1
|
Saw PE, Song E. The 'inflammazone' in chronic inflammatory diseases: psoriasis and sarcoidosis. Trends Immunol 2025:S1471-4906(25)00002-X. [PMID: 39875239 DOI: 10.1016/j.it.2025.01.002] [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: 11/12/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
Chronic inflammatory diseases show significant heterogeneity in their phenotypes, with diverse immune cells and mediators interacting in response to various stimuli. This review proposes the concept of the 'inflammazone' framework - which maps the distribution of immune components driving disease pathogenesis - using sarcoidosis and psoriasis as examples. Sarcoidosis features granulomatous inflammation with macrophages and CD4+ T cells, which can spread to lymph nodes and other organs. Psoriasis, affecting primarily the skin, involves Th1, Th17, and Th22 pathways with CD8+ T cells and dendritic cells. Human sarcoidosis exhibits geographic and racial variability, while psoriasis shows varying morphologies and disease courses. Sarcoidosis has more extensive distal immune signaling, whereas psoriasis remains more localized. Understanding the inflammazone is crucial for advancing personalized treatments for inflammatory diseases.
Collapse
Affiliation(s)
- Phei Er Saw
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Nanhai Clinical Translational Center, Sun Yat-sen Memorial Hospital, Foshan, China; Department of General Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Erwei Song
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Nanhai Clinical Translational Center, Sun Yat-sen Memorial Hospital, Foshan, China; Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Zenith Institute of Medical Sciences, Guangzhou 510120, China.
| |
Collapse
|
2
|
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
|
3
|
Wang B, Cao X, Garcia-Mansfield K, Zhou J, Manousopoulou A, Pirrotte P, Wang Y, Wang LD, Feng M. Phosphoproteomic Profiling Reveals mTOR Signaling in Sustaining Macrophage Phagocytosis of Cancer Cells. Cancers (Basel) 2024; 16:4238. [PMID: 39766137 PMCID: PMC11674635 DOI: 10.3390/cancers16244238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Macrophage-mediated cancer cell phagocytosis has demonstrated considerable therapeutic potential. While the initiation of phagocytosis, facilitated by interactions between cancer cell surface signals and macrophage receptors, has been characterized, the mechanisms underlying its sustentation and attenuation post-initiation remain poorly understood. Methods: Through comprehensive phosphoproteomic profiling, we interrogated the temporal evolution of the phosphorylation profiles within macrophages during cancer cell phagocytosis. Results: Our findings reveal that activation of the mTOR pathway occurs following the initiation of phagocytosis and is crucial in sustaining phagocytosis of cancer cells. mTOR inhibition impaired the phagocytic capacity, but not affinity, of the macrophages toward the cancer cells by delaying phagosome maturation and impeding the transition between non-phagocytic and phagocytic states of macrophages. Conclusions: Our findings delineate the intricate landscape of macrophage phagocytosis and highlight the pivotal role of the mTOR pathway in mediating this process, offering valuable mechanistic insights for therapeutic interventions.
Collapse
Affiliation(s)
- Bixin Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Xu Cao
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Krystine Garcia-Mansfield
- Cancer and Cell Biology Division, Translational Genomics Institute, Phoenix, AZ 85004, USA
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Jingkai Zhou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Antigoni Manousopoulou
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Patrick Pirrotte
- Cancer and Cell Biology Division, Translational Genomics Institute, Phoenix, AZ 85004, USA
- Integrated Mass Spectrometry Shared Resource, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Yingyu Wang
- Center for Informatics, City of Hope, Duarte, CA 91010, USA
| | - Leo D. Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
- Department of Pediatrics, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mingye Feng
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
4
|
Damsky W, King B. Cutaneous sarcoidosis: clinical and pathologic features, molecular pathogenesis, and treatment. Clin Dermatol 2024:S0738-081X(24)00282-7. [PMID: 39694198 DOI: 10.1016/j.clindermatol.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Sarcoidosis is a multisystem inflammatory disorder that most commonly affects the lungs, lymphatic system, eyes, and skin. Cutaneous involvement is present in approximately 20-30% of patients. Prednisone and corticotropin repositories are the only FDA-approved therapies for sarcoidosis. We review the varied cutaneous manifestations of sarcoidosis and the recommended evaluation for this disease. We also discuss histopathologic findings and outline disease pathogenesis, incorporating newer molecular data. Treatment approaches for cutaneous sarcoidosis and their associated levels of evidence are also delineated.
Collapse
Affiliation(s)
- William Damsky
- Department of Dermatology, Yale School of Medicine, 333 Cedar St, LMP 5040, PO Box 208059, New Haven, Connecticut, 06510, United States; Department of Pathology, Yale School of Medicine, 310 Cedar Street, LH 108, PO Box 208023, New Haven, Connecticut, 06520, United States.
| | - Brett King
- Department of Dermatology, Yale School of Medicine, 333 Cedar St, LMP 5040, PO Box 208059, New Haven, Connecticut, 06510, United States.
