channel Cystic Fibrosis Transmembrane conductance Regulator (CFTR), resulting in ionic imbalance of surface fluid. Although affecting multiple organs, the progressive deterioration of respiratory function by recurrent infections and chronic inflammation represents the main cause of morbidity and mortality in CF patients. The development of modulators targeting the basic defect of CFTR has represented a major breakthrough in CF therapy, but the impact on inflammation has remained enigmatic. The emerging scenario taking hold in the field points to inflammation as a major, somehow missed, therapeutic target for prevention of lung decline. Not surprisingly, the development of anti-inflammatory drugs is taking its share in the drug development pipeline. But the path is not straightforward and targeting inflammation should be balanced with the increased risk of infection. The strategy to restore the homeostatic regulation of inflammation to efficiently respond to infection while preventing lung damage needs to be based on identifying and targeting endogenous immunoregulatory pathways that are defective in CF. We herein provide an overview of anti-inflammatory drugs currently approved or under investigation in CF patients, and present our recent studies on how the knowledge on defective immune pathways in CF may translate into innovative and selective anti-inflammatory therapeutics. Through the discovery of naturally occurring molecules or their synthetic mimics, this review emphasizes the critical importance of selectively targeting key inflammatory pathways to preserve immunocompetence in CF patients.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Matteo Puccetti
- Department of Pharmaceutical Science, University of Perugia, Perugia, 06132, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Claudia Stincardini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Fiorella D'Onofrio
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Barbara Cellini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Stefano Giovagnoli
- Department of Pharmaceutical Science, University of Perugia, Perugia, 06132, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.
| |
Collapse
|
34
|
Ayroldi E, Grohmann U. Exemplifying complexity of immune suppression by a "canonical" speech: A glimpse into TNFRSF-activated signaling pathways in Treg cells. Eur J Immunol 2020; 50:944-948. [PMID: 32428262 DOI: 10.1002/eji.202048711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Indexed: 01/03/2023]
Abstract
Regulatory T (Treg) cells are crucial mediators of immune tolerance suppressing self-reactive T cells and preventing autoimmune diseases. Besides activation of the T cell receptor (TCR), empowerment of Treg cell functions requires co-accessory signals, such as those released by the TNF receptor superfamily (TNFRSF) that, however, can also promote immunostimulatory responses when engaged by effector T cells. In this issue of European Journal of Immunology, Lubrano di Ricco et al. [Eur. J. Immunol. 2020. 50: 972-985] have taken a closer look at the important question of the functional meaning of TNFRSF-activated signaling pathways in Treg cells. They have demonstrated that costimulation by TNFR2, 4-1BB, GITR, DR3, but not OX40 in mouse Foxp3+ Treg cells activates the same and unique signaling pathway, i.e., canonical NF-κB, which in turn leads to Foxp3 gene upregulation, cell expansion in vitro and in vivo, and suppressive activity in an experimental model of colitis. Moreover, induction of markers of T helper 2 (Th2) and Th17 as well as of genes encoding proteins involved in noncanonical NF-κΒ was also observed. We here discussed how these findings further highlight the emerging concept of Treg cell plasticity in immune tolerance.
Collapse
Affiliation(s)
- Emira Ayroldi
- Department of Medicine, University of Perugia, Perugia, Italy
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
35
|
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
|
36
|
Renga G, Oikonomou V, Moretti S, Stincardini C, Bellet MM, Pariano M, Bartoli A, Brancorsini S, Mosci P, Finocchi A, Rossi P, Costantini C, Garaci E, Goldstein AL, Romani L. Thymosin β4 promotes autophagy and repair via HIF-1α stabilization in chronic granulomatous disease. Life Sci Alliance 2019; 2:2/6/e201900432. [PMID: 31719116 PMCID: PMC6851533 DOI: 10.26508/lsa.201900432] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 02/07/2023] Open
Abstract
This study demonstrates that thymosin β4 stabilizes HIF-1a to promote autophagy and up-regulate genes involved in tissue and mucosal barrier protection in chronic granulomatous disease. Chronic granulomatous disease (CGD) is a genetic disorder of the NADPH oxidase characterized by increased susceptibility to infections and hyperinflammation associated with defective autophagy and increased inflammasome activation. Herein, we demonstrate that thymosin β4 (Tβ4), a g-actin sequestering peptide with multiple and diverse intracellular and extracellular activities affecting inflammation, wound healing, fibrosis, and tissue regeneration, promoted in human and murine cells noncanonical autophagy, a form of autophagy associated with phagocytosis and limited inflammation via the death-associated protein kinase 1. We further show that the hypoxia inducible factor-1 (HIF-1)α was underexpressed in CGD but normalized by Tβ4 to promote autophagy and up-regulate genes involved in mucosal barrier protection. Accordingly, inflammation and granuloma formation were impaired and survival increased in CGD mice with colitis or aspergillosis upon Tβ4 treatment or HIF-1α stabilization. Thus, the promotion of endogenous pathways of inflammation resolution through HIF-1α stabilization is druggable in CGD by Tβ4.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Silvia Moretti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Andrea Bartoli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Paolo Mosci
- Internal Medicine, Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Andrea Finocchi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Paolo Rossi
- Department of Pediatrics, Unit of Immune and Infectious Diseases, Children's Hospital Bambino Gesù, Rome, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Rome, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, the George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
37
|
Napolioni V, Pariano M, Borghi M, Oikonomou V, Galosi C, De Luca A, Stincardini C, Vacca C, Renga G, Lucidi V, Colombo C, Fiscarelli E, Lass-Flörl C, Carotti A, D'Amico L, Majo F, Russo MC, Ellemunter H, Spolzino A, Mosci P, Brancorsini S, Aversa F, Velardi A, Romani L, Costantini C. Genetic Polymorphisms Affecting IDO1 or IDO2 Activity Differently Associate With Aspergillosis in Humans. Front Immunol 2019; 10:890. [PMID: 31134053 PMCID: PMC6514051 DOI: 10.3389/fimmu.2019.00890] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/08/2019] [Indexed: 12/29/2022] Open
Abstract
Aspergillus is the causative agent of human diseases ranging from asthma to invasive infection. Genetic and environmental factors are crucial in regulating the interaction between the host and Aspergillus. The role played by the enzyme indoleamine 2,3-dioxygenase 1 (IDO1), which catalyzes the first and rate-limiting step of tryptophan catabolism along the kynurenine pathway, is increasingly being recognized, but whether and how genetic variation of IDO1 influences the risk of aspergillosis in susceptible patients is incompletely understood. In addition, whether the closely related protein IDO2 plays a similar role remains unexplored. In the present study, we performed genetic association studies in two different cohorts of susceptible patients [cystic fibrosis (CF) patients and recipients of hematopoietic stem cell transplantation (HSCT)], and identified IDO1 polymorphisms that associate with the risk of infection in both cohorts. By using human bronchial epithelial cells and PBMC from CF and HSCT patients, respectively, we could show that the IDO1 polymorphisms appeared to down-modulate IDO1 expression and function in response to IFNγ or Aspergillus conidia, and to associate with an increased inflammatory response. In contrast, IDO2 polymorphisms, including variants known to profoundly affect protein expression and function, were differently associated with the risk of aspergillosis in the two cohorts of patients as no association was found in CF patients as opposed to recipients of HSCT. By resorting to a murine model of bone marrow transplantation, we could show that the absence of IDO2 more severely affected fungal burden and lung pathology upon infection with Aspergillus as compared to IDO1, and this effect appeared to be linked to a deficit in the antifungal effector phagocytic activity. Thus, our study confirms and extends the role of IDO1 in the response to Aspergillus, and shed light on the possible involvement of IDO2 in specific clinical settings.
Collapse
Affiliation(s)
- Valerio Napolioni
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University Stanford, CA, United States
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Monica Borghi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudia Galosi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Antonella De Luca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Carmine Vacca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vincenzina Lucidi
- Unit of Endocrinology and Diabetes, Bambino Gesù Children's Hospital, Rome, Italy
| | - Carla Colombo
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Cornelia Lass-Flörl
- Division of Hygiene and Medical Microbiology, Innsbruck Medical University, Innsbruck, Austria
| | - Alessandra Carotti
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche, University of Perugia, Perugia, Italy
| | - Lucia D'Amico
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche, University of Perugia, Perugia, Italy
| | - Fabio Majo
- Unit of Endocrinology and Diabetes, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Chiara Russo
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Angelica Spolzino
- Division of Hematology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Paolo Mosci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | | | - Franco Aversa
- Division of Hematology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Andrea Velardi
- Institute of Hematology-Centro di Ricerche Emato-Oncologiche, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
38
|
Warris A. Immunopathology of Aspergillus Infections in Children With Chronic Granulomatous Disease and Cystic Fibrosis. Pediatr Infect Dis J 2019; 38:e96-e98. [PMID: 30531311 PMCID: PMC6499723 DOI: 10.1097/inf.0000000000002265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Adilia Warris
- From the MRC Centre for Medical Mycology, Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, United Kingdom
| |
Collapse
|
39
|
Wong AYW, Fric J, Zelante T. Learning to control tissue damage while fighting Aspergillus. Med Mycol 2019; 57:S189-S195. [PMID: 30816972 DOI: 10.1093/mmy/myy053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022] Open
Abstract
Aspergillus moulds are increasingly being recognised as significant human pathogens that can cause life-threatening infections in the context of host immune dysregulation, particularly in the lung. It is now clear that there is a close relationship between infection susceptibility and the fine regulation of pulmonary immunity and inflammation. While the contribution of IL-17/Th17 responses to both physiological and pathological lung inflammation is now well established, the cellular interactions, soluble factors, and signalling pathways that determine Th17 cell responses to fungal infection remain unclear. Here, we identify potential key mediators of fungus-DC-T cell interactions in the respiratory tract, with a focus on the DC-derived cytokines thought to exert a major influence on generation of pathological Th17 cells. We review recent data indicating a crucial role for Aspergillus-induced autophagy in lung DCs on subsequent T-cell polarization and modulation of 'stemness', which appears critical for avoiding pathological lung inflammation and promoting disease resolution.
Collapse
Affiliation(s)
- Alicia Yoke Wei Wong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jan Fric
- Center for Translational Medicine, International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Teresa Zelante
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
40
|
Warris A, Bercusson A, Armstrong-James D. Aspergillus colonization and antifungal immunity in cystic fibrosis patients. Med Mycol 2019; 57:S118-S126. [PMID: 30816976 DOI: 10.1093/mmy/myy074] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/20/2018] [Accepted: 07/26/2018] [Indexed: 12/11/2022] Open
Abstract
Cystic fibrosis (CF), caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, is the most common inherited life-limiting disease in North European people affecting 90,000 people worldwide. Progressive lung damage caused by recurrent infection and chronic airway inflammation is the major determinant of survival with a median age at death of 29 years. Approximately 60% of CF patients are infected with Aspergillus fumigatus, a ubiquitous environmental fungus, and its presence has been associated with accelerated lung function decline. Half of the patients infected with Aspergillus are <18 years of age. Yet time of acquisition of this fungus and determinants of CF-related Aspergillus disease severity and progression are not known. CFTR expression has been demonstrated in cells of the innate and adaptive immune system and has shown to be critical for normal function. Research delineating the role of CFTR-deficient phagocytes in Aspergillus persistence and infection in the CF lung, has only recently received attention. In this concise review we aim to present the current understanding with respect to when people with CF acquire infection with A. fumigatus and antifungal immune responses by CF immune cells.
Collapse
Affiliation(s)
- Adilia Warris
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, University of Aberdeen, United Kingdom
| | - Amelia Bercusson
- National Heart and Lung Institute, Imperial College London, United Kingdom
| | | |
Collapse
|
41
|
Roesch EA, Nichols DP, Chmiel JF. Inflammation in cystic fibrosis: An update. Pediatr Pulmonol 2018; 53:S30-S50. [PMID: 29999593 DOI: 10.1002/ppul.24129] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
Inflammation plays a critical role in cystic fibrosis (CF) lung pathology and disease progression making it an active area of research and important therapeutic target. In this review, we explore the most recent research on the major contributors to the exuberant inflammatory response seen in CF as well as potential therapeutics to combat this response. Absence of functional cystic fibrosis transmembrane conductance regulator (CFTR) alters anion transport across CF airway epithelial cells and ultimately results in dehydration of the airway surface liquid. The dehydrated airway surface liquid in combination with abnormal mucin secretion contributes to airway obstruction and subsequent infection that may serve as a trigger point for inflammation. There is also evidence to suggest that airway inflammation may be excessive and sustained relative to the infectious stimuli. Studies have shown dysregulation of both pro-inflammatory mediators such as IL-17 and pro-resolution mediators including metabolites of the eicosanoid pathway. Recently, CFTR potentiators and correctors have garnered much attention in the CF community. Although these modulators address the underlying defect in CF, their impact on downstream consequences such as inflammation are not known. Here, we review pre-clinical and clinical data on the impact of CFTR modulators on inflammation. In addition, we examine other cell types including neutrophils, macrophages, and T-lymphocytes that express CFTR and contribute to the CF inflammatory response. Finally, we address challenges in developing anti-inflammatory therapies and highlight some of the most promising anti-inflammatory drugs under development for CF.
Collapse
Affiliation(s)
- Erica A Roesch
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| | - David P Nichols
- Department of Pediatrics, Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - James F Chmiel
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| |
Collapse
|
42
|
Yang SL, Niu TT, Li XL, Li DJ, Li MQ, Wang HY. Bu-Shen-Yi-Qi formula impairs cytotoxicity of NK cells by up-regulating IDO expression in trophoblasts. Gynecol Endocrinol 2018; 34:675-679. [PMID: 29334801 DOI: 10.1080/09513590.2018.1425988] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Recurrent spontaneous abortion (RSA) is a common health problem that affects about 5% of fertile women, when it occurs for unknown reasons, it is called unexplained recurrent spontaneous abortion (URSA). Traditional Chinese medicine, such as Bu-Shen-Yi-Qi formula which consists of Dangshen, Tusizi, Baizhu, Baishuo, Duzhong, Sangjisheng, Sugeng, and Tiaohuangqin, has played an invaluable role in the treatment of RSA since ancient times. However, the mechanism of how it takes effect is still not clear. To identify Bu-Shen-Yi-Qi formula could modulate immune condition at maternal-fetal interface via its effect on trophoblasts, HTR-8 of different treatment were co-cultured with peripheral or decidual natural killer (NK) cells, and the receptors such as NKP30 and NKP46 expression on NK cells were measured by flow cytometry (FCM). In this study, we found that herb medium could increase the IDO expression at appropriate concentrations. As an inhibitor of IDO, 1-MT could impair the inhibitory function of trophoblasts on NK cells. Furthermore, Bu-Shen-Yi-Qi formula could enhance the inhibitory function of trophoblasts on NK cells. In conclusion, Bu-Shen-Yi-Qi formula can inhibit NK cytotoxicity by up-regulating IDO expression in trophoblasts and play a role in the treatment of URSA patients.
Collapse
Affiliation(s)
- Shao-Liang Yang
- a Department of Gynecology of Integrated Traditional Chinese and Western Medicine , Hospital of Obstetrics and Gynecology, Fudan University , Shanghai , People's Republic of China
| | - Tian-Tian Niu
- a Department of Gynecology of Integrated Traditional Chinese and Western Medicine , Hospital of Obstetrics and Gynecology, Fudan University , Shanghai , People's Republic of China
| | - Xue-Lian Li
- a Department of Gynecology of Integrated Traditional Chinese and Western Medicine , Hospital of Obstetrics and Gynecology, Fudan University , Shanghai , People's Republic of China
| | - Da-Jin Li
- b Laboratory for Reproductive Immunology , Hospital of Obstetrics and Gynecology, Fudan University , Shanghai , People's Republic of China
- c Key Laboratory of Reproduction Regulation of NPFPC , SIPPR, IRD, Fudan University , Shanghai , People's Republic of China
| | - Ming-Qing Li
- b Laboratory for Reproductive Immunology , Hospital of Obstetrics and Gynecology, Fudan University , Shanghai , People's Republic of China
- c Key Laboratory of Reproduction Regulation of NPFPC , SIPPR, IRD, Fudan University , Shanghai , People's Republic of China
- d Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , People's Republic of China
| | - Hai-Yan Wang
- a Department of Gynecology of Integrated Traditional Chinese and Western Medicine , Hospital of Obstetrics and Gynecology, Fudan University , Shanghai , People's Republic of China
- d Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases , Shanghai , People's Republic of China
| |
Collapse
|
43
|
Garaci E. From thymus to cystic fibrosis: the amazing life of thymosin alpha 1. Expert Opin Biol Ther 2018; 18:9-11. [PMID: 30063868 DOI: 10.1080/14712598.2018.1484447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Enrico Garaci
- a University San Raffaele and IRCCS San Raffaele , Rome , Italy
| |
Collapse
|
44
|
Stincardini C, Renga G, Villella V, Pariano M, Oikonomou V, Borghi M, Bellet MM, Sforna L, Costantini C, Goldstein AL, Garaci E, Romani L. Cellular proteostasis: a new twist in the action of thymosin α1. Expert Opin Biol Ther 2018; 18:43-48. [DOI: 10.1080/14712598.2018.1484103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Valeria Villella
- European Institute for Research in Cystic Fibrosis, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Vasilis Oikonomou
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Monica Borghi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marina M. Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Luigi Sforna
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Allan L. Goldstein
- Department of Biochemistry and Molecular Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC, USA
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, Rome, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
45
|
|
46
|
Choera T, Zelante T, Romani L, Keller NP. A Multifaceted Role of Tryptophan Metabolism and Indoleamine 2,3-Dioxygenase Activity in Aspergillus fumigatus-Host Interactions. Front Immunol 2018; 8:1996. [PMID: 29403477 PMCID: PMC5786828 DOI: 10.3389/fimmu.2017.01996] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022] Open
Abstract
Aspergillus fumigatus is the most prevalent filamentous fungal pathogen of humans, causing either severe allergic bronchopulmonary aspergillosis or often fatal invasive pulmonary aspergillosis (IPA) in individuals with hyper- or hypo-immune deficiencies, respectively. Disease is primarily initiated upon the inhalation of the ubiquitous airborne conidia—the initial inoculum produced by A. fumigatus—which are complete developmental units with an ability to exploit diverse environments, ranging from agricultural composts to animal lungs. Upon infection, conidia initially rely on their own metabolic processes for survival in the host’s lungs, a nutritionally limiting environment. One such nutritional limitation is the availability of aromatic amino acids (AAAs) as animals lack the enzymes to synthesize tryptophan (Trp) and phenylalanine and only produce tyrosine from dietary phenylalanine. However, A. fumigatus produces all three AAAs through the shikimate–chorismate pathway, where they play a critical role in fungal growth and development and in yielding many downstream metabolites. The downstream metabolites of Trp in A. fumigatus include the immunomodulatory kynurenine derived from indoleamine 2,3-dioxygenase (IDO) and toxins such as fumiquinazolines, gliotoxin, and fumitremorgins. Host IDO activity and/or host/microbe-derived kynurenines are increasingly correlated with many Aspergillus diseases including IPA and infections of chronic granulomatous disease patients. In this review, we will describe the potential metabolic cross talk between the host and the pathogen, specifically focusing on Trp metabolism, the implications for therapeutics, and the recent studies on the coevolution of host and microbe IDO activation in regulating inflammation, while controlling infection.
Collapse
Affiliation(s)
- Tsokyi Choera
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| | - Teresa Zelante
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Nancy P Keller
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States.,Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
47
|
Bodas M, Mazur S, Min T, Vij N. Inhibition of histone-deacetylase activity rescues inflammatory cystic fibrosis lung disease by modulating innate and adaptive immune responses. Respir Res 2018; 19:2. [PMID: 29301535 PMCID: PMC5755330 DOI: 10.1186/s12931-017-0705-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/15/2017] [Indexed: 12/25/2022] Open
Abstract
Background Chronic lung disease resulting from dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) and NFκB-mediated neutrophilic-inflammation forms the basis of CF-related mortality. Here we aimed to evaluate if HDAC inhibition controls Pseudomonas-aeruginosa-lipopolysaccharide (Pa-LPS) induced airway inflammation and CF-lung disease. Methods For in vitro experiments, HEK293-cells were transfected with IL-8 or NFκB-firefly luciferase, and SV40-renilla- luciferase reporter constructs or ΔF508-CFTR-pCEP, followed by treatment with suberoylanilide hydroxamic acid (SAHA), Trichostatin-A (TSA) and/or TNFα. For murine studies, Cftr+/+ or Cftr−/− mice (n = 3) were injected/instilled with Pa-LPS and/or treated with SAHA or vehicle control. The progression of lung disease was monitored by quantifying changes in inflammatory markers (NFκB), cytokines (IL-6/IL-10), neutrophil activity (MPO, myeloperoxidase and/or NIMP-R14) and T-reg numbers. Results SAHA treatment significantly (p < 0.05) suppresses TNFα-induced NFκB and IL-8 reporter activities in HEK293-cells. Moreover, SAHA, Tubacin (selective HDAC6-inhibitor) or HDAC6-shRNAs controls CSE-induced ER-stress activities (p < 0.05). In addition, SAHA restores trafficking of misfolded-ΔF508-CFTR, by inducing protein levels of both B and C forms of CFTR. Murine studies using Cftr+/+ or Cftr−/− mice verified that SAHA controls Pa-LPS induced IL-6 levels, and neutrophil (MPO levels and/or NIMP-R14), NFκB-(inflammation) and Nrf2 (oxidative-stress marker) activities, while promoting FoxP3+ T-reg activity. Conclusion In summary, SAHA-mediated HDAC inhibition modulates innate and adaptive immune responses involved in pathogenesis and progression of inflammatory CF-lung disease. Electronic supplementary material The online version of this article (10.1186/s12931-017-0705-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manish Bodas
- College of Medicine, Central Michigan University, 2630 Denison Drive, Room# 120 (Office) & 126-127 (Lab), Mt Pleasant, MI, USA.,Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven Mazur
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,National Institute of Allergy and Infectious Diseases, National Institutes of Health, Integrated Research Facility at Fort Detrick, Fort Detrick, Frederick, MD, USA
| | - Taehong Min
- Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Genentech, 1 DNA Way, San Francisco, CA, USA
| | - Neeraj Vij
- College of Medicine, Central Michigan University, 2630 Denison Drive, Room# 120 (Office) & 126-127 (Lab), Mt Pleasant, MI, USA. .,Department of Pediatrics and Pulmonary Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,VIJ Biotech LLC, Baltimore, Maryland, USA.
| |
Collapse
|
48
|
Abstract
Inflammasomes are large innate cytoplasmic complexes that play a major role in promoting inflammation in the lung in response to a range of environmental and infectious stimuli. Inflammasomes are critical for driving acute innate immune responses that resolve infection and maintain tissue homeostasis. However, dysregulated or excessive inflammasome activation can be detrimental. Here, we discuss the plethora of recent data from clinical studies and small animal disease models that implicate excessive inflammasome responses in the pathogenesis of a number of acute and chronic respiratory inflammatory diseases. Understanding of the role of inflammasomes in lung disease is of great therapeutic interest.
Collapse
Affiliation(s)
- Saleela M Ruwanpura
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Sarah Rosli
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
49
|
Zhang P, Xin X, Fang L, Jiang H, Xu X, Su X, Shi Y. HMGB1 mediates Aspergillus fumigatus -induced inflammatory response in alveolar macrophages of COPD mice via activating MyD88/NF-κB and syk/PI3K signalings. Int Immunopharmacol 2017; 53:125-132. [DOI: 10.1016/j.intimp.2017.10.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/23/2017] [Accepted: 10/05/2017] [Indexed: 12/20/2022]
|
50
|
Scheffold A, Schwarz C, Bacher P. Fungus-Specific CD4 T Cells as Specific Sensors for Identification of Pulmonary Fungal Infections. Mycopathologia 2017; 183:213-226. [PMID: 29168073 DOI: 10.1007/s11046-017-0229-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 11/14/2017] [Indexed: 12/24/2022]
Abstract
Patients with cystic fibrosis (CF) suffer from chronic lung infections, caused by bacterial, viral or fungal pathogens, which determine morbidity and mortality. The contribution of individual pathogens to chronic disease and acute lung exacerbations is often difficult to determine due to the complex composition of the lung microbiome in CF. In particular, the relevance of fungal pathogens in CF airways remains poorly understood due to limitations of current diagnostics to identify the presence of fungal pathogens and to resolve the individual host-pathogen interaction status. T-lymphocytes play an essential role in host defense against pathogens, but also in inappropriate immune reactions such as allergies. They have the capacity to specifically recognize and discriminate the different pathogens and orchestrate a diverse array of effector functions. Thus, the analysis of the fungus-specific T cell status of an individual can in principle provide detailed information about the identity of the fungal pathogen(s) encountered and the actual fungus-host interaction status. This may allow to classify patients, according to appropriate (protective) or inappropriate (pathology-associated) immune reactions against individual fungal pathogens. However, T cell-based diagnostics are currently not part of the clinical routine. The identification and characterization of fungus-specific T cells in health and disease for diagnostic purposes are associated with significant challenges. Recent technological developments in the field of fungus-specific T helper cell detection provide new insights in the host T cell-fungus interaction. In this review, we will discuss basic principles and the potential of T cell-based diagnostics, as well as the perspectives and further needs for use of T cells for improved clinical diagnostics of fungal diseases.
Collapse
Affiliation(s)
- Alexander Scheffold
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, Leibniz Association, Berlin, Germany.
| | - Carsten Schwarz
- Department of Pediatric Pneumology and Immunology, Cystic Fibrosis Centre Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Bacher
- Department of Cellular Immunology, Clinic for Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|