1
|
Belfeki N, Kammoun S, Ghriss N, Eldirani C, Mekinian A. Current concepts on pathogenesis, diagnosis and management of hepatic sarcoidosis. Rheumatol Int 2025; 45:130. [PMID: 40304755 DOI: 10.1007/s00296-025-05888-2] [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: 03/02/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025]
Abstract
Sarcoidosis is an inflammatory multisystemic granulomatosis of unknown cause, with a wide range of clinical features, characterized by the presence of non-caseating granulomas. Liver is the third involved organ after lungs and lymph nodes, with a reported prevalence of hepatic involvement in 5 to 25% of systemic symptomatic sarcoidosis. The immunopathogenesis of sarcoidosis is still unknown but it seems to involve the innate and adaptive immune actors in response to a putative antigen in genetically predisposed individuals. Because of its paucisymptomatic presentation, hepatic sarcoidosis may be underdiagnosed. Unspecified impaired general condition, fever, abdominal pain, and jaundice are the main symptoms associated with liver sarcoidosis. Frequently, laboratory liver tests are abnormal. Imaging tools may reveal liver nodular enlargement but can be totally normal. Liver biopsy is often required to confirm the diagnosis. A meticulous workup is mandatory to rule out differential diagnosis of hepatic granuloma. Portal hypertension and liver cirrhosis are the most prevalent complications of hepatic sarcoidosis. Treatment is not necessary in all cases and first line treatment in symptomatic patients requires corticosteroids and/or ursodeoxycholic acid. Immunosuppressants and biologics could be used as second line agents. In severe cases, liver transplantation is indicated.
Collapse
Affiliation(s)
- Nabil Belfeki
- Department of Internal Medicine and Clinical Immunology, Groupe Hospitalier Sud-Ile de France, 270 Avenue Marc Jacquet, Melun, 77 000, France.
| | - Sonia Kammoun
- Department of Internal Medicine and Clinical Immunology, Groupe Hospitalier Sud-Ile de France, 270 Avenue Marc Jacquet, Melun, 77 000, France
| | - Nouha Ghriss
- Department of Internal Medicine and Clinical Immunology, Groupe Hospitalier Sud-Ile de France, 270 Avenue Marc Jacquet, Melun, 77 000, France
| | - Charif Eldirani
- Departement of Gastroenterology and Hepatology, Groupe Hospitalier Sud-Ile de France, Melun, France
| | - Arsene Mekinian
- Department of Internal Medicine, Inflammation-Immunopathology-Biotherapy, Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine Paris, Paris, France
- Sorbonne University, UMPC University Paris 06, INSERM U938, Centre de Recherche Saint- Antoine (CRSA), Paris, France
| |
Collapse
|
2
|
Bahetjan K, Yu-Xia, Lin S, Aili N, Yang H, Du S. Analysis of the bronchoalveolar lavage fluid microbial flora in COPD patients at different lung function during acute exacerbation. Sci Rep 2025; 15:13179. [PMID: 40240456 PMCID: PMC12003667 DOI: 10.1038/s41598-025-96746-5] [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: 08/05/2024] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
There is a correlation between the dysbiosis of the respiratory microbiota and the occurrence, severity, frequency, and mortality of Chronic Obstructive Pulmonary Disease (COPD). However, it is not unclear if there are differences in the bronchoalveolar lavage fluid (BALF) microbiota among patients at differente lung function. In this study, BALF samples were collected from 70 COPD patients experiencing acute exacerbations (AECOPD). The patients were divided into a mild group (FEV1/pre ≥ 50; PFT I, n = 50) and a severe group (FEV1/pre < 50; PFT II, n = 20) according to the lung function: or a frequent exacerbation (FE, n = 41) group and a non-frequent exacerbation (NFE, n = 29) group according to their exacerbation history. Microbiota analysis of BALF samples was conducted using mNGS and bioinfromatic analysis. Compared to PFT I group, PFT II group exhibited a significant decrease in species diversity (Shannon index), as well as a significant reduction in total species count and richness (Chao1, ACE indices). NFE group demonstrated diversity similar to that of FE group. Conversely, the microbial diversity of NFE group was comparable to that of FE group. The most abundant bacterial genera were Streptococcus, Prevotella, Veillonella, Rod-shaped Bacillus, and Rothia. Aspergillus was the most dominant fungal genus in AECOPD. Lymphocryptovirus was the most prevalent virus in AECOPD.Compared to the PFT I group, Corynebacterium's abundance significantly increased in PFT II group. Furthermore, FE group showed a notable increase in Streptococcus mitis abundance relative to NFE group. Bubble plot analysis revealed a significant increase in Moraxella, Fusobacterium, Haemophilus, Pseudomonas, Streptomyces, and Klebsiella in PFT II group, including a notable increase in typical Veillonella, Actinomyces, and Gordonia. The NFE group exhibited a significant increase in Bacteroides and Prevotella's relative abundance. Spearman correlation analysis revealed strong positive correlations among certain microbial communities. There exists a significant variation in microbial composition across groups of AECOPD patients at different lung function. Specifically, patients with severe airflow limitations exhibit a significant reduction in microbial diversity. Additionally, distinct bacterial taxa are enriched in patients characterized by varying levels of airflow limitation and exacerbation frequency. These observations offer vital insights into the pathogenesis of AECOPD, suggesting a potentially crucial role for the microbiota in its development. Such findings pave the way for identifying potential therapeutic targets and intervention strategies, ultimately aiming to improve treatment outcomes for AECOPD patients.
Collapse
Affiliation(s)
- Kawsar Bahetjan
- Respiratory and Respiratory Critical Care Center, School of Medicine, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Yu-Xia
- Second Ward, Respiratory and Respiratory Critical Care Center, First Affiliated Hospital of Xinjiang Medical University, Urumchi, 830011, China.
| | - Shijun Lin
- Respiratory and Respiratory Critical Care Center, School of Medicine, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Nuerziba Aili
- Second Ward, Respiratory and Respiratory Critical Care Center, First Affiliated Hospital of Xinjiang Medical University, Urumchi, 830011, China
| | - Haiyan Yang
- Second Ward, Respiratory and Respiratory Critical Care Center, First Affiliated Hospital of Xinjiang Medical University, Urumchi, 830011, China
| | - Shijun Du
- Second Ward, Respiratory and Respiratory Critical Care Center, First Affiliated Hospital of Xinjiang Medical University, Urumchi, 830011, China
| |
Collapse
|
3
|
Xu D, Tao X, Fan Y, Teng Y. Sarcoidosis: molecular mechanisms and therapeutic strategies. MOLECULAR BIOMEDICINE 2025; 6:6. [PMID: 39904950 PMCID: PMC11794924 DOI: 10.1186/s43556-025-00244-z] [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: 08/27/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
Sarcoidosis, a multisystemic granulomatous disease with unknown etiology, is characterized by formation of noncaseating granulomas, which can affect all organs. Recent studies have made outstanding achievement in understanding the pathology, etiology, genetics, and immune dysregulation involved in granuloma formation of sarcoidosis. Antigen stimulation in genetically predisposed individuals enhances the phagocytic activity of antigen-presenting cells, including macrophages and dendritic cells. CD4 + T cells initiate dysregulated immune responses and secrete significant quantities of inflammatory cytokines, including interleukin (IL)-2 and interferon-gamma (IFN-γ), which play a crucial role in modulating the aggregation and fusion of macrophages to form granulomas. The current therapeutic strategies focus on blocking the formation and spread of granulomas to protect organ function and alleviate symptoms. The efficacy of traditional treatments, such as glucocorticoids and immunosuppressants, has been confirmed in the management of sarcoidosis. Promising therapeutic agents encompass inhibitors of cytokines, like those targeting tumor necrosis factor (TNF)-α, as well as inhibitors of signaling pathways, such as Janus kinase (JAK) inhibitors, which exhibit favorable prospects for application. Although there has been progress in the identification of biomarkers for the diagnosis, prognosis, activity and severity of sarcoidosis, specific and sensitive biomarkers have yet to be identified. This review outlines recent advancements in the molecular mechanisms and therapeutic strategies for the sarcoidosis.
Collapse
Affiliation(s)
- Danfeng Xu
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaohua Tao
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yibin Fan
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Teng
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Moore C, Liao SY, Wood C, Sarkar A, Cardwell J, MacPhail K, Mroz MM, Riley C, Mould K, Restrepo C, Li L, Maier LA, Yang IV. Single Cell Transcriptome Signatures of Sarcoidosis in Lung Immune Cell Populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633917. [PMID: 39896662 PMCID: PMC11785102 DOI: 10.1101/2025.01.20.633917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Rationale To identify cell specific molecular changes associated with sarcoidosis risk and progression, we aimed to characterize the cellular composition, gene expression patterns, and cell-cell interactions in BAL cells from patients with sarcoidosis (both progressive and non-progressive) and healthy controls. Methods Single cell RNA-seq data were collected on 12 sarcoidosis and 4 control participants. We combined scRNA-seq data from these participants with our previously collected data on 4 sarcoidosis and 10 control participants for a final sample size of 16 sarcoidosis cases (8 progressive and 8 non-progressive) and 14 controls. Following initial preprocessing in CellRanger, data were quality controlled, combined, and clustered in Seurat. We tested differences in cell proportions by disease group using F-tests on cell proportions and differences in gene expression using pseudobulk analysis. Cell to cell communication and pathway analysis were performed using CellChat. Results We identified five macrophage populations: resident, high metallothionein (MT) resident, recruited, profibrotic recruited, and proliferating macrophages. Each subpopulation displayed unique gene expression profiles, with notable differential expression of genes and pathways linked to sarcoidosis in resident macrophages, recruited macrophages, and proliferating macrophages. We also observed changes in gene expression associated with disease progression in resident and recruited macrophages. In non-macrophages cells, we observed a significant reduction in the number of B cells in sarcoidosis patients compared to controls. Among T cell populations, we identified specific transcriptional alterations at gene and pathway level. Additionally, we observed distinct differences in cell-to-cell interactions of macrophages and T cells between sarcoidosis patients and healthy controls. Conclusions These findings underscore the complexity of immune cell involvement in sarcoidosis and highlight potential cellular and molecular targets for further investigation.
Collapse
|
5
|
Miedema J, Cinetto F, Smed-Sörensen A, Spagnolo P. The immunopathogenesis of sarcoidosis. J Autoimmun 2024; 149:103247. [PMID: 38734536 DOI: 10.1016/j.jaut.2024.103247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Sarcoidosis is a granulomatous multiorgan disease, thought to result from exposure to yet unidentified antigens in genetically susceptible individuals. The exaggerated inflammatory response that leads to granuloma formation is highly complex and involves the innate and adaptive immune system. Consecutive immunological studies using advanced technology have increased our understanding of aberrantly activated immune cells, mediators and pathways that influence the formation, maintenance and resolution of granulomas. Over the years, it has become increasingly clear that disease immunopathogenesis can only be understood if the clinical heterogeneity of sarcoidosis is taken into consideration, along with the distribution of immune cells in peripheral blood and involved organs. Most studies offer an immunological snapshot during disease course, while the cellular composition of both the circulation and tissue microenvironment may change over time. Despite these challenges, novel insights on the role of the immune system are continuously published, thus bringing the field forward. This review highlights current knowledge on the innate and adaptive immune responses involved in sarcoidosis pathogenesis, as well as the pathways involved in non-resolving disease and fibrosis development. Additionally, we describe proposed immunological mechanisms responsible for drug-induced sarcoid like reactions. Although many aspects of disease immunopathogenesis remain to be unraveled, the identification of crucial immune reactions in sarcoidosis may help identify new treatment targets. We therefore also discuss potential therapies and future strategies based on the latest immunological findings.
Collapse
Affiliation(s)
- Jelle Miedema
- Department of Pulmonary Medicine, Center of Expertise for Interstitial Lung Disease, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Italy; Department of Medicine - DIMED, University of Padova, Padova, Italy.
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy.
| |
Collapse
|
6
|
Yang J, Ma Y, Yu J, Liu Y, Xia J, Kong X, Jin X, Li J, Lin S, Ruan Y, Yang F, Pi J. Advancing Roles and Therapeutic Potentials of Pyroptosis in Host Immune Defenses against Tuberculosis. Biomolecules 2024; 14:1255. [PMID: 39456188 PMCID: PMC11505957 DOI: 10.3390/biom14101255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (Mtb) infection, remains a deadly global public health burden. The use of recommended drug combinations in clinic has seen an increasing prevalence of drug-resistant TB, adding to the impediments to global control of TB. Therefore, control of TB and drug-resistant TB has become one of the most pressing issues in global public health, which urges the exploration of potential therapeutic targets in TB and drug-resistant TB. Pyroptosis, a form of programmed cell death characterized by cell swelling and rupture, release of cellular contents and inflammatory responses, has been found to promote pathogen clearance and adopt crucial roles in the control of bacterial infections. It has been demonstrated that Mtb can cause host cell pyroptosis, and these host cells, which are infected by Mtb, can kill Mtb accompanied by pyroptosis, while, at the same time, pyroptosis can also release intracellular Mtb, which may potentially worsen the infection by exacerbating the inflammation. Here, we describe the main pathways of pyroptosis during Mtb infection and summarize the identified effectors of Mtb that regulate pyroptosis to achieve immune evasion. Moreover, we also discuss the potentials of pyroptosis to serve as an anti-TB therapeutic target, with the aim of providing new ideas for the development of TB treatments.
Collapse
Affiliation(s)
- Jiayi Yang
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yuhe Ma
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaqi Yu
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yilin Liu
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaojiao Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China;
| | - Xinen Kong
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Xiaoying Jin
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiaxiang Li
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Siqi Lin
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Yongdui Ruan
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Fen Yang
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| | - Jiang Pi
- Acupuncture and Moxibustion Department, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan 523808, China; (J.Y.); (Y.M.); (J.Y.); (Y.L.); (X.K.); (X.J.); (J.L.); (S.L.); (Y.R.)
| |
Collapse
|
7
|
Gogoi D, Yu H, Casey M, Baird R, Yusuf A, Forde L, O' Brien ME, West JR, Flagg T, McElvaney NG, Eden E, Mueller C, Brantly ML, Geraghty P, Reeves EP. Monocyte NLRP3 inflammasome and interleukin-1β activation modulated by alpha-1 antitrypsin therapy in deficient individuals. Thorax 2024; 79:822-833. [PMID: 38418195 PMCID: PMC11347198 DOI: 10.1136/thorax-2023-221071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/02/2024] [Indexed: 03/01/2024]
Abstract
INTRODUCTION Altered complement component 3 (C3) activation in patients with alpha-1 antitrypsin (AAT) deficiency (AATD) has been reported. To understand the potential impact on course of inflammation, the aim of this study was to investigate whether C3d, a cleavage-product of C3, triggers interleukin (IL)-1β secretion via activation of NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome. The objective was to explore the effect of AAT augmentation therapy in patients with AATD on the C3d/complement receptor 3 (CR3) signalling axis of monocytes and on circulating pro-inflammatory markers. METHODS Inflammatory mediators were detected in blood from patients with AATD (n=28) and patients with AATD receiving augmentation therapy (n=19). Inflammasome activation and IL-1β secretion were measured in monocytes of patients with AATD, and following C3d stimulation in the presence or absence of CR3 or NLRP3 inhibitors. RESULTS C3d acting via CR3 induces NLRP3 and pro-IL-1β production, and through induction of endoplasmic reticulum (ER) stress and calcium flux, triggers caspase-1 activation and IL-1β secretion. Treatment of individuals with AATD with AAT therapy results in decreased plasma levels of C3d (3.0±1.2 µg/mL vs 1.3±0.5 µg/mL respectively, p<0.0001) and IL-1β (115.4±30 pg/mL vs 73.3±20 pg/mL, respectively, p<0.0001), with a 2.0-fold decrease in monocyte NLRP3 protein expression (p=0.0303), despite continued ER stress activation. DISCUSSION These results provide strong insight into the mechanism of complement-driven inflammation associated with AATD. Although the described variance in C3d and NLRP3 activation decreased post AAT augmentation therapy, results demonstrate persistent C3d and monocyte ER stress, with implications for new therapeutics and clinical practice.
Collapse
Affiliation(s)
- Debananda Gogoi
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Howard Yu
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Michelle Casey
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rory Baird
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Azeez Yusuf
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Luke Forde
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Michael E O' Brien
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jesse R West
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Tammy Flagg
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Edward Eden
- Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Christian Mueller
- The Li Weibo Institute for Rare Diseases Research, Horae Gene Therapy Center, Worcester, MA, USA
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mark L Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, J. Hillis Miller Health Science Center, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Emer P Reeves
- Pulmonary Clinical Science, Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
8
|
Heymans S, Van Linthout S, Kraus SM, Cooper LT, Ntusi NAB. Clinical Characteristics and Mechanisms of Acute Myocarditis. Circ Res 2024; 135:397-411. [PMID: 38963866 DOI: 10.1161/circresaha.124.324674] [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] [Indexed: 07/06/2024]
Abstract
REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT05335928.
Collapse
Affiliation(s)
- Stephane Heymans
- Centre for Heart Failure Research, Department of Cardiology, Maastricht University, The Netherlands (S.H.)
- Department of Cardiovascular Sciences, University of Leuven, Belgium (S.H.)
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Germany (S.V.L.)
- German Centre for Cardiovascular Research, partner site Berlin, Germany (S.V.L.)
| | - Sarah Mignon Kraus
- Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, South Africa (S.M.K., N.A.B.N.)
- South African Medical Research Council Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, Cape Town, South Africa (S.M.K., N.A.B.N.)
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL (L.T.C.)
| | - Ntobeko A B Ntusi
- Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, South Africa (S.M.K., N.A.B.N.)
- South African Medical Research Council Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, Cape Town, South Africa (S.M.K., N.A.B.N.)
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, South Africa (N.A.B.N.)
- ARUA/Guild Cluster of Research Excellence on Noncommunicable Diseases and Associated Multiborbidity, South Africa (N.A.B.N.)
| |
Collapse
|
9
|
Ma H, Ou ZL, Alaeiilkhchi N, Cheng YQ, Chen K, Chen JY, Guo RQ, He MY, Tang SY, Zhang X, Huang ZP, Liu J, Liu J, Zhu QA, Huang ZC, Jiang H. MiR-223 enhances lipophagy by suppressing CTSB in microglia following lysolecithin-induced demyelination in mice. Lipids Health Dis 2024; 23:194. [PMID: 38909243 PMCID: PMC11193262 DOI: 10.1186/s12944-024-02185-y] [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: 02/07/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Lipid droplet (LD)-laden microglia is a key pathological hallmark of multiple sclerosis. The recent discovery of this novel microglial subtype, lipid-droplet-accumulating microglia (LDAM), is notable for increased inflammatory factor secretion and diminished phagocytic capability. Lipophagy, the autophagy-mediated selective degradation of LDs, plays a critical role in this context. This study investigated the involvement of microRNAs (miRNAs) in lipophagy during demyelinating diseases, assessed their capacity to modulate LDAM subtypes, and elucidated the potential underlying mechanisms involved. METHODS C57BL/6 mice were used for in vivo experiments. Two weeks post demyelination induction at cervical level 4 (C4), histological assessments and confocal imaging were performed to examine LD accumulation in microglia within the lesion site. Autophagic changes were observed using transmission electron microscopy. miRNA and mRNA multi-omics analyses identified differentially expressed miRNAs and mRNAs under demyelinating conditions and the related autophagy target genes. The role of miR-223 in lipophagy under these conditions was specifically explored. In vitro studies, including miR-223 upregulation in BV2 cells via lentiviral infection, validated the bioinformatics findings. Immunofluorescence staining was used to measure LD accumulation, autophagy levels, target gene expression, and inflammatory mediator levels to elucidate the mechanisms of action of miR-223 in LDAM. RESULTS Oil Red O staining and confocal imaging revealed substantial LD accumulation in the demyelinated spinal cord. Transmission electron microscopy revealed increased numbers of autophagic vacuoles at the injury site. Multi-omics analysis revealed miR-223 as a crucial regulatory gene in lipophagy during demyelination. It was identified that cathepsin B (CTSB) targets miR-223 in autophagy to integrate miRNA, mRNA, and autophagy gene databases. In vitro, miR-223 upregulation suppressed CTSB expression in BV2 cells, augmented autophagy, alleviated LD accumulation, and decreased the expression of the inflammatory mediator IL-1β. CONCLUSION These findings indicate that miR-223 plays a pivotal role in lipophagy under demyelinating conditions. By inhibiting CTSB, miR-223 promotes selective LD degradation, thereby reducing the lipid burden and inflammatory phenotype in LDAM. This study broadens the understanding of the molecular mechanisms of lipophagy and proposes lipophagy induction as a potential therapeutic approach to mitigate inflammatory responses in demyelinating diseases.
Collapse
Grants
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No.81902217 National Natural Science Foundation of China
- No. 81972064 National Natural Science Foundation of China
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- 2020A1515011415 Natural Science Foundation of Guangdong Province
- 2023A1515010565 Natural Science Foundation of Guangdong Province
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- No.2023A024 President Foundation of Nanfang Hospital, Southern Medical University
- 202102021244 Guangzhou Science and Technology Plan Project
Collapse
Affiliation(s)
- Hao Ma
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhi-Lin Ou
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Nima Alaeiilkhchi
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, Vancouver, Canada
| | - Yong-Quan Cheng
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Kai Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jia-Yu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ru-Qin Guo
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Min-Yue He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shi-Yi Tang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Xin Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhi-Ping Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Junhao Liu
- Division of Spine Surgery, Department of Orthopaedics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 51000, China
| | - Jie Liu
- The 3rd Affiliated Hospital of Xinxiang Medical University, Henan, China
| | - Qing-An Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zu-Cheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Hui Jiang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
10
|
Santos-Ribeiro D, Cunha C, Carvalho A. Humoral pathways of innate immune regulation in granuloma formation. Trends Immunol 2024; 45:419-427. [PMID: 38762333 DOI: 10.1016/j.it.2024.04.008] [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: 03/30/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/20/2024]
Abstract
The humoral arm of mammalian innate immunity regulates several molecular mechanisms involved in resistance to pathogens, inflammation, and tissue repair. Recent studies highlight the crucial role played by humoral mediators in granulomatous inflammation. However the molecular mechanisms linking the function of these soluble molecules to the initiation and maintenance of granulomas remain elusive. We propose that humoral innate immunity coordinates fundamental physiological processes in macrophages which, in turn, initiate activation and transformation events that enable granuloma formation. We discuss the involvement of humoral mediators in processes such as immune activation, phagocytosis, metabolism, and tissue remodeling, and how these can dictate macrophage functionality during granuloma formation. These advances present opportunities for discovering novel disease factors and developing targeted, more effective treatments for granulomatous diseases.
Collapse
Affiliation(s)
- Diana Santos-Ribeiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 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, 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, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
11
|
Toldo S, Abbate A. The role of the NLRP3 inflammasome and pyroptosis in cardiovascular diseases. Nat Rev Cardiol 2024; 21:219-237. [PMID: 37923829 PMCID: PMC11550901 DOI: 10.1038/s41569-023-00946-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2023] [Indexed: 11/06/2023]
Abstract
An intense, stereotyped inflammatory response occurs in response to ischaemic and non-ischaemic injury to the myocardium. The NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome is a finely regulated macromolecular protein complex that senses the injury and triggers and amplifies the inflammatory response by activation of caspase 1; cleavage of pro-inflammatory cytokines, such as pro-IL-1β and pro-IL-18, to their mature forms; and induction of inflammatory cell death (pyroptosis). Inhibitors of the NLRP3 inflammasome and blockers of IL-1β and IL-18 activity have been shown to reduce injury to the myocardium and pericardium, favour resolution of the inflammation and preserve cardiac function. In this Review, we discuss the components of the NLRP3 inflammasome and how it is formed and activated in various ischaemic and non-ischaemic cardiac pathologies (acute myocardial infarction, cardiac dysfunction and remodelling, atherothrombosis, myocarditis and pericarditis, cardiotoxicity and cardiac sarcoidosis). We also summarize current preclinical and clinical evidence from studies of agents that target the NLRP3 inflammasome and related cytokines.
Collapse
Affiliation(s)
- Stefano Toldo
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW There has been a rapid increase in silicosis cases, particularly related to artificial stone. The key to management is avoidance of silica exposure. Despite this, many develop progressive disease and there are no routinely recommended treatments. This review provides a summary of the literature pertaining to pharmacological therapies for silicosis and examines the plausibility of success of such treatments given the disease pathogenesis. RECENT FINDINGS In-vitro and in-vivo models demonstrate potential efficacy for drugs, which target inflammasomes, cytokines, effector cells, fibrosis, autophagy, and oxidation. SUMMARY There is some evidence for potential therapeutic targets in silicosis but limited translation into human studies. Treatment of silicosis likely requires a multimodal approach, and there is considerable cross-talk between pathways; agents that modulate both inflammation, fibrosis, autophagy, and ROS production are likely to be most efficacious.
Collapse
Affiliation(s)
- Hayley Barnes
- Monash Centre for Occupational and Environmental Health, Monash University
- Department of Respiratory Medicine, Alfred Health
- Central Clinical School, Monash University, Melbourne
| | - Maggie Lam
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Michelle D Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Ryan Hoy
- Monash Centre for Occupational and Environmental Health, Monash University
- Department of Respiratory Medicine, Alfred Health
| |
Collapse
|
13
|
Kim J, Dwivedi G, Boughton BA, Sharma A, Lee S. Advances in cellular and tissue-based imaging techniques for sarcoid granulomas. Am J Physiol Cell Physiol 2024; 326:C10-C26. [PMID: 37955119 DOI: 10.1152/ajpcell.00507.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Sarcoidosis embodies a complex inflammatory disorder spanning multiple systems, with its origin remaining elusive. It manifests as the infiltration of inflammatory cells that coalesce into distinctive noncaseous granulomas within afflicted organs. Unraveling this disease necessitates the utilization of cellular or tissue-based imaging methods to both visualize and characterize the biochemistry of these sarcoid granulomas. Although hematoxylin and eosin stain, standard in routine use alongside cytological stains have found utility in diagnosis within clinical contexts, special stains such as Masson's trichrome, reticulin, methenamine silver, and Ziehl-Neelsen provide additional varied perspectives of sarcoid granuloma imaging. Immunohistochemistry aids in pinpointing specific proteins and gene expressions further characterizing these granulomas. Finally, recent advances in spatial transcriptomics promise to divulge profound insights into their spatial orientation and three-dimensional (3-D) molecular mapping. This review focuses on a range of preexisting imaging methods employed for visualizing sarcoid granulomas at the cellular level while also exploring the potential of the latest cutting-edge approaches like spatial transcriptomics and matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI), with the overarching goal of shedding light on the trajectory of sarcoidosis research.
Collapse
Affiliation(s)
- Junwoo Kim
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
- Department of Cardiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Berin A Boughton
- Australian National Phenome Centre, Murdoch University, Murdoch, Western Australia, Australia
| | - Ankur Sharma
- Onco-Fetal Ecosystem Laboratory, Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| | - Silvia Lee
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia, Australia
- Curtin Medical School, Curtin University, Perth, Western Australia, Australia
| |
Collapse
|
14
|
Harvest CK, Abele TJ, Yu C, Beatty CJ, Amason ME, Billman ZP, DePrizio MA, Souza FW, Lacey CA, Maltez VI, Larson HN, McGlaughon BD, Saban DR, Montgomery SA, Miao EA. An innate granuloma eradicates an environmental pathogen using Gsdmd and Nos2. Nat Commun 2023; 14:6686. [PMID: 37865673 PMCID: PMC10590453 DOI: 10.1038/s41467-023-42218-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/21/2023] [Indexed: 10/23/2023] Open
Abstract
Granulomas often form around pathogens that cause chronic infections. Here, we discover an innate granuloma model in mice with an environmental bacterium called Chromobacterium violaceum. Granuloma formation not only successfully walls off, but also clears, the infection. The infected lesion can arise from a single bacterium that replicates despite the presence of a neutrophil swarm. Bacterial replication ceases when macrophages organize around the infection and form a granuloma. This granuloma response is accomplished independently of adaptive immunity that is typically required to organize granulomas. The C. violaceum-induced granuloma requires at least two separate defense pathways, gasdermin D and iNOS, to maintain the integrity of the granuloma architecture. This innate granuloma successfully eradicates C. violaceum infection. Therefore, this C. violaceum-induced granuloma model demonstrates that innate immune cells successfully organize a granuloma and thereby resolve infection by an environmental pathogen.
Collapse
Affiliation(s)
- Carissa K Harvest
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Taylor J Abele
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Chen Yu
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Cole J Beatty
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Megan E Amason
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zachary P Billman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Morgan A DePrizio
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Fernando W Souza
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Carolyn A Lacey
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Vivien I Maltez
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather N Larson
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Benjamin D McGlaughon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel R Saban
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie A Montgomery
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edward A Miao
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA.
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
15
|
Hodzhev Y, Tsafarova B, Tolchkov V, Youroukova V, Ivanova S, Kostadinov D, Yanev N, Zhelyazkova M, Tsonev S, Kalfin R, Panaiotov S. Visualization of the individual blood microbiome to study the etiology of sarcoidosis. Comput Struct Biotechnol J 2023; 22:50-57. [PMID: 37928975 PMCID: PMC10624578 DOI: 10.1016/j.csbj.2023.10.027] [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: 06/19/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Single microbial pathogens or host-microbiome dysbiosis are the causes of lung diseases with suspected infectious etiology. Metagenome sequencing provides an overview of the microbiome content. Due to the rarity of most granulomatous lung diseases collecting large systematic datasets is challenging. Thus, single-patient data often can only be summarized visually. Objective To increase the information gain from a single-case metagenome analysis we suggest a quantitative and qualitative approach. Results The 16S metagenomic results of 7 patients with pulmonary sarcoidosis were compared with those of 22 healthy individuals. From lysed blood, total microbial DNA was extracted and sequenced. Cleaned data reads were identified taxonomically using Kraken 2 software. Individual metagenomic data were visualized with a Sankey diagram, Krona chart, and a heat-map. We identified five genera that were exclusively present or significantly enhanced in patients with sarcoidosis - Veillonella, Prevotella, Cutibacterium, Corynebacterium, and Streptococcus. Conclusions Our approach can characterize the blood microbiome composition and diversity in rare diseases at an individual level. Investigation of the blood microbiome in patients with granulomatous lung diseases of unknown etiology, such as sarcoidosis could enhance our comprehension of their origin and pathogenesis and potentially uncover novel personalized therapeutics.
Collapse
Affiliation(s)
- Yordan Hodzhev
- Microbiology Department, National Center of Infectious and Parasitic Diseases, Yanko Sakazov 26 Blvd., Sofia 1504, Bulgaria
| | - Borislava Tsafarova
- Microbiology Department, National Center of Infectious and Parasitic Diseases, Yanko Sakazov 26 Blvd., Sofia 1504, Bulgaria
| | - Vladimir Tolchkov
- Microbiology Department, National Center of Infectious and Parasitic Diseases, Yanko Sakazov 26 Blvd., Sofia 1504, Bulgaria
| | - Vania Youroukova
- Department of Pulmonary Diseases, University Hospital for Pulmonary Diseases “St. Sofia”, Medical University of Sofia, Akad. Ivan Evstratiev Geshov 17 Blvd., Sofia 1431, Bulgaria
| | - Silvia Ivanova
- Department of Pulmonary Diseases, University Hospital for Pulmonary Diseases “St. Sofia”, Medical University of Sofia, Akad. Ivan Evstratiev Geshov 17 Blvd., Sofia 1431, Bulgaria
| | - Dimitar Kostadinov
- Department of Pulmonary Diseases, University Hospital for Pulmonary Diseases “St. Sofia”, Medical University of Sofia, Akad. Ivan Evstratiev Geshov 17 Blvd., Sofia 1431, Bulgaria
| | - Nikolay Yanev
- Department of Pulmonary Diseases, University Hospital for Pulmonary Diseases “St. Sofia”, Medical University of Sofia, Akad. Ivan Evstratiev Geshov 17 Blvd., Sofia 1431, Bulgaria
| | - Maya Zhelyazkova
- Faculti of Mathematics and Informatics, Sofia University St. Kliment Ohridski, 5 James Bourchier Blvd., 1164 Sofia, Bulgaria
| | - Stefan Tsonev
- Agrobioinstitute (ABI), 8 Dragan Tsankov, Blvd, Sofia 1164, Bulgaria
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia 1113, Bulgaria
- Department of Health Care, South-West University “Neofit Rilski”, Blagoevgrad 2700, Bulgaria
| | - Stefan Panaiotov
- Microbiology Department, National Center of Infectious and Parasitic Diseases, Yanko Sakazov 26 Blvd., Sofia 1504, Bulgaria
| |
Collapse
|
16
|
Chai Q, Lei Z, Liu CH. Pyroptosis modulation by bacterial effector proteins. Semin Immunol 2023; 69:101804. [PMID: 37406548 DOI: 10.1016/j.smim.2023.101804] [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: 03/21/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/07/2023]
Abstract
Pyroptosis is a proinflammatory form of programmed cell death featured with membrane pore formation that causes cellular swelling and allows the release of intracellular inflammatory mediators. This cell death process is elicited by the activation of the pore-forming proteins named gasdermins, and is intricately orchestrated by diverse regulatory factors in mammalian hosts to exert a prompt immune response against infections. However, growing evidence suggests that bacterial pathogens have evolved to regulate host pyroptosis for evading immune clearance and establishing progressive infection. In this review, we highlight current understandings of the functional role and regulatory network of pyroptosis in host antibacterial immunity. Thereafter, we further discuss the latest advances elucidating the mechanisms by which bacterial pathogens modulate pyroptosis through adopting their effector proteins to drive infections. A better understanding of regulatory mechanisms underlying pyroptosis at the interface of host-bacterial interactions will shed new light on the pathogenesis of infectious diseases and contribute to the development of promising therapeutic strategies against bacterial pathogens.
Collapse
Affiliation(s)
- Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zehui Lei
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China.
| |
Collapse
|
17
|
Kraaijvanger R, Ambarus CA, Damen J, van der Vis JJ, Kazemier KM, Grutters JC, van Moorsel CHM, Veltkamp M. Simultaneous Assessment of mTORC1, JAK/STAT, and NLRP3 Inflammasome Activation Pathways in Patients with Sarcoidosis. Int J Mol Sci 2023; 24:12792. [PMID: 37628972 PMCID: PMC10454122 DOI: 10.3390/ijms241612792] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/08/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
The unknown etiology of sarcoidosis, along with the variability in organ involvement and disease course, complicates the effective treatment of this disease. Based on recent studies, the cellular inflammatory pathways involved in granuloma formation are of interest regarding possible new treatment options, such as the mechanistic (formerly mammalian) target of rapamycin complex 1 (mTORC1) pathway, the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway, and the nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome pathway. The aim of this study was to explore the potential coexpression of these three inflammatory pathways in patients with sarcoidosis and see whether possible differences were related to disease outcome. The tissue of 60 patients with sarcoidosis was used to determine the activity of these three signaling pathways using immunohistochemistry. The activation of NLRP3 was present in 85% of all patients, and the activation of mTORC1 and JAK/STAT was present in 49% and 50% of patients, respectively. Furthermore, the presence of NLRP3 activation at diagnosis was associated with a chronic disease course of sarcoidosis. Our finding of different new conceptual inflammatory tissue phenotypes in sarcoidosis could possibly guide future treatment studies using the available inhibitors of either NLRP3, JAK-STAT, and mTORC1 inhibitors in a more personalized medicine approach.
Collapse
Affiliation(s)
- Raisa Kraaijvanger
- Interstitial Lung Diseases Center of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.)
| | - Carmen A. Ambarus
- Interstitial Lung Diseases Center of Excellence, Pathologie DNA, Department of Pathology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Jan Damen
- Pathologie DNA, Department of Pathology, Jeroen Bosch Hospital, 5223 GZ ‘s-Hertogenbosch, The Netherlands
| | - Joanne J. van der Vis
- Interstitial Lung Diseases Center of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.)
- Department of Clinical Chemistry, St Antonius ILD Center of Excellence, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Karin M. Kazemier
- Center of Translational Immunology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
- Division of Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Jan C. Grutters
- Interstitial Lung Diseases Center of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.)
- Division of Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| | - Coline H. M. van Moorsel
- Interstitial Lung Diseases Center of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.)
| | - Marcel Veltkamp
- Interstitial Lung Diseases Center of Excellence, Department of Pulmonology, St. Antonius Hospital, 3435 CM Nieuwegein, The Netherlands; (R.K.)
- Division of Heart and Lungs, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
18
|
Shi M, Lu Q, Zhao Y, Ding Z, Yu S, Li J, Ji M, Fan H, Hou S. miR-223: a key regulator of pulmonary inflammation. Front Med (Lausanne) 2023; 10:1187557. [PMID: 37465640 PMCID: PMC10350674 DOI: 10.3389/fmed.2023.1187557] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
Small noncoding RNAs, known as microRNAs (miRNAs), are vital for the regulation of diverse biological processes. miR-223, an evolutionarily conserved anti-inflammatory miRNA expressed in cells of the myeloid lineage, has been implicated in the regulation of monocyte-macrophage differentiation, proinflammatory responses, and the recruitment of neutrophils. The biological functions of this gene are regulated by its expression levels in cells or tissues. In this review, we first outline the regulatory role of miR-223 in granulocytes, macrophages, endothelial cells, epithelial cells and dendritic cells (DCs). Then, we summarize the possible role of miR-223 in chronic obstructive pulmonary disease (COPD), acute lung injury (ALI), coronavirus disease 2019 (COVID-19) and other pulmonary inflammatory diseases to better understand the molecular regulatory networks in pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Mingyu Shi
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Ziling Ding
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Sifan Yu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Junfeng Li
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Mengjun Ji
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
| | - Shike Hou
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
| |
Collapse
|
19
|
Chang X, Song YH, Xia T, He ZX, Zhao SB, Wang ZJ, Gu L, Li ZS, Xu C, Wang SL, Bai Y. Macrophage-derived exosomes promote intestinal mucosal barrier dysfunction in inflammatory bowel disease by regulating TMIGD1 via mircroRNA-223. Int Immunopharmacol 2023; 121:110447. [PMID: 37301121 DOI: 10.1016/j.intimp.2023.110447] [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: 03/24/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND & AIM Exosomes are effective mediators of cell-to-cell interactions and transport several regulatory molecules, including microRNAs (miRNAs), involved in diverse fundamental biological processes. The role of macrophage-derived exosomes in the development of inflammatory bowel disease (IBD) has not been previously reported. This study investigated specific miRNAs in macrophage-derived exosomes in IBD and their molecular mechanism. METHODS A dextran sulfate sodium (DSS)-induced IBD mouse model was established. The culture supernatant of murine bone marrow-derived macrophages (BMDMs) cultured with or without lipopolysaccharide (LPS) was used for isolating exosomes, which were subjected to miRNA sequencing. Lentiviruses were used to alter miRNA expression and investigate the role of macrophage-derived exosomal miRNAs. Both mouse and human organoids were co-cultured with macrophages in a Transwell system to model cellular IBD in vitro. RESULTS LPS-induced macrophages released exosomes containing various miRNAs and exacerbated IBD. Based on miRNA sequencing of macrophage-derived exosomes, miR-223 was selected for further analysis. Exosomes with upregulated miR-223 expression contributed to the exacerbation of intestinal barrier dysfunction in vivo, which was further verified using both mouse and human colon organoids. Furthermore, time-dependent analysis of the mRNAs in DSS-induced colitis mouse tissue and miR-223 target gene prediction were performed to select the candidate gene, resulting in the identification of the barrier-related factor Tmigd1. CONCLUSION Macrophage-derived exosomal miR-223 has a novel role in the progression of DSS-induced colitis by inducing intestinal barrier dysfunction through the inhibition of TMIGD1.
Collapse
Affiliation(s)
- Xin Chang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China; Department of Gastroenterology, the General Hospital of Central Theater Command, Wuhan, China
| | - Yi-Hang Song
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Tian Xia
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zi-Xuan He
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Sheng-Bing Zhao
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhi-Jie Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Lun Gu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Can Xu
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Shu-Ling Wang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China.
| | - Yu Bai
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
20
|
Harvest CK, Abele TJ, Yu C, Beatty CJ, Amason ME, Billman ZP, DePrizio MA, Lacey CA, Maltez VI, Larson HN, McGlaughon BD, Saban DR, Montgomery SA, Miao EA. An innate granuloma eradicates an environmental pathogen using Gsdmd and Nos2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531568. [PMID: 36945446 PMCID: PMC10028874 DOI: 10.1101/2023.03.07.531568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Granulomas often form around pathogens that cause chronic infections. Here, we discover a novel granuloma model in mice. Chromobacterium violaceum is an environmental bacterium that stimulates granuloma formation that not only successfully walls off but also clears the infection. The infected lesion can arise from a single bacterium that replicates in the presence of a neutrophil swarm. Bacterial replication ceases when macrophages organize around the infection and form a granuloma. This granuloma response is accomplished independently of adaptive immunity that is typically required to organize granulomas. The C. violaceum -induced granuloma requires at least two separate defense pathways, gasdermin D and iNOS, to maintain the integrity of the granuloma architecture. These innate granulomas successfully eradicate C. violaceum infection. Therefore, this new C. violaceum -induced granuloma model demonstrates that innate immune cells successfully organize a granuloma and thereby eradicate infection by an environmental pathogen.
Collapse
|
21
|
Abstract
Sarcoidosis is a systemic inflammatory disease of unknown etiology. The pathogenesis rests on an aberrant T cell response to unidentified antigens in individuals predisposed by genetic and environmental factors. Increased expression of polarized macrophages and disequilibrium between effector and regulator T cells contribute to the formation of noncaseating granulomas, that are frequently found in affected organs. The main kidney abnormalities in sarcoidosis are granulomatous interstitial nephritis (GIN) and hypercalcemia-related disorders. The clinical diagnosis is difficult. The outcome is variable, ranging from spontaneous remission to end-stage kidney disease (ESKD). Early diagnosis and prompt treatment with corticosteroids can improve the prognosis. Hypercalcemia may be responsible for acute kidney injury (AKI) caused by vasoconstriction of afferent arterioles. Complications of persistent hypercalcemia include nephrocalcinosis and renal stones. In patients with ESKD, dialysis and transplantation can offer results comparable to those observed in patients with other causes of kidney failure. Based on a review of the literature, we present an overview of the etiopathogenesis, the renal manifestations of sarcoidosis and their complications, management and prognosis.
Collapse
|
22
|
Offman E, Singh N, Julian MW, Locke LW, Bicer S, Mitchell J, Matthews T, Anderson K, Crouser ED. Leveraging in vitro and pharmacokinetic models to support bench to bedside investigation of XTMAB-16 as a novel pulmonary sarcoidosis treatment. Front Pharmacol 2023; 14:1066454. [PMID: 37021060 PMCID: PMC10067675 DOI: 10.3389/fphar.2023.1066454] [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: 10/20/2022] [Accepted: 02/24/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Sarcoidosis is a chronic, multisystem inflammatory disorder characterized by non-caseating epithelioid granulomas; infiltration of mononuclear cells; and destruction of microarchitecture in the skin, eye, heart, and central nervous system, and the lung in >90% of cases. XTMAB-16 is a chimeric anti-tumor necrosis factor alpha (TNFα) antibody, distinct from other anti-TNF antibodies based on its molecular structure. The efficacy of XTMAB-16 has not been clinically demonstrated, and it is still undergoing clinical development as a potential treatment for sarcoidosis. The current study demonstrates the activity of XTMAB-16 in a well-established in vitro sarcoidosis granuloma model, although XTMAB-16 is not yet approved by the United States Food and Drug Administration (FDA) for treatment of sarcoidosis, or any other disease. Objective: To provide data to guide safe and efficacious dose selection for the ongoing clinical development of XTMAB-16 as a potential treatment for sarcoidosis. Methods: First, XTMAB-16 activity was evaluated in an established in vitro model of granuloma formation using peripheral blood mononuclear cells from patients with active pulmonary sarcoidosis to determine a potentially efficacious dose range. Second, data obtained from the first-in-human study of XTMAB-16 (NCT04971395) were used to develop a population pharmacokinetic (PPK) model to characterize the pharmacokinetics (PK) of XTMAB-16. Model simulations were performed to evaluate the sources of PK variability and to predict interstitial lung exposure based on concentrations in the in vitro granuloma model. Results: XTMAB-16 dose levels of 2 and 4 mg/kg, once every 2 weeks (Q2W) or once every 4 weeks (Q4W) for up to 12 weeks, were supported by data from the non-clinical, in vitro secondary pharmacology; the Phase 1 clinical study; and the PPK model developed to guide dose level and frequency assumptions. XTMAB-16 inhibited granuloma formation and suppressed interleukin-1β (IL-1β) secretion in the in vitro granuloma model with a half maximal inhibitory concentration (IC50) of 5.2 and 3.5 μg/mL, respectively. Interstitial lung concentrations on average, following 2 or 4 mg/kg administered Q2W or Q4W, are anticipated to exceed the in vitro IC50 concentrations. Conclusion: The data presented in this report provide a rationale for dose selection and support the continued clinical development of XTMAB-16 for patients with pulmonary sarcoidosis.
Collapse
Affiliation(s)
| | | | - Mark W. Julian
- Division of Pulmonary, Critical Care and Sleep Medicine, The Dorothy M. Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Landon W. Locke
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | - Sabahattin Bicer
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | - Jonah Mitchell
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | | | | | - Elliott D. Crouser
- Division of Pulmonary, Critical Care and Sleep Medicine, The Dorothy M. Davis Heart and Lung Research Institute, Columbus, OH, United States
- *Correspondence: Elliott D. Crouser,
| |
Collapse
|
23
|
Feng Y, Li M, Yangzhong X, Zhang X, Zu A, Hou Y, Li L, Sun S. Pyroptosis in inflammation-related respiratory disease. J Physiol Biochem 2022; 78:721-737. [PMID: 35819638 PMCID: PMC9684248 DOI: 10.1007/s13105-022-00909-1] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/30/2022] [Indexed: 12/15/2022]
Abstract
Pyroptosis is commonly induced by the gasdermin (GSDM) family and is accompanied by the release of inflammatory cytokines such as IL-1β and IL-18. Recently, increasing evidence suggests that pyroptosis plays a role in respiratory diseases. This review aimed to summarize the roles and mechanisms of pyroptosis in inflammation-related respiratory diseases. There are several pathways involved in pyroptosis, such as the canonical inflammasome-induced pathway, non-canonical inflammasome-induced pathway, caspase-1/3/6/7/GSDMB pathway, caspase-8/GSDMC pathway, caspase-8/GSDMD pathway, and caspase-3/GSEME pathway. Pyroptosis may be involved in asthma, chronic obstructive pulmonary disease (COPD), lung cancer, acute lung injury (ALI), silicosis, pulmonary hypertension (PH), and tuberculosis (TB), in which the NLRP3 inflammasome-induced pathway is mostly highlighted. Pyroptosis contributes to the deterioration of asthma, COPD, ALI, silicosis, and PH. In addition, pyroptosis has dual effects on lung cancer and TB. Additionally, whether pyroptosis participates in cystic fibrosis (CF) and sarcoidosis or not is largely unknown, though the activation of NLRP3 inflammasome is found in CF and sarcoidosis. In conclusion, pyroptosis may play a role in inflammation-related respiratory diseases, providing new therapeutic targets.
Collapse
Affiliation(s)
- Yuanyu Feng
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, China
- Clinical Medicine, Innovation Class, 2019 Grade, Kunming Medical University, Kunming, China
| | - Min Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, China
| | - Xiaoting Yangzhong
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, China
- Pediatrics, One Class, 2020 Grade, Kunming Medical University, Kunming, China
| | - Xifeng Zhang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, China
| | - Anju Zu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, China
| | - Yunjiao Hou
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, China
| | - Lin Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, No.295, Xichang Road, Wuhua District, Kunming, China.
| |
Collapse
|
24
|
Chai Q, Yu S, Zhong Y, Lu Z, Qiu C, Yu Y, Zhang X, Zhang Y, Lei Z, Qiang L, Li BX, Pang Y, Qiu XB, Wang J, Liu CH. A bacterial phospholipid phosphatase inhibits host pyroptosis by hijacking ubiquitin. Science 2022; 378:eabq0132. [PMID: 36227980 DOI: 10.1126/science.abq0132] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The inflammasome-mediated cleavage of gasdermin D (GSDMD) causes pyroptosis and inflammatory cytokine release to control pathogen infection, but how pathogens evade this immune response remains largely unexplored. Here we identify the known protein phosphatase PtpB from Mycobacterium tuberculosis as a phospholipid phosphatase inhibiting the host inflammasome-pyroptosis pathway. Mechanistically, PtpB dephosphorylated phosphatidylinositol-4-monophosphate and phosphatidylinositol-(4,5)-bisphosphate in host cell membrane, thus disrupting the membrane localization of the cleaved GSDMD to inhibit cytokine release and pyroptosis of macrophages. Notably, this phosphatase activity requires PtpB binding to ubiquitin. Disrupting phospholipid phosphatase activity or the ubiquitin-interacting motif of PtpB enhanced host GSDMD-dependent immune responses and reduced intracellular pathogen survival. Thus, pathogens inhibit pyroptosis and counteract host immunity by altering host membrane composition.
Collapse
Affiliation(s)
- Qiyao Chai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shanshan Yu
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Yanzhao Zhong
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Zhe Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Changgen Qiu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yang Yu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xinwen Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yong Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zehui Lei
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Lihua Qiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Bing-Xi Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yu Pang
- Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, China
| | - Xiao-Bo Qiu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.,Ministry of Education Key Laboratory of Cell Proliferation and Regulation Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China
| | - Jing Wang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.,Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
25
|
Uchiyama A, Kosaka K, Okada K, Motegi SI. Possible suppressive effects of tranilast on NLRP3 inflammasome activation in necrobiosis lipoidica. J Dermatol 2022; 49:e433-e434. [PMID: 35848097 DOI: 10.1111/1346-8138.16521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/04/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Akihiko Uchiyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Keiji Kosaka
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan.,Kiryu Kosei General Hospital, Kiryu, Gunma, Japan
| | | | - Sei-Ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| |
Collapse
|
26
|
Neutrophils and Asthma. Diagnostics (Basel) 2022; 12:diagnostics12051175. [PMID: 35626330 PMCID: PMC9140072 DOI: 10.3390/diagnostics12051175] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/04/2023] Open
Abstract
Although eosinophilic inflammation is characteristic of asthma pathogenesis, neutrophilic inflammation is also marked, and eosinophils and neutrophils can coexist in some cases. Based on the proportion of sputum cell differentiation, asthma is classified into eosinophilic asthma, neutrophilic asthma, neutrophilic and eosinophilic asthma, and paucigranulocytic asthma. Classification by bronchoalveolar lavage is also performed. Eosinophilic asthma accounts for most severe asthma cases, but neutrophilic asthma or a mixture of the two types can also present a severe phenotype. Biomarkers for the diagnosis of neutrophilic asthma include sputum neutrophils, blood neutrophils, chitinase-3-like protein, and hydrogen sulfide in sputum and serum. Thymic stromal lymphoprotein (TSLP)/T-helper 17 pathways, bacterial colonization/microbiome, neutrophil extracellular traps, and activation of nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 pathways are involved in the pathophysiology of neutrophilic asthma and coexistence of obesity, gastroesophageal reflux disease, and habitual cigarette smoking have been associated with its pathogenesis. Thus, targeting neutrophilic asthma is important. Smoking cessation, neutrophil-targeting treatments, and biologics have been tested as treatments for severe asthma, but most clinical studies have not focused on neutrophilic asthma. Phosphodiesterase inhibitors, anti-TSLP antibodies, azithromycin, and anti-cholinergic agents are promising drugs for neutrophilic asthma. However, clinical research targeting neutrophilic inflammation is required to elucidate the optimal treatment.
Collapse
|
27
|
Liu Y, Chen S, Yu L, Deng Y, Li D, Yu X, Chen D, Lu Y, Liu S, Chen R. Pemafibrate attenuates pulmonary fibrosis by inhibiting myofibroblast differentiation. Int Immunopharmacol 2022; 108:108728. [PMID: 35397395 DOI: 10.1016/j.intimp.2022.108728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/26/2022] [Accepted: 03/18/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND OBJECTIVE Idiopathic pulmonary fibrosis is a chronic progressive disease associated with substantial morbidity and mortality despite advances in medical therapy. Increasing evidence suggests that peroxisome proliferator-activated receptors (PPARs) play important roles in the fibrosis-related diseases and their agonists may become effective therapeutic targets. Pemafibrate is a selective PPARα agonist, but the efficacy against pulmonary fibrosis and mechanisms involved have not been systematically evaluated. Thus, the aims of this study were to explore the role of PPARα in the pulmonary fibrosis and to assess the effect of pemafibrate in vivo and in vitro. METHODS The effects of pemafibrate were evaluated in bleomycin-challenged murine pulmonary fibrosis model and transforming growth factor-beta 1 (TGF-β1) stimulated lung fibroblasts. RESULTS Bleomycin instillation induced body weight loss, declined lung function, pulmonary fibrosis, and extensive collagen deposition in the mice, accompanied with decreased pulmonary expression of PPARα, all of which were partially improved by pemafibrate at a dose of 2 mg/kg. Besides, pemafibrate effectively inhibits TGF-β1-induced myofibroblast differentiation and extracellular matrix (ECM) production in vivo and in vitro. Furthermore, we showed that pemafibrate not only inhibited pulmonary expression of NLRP3 and cleaved caspase-1 in bleomycin-inhaled mice, but also repressed activation of NLRP3/caspase-1 axis in TGF-β1 stimulated lung fibroblasts. CONCLUSION Our data suggest that pemafibrate exerts a marked protection against from the development of pulmonary fibrosis, which could constitute a novel candidate for the treatment for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China; Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shuyu Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China; Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Li Yu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Yao Deng
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Difei Li
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Xiu Yu
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Dandan Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Ye Lu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shengming Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), The First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China.
| |
Collapse
|
28
|
Zohny MH, Cavalu S, Youssef ME, Kaddah MM, Mourad AA, Gaafar AGA, El-Ahwany E, Amin NA, Arakeep HM, Shata A, Saleh S, Hafez MM, Elazab ST, Abdelhady R, El Shahat RM, Yahya G, Saber S. Coomassie brilliant blue G-250 dye attenuates bleomycin-induced lung fibrosis by regulating the NF-κB and NLRP3 crosstalk: A novel approach for filling an unmet medical need. Pharmacotherapy 2022; 148:112723. [DOI: 10.1016/j.biopha.2022.112723] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 02/06/2023]
|
29
|
Randhawa KS, Lee BW, Micali G, Benson BE, Schwartz RA. Cutaneous sarcoidosis: Lupus pernio and more. Ital J Dermatol Venerol 2022; 157:220-227. [PMID: 35274876 DOI: 10.23736/s2784-8671.21.07027-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Sarcoidosis is a multi-organ disease commonly evident with skin involvement. Cutaneous manifestations occur in about 25% of sarcoid patients and are of two types: histologically specific sarcoidal infiltrations and a cutaneous reaction pattern not containing sarcoidal changes, usually erythema nodosum. Cutaneous plaques, nodules, and tumors, sometimes with disfiguring facial features associated with pain and paresthesia. The disease itself may produce substantial morbidity due to visceral involvement. Advances in therapeutic options include tocilizumab, an IL-6 inhibitor, and tofacitinib, a Janus kinase inhibitor. This review discusses sarcoidosis etiology and pathogenesis, its clinical features, differential diagnosis, and management.
Collapse
Affiliation(s)
| | - Brian W Lee
- Rutgers-New Jersey Medical School, Newark, NJ, USA
| | | | | | | |
Collapse
|
30
|
Affiliation(s)
- Marjolein Drent
- From the Interstitial Lung Diseases (ILD) Center of Excellence, Department of Pulmonology, St. Antonius Hospital, Nieuwegein, the Department of Pharmacology and Toxicology, Faculty of Health and Life Sciences, Maastricht University, Maastricht, and the ILD Care Foundation Research Team, Ede - all in the Netherlands (M.D.); the Division of Pulmonary, Critical Care, and Sleep Medicine, Ohio State University, Columbus (E.D.C.); and the Respiratory Medicine Division, Department of Medicine Solna, and the Center for Molecular Medicine, Karolinska Institutet, and Respiratory Medicine, Theme Inflammation and Aging, Karolinska University Hospital - both in Stockholm (J.G.)
| | - Elliott D Crouser
- From the Interstitial Lung Diseases (ILD) Center of Excellence, Department of Pulmonology, St. Antonius Hospital, Nieuwegein, the Department of Pharmacology and Toxicology, Faculty of Health and Life Sciences, Maastricht University, Maastricht, and the ILD Care Foundation Research Team, Ede - all in the Netherlands (M.D.); the Division of Pulmonary, Critical Care, and Sleep Medicine, Ohio State University, Columbus (E.D.C.); and the Respiratory Medicine Division, Department of Medicine Solna, and the Center for Molecular Medicine, Karolinska Institutet, and Respiratory Medicine, Theme Inflammation and Aging, Karolinska University Hospital - both in Stockholm (J.G.)
| | - Johan Grunewald
- From the Interstitial Lung Diseases (ILD) Center of Excellence, Department of Pulmonology, St. Antonius Hospital, Nieuwegein, the Department of Pharmacology and Toxicology, Faculty of Health and Life Sciences, Maastricht University, Maastricht, and the ILD Care Foundation Research Team, Ede - all in the Netherlands (M.D.); the Division of Pulmonary, Critical Care, and Sleep Medicine, Ohio State University, Columbus (E.D.C.); and the Respiratory Medicine Division, Department of Medicine Solna, and the Center for Molecular Medicine, Karolinska Institutet, and Respiratory Medicine, Theme Inflammation and Aging, Karolinska University Hospital - both in Stockholm (J.G.)
| |
Collapse
|
31
|
Jeny F, Bernaudin JF, Valeyre D, Kambouchner M, Pretolani M, Nunes H, Planès C, Besnard V. Hypoxia Promotes a Mixed Inflammatory-Fibrotic Macrophages Phenotype in Active Sarcoidosis. Front Immunol 2021; 12:719009. [PMID: 34456926 PMCID: PMC8385772 DOI: 10.3389/fimmu.2021.719009] [Citation(s) in RCA: 21] [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: 06/01/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022] Open
Abstract
Background Macrophages are pivotal cells in sarcoidosis. Monocytes-derived (MD) macrophages have recently been demonstrated to play a major role especially in pulmonary sarcoidosis. From inflammatory tissues to granulomas, they may be exposed to low oxygen tension environments. As hypoxia impact on sarcoidosis immune cells has never been addressed, we designed the present study to investigate MD-macrophages from sarcoidosis patients in this context. We hypothesized that hypoxia may induce functional changes on MD-macrophages which could have a potential impact on the course of sarcoidosis. Methods We studied MD-macrophages, from high active sarcoidosis (AS) (n=26), low active or inactive sarcoidosis (IS) (n=24) and healthy controls (n=34) exposed 24 hours to normoxia (21% O2) or hypoxia (1.5% O2). Different macrophage functions were explored: hypoxia-inducible factor-1α (HIF-1α) and nuclear factor-kappa B (NF-κB) activation, cytokines secretion, phagocytosis, CD80/CD86/HLA-DR expression, profibrotic response. Results We observed that hypoxia, with a significantly more pronounced effect in AS compared with controls and IS, increased the HIF-1α trans-activity, promoted a proinflammatory response (TNFα, IL1ß) without activating NF-κB pathway and a profibrotic response (TGFß1, PDGF-BB) with PAI-1 secretion associated with human lung fibroblast migration inhibition. These results were confirmed by immunodetection of HIF-1α and PAI-1 in granulomas observed in pulmonary biopsies from patients with sarcoidosis. Hypoxia also decreased the expression of CD80/CD86 and HLA-DR on MD-macrophages in the three groups while it did not impair phagocytosis and the expression of CD36 expression on cells in AS and IS at variance with controls. Conclusions Hypoxia had a significant impact on MD-macrophages from sarcoidosis patients, with the strongest effect seen in patients with high active disease. Therefore, hypoxia could play a significant role in sarcoidosis pathogenesis by increasing the macrophage proinflammatory response, maintaining phagocytosis and reducing antigen presentation, leading to a deficient T cell response. In addition, hypoxia could favor fibrosis by promoting profibrotic cytokines response and by sequestering fibroblasts in the vicinity of granulomas.
Collapse
Affiliation(s)
- Florence Jeny
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Pulmonology Department, Avicenne Hospital, Bobigny, France
| | - Jean-François Bernaudin
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- Faculty of Medicine, Sorbonne University, Paris, France
| | - Dominique Valeyre
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Pulmonology Department, Avicenne Hospital, Bobigny, France
| | - Marianne Kambouchner
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Pathology Department, Avicenne Hospital, Bobigny, France
| | - Marina Pretolani
- Inserm UMR1152, Physiopathology and Epidemiology of Respiratory Diseases, Paris, France
- Faculty of Medicine, Bichat Hospital, Paris University, Paris, France
- Laboratory of Excellence, INFLAMEX, Paris University, DHU FIRE, Paris, France
| | - Hilario Nunes
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Pulmonology Department, Avicenne Hospital, Bobigny, France
| | - Carole Planès
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
- AP-HP, Physiology Department, Avicenne Hospital, Bobigny, France
| | - Valérie Besnard
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, France
| |
Collapse
|
32
|
Belperio JA, Shaikh F, Abtin F, Fishbein MC, Saggar R, Tsui E, Lynch JP. Extrapulmonary sarcoidosis with a focus on cardiac, nervous system, and ocular involvement. EClinicalMedicine 2021; 37:100966. [PMID: 34258571 PMCID: PMC8254127 DOI: 10.1016/j.eclinm.2021.100966] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023] Open
Abstract
Sarcoidosis is a poorly understood granulomatous disease that involves the lungs and/or intrathoracic lymph nodes in more than 90% of cases. Although pulmonary sarcoidosis is the leading cause of mortality in this disease, this review focuses on three sites of extrapulmonary involvement (heart, nervous system, and eyes), since involvement of any of these sites can be catastrophic, leading to death, debilitation, or blindness. Patients with cardiac, ocular and neurosarcoidosis necessitate a multidisciplinary approach with careful and long-term follow-up. Prompt diagnosis with imaging and/or biopsy and treatment is required to avoid irreversible damage. Corticosteroids are the mainstay of therapy and are often associated with rapid and durable remissions. Immunosuppressive or biologic agents are reserved for patients failing or experiencing side effects from steroids. Managing sarcoidosis requires vigilance, judgement, and awareness of the vagaries of this fascinating disease.
Collapse
Affiliation(s)
- John A. Belperio
- The Division of Pulmonary and Critical Care Medicine, Holt and Jo Hickman Endowed Chair of Advanced Lung Disease and Lung Transplantation, Clinical Immunology, and Allergy, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 37-131 CHS, Los Angeles, CA 90095, United States
| | - Faisal Shaikh
- The Division of Pulmonary and Critical Care Medicine, Holt and Jo Hickman Endowed Chair of Advanced Lung Disease and Lung Transplantation, Clinical Immunology, and Allergy, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 37-131 CHS, Los Angeles, CA 90095, United States
| | - Fereidoun Abtin
- Department of Radiology, Thoracic and Interventional Section, David Geffen School of Medicine at UCLA, United States
| | - Michael C. Fishbein
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, United States
| | - Rajan Saggar
- The Division of Pulmonary and Critical Care Medicine, Holt and Jo Hickman Endowed Chair of Advanced Lung Disease and Lung Transplantation, Clinical Immunology, and Allergy, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 37-131 CHS, Los Angeles, CA 90095, United States
| | - Edmund Tsui
- Department of Ophthalmology, David Geffen School of Medicine at UCLA, United States
| | - Joseph P. Lynch
- The Division of Pulmonary and Critical Care Medicine, Holt and Jo Hickman Endowed Chair of Advanced Lung Disease and Lung Transplantation, Clinical Immunology, and Allergy, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave, Room 37-131 CHS, Los Angeles, CA 90095, United States
| |
Collapse
|
33
|
Corcoran SE, Halai R, Cooper MA. Pharmacological Inhibition of the Nod-Like Receptor Family Pyrin Domain Containing 3 Inflammasome with MCC950. Pharmacol Rev 2021; 73:968-1000. [PMID: 34117094 DOI: 10.1124/pharmrev.120.000171] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Activation of the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome drives release of the proinflammatory cytokines interleukin (IL)-1β and IL-18 and induces pyroptosis (lytic cell death). These events drive chronic inflammation, and as such, NLRP3 has been implicated in a large number of human diseases. These range from autoimmune conditions, the simplest of which is NLRP3 gain-of-function mutations leading to an orphan disease, cryopyrin-associated period syndrome, to large disease burden indications, such as atherosclerosis, heart failure, stroke, neurodegeneration, asthma, ulcerative colitis, and arthritis. The potential clinical utility of NLRP3 inhibitors is substantiated by an expanding list of indications in which NLRP3 activation has been shown to play a detrimental role. Studies of pharmacological inhibition of NLRP3 in nonclinical models of disease using MCC950 in combination with human genetics, epigenetics, and analyses of the efficacy of biologic inhibitors of IL-1β, such as anakinra and canakinumab, can help to prioritize clinical trials of NLRP3-directed therapeutics. Although MCC950 shows excellent (nanomolar) potency and high target selectivity, its pharmacokinetic and toxicokinetic properties limited its therapeutic development in the clinic. Several improved, next-generation inhibitors are now in clinical trials. Hence the body of research in a plethora of conditions reviewed herein may inform analysis of the potential translational value of NLRP3 inhibition in diseases with significant unmet medical need. SIGNIFICANCE STATEMENT: The nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome is one of the most widely studied and best validated biological targets in innate immunity. Activation of NLRP3 can be inhibited with MCC950, resulting in efficacy in more than 100 nonclinical models of inflammatory diseases. As several next-generation NLRP3 inhibitors are entering proof-of-concept clinical trials in 2020, a review of the pharmacology of MCC950 is timely and significant.
Collapse
Affiliation(s)
- Sarah E Corcoran
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| | - Reena Halai
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| | - Matthew A Cooper
- Trinity College Dublin, Dublin, Ireland (S.E.C.); Inflazome, D6 Grain House, Mill Court, Great Shelford, Cambridge, United Kingdom (R.H., M.A.C.); and Institute for Molecular Bioscience, University of Queensland, Queensland, Australia (M.A.C.)
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Sarcoidosis remains a mysterious disease that presents many challenges both in pathogenetic understanding and in the management of patients. This review presents experimental models for sarcoidosis developed since 2016 and discusses their strengths and weaknesses and how they have contributed to the understanding and therapeutic approaches in this disease. In addition, future directions are proposed to overcome the limitations of current models. RECENT FINDINGS New cellular models using infectious antigen as trigger, and transgenic models in mice have recently been developed to study signaling pathways potentially implicated in the pathogenesis of sarcoidosis. SUMMARY No model fully reproduces sarcoidosis, but most of them generate data supporting key concepts and some open up therapeutic perspectives, like mTOR inhibition or IL-1β blocking. However, there are still many limitations to these models, largely related to the complexity of sarcoidosis, which might be overcome with new technologies, such as mathematical modeling.
Collapse
|
35
|
Proteome Characterization of BALF Extracellular Vesicles in Idiopathic Pulmonary Fibrosis: Unveiling Undercover Molecular Pathways. Int J Mol Sci 2021; 22:ijms22115696. [PMID: 34071777 PMCID: PMC8199247 DOI: 10.3390/ijms22115696] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/24/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
In the longtime challenge of identifying specific, easily detectable and reliable biomarkers of IPF, BALF proteomics is providing interesting new insights into its pathogenesis. To the best of our knowledge, the present study is the first shotgun proteomic investigation of EVs isolated from BALF of IPF patients. Our main aim was to characterize the proteome of the vesicular component of BALF and to explore its individual impact on the pathogenesis of IPF. To this purpose, ultracentrifugation was chosen as the EVs isolation technique, and their purification was assessed by TEM, 2DE and LC-MS/MS. Our 2DE data and scatter plots showed considerable differences between the proteome of EVs and that of whole BALF and of its fluid component. Analysis of protein content and protein functions evidenced that EV proteins are predominantly involved in cytoskeleton remodeling, adenosine signaling, adrenergic signaling, C-peptide signaling and lipid metabolism. Our findings may suggest a wider system involvement in the disease pathogenesis and support the importance of pre-fractioning of complex samples, such as BALF, in order to let low-abundant proteins-mediated pathways emerge.
Collapse
|
36
|
Barna BP, Judson MA, Thomassen MJ. Inflammatory Pathways in Sarcoidosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:39-52. [PMID: 34019262 DOI: 10.1007/978-3-030-68748-9_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Concepts regarding etiology and pathophysiology of sarcoidosis have changed remarkably within the past 5 years. Sarcoidosis is now viewed as a complex multi-causation disease related to a diverse collection of external environmental or infectious signals. It is generally accepted that the cause of sarcoidosis is unknown. Moreover, concepts of the inflammatory pathway have been modified by the realization that intrinsic genetic factors and innate immunity may modify adaptive immune responses to external triggers. With those potential regulatory pathways in mind, we will attempt to discuss the current understanding of the inflammatory response in sarcoidosis with emphasis on development of pulmonary granulomatous pathology. In that context, we will emphasize that both macrophages and T lymphocytes play key roles, with sometimes overlapping cytokine production (i.e., TNFα and IFN-γ) but also with unique mediators that influence the pathologic picture. Numerous studies have shown that in a sizable number of sarcoidosis patients, granulomas spontaneously resolve, usually within 3 years. Other sarcoidosis patients, however, may develop a chronic granulomatous disease which may subsequently lead to fibrosis. This chapter will outline our current understanding of inflammatory pathways in sarcoidosis which initiate and mediate granulomatous changes or onset of pulmonary fibrosis.
Collapse
Affiliation(s)
- Barbara P Barna
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary and Critical Care Medicine, East Carolina University, Greenville, NC, USA
| | - Marc A Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Mary Jane Thomassen
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary and Critical Care Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
37
|
Jäger B, Seeliger B, Terwolbeck O, Warnecke G, Welte T, Müller M, Bode C, Prasse A. The NLRP3-Inflammasome-Caspase-1 Pathway Is Upregulated in Idiopathic Pulmonary Fibrosis and Acute Exacerbations and Is Inducible by Apoptotic A549 Cells. Front Immunol 2021; 12:642855. [PMID: 33968032 PMCID: PMC8104027 DOI: 10.3389/fimmu.2021.642855] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a relentlessly progressive disease harboring significant morbidity and mortality despite recent advances in therapy. Regardless of disease severity acute exacerbations (IPF-AEs) may occur leading to considerable loss of function and are the leading cause of death in IPF. Histologic features of IPF-AE are very similar to acute respiratory distress syndrome (ARDS), but the underlying mechanisms are incompletely understood. We investigated the role of the NLRP3 inflammasome in IPF and IPF-AE. Bronchoalveolar lavage (BAL) cells were sampled from patients with IPF (n = 32), IPF-AE (n = 10), ARDS (n = 7) and healthy volunteers (HV, n = 37) and the NLRP3-inflammasome was stimulated in-vitro. We found the NLRP3 inflammasome to be hyper-inducible in IPF compared to HV with increased IL-1ß and pro-IL-1ß levels on ELISA upon stimulation as well as increased caspase-1 activity measured by caspase-1p20 immunoblotting. In IPF-AE, IL-1ß was massively elevated to an extent similar to ARDS. To evaluate potential mechanisms, we co-cultured BAL cells with radiated A549 cells (a model to simulate apoptotic alveolar epithelial cells), which led to increased NLRP3 mRNA expression and increased caspase-1 dependent IL-1ß production. In the presence of a reactive oxygen species (ROS) inhibitor (diphenyleneiodonium) and a cathepsin B inhibitor (E64D), NLRP3 expression was suppressed indicating that induction of NLRP3 activation following efferocytosis of apoptotic A549 cells is mediated via ROS and cathepsin-B. In summary, we present evidence of involvement of the NLRP3 inflammasome-caspase pathway in the pathogenesis of IPF-AE, similarly to ARDS, which may be mediated by efferocytosis of apoptotic alveolar epithelial cells in IPF.
Collapse
Affiliation(s)
- Benedikt Jäger
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.,Department of Respiratory Medicine, University Medical Center, Freiburg, Germany.,Faculty of Biology, Albert Ludwig University, Freiburg, Germany
| | - Benjamin Seeliger
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Oliver Terwolbeck
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Gregor Warnecke
- Department of Heart, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| | - Meike Müller
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Bonn, Germany
| | - Antje Prasse
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany.,Department of Respiratory Medicine, University Medical Center, Freiburg, Germany.,Department of Respiratory Medicine, Hannover Medical School and Biomedical Research in End-stage and Obstructive Lung Disease (BREATH), German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
38
|
Minasyan M, Sharma L, Pivarnik T, Liu W, Adams T, Bermejo S, Peng X, Liu A, Ishikawa G, Perry C, Kaminski N, Gulati M, Herzog EL, Dela Cruz CS, Ryu C. Elevated IL-15 concentrations in the sarcoidosis lung are independent of granuloma burden and disease phenotypes. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1137-L1146. [PMID: 33851886 DOI: 10.1152/ajplung.00575.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Sarcoidosis is a systemic granulomatous disease predominantly affecting the lungs. The mechanisms promoting disease pathogenesis and progression are unknown, although interleukin-15 (IL-15) has been associated with the immune-mediated inflammation of sarcoidosis. Because the identification of a mechanistically based, clinically relevant biomarker for sarcoidosis remains elusive, we hypothesized this role for IL-15. Pulmonary sarcoidosis granuloma formation was modeled using trehalose 6,6'-dimicolate (TDM), which was administered into wild-type and three lineages of mice: those overexpressing IL-15, deficient in IL-15, and deficient in IL-15 receptor α. The number of granulomas per lung was counted and normalized to the wild type. IL-15 concentrations were measured in the bronchoalveolar lavage (BAL) from healthy controls and subjects with sarcoidosis in our cohort, where associations between IL-15 levels and clinical manifestations were sought. Findings were validated in another independent sarcoidosis cohort. TDM administration resulted in similar granuloma numbers across all lineages of mice. IL-15 concentrations were elevated in the BAL of both human cohorts, irrespective of disease phenotypes. In exploratory analysis, an association with obesity was observed, and various other soluble mediators were identified in the BAL of both cohorts. Although IL-15 is enriched in the sarcoidosis lung, it was independent of disease pathogenesis or clinical manifestations in our mouse model and human cohorts of sarcoidosis. An association with obesity perhaps reflects the ongoing inflammatory processes of these comorbid conditions. Our findings showed that IL-15 is redundant for disease pathogenesis and clinical progression of sarcoidosis.
Collapse
Affiliation(s)
- Maksym Minasyan
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Taylor Pivarnik
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Wei Liu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Taylor Adams
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Santos Bermejo
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Xiaohua Peng
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Angela Liu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Carrighan Perry
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Mridu Gulati
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Erica L Herzog
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Changwan Ryu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| |
Collapse
|
39
|
Weber ANR. Targeting the NLRP3 Inflammasome via BTK. Front Cell Dev Biol 2021; 9:630479. [PMID: 33718366 PMCID: PMC7947255 DOI: 10.3389/fcell.2021.630479] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/14/2021] [Indexed: 12/28/2022] Open
Abstract
The NLRP3 inflammasome represents a critical inflammatory machinery driving pathology in many acute (e. g., myocardial infarction or stroke) and chronic (Alzheimer's disease, atherosclerosis) human disorders linked to the activity of IL-1 cytokines. Although the therapeutic potential of NLRP3 is undisputed, currently no clinically approved therapies exist to target the NLRP3 inflammasome directly. The recent discovery of BTK as a direct and positive regulator of the NLRP3 inflammasome has, however, raised the intriguing possibility of targeting the NLRP3 inflammasome via existing or future BTK inhibitors. Here, I review the mechanistic basis for this notion and discuss the molecular and cellular role of BTK in the inflammasome process. Specific attention will be given to cell-type dependent characteristics and differences that may be relevant for targeting approaches. Furthermore, I review recent (pre-)clinical evidence for effects of BTK inhibitors on NLRP3 activity and highlight and discuss open questions and future research directions. Collectively, the concept of targeting BTK to target NLRP3-dependent inflammation will be explored comprehensively at the molecular, cellular and therapeutic levels.
Collapse
Affiliation(s)
- Alexander N. R. Weber
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
- iFIT – Cluster of Excellence (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- CMFI – Cluster of Excellence (EXC 2124) “Controlling Microbes to Fight Infection”, University of Tübingen, Tübingen, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK; German Cancer Consortium), Partner Site Tübingen, Department of Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
40
|
Chandra S, Ehrlich KC, Lacey M, Baribault C, Ehrlich M. Epigenetics and expression of key genes associated with cardiac fibrosis: NLRP3, MMP2, MMP9, CCN2/CTGF and AGT. Epigenomics 2021; 13:219-234. [PMID: 33538177 PMCID: PMC7907962 DOI: 10.2217/epi-2020-0446] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Aims: Excessive inflammatory signaling and pathological remodeling of the extracellular matrix drive cardiac fibrosis and require changes in gene expression. Materials and methods: Using bioinformatics, both tissue-specific expression profiles and epigenomic profiles of some genes critical for cardiac fibrosis were examined, namely, NLRP3, MMP2, MMP9, CCN2/CTGF, AGT (encodes angiotensin II precursors) and hsa-mir-223 (post-transcriptionally regulates NLRP3). Results: In monocytes, neutrophils, fibroblasts, venous cells, liver and brain, enhancers or super-enhancers were found that correlate with high expression of these genes. One enhancer extended into a silent gene neighbor. These enhancers harbored tissue-specific foci of DNA hypomethylation, open chromatin and transcription factor binding. Conclusions: This study identified previously undescribed enhancers containing hypomethylated transcription factor binding subregions that are predicted to regulate expression of these cardiac fibrosis-inducing genes.
Collapse
Affiliation(s)
- Sruti Chandra
- Tulane Research Innovation for Arrhythmia Discoveries, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Kenneth C Ehrlich
- Tulane Center for Biomedical Informatics & Genomics, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Michelle Lacey
- Department of Mathematics, Tulane University, New Orleans, LA, 70112, USA.,Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Carl Baribault
- Center for Research & Scientific Computing, Tulane University Information Technology, New Orleans, LA, 70112, USA
| | - Melanie Ehrlich
- Tulane Center for Biomedical Informatics & Genomics, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA.,Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA.,Hayward Genetics Center, Tulane University Health Sciences Center, New Orleans, LA, 70112, USA
| |
Collapse
|
41
|
Obi ON, Lower EE, Baughman RP. Biologic and advanced immunomodulating therapeutic options for sarcoidosis: a clinical update. Expert Rev Clin Pharmacol 2021; 14:179-210. [PMID: 33487042 DOI: 10.1080/17512433.2021.1878024] [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] [Indexed: 12/15/2022]
Abstract
Introduction: Sarcoidosis is a multi-organ disease with a wide range of clinical manifestations and outcomes. A quarter of sarcoidosis patients require long-term treatment for chronic disease. In this group, corticosteroids and cytotoxic agents be insufficient to control diseaseAreas covered: Several biologic agents have been studied for treatment of chronic pulmonary and extra-pulmonary disease. A review of the available literature was performed searching PubMed and an expert opinion regarding specific therapy was developed.Expert opinion: These agents have the potential of treating patients who have progressive disease. Many of these agents have different mechanisms of action, response rates, and toxicity profiles.
Collapse
Affiliation(s)
- Ogugua Ndili Obi
- Division of Pulmonary Critical Care and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Elyse E Lower
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Robert P Baughman
- Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
42
|
Valeyre D, Bernaudin JF. Management of Sarcoidosis, a Selection of Topical Items Updating. J Clin Med 2020; 9:jcm9103220. [PMID: 33036457 PMCID: PMC7599542 DOI: 10.3390/jcm9103220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/01/2022] Open
Affiliation(s)
- Dominique Valeyre
- UMR INSERM 1272 Université Sorbonne Paris Nord, 93000 Bobigny, France;
- APHP Hôpital Avicenne, 93000 Bobigny, France
- Groupe Hospitalier Paris-Saint Joseph, 75014 Paris, France
- Correspondence:
| | - Jean-François Bernaudin
- UMR INSERM 1272 Université Sorbonne Paris Nord, 93000 Bobigny, France;
- APHP Hôpital Avicenne, 93000 Bobigny, France
- Faculté de Médecine, Sorbonne Université, 75013 Paris, France
| |
Collapse
|
43
|
Current perspectives on the immunopathogenesis of sarcoidosis. Respir Med 2020; 173:106161. [PMID: 32992264 DOI: 10.1016/j.rmed.2020.106161] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022]
Abstract
Sarcoidosis is an inflammatory systemic disease that commonly affects the lungs or lymph nodes but can manifest in other organs. Herein, we review the latest evidence establishing how innate and adaptive immune responses contribute to the pathogenesis and clinical course of sarcoidosis. We discuss the possible role of microbial organisms as etiologic agents in sarcoidosis and the evidence supporting sarcoidosis as an autoimmune disease. We also discuss how animal and in vitro human models have advanced our understanding of the immunopathogenesis of sarcoidosis. Finally, we discuss therapeutics for sarcoidosis and the effects on the immune system.
Collapse
|
44
|
Garman L, Montgomery CG, Rivera NV. Recent advances in sarcoidosis genomics: epigenetics, gene expression, and gene by environment (G × E) interaction studies. Curr Opin Pulm Med 2020; 26:544-553. [PMID: 32701681 PMCID: PMC7735660 DOI: 10.1097/mcp.0000000000000719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW We aim to review the most recent findings in genomics of sarcoidosis and highlight the gaps in the field. RECENT FINDINGS Original explorations of sarcoidosis subphenotypes, including cases associated with the World Trade Center and ocular sarcoidosis, have identified novel risk loci. Innovative gene--environment interaction studies utilizing modern analytical techniques have discovered risk loci associated with smoking and insecticide exposure. The application of whole-exome sequencing has identified genetic variants associated with persistent sarcoidosis and rare functional variations. A single epigenomics study has provided background knowledge of DNA methylation mechanisms in comparison with gene expression data. The application of machine-learning techniques has suggested new drug repositioning for the treatment of sarcoidosis. Several gene expression studies have identified prominent inflammatory pathways enriched in the affected tissue. SUMMARY Certainly, sarcoidosis research has recently advanced in the exploration of disease subphenotypes, utilizing novel analytical techniques, and including measures of clinical variation. Nevertheless, large-scale and diverse cohorts investigated with advanced sequencing methods, such as whole-genome and single-cell RNA sequencing, epigenomics, and meta-analysis coupled with cutting-edge analytic approaches, when employed, will broaden and translate genomics findings into clinical applications, and ultimately open venues for personalized medicine.
Collapse
Affiliation(s)
- Lori Garman
- Department of Genes and Human Disease, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Courtney G. Montgomery
- Department of Genes and Human Disease, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Natalia V. Rivera
- Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Rheumatology Division, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine (CMM), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
45
|
Derlin T, Jaeger B, Jonigk D, Apel RM, Freise J, Shin HO, Weiberg D, Warnecke G, Ross TL, Wester HJ, Seeliger B, Welte T, Bengel FM, Prasse A. Clinical Molecular Imaging of Pulmonary CXCR4 Expression to Predict Outcome of Pirfenidone Treatment in Idiopathic Pulmonary Fibrosis. Chest 2020; 159:1094-1106. [PMID: 32822674 DOI: 10.1016/j.chest.2020.08.2043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/03/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive disease for which two antifibrotic drugs recently were approved. However, an unmet need exists to predict responses to antifibrotic treatment, such as pirfenidone. Recent data suggest that upregulated expression of CXCR4 is indicative of outcomes in IPF. RESEARCH QUESTION Can quantitative, molecular imaging of pulmonary CXCR4 expression as a biomarker for disease activity predict response to the targeted treatment pirfenidone and prognosis in patients with IPF? STUDY DESIGN AND METHODS CXCR4 expression was analyzed by immunohistochemistry examination of lung tissues and reverse-transcriptase polymerase chain reaction analysis of BAL. PET-CT scanning with the specific CXCR4 ligand 68Ga-pentixafor was performed in 28 IPF patients and compared with baseline clinical characteristics. In 16 patients, a follow-up scan was obtained 6 to 12 weeks after initiation of treatment with pirfenidone. Patients were followed up in our outpatient clinic for ≥ 12 months. RESULTS Immunohistochemistry analysis showed high CXCR4 staining of epithelial cells and macrophages in areas with vast fibrotic remodeling. Targeted PET scanning revealed CXCR4 upregulation in fibrotic areas of the lungs, particularly in zones with subpleural honeycombing. Baseline CXCR4 signal demonstrated a significant correlation with Gender Age Physiology stage (r = 0.44; P = .02) and with high-resolution CT scan score (r = 0.38; P = .04). Early changes in CXCR4 signal after initiation of pirfenidone treatment correlated with the long-term course of FVC after 12 months (r = -0.75; P = .0008). Moreover, patients with a high pulmonary CXCR4 signal on follow-up PET scan after 6 weeks into treatment demonstrated a statistically significant worse outcome at 12 months (P = .002). In multiple regression analysis, pulmonary CXCR4 signal on follow-up PET scan emerged as the only independent predictor of long-term outcome (P = .0226). INTERPRETATION CXCR4-targeted PET imaging identified disease activity and predicted outcome of IPF patients treated with pirfenidone. It may serve as a future biomarker for personalized guidance of antifibrotic treatment.
Collapse
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Benedikt Jaeger
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Rosa M Apel
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Julia Freise
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Hoen-Oh Shin
- Institute of Radiology, Hannover Medical School, Hannover, Germany
| | - Desiree Weiberg
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Gregor Warnecke
- Department of Heart, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Hans-Jürgen Wester
- Institute of Radiopharmaceutical Chemistry, Technical University Munich, Garching, Germany
| | - Benjamin Seeliger
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Tobias Welte
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Antje Prasse
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; DZL-BREATH, Hannover, Germany.
| |
Collapse
|
46
|
Models Contribution to the Understanding of Sarcoidosis Pathogenesis: "Are There Good Models of Sarcoidosis?". J Clin Med 2020; 9:jcm9082445. [PMID: 32751786 PMCID: PMC7464295 DOI: 10.3390/jcm9082445] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 12/29/2022] Open
Abstract
Sarcoidosis is a systemic, granulomatous, and noninfectious disease of unknown etiology. The clinical heterogeneity of the disease (targeted tissue(s), course of the disease, and therapy response) supports the idea that a multiplicity of trigger antigens may be involved. The pathogenesis of sarcoidosis is not yet completely understood, although in recent years, considerable efforts were put to develop novel experimental research models of sarcoidosis. In particular, sarcoidosis patient cells were used within in vitro 3D models to study their characteristics compared to control patients. Likewise, a series of transgenic mouse models were developed to highlight the role of particular signaling pathways in granuloma formation and persistence. The purpose of this review is to put in perspective the contributions of the most recent models in the understanding of sarcoidosis.
Collapse
|
47
|
|
48
|
Yang HH, Duan JX, Liu SK, Xiong JB, Guan XX, Zhong WJ, Sun CC, Zhang CY, Luo XQ, Zhang YF, Chen P, Hammock BD, Hwang SH, Jiang JX, Zhou Y, Guan CX. A COX-2/sEH dual inhibitor PTUPB alleviates lipopolysaccharide-induced acute lung injury in mice by inhibiting NLRP3 inflammasome activation. Theranostics 2020; 10:4749-4761. [PMID: 32308747 PMCID: PMC7163435 DOI: 10.7150/thno.43108] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/08/2020] [Indexed: 01/11/2023] Open
Abstract
Rationale: Dysregulation of arachidonic acid (ARA) metabolism results in inflammation; however, its role in acute lung injury (ALI) remains elusive. In this study, we addressed the role of dysregulated ARA metabolism in cytochromes P450 (CYPs) /cyclooxygenase-2 (COX-2) pathways in the pathogenesis of lipopolysaccharide (LPS)-induced ALI in mice. Methods: The metabolism of CYPs/COX-2-derived ARA in the lungs of LPS-induced ALI was investigated in C57BL/6 mice. The COX-2/sEH dual inhibitor PTUPB was used to establish the function of CYPs/COX-2 dysregulation in ALI. Primary murine macrophages were used to evaluate the underlying mechanism of PTUPB involved in the activation of NLRP3 inflammasome in vitro. Results: Dysregulation of CYPs/COX-2 metabolism of ARA occurred in the lungs and in primary macrophages under the LPS challenge. Decrease mRNA expression of Cyp2j9, Cyp2j6, and Cyp2j5 was observed, which metabolize ARA into epoxyeicosatrienoic acids (EETs). The expressions of COX-2 and soluble epoxide hydrolase (sEH), on the other hand, was significantly upregulated. Pre-treatment with the dual COX-2 and sEH inhibitor, PTUPB, attenuated the pathological injury of lung tissues and reduced the infiltration of inflammatory cells. Furthermore, PTUPB decreased the pro-inflammatory factors, oxidative stress, and activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome in LPS-induced ALI mice. PTUPB pre-treatment remarkably reduced the activation of macrophages and NLRP3 inflammasome in vitro. Significantly, both preventive and therapeutic treatment with PTUPB improved the survival rate of mice receiving a lethal dose of LPS. Conclusion: The dysregulation of CYPs/COX-2 metabolized ARA contributes to the uncontrolled inflammatory response in ALI. The dual COX-2 and sEH inhibitor PTUPB exerts anti-inflammatory effects in treating ALI by inhibiting the NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Hui-Hui Yang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jia-Xi Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Shao-Kun Liu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Jian-Bing Xiong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xin-Xin Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Chen Sun
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xiao-Qin Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yan-Feng Zhang
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Ping Chen
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Hunan Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Bruce D. Hammock
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Sung Hee Hwang
- Department of Entomology and Nematology and UC Davis Comprehensive Cancer Center, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jian-Xin Jiang
- State Key Laboratory of Trauma, Burns, and Combined Injury, Army Medical University, Chongqing, 400038, China
| | - Yong Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
49
|
Riteau N, Bernaudin JF. In addition to mTOR and JAK/STAT, NLRP3 inflammasome is another key pathway activated in sarcoidosis. Eur Respir J 2020; 55:55/3/2000149. [PMID: 32217622 DOI: 10.1183/13993003.00149-2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/10/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Nicolas Riteau
- CNRS, INEM-UMR7355, University of Orleans, Orleans, France
| | - Jean-François Bernaudin
- Sorbonne Université, Paris, France.,INSERM UMR 1272 Université Paris 13, Bobigny, France.,Pneumology Dept, Hôpital Avicenne APHP, Bobigny, France
| |
Collapse
|