| |
Collapse
|
5
|
Pozzan R, Salton F, Confalonieri P, Trotta L, Barbieri M, Reccardini N, Torregiani C, Screm G, Hughes M, Baratella E, Confalonieri M, Mondini L, Ruaro B. Autoantibodies in sarcoidosis: Innocent bystander or promising biomarker for organ involvement? SARCOIDOSIS, VASCULITIS, AND DIFFUSE LUNG DISEASES : OFFICIAL JOURNAL OF WASOG 2024; 41:e2024056. [PMID: 39655585 PMCID: PMC11708945 DOI: 10.36141/svdld.v41i4.16043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/12/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND AND AIM Sarcoidosis is a rare inflammatory disease that can affect any organ in the body, but most commonly involves lungs and lymph nodes. Sarcoidosis is often considered an autoimmune disease, attributed to many factors, including autoantigen-specific T cells, antibodies producing B lymphocytes, autoimmune inflammation, although its exact cause and classification are still under debate.The aim of our study was to evaluate the possible role of autoantibodies, such as anti-nuclear (ANA), extractable nuclear antigen (ENA) and antiphospholipids, in sarcoidosis patients. METHODS We conduct a retrospective study on our patients with confirmed diagnosis of sarcoidosis involving lungs, lymph nodes and multiple organs, and we collected and analyzed data on blood and urine tests (C-reactive protein, CRP, amount of calcium in blood and urine, CD4/CD8 ratio, lymphocyte count), lung function, radiological patterns, ongoing treatments (steroid therapy, hydroxychloroquine or methotrexate, other immunosuppressive agents). RESULTS We enrolled 328 sarcoidosis patients, and we focused our attention on 32 patients with positive ANA antibodies (11%), observing a high percentage of them with sarcoidosis involving the lungs (77%), but more specifically a significant discrepancy, in percentage terms, in the blood CD4/CD8 ratio. In the ANA-positive group we observed 26% of patients with a high blood CD4/CD8 ratio (average CD4/CD8 ratio of 2.41), whereas in the ANA-negative group, patients with a high CD4/CD8 ratio (average ratio 1.78) represented a much smaller percentage (13%). This finding may be a source of further investigation for other studies on the topic. CONCLUSIONS Analysis of autoantibodies expressed in our case series did not identify a specific autoantibodies pattern in sarcoidosis. Few studies have analyzed autoantibody patterns in sarcoidosis patients and involved smaller populations. In conclusion, our study evaluates a sizable population, and underlines the need for further, larger clinical studies to evaluate possible associations between sarcoidosis and autoimmunity.
Collapse
Affiliation(s)
- Riccardo Pozzan
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
- These authors contributed equally to this work
| | - Francesco Salton
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
- These authors contributed equally to this work
| | - Paola Confalonieri
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
| | - Liliana Trotta
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
| | - Mariangela Barbieri
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
| | - Nicolò Reccardini
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
| | - Chiara Torregiani
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
| | - Gianluca Screm
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
| | - Michael Hughes
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester, UK
| | - Elisa Baratella
- Department of Radiology, Department of Medicine, Surgery and Health Science, University of Trieste, Trieste, Italy
| | - Marco Confalonieri
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
| | - Lucrezia Mondini
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
- These authors contributed equally to this work
| | - Barbara Ruaro
- Pulmonology Unit, University of Trieste, Department of Medical Surgical and Health Sciences, Hospital of Cattinara, Trieste, Italy
- These authors contributed equally to this work
| |
Collapse
|
6
|
van Stigt AC, Gualtiero G, Cinetto F, Dalm VA, IJspeert H, Muscianisi F. The biological basis for current treatment strategies for granulomatous disease in common variable immunodeficiency. Curr Opin Allergy Clin Immunol 2024; 24:479-487. [PMID: 39431514 PMCID: PMC11537477 DOI: 10.1097/aci.0000000000001032] [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] [Indexed: 10/22/2024]
Abstract
PURPOSE OF REVIEW The pathogenesis of granulomatous disease in common variable immunodeficiency (CVID) is still largely unknown, which hampers effective treatment. This review describes the current knowledge on the pathogenesis of granuloma formation in CVID and the biological basis of the current treatment options. RECENT FINDINGS Histological analysis shows that T and B cells are abundantly present in the granulomas that are less well organized and are frequently associated with lymphoid hyperplasia. Increased presence of activation markers such as soluble IL-2 receptor (sIL-2R) and IFN-ɣ, suggest increased Th1-cell activity. Moreover, B-cell abnormalities are prominent in CVID, with elevated IgM, BAFF, and CD21low B cells correlating with granulomatous disease progression. Innate immune alterations, as M2 macrophages and neutrophil dysregulation, indicate chronic inflammation. Therapeutic regimens include glucocorticoids, DMARDs, and biologicals like rituximab. SUMMARY Our review links the biological context of CVID with granulomatous disease or GLILD to currently prescribed therapies and potential targeted treatments.
Collapse
Affiliation(s)
- Astrid C. van Stigt
- Laboratory Medical Immunology, Department of Immunology
- Division of Allergy & Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Giulia Gualtiero
- Hematology and Clinical Immunology Unit, Department of Medicine (DIMED)
- Veneto Institute of Molecular Medicine (VIMM)
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, Padova, Italy
| | - Virgil A.S.H. Dalm
- Laboratory Medical Immunology, Department of Immunology
- Division of Allergy & Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Francesco Muscianisi
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, Padova, Italy
| |
Collapse
|
7
|
Kim JS, Gupta R. Lung transplantation in pulmonary sarcoidosis. J Autoimmun 2024; 149:103135. [PMID: 37923622 DOI: 10.1016/j.jaut.2023.103135] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 11/07/2023]
Abstract
Sarcoidosis is a systemic inflammatory disease of unknown etiology and variable clinical course. Pulmonary sarcoidosis is the most common presentation and accounts for most morbidity and mortality related to sarcoidosis. While sarcoidosis generally has good outcomes, few patients experience chronic disease. A minority of patients progress to a specific phenotype of sarcoidosis referred to advanced pulmonary sarcoidosis (APS) which includes advanced fibrosis, pulmonary hypertension and respiratory failure, leading to high morbidity and mortality. In patients with advanced disease despite medical therapy, lung transplantation may be the last viable option for improvement in quality of life. Though post-transplant survival is similar to that of other end-stage lung diseases, it is imperative that patients are evaluated and referred early to transplant centers with experience in APS. A multidisciplinary approach and clinical experience are crucial in detecting the optimal timing of referral, initiating comprehensive transplantation evaluation and listing, discussing surgical approach, and managing perioperative and post-transplant care. This review article seeks to address these aspects of lung transplantation in APS.
Collapse
Affiliation(s)
- Jin Sun Kim
- Lewis Katz School of Medicine, Department of Thoracic Medicine and Surgery, Philadelphia, PA, USA.
| | - Rohit Gupta
- Lewis Katz School of Medicine, Department of Thoracic Medicine and Surgery, Philadelphia, PA, USA
| |
Collapse
|
8
|
Liu Z, Wang H, Liang Y, Liu M, Huang Q, Wang M, Zhou J, Bu Q, Zhou H, Lu L. E2F2 Reprograms Macrophage Function By Modulating Material and Energy Metabolism in the Progression of Metabolic Dysfunction-Associated Steatohepatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2410880. [PMID: 39465673 DOI: 10.1002/advs.202410880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/30/2024] [Indexed: 10/29/2024]
Abstract
Macrophages are essential for the development of steatosis, hepatic inflammation, and fibrosis in metabolic dysfunction-associated steatohepatitis(MASH). However, the roles of macrophage E2F2 in the progression of MASH have not been elucidated. This study reveals that the expression of macrophage E2F2 is dramatically downregulated in MASH livers from mice and humans, and that this expression is adversely correlated with the severity of the disease. Myeloid-specific E2F2 depletion aggravates intrahepatic inflammation, hepatic stellate cell activation, and hepatocyte lipid accumulation during MASH progression. Mechanistically, E2F2 can inhibit the SLC7A5 transcription directly. E2F2 deficiency upregulates the expression of SLC7A5 to mediate amino acids flux, resulting in enhanced glycolysis, impaired mitochondrial function, and increased macrophages proinflammatory response in a Leu-mTORC1-dependent manner. Moreover, bioinformatics analysis and CUT &Tag assay identify the direct binding of Nrf2 to E2F2 promoter to promote its transcription and nuclear translocation. Genetic or pharmacological activation of Nrf2 effectively activates E2F2 to attenuate the MASH progression. Finally, patients treated with CDK4/6 inhibitors demonstrate reduced E2F2 activity but increased SLC7A5 activity in PBMCs. These findings indicated macrophage E2F2 suppresses MASH progression by reprogramming amino acid metabolism via SLC7A5- Leu-mTORC1 signaling pathway. Activating E2F2 holds promise as a therapeutic strategy for MASH.
Collapse
Affiliation(s)
- Zheng Liu
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hao Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuan Liang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Mu Liu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, 210029, China
| | - Qiyuan Huang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, 210029, China
| | - Mingming Wang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jinren Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, 210029, China
| | - Qingfa Bu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, 210029, China
| | - Haoming Zhou
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, 210029, China
| | - Ling Lu
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, 220005, China
| |
Collapse
|
9
|
Obi ON, Saketkoo LA, Maier LA, Baughman RP. Developmental drugs for sarcoidosis. J Autoimmun 2024; 149:103179. [PMID: 38548579 DOI: 10.1016/j.jaut.2024.103179] [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: 09/10/2023] [Revised: 12/04/2023] [Accepted: 02/08/2024] [Indexed: 12/15/2024]
Abstract
Sarcoidosis is a multi-organ granulomatous inflammatory disease of unknown etiology. Over 50% of patients will require treatment at some point in their disease and 10%-30% will develop a chronic progressive disease with pulmonary fibrosis leading to significant morbidity and mortality. Recently published guidelines recommend immunosuppressive therapy for sarcoidosis patients at risk of increased disease-related morbidity and mortality, and in whom disease has negatively impacted quality of life. Prednisone the currently recommended first line therapy is associated with significant toxicity however none of the other guideline recommended steroid sparing therapy is approved by regulatory agencies for use in sarcoidosis, and data in support of their use is weak. For patients with severe refractory disease requiring prolonged therapy, treatment options are limited. The need for expanding treatment options in sarcoidosis has been emphasized. Well conducted large, randomized trials evaluating currently available therapeutic options as well as novel pathways for targeting disease are necessary to better guide treatment decisions. These trials will not be without significant challenges. Sarcoidosis is a rare disease with heterogenous presentation and variable progression and clinical outcome. There are no universally agreed upon biomarkers of disease activity and measurement of outcomes is confounded by the need to balance patient centric measures and objective measures of disease activity. Our paper provides an update on developmental drugs in sarcoidosis and outlines several novel pathways that may be targeted for future drug development. Currently available trials are highlighted and ongoing challenges to drug development and clinical trial design are briefly discussed.
Collapse
Affiliation(s)
- Ogugua Ndili Obi
- Division of Pulmonary Critical Care and Sleep Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| | - Lesley Ann Saketkoo
- New Orleans Scleroderma and Sarcoidosis Patient Care and Research Center, New Orleans, USA; University Medical Center - Comprehensive Pulmonary Hypertension Center and Interstitial Lung Disease Clinic Programs, New Orleans, USA; Louisiana State University School of Medicine, Section of Pulmonary Medicine, New Orleans, LA, USA; Tulane University School of Medicine, Undergraduate Honors Department, New Orleans, LA, USA
| | - Lisa A Maier
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA; Division of Pulmonary and Critical Care Sciences, Department of Medicine, University of Colorado School of Medicine, Denver, CO, USA
| | - Robert P Baughman
- Emeritus Professor of Medicine, Department of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
10
|
Bhargava M, Crouser ED. Application of laboratory models for sarcoidosis research. J Autoimmun 2024; 149:103184. [PMID: 38443221 DOI: 10.1016/j.jaut.2024.103184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/15/2024] [Indexed: 03/07/2024]
Abstract
This manuscript will review the implications and applications of sarcoidosis models towards advancing our understanding of sarcoidosis disease mechanisms, identification of biomarkers, and preclinical testing of novel therapies. Emerging disease models and innovative research tools will also be considered.
Collapse
Affiliation(s)
- Maneesh Bhargava
- University of Minnesota Medical Center, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, 420 Delaware Street SE, MMC 276. Minneapolis, MN 55455, USA
| | - Elliott D Crouser
- Ohio State University Wexner Medicine Center, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, 241 W. 11th Street, Suite 5000, Columbus, OH 43201, USA.
| |
Collapse
|
11
|
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
|
12
|
Pei C, Shen Z, Wu Y, Zhao S, Wang Y, Shi S, Huang D, Jia N, Liu J, Wang X, He Y, Wang Z. Eleutheroside B Pretreatment Attenuates Hypobaric Hypoxia-Induced High-Altitude Pulmonary Edema by Regulating Autophagic Flux via the AMPK/mTOR Pathway. Phytother Res 2024; 38:5657-5671. [PMID: 39307910 DOI: 10.1002/ptr.8333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 05/22/2024] [Accepted: 08/18/2024] [Indexed: 12/13/2024]
Abstract
High-altitude pulmonary edema (HAPE) is a life-threatening disease, and autophagy deficiency is implicated in the pathogenesis of HAPE. Eleutheroside B (EB), which is the main bioactive component of Acanthopanax senticosus, exhibits various pharmacological activities. Our previous research demonstrated that autophagic structures were widely found in the ultrastructure of lung tissue in HAPE rats. However, whether EB regulates autophagy deficiency in HAPE remains unknown. This study aimed to investigate the protective effects of EB on hypobaric hypoxia-induced HAPE and explore the underlying molecular mechanism of regulating autophagy. The rat model of high-altitude pulmonary edema was replicated using a hypobaric hypoxic chamber. Rats were pretreated with EB or in combination with chloroquine or compound C. The pulmonary edema was assessed by the lung wet/dry ratio, total protein concentration in bronchoalveolar lavage fluid, and histological analysis. Inflammation and oxidative stress were measured using commercial biochemical kits. Autophagy and autophagic flux were evaluated by western blotting, transmission electron microscopy, and adeno-associated virus-mRFP-GFP-labeled tandem fluorescence LC3. The AMPK/mTOR signaling pathway was detected by western blotting. EB alleviated hypobaric hypoxia-induced pulmonary edema, hypoxemia, acid-base imbalance in the blood, inflammation, and oxidative stress in a dose-dependent manner. EB restored impaired autophagic flux by activating the AMPK/mTOR signaling pathway. However, chloroquine or compound C abolished eleutheroside B-mediated autophagy flux restoration. EB has the potential to restore impaired autophagic flux in the lung of hypobaric hypoxia-induced HAPE rats, which could be attributed to the activation of AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Caixia Pei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zherui Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yongcan Wu
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, Chongqing, China
- College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, China
| | - Sijing Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yilan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shihua Shi
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Demei Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Nan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Junling Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaomin Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yacong He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- State Key Laboratory of Southwestern Chinese Medicine Resources School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenxing Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Richards D, Fujito H, Shanbhag A, Akhavan B, Frame E, Hayes S, Friedman J, Thomson L, Slomka P, Berman D, Kransdorf EP. Utility of Mechanistic Target of Rapamycin Inhibitors in Cardiac Sarcoidosis. J Card Fail 2024:S1071-9164(24)00929-1. [PMID: 39615743 DOI: 10.1016/j.cardfail.2024.10.444] [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: 05/08/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 12/22/2024]
Abstract
BACKGROUND Cardiac sarcoidosis is an uncommon but potentially devastating manifestation of sarcoidosis, which is a multisystem inflammatory granulomatous disease. Although corticosteroids are the mainstay of treatment, given the number of complications associated with their long-term use, there is increasing interest in the use of steroid-sparing agents. Recent basic and translational studies have suggested a role for the mechanistic target of rapamycin (mTOR) pathway in cardiac sarcoidosis. METHODS We identified 4 patients treated at the Cedars-Sinai Cardiac Sarcoidosis Clinic who had active cardiac sarcoidosis and contraindications to corticosteroid intensification. We sought to evaluate the role of mechanistic target of mTOR inhibitors on the change in cardiac inflammation via cardiac 18 F-fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging. RESULTS Of the 4 patients, 2 showed substantial improvement in cardiac inflammation on follow-up FDG-PET imaging after 6 months of treatment with an mTOR inhibitor but without corticosteroid intensification. There was a greater than 80% reduction in the cardiometabolic activity. The other 2 patients treated with an mTOR inhibitor had persistent evidence of cardiac inflammation on follow-up FDG-PET, necessitating an augmented treatment regimen. DISCUSSION This case series represents the first clinical use of mTOR inhibitors for cardiac sarcoidosis, and it suggests that these agents may have a role in the management of cardiac sarcoidosis.
Collapse
Affiliation(s)
- Donald Richards
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Hidesato Fujito
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA; Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Aakash Shanbhag
- Division of Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Bobak Akhavan
- Division of Pulmonary & Critical Care Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Elizabeth Frame
- Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Sean Hayes
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - John Friedman
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Louise Thomson
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Piotr Slomka
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA; Division of Artificial Intelligence in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Daniel Berman
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA; Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Evan P Kransdorf
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA.
| |
Collapse
|
14
|
Starshinova A, Berg E, Rubinstein A, Kulpina A, Kudryavtsev I, Kudlay D. Chronic Sarcoidosis: Diagnostic Difficulties and Search for New Criteria of Inflammatory Activity (A Case Report and Literature Review). J Clin Med 2024; 13:6974. [PMID: 39598118 PMCID: PMC11594891 DOI: 10.3390/jcm13226974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/05/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Sarcoidosis is a systemic inflammatory disease with an unknown etiology and a wide range of clinical manifestations. The incidence of sarcoidosis ranges from approximately 1 to 15 cases per 100,000 individuals per year worldwide. The significant variability in clinical presentations and target organs, as well as concomitant diseases, greatly complicates diagnosis. We analyzed articles in PubMed, Scopus, Cochrane Library, and Embase, where databases were searched using the keywords "chronic sarcoidosis", "diagnosis of sarcoidosis", "course of sarcoidosis", "pulmonary sarcoidosis", "cardiac sarcoidosis", "skin sarcoidosis", "neurosarcoidosis", "ocular sarcoidosis", and "autoimmune inflammation". Studies on the course and diagnosis of sarcoidosis with a deep search of ten years were included. In this review, we present an analysis of publications on the course and diagnosis of chronic sarcoidosis, as well as a clinical case. We have noted that the diagnosis of chronic sarcoidosis is particularly difficult due to the lack of specific biomarkers or their combination. The development and introduction of new diagnostic criteria for this disease will contribute to increasing the level of efficiency, not only of the diagnostic complex, but also the prognosis of the development and course of the pathological process. Conclusion: For the most accurate diagnosis and determination of prognosis, the existence of a single immunological or imaging marker with sufficient sensitivity and specificity is necessary.
Collapse
Affiliation(s)
- Anna Starshinova
- Department of Mathematics and Computer Science, St. Petersburg State University, St. Petersburg 199034, Russia;
- Almazov National Medical Research Centre, St. Petersburg 197341, Russia; (E.B.); (I.K.)
| | - Elizaveta Berg
- Almazov National Medical Research Centre, St. Petersburg 197341, Russia; (E.B.); (I.K.)
| | - Artem Rubinstein
- Department of Immunology, Institution of Experimental Medicine, St. Petersburg 197376, Russia;
| | - Anastasia Kulpina
- Department of Mathematics and Computer Science, St. Petersburg State University, St. Petersburg 199034, Russia;
- Almazov National Medical Research Centre, St. Petersburg 197341, Russia; (E.B.); (I.K.)
| | - Igor Kudryavtsev
- Almazov National Medical Research Centre, St. Petersburg 197341, Russia; (E.B.); (I.K.)
- Department of Immunology, Institution of Experimental Medicine, St. Petersburg 197376, Russia;
| | - Dmitry Kudlay
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia;
- Institute of Immunology, Moscow 115478, Russia
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow 119991, Russia
| |
Collapse
|
15
|
Becher B, Derfuss T, Liblau R. Targeting cytokine networks in neuroinflammatory diseases. Nat Rev Drug Discov 2024; 23:862-879. [PMID: 39261632 DOI: 10.1038/s41573-024-01026-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
In neuroinflammatory diseases, systemic (blood-borne) leukocytes invade the central nervous system (CNS) and lead to tissue damage. A causal relationship between neuroinflammatory diseases and dysregulated cytokine networks is well established across several preclinical models. Cytokine dysregulation is also observed as an inadvertent effect of cancer immunotherapy, where it often leads to neuroinflammation. Neuroinflammatory diseases can be separated into those in which a pathogen is at the centre of the immune response and those of largely unknown aetiology. Here, we discuss the pathophysiology, cytokine networks and therapeutic landscape of 'sterile' neuroinflammatory diseases such as multiple sclerosis (MS), neuromyelitis optica spectrum disorder (NMOSD), neurosarcoidosis and immune effector cell-associated neurotoxicity syndrome (ICANS) triggered by cancer immunotherapy. Despite successes in targeting cytokine networks in preclinical models of neuroinflammation, the clinical translation of targeting cytokines and their receptors has shown mixed and often paradoxical responses.
Collapse
Affiliation(s)
- Burkhard Becher
- Institute of experimental Immunology, University of Zurich, Zurich, Switzerland.
| | - Tobias Derfuss
- Department of Neurology and Biomedicine, Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Roland Liblau
- Institute for inflammatory and infectious diseases, INSERM UMR1291 - CNRS UMR505, Toulouse, France.
| |
Collapse
|
16
|
Fraser SD, Thackray-Nocera S, Wright C, Flockton R, James SR, Crooks MG, Kaye PM, Hart SP. Effects of Azithromycin on Blood Inflammatory Gene Expression and Cytokine Production in Sarcoidosis. Lung 2024; 202:683-693. [PMID: 39284999 PMCID: PMC11427505 DOI: 10.1007/s00408-024-00743-w] [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: 07/01/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION In sarcoidosis granulomas, monocyte-derived macrophages are activated by pro-inflammatory cytokines including TNF and IL-6. Current drug treatment for sarcoidosis aims to suppress inflammation but disabling side effects can ensue. The macrolide azithromycin may be anti-inflammatory. We aimed to determine whether treatment with azithromycin affects blood inflammatory gene expression and monocyte functions in sarcoidosis. METHODS Blood samples were collected from patients with chronic pulmonary sarcoidosis enrolled in a single arm, open label clinical trial who received oral azithromycin 250 mg once daily for 3 months. Whole blood inflammatory gene expression with or without LPS stimulation was measured using a 770-mRNA panel. Phenotypic analysis and cytokine production were conducted by flow cytometry and ELISA after 24h stimulation with growth factors and TLR ligands. mTOR activity was assessed by measuring phosphorylated S6RP. RESULTS Differential gene expression analysis indicated a state of heightened myeloid cell activation in sarcoidosis. Compared with controls, sarcoidosis patients showed increased LPS responses for several cytokines and chemokines. Treatment with azithromycin had minimal effect on blood gene expression overall, but supervised clustering analysis identified several chemokine genes that were upregulated. At the protein level, azithromycin treatment increased LPS-stimulated TNF and unstimulated IL-8 production. No other cytokines showed significant changes following azithromycin. Blood neutrophil counts fell during azithromycin treatment whereas mononuclear cells remained stable. Azithromycin had no detectable effects on mTOR activity or activation markers. CONCLUSION Blood myeloid cells are activated in sarcoidosis, but azithromycin therapy did not suppress inflammatory gene expression or cytokine production in blood. TRIAL REGISTRATION EudraCT 2019-000580-24 (17 May 2019).
Collapse
Affiliation(s)
- Simon D Fraser
- Respiratory Research Group, Hull York Medical School, Castle Hill Hospital, Cottingham, HU16 5JQ, UK
| | - Susannah Thackray-Nocera
- Respiratory Clinical Trials Unit, Hull University Teaching Hospitals NHS Trust, Castle Hill Hospital, Cottingham, HU16 5JQ, UK
| | - Caroline Wright
- Respiratory Clinical Trials Unit, Hull University Teaching Hospitals NHS Trust, Castle Hill Hospital, Cottingham, HU16 5JQ, UK
| | - Rachel Flockton
- Respiratory Clinical Trials Unit, Hull University Teaching Hospitals NHS Trust, Castle Hill Hospital, Cottingham, HU16 5JQ, UK
| | - Sally R James
- Biosciences Technology Facility, Dept. of Biology, University of York, York, UK
| | - Michael G Crooks
- Respiratory Research Group, Hull York Medical School, Castle Hill Hospital, Cottingham, HU16 5JQ, UK
| | - Paul M Kaye
- York Biomedical Research Institute, University of York, York, YO10 5DD, UK
| | - Simon P Hart
- Respiratory Research Group, Hull York Medical School, Castle Hill Hospital, Cottingham, HU16 5JQ, UK.
| |
Collapse
|
17
|
Sterniste G, Hackner K, Moazedi-Fürst F, Grasl M, Idzko M, Shao G, Guttmann-Ducke C, Talakić E, Prosch H, Lohfink-Schumm S, Gabriel M, Lim C, Hochreiter J, Bucher B, Böckle BC, Kiener HP, Duftner C, Kastrati K, Rath E, Funk M, Löffler-Ragg J, Steinmaurer M, Kovacs G, Verheyen N, Flick H, Antlanger M, Traxler G, Tatscher E, Zwick RH, Lang D. [Position paper of the Austrian Society for Rheumatology and the Austrian Society for Pneumology on the diagnosis and treatment of sarcoidosis 2024]. Wien Klin Wochenschr 2024; 136:669-687. [PMID: 39382646 PMCID: PMC11464578 DOI: 10.1007/s00508-024-02444-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] [Accepted: 08/28/2024] [Indexed: 10/10/2024]
Abstract
In many cases sarcoidosis is a multisystemic disease that requires interdisciplinary medical cooperation in the diagnostics, treatment and medical care during follow-up. Due to the often chronic course, it is of utmost importance to include patients with their priorities and wishes at an early stage and extensively in disease management and to establish a shared decision making whenever possible. In the process of writing this joint position paper, the expert group on interstitial and orphan lung diseases of the Austrian Society for Pulmonology and the working group on rheumatological lung disorders of the Austrian Society for Rheumatology and Rehabilitation sought to include patient advocacy groups as well as experts for rare organ manifestations of sarcoidosis. This position paper is not only meant to reflect current scientific and clinical standards but should also focus the national expertise and by networking and exchange to be a first step to strengthen cooperation between stakeholders to ultimately improve care for patients with sarcoidosis.
Collapse
Affiliation(s)
- Georg Sterniste
- Abteilung für Innere Medizin und Pneumologie, Klinik Floridsdorf, 1210, Wien, Österreich
| | - Klaus Hackner
- Klinische Abteilung für Pneumologie, Universitätsklinikum Krems, Karl Landsteiner Privatuniversität für Gesundheitswissenschaften, 3500, Krems, Österreich
| | - Florentine Moazedi-Fürst
- Klinische Abteilung für Rheumatologie und Immunologie, Medizinische Universität Graz, 8036, Graz, Österreich
| | - Marie Grasl
- Abteilung für Atemwegs- und Lungenkrankheiten, Klinik Penzing, Ludwig Boltzmann Institut für Lungengesundheit, Wien, Österreich, 1140, Wien, Österreich
| | - Marco Idzko
- Univ. Klinik für Innere Medizin II, Klin. Abteilung für Pulmologie, Medizinische Universität Wien, Wien, Österreich
| | - Guangyu Shao
- Universitätsklinikum für Innere Medizin 4/Pneumologie, Kepler Universitätsklinikum, Johannes Kepler Universität, Linz, Österreich
| | - Claudia Guttmann-Ducke
- Univ. Klinik für Innere Medizin II, Klin. Abteilung für Pulmologie, Medizinische Universität Wien, Wien, Österreich
| | - Emina Talakić
- Klinische Abteilung für Allgemeine Radiologische Diagnostik, Universitätsklinik für Radiologie, Medizinische Universität Graz, Graz, Österreich
| | - Helmut Prosch
- Univ. Klinik für Radiologie und Nuklearmedizin, Medizinische Universität Wien, Wien, Österreich
| | - Sylvia Lohfink-Schumm
- Institut für Pathologie und Molekularpathologie, Kepler Universitätsklinikum, Johannes Kepler Universität, Linz, Österreich
| | - Michael Gabriel
- Institut für Nuklearmedizin und Endokrinologie, Kepler Universitätsklinikum, Johannes Kepler Universität, Linz, Österreich
| | - Clarice Lim
- Abteilung für Atemwegs- und Lungenkrankheiten, Klinik Penzing, Ludwig Boltzmann Institut für Lungengesundheit, Wien, Österreich, 1140, Wien, Österreich
| | | | - Brigitte Bucher
- Abteilung Pneumologie, LKH Hochzirl Natters, Natters, Medizinische Universität Innsbruck, Innsbruck, Österreich
| | - Barbara C Böckle
- Universitätsklinik für Dermatologie, Venerologie & Allergologie, Medizinische Universität Innsbruck, Innsbruck, Österreich
| | - Hans Peter Kiener
- Universitätsklinik für Innere Medizin III, Klinische Abteilung für Rheumatologie, Medizinische Universität Wien, Wien, Österreich
| | - Christina Duftner
- Universitätsklinik für Innere Medizin II, Medizinische Universität Innsbruck, Innsbruck, Österreich
| | - Kastriot Kastrati
- Universitätsklinik für Innere Medizin III, Klinische Abteilung für Rheumatologie, Medizinische Universität Wien, Wien, Österreich
| | - Eva Rath
- 1. Medizinische Abteilung, Hanusch Krankenhaus, Heinrich-Collin-Str. 30, 1140, Wien, Österreich
| | - Marion Funk
- Universitätsklinik für Augenheilkunde und Optometrie, Medizinische Universität Wien, Wien, Österreich
| | - Judith Löffler-Ragg
- Abteilung Pneumologie, LKH Hochzirl Natters, Natters, Medizinische Universität Innsbruck, Innsbruck, Österreich
| | - Monika Steinmaurer
- Abteilung für Lungenkrankheiten, Klinikum Wels-Grieskirchen, 4600, Wels, Österreich
| | - Gabor Kovacs
- Universitätsklinik für Innere Medizin, Klinische Abteilung für Pulmonologie, Medizinische Universität Graz, Graz, Österreich
| | - Nicolas Verheyen
- Universitätsklinik für Innere Medizin, Klinische Abteilung für Kardiologie, Medizinische Universität Graz, Graz, Österreich
| | - Holger Flick
- Universitätsklinik für Innere Medizin, Klinische Abteilung für Pulmonologie, Medizinische Universität Graz, Graz, Österreich
| | - Marlies Antlanger
- Universitätsklinik für Innere Medizin 2, Kepler Universitätsklinikum, Johannes Kepler Universität, Linz, Österreich
| | - Gerhard Traxler
- Universitätsklinik für Neurologie, Kepler Universitätsklinikum, Johannes Kepler Universität, Linz, Österreich
| | - Elisabeth Tatscher
- Universitätsklinik für Innere Medizin, Klinische Abteilung für Gastroenterologie und Hepatologie, Medizinische Universität Graz, Graz, Österreich
| | - Ralf Harun Zwick
- Ambulante Rehabilitation, Ludwig Boltzmann Institute for Rehabilitation Research, Therme Wien Med, Wien, Österreich
| | - David Lang
- Universitätsklinikum für Innere Medizin 4/Pneumologie, Kepler Universitätsklinikum, Johannes Kepler Universität, Linz, Österreich.
| |
Collapse
|
18
|
Wang W, Jia H, Hua X, Song J. New insights gained from cellular landscape changes in myocarditis and inflammatory cardiomyopathy. Heart Fail Rev 2024; 29:883-907. [PMID: 38896377 DOI: 10.1007/s10741-024-10406-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2024] [Indexed: 06/21/2024]
Abstract
Advances in the etiological classification of myocarditis and inflammatory cardiomyopathy (ICM) have reached a consensus. However, the mechanism of myocarditis/ICM remains unclear, which affects the development of treatment and the improvement of outcome. Cellular transcription and metabolic reprogramming, and the interactions between cardiomyocytes and non-cardiomyocytes, such as the immune cells, contribute to the process of myocarditis/ICM. Recent efforts have been made by multi-omics techniques, particularly in single-cell RNA sequencing, to gain a better understanding of the cellular landscape alteration occurring in disease during the progression. This article aims to provide a comprehensive overview of the latest studies in myocarditis/ICM, particularly as revealed by single-cell sequencing.
Collapse
Affiliation(s)
- Weiteng Wang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10037, China
| | - Xiumeng Hua
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10037, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Fuwai Hospital, National Centre for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 167 Beilishi Road, Xicheng District, Beijing, 100037, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, 518057, China.
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 10037, China.
| |
Collapse
|
19
|
Crouser ED, Julian MW, Locke LW, Bicer S, Mitchell JR, Singha A, Kramer PJ, Rajaram MVS, Raman SV. The Renin-Angiotensin-Aldosterone System Regulates Sarcoidosis Granulomatous Inflammation. Am J Respir Crit Care Med 2024; 210:497-507. [PMID: 38941161 PMCID: PMC11351795 DOI: 10.1164/rccm.202402-0265oc] [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: 02/01/2024] [Accepted: 06/28/2024] [Indexed: 06/30/2024] Open
Abstract
Rationale: Sarcoidosis is a granulomatous disorder of unclear cause notable for abnormal elevation of blood and tissue ACE1 (angiotensin converting enzyme 1) levels and activity. ACE1 regulates the renin-angiotensin-aldosterone system (RAAS), the terminal product of which is aldosterone, which selectively engages mineralocorticoid receptors to promote inflammation. Objectives: We sought to determine whether the RAAS promotes sarcoidosis granuloma formation and related inflammatory responses. Methods: Using an established ex vivo model, we first determined whether aldosterone was produced by sarcoidosis granulomas and verified the presence of CYP11B2, the enzyme required for its production. We then evaluated the effects of selective inhibitors of ACE1 (captopril), angiotensin type 1 receptor (losartan), and mineralocorticoid receptors (spironolactone, eplerenone) on granuloma formation, reflected by computer image analysis-generated granuloma area, and selected cytokines incriminated in sarcoidosis pathogenesis. Measurements and Main Results: Aldosterone was spontaneously produced by sarcoidosis peripheral blood mononuclear cells, and both intra- and extracellular levels steadily increased during granuloma formation. In parallel, peripheral blood mononuclear cells were shown to express more CYP11B2 during granuloma formation. Significant inhibition of sarcoidosis granulomas and related cytokines (TNFα, IL-1β, IFNγ, IL-10) was observed in response to pretreatments with captopril, losartan, spironolactone, or eplerenone, comparable to that of prednisone. Conclusions: The RAAS is intact in sarcoidosis granulomas and contributes significantly to early granuloma formation and to related inflammatory mediator responses, with important implications for clinical management.
Collapse
Affiliation(s)
- Elliott D. Crouser
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Dorothy M. Davis Heart and Lung Research Institute
| | - Mark W. Julian
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Dorothy M. Davis Heart and Lung Research Institute
| | | | - Sabahattin Bicer
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Dorothy M. Davis Heart and Lung Research Institute
- Department of Biomedical Engineering
| | | | - Arindam Singha
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Dorothy M. Davis Heart and Lung Research Institute
| | - Patrick J. Kramer
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Dorothy M. Davis Heart and Lung Research Institute
| | - Murugesan V. S. Rajaram
- Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | | |
Collapse
|
20
|
Atif SM, Drake WP. Renin-Angiotensin-Aldosterone System: A Potential Source of Biomarkers and Therapeutic Targets for Sarcoidosis. Am J Respir Crit Care Med 2024; 210:387-389. [PMID: 38990737 PMCID: PMC11351798 DOI: 10.1164/rccm.202406-1277ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024] Open
Affiliation(s)
- Shaikh M Atif
- Department of Medicine University of Colorado Anschutz Medical Campus Aurora, Colorado
| | - Wonder P Drake
- Department of Medicine University of Maryland School of Medicine Baltimore, Maryland
| |
Collapse
|
21
|
Konigsberg IR, Lin NW, Liao SY, Liu C, MacPhail K, Mroz MM, Davidson E, Restrepo CI, Sharma S, Li L, Maier LA, Yang IV. Multi-omic signatures of sarcoidosis and progression in bronchoalveolar lavage cells. Respir Res 2024; 25:289. [PMID: 39080656 PMCID: PMC11290275 DOI: 10.1186/s12931-024-02919-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Sarcoidosis is a heterogeneous granulomatous disease with no accurate biomarkers of disease progression. Therefore, we profiled and integrated the DNA methylome, mRNAs, and microRNAs to identify molecular changes associated with sarcoidosis and disease progression that might illuminate underlying mechanisms of disease and potential biomarkers. METHODS Bronchoalveolar lavage cells from 64 sarcoidosis subjects and 16 healthy controls were used. DNA methylation was profiled on Illumina HumanMethylationEPIC arrays, mRNA by RNA-sequencing, and miRNAs by small RNA-sequencing. Linear models were fit to test for effect of sarcoidosis diagnosis and progression phenotype, adjusting for age, sex, smoking, and principal components of the data. We built a supervised multi-omics model using a subset of features from each dataset. RESULTS We identified 1,459 CpGs, 64 mRNAs, and five miRNAs associated with sarcoidosis versus controls and four mRNAs associated with disease progression. Our integrated model emphasized the prominence of the PI3K/AKT1 pathway, which is important in T cell and mTOR function. Novel immune related genes and miRNAs including LYST, RGS14, SLFN12L, and hsa-miR-199b-5p, distinguished sarcoidosis from controls. Our integrated model also demonstrated differential expression/methylation of IL20RB, ABCC11, SFSWAP, AGBL4, miR-146a-3p, and miR-378b between non-progressive and progressive sarcoidosis. CONCLUSIONS Leveraging the DNA methylome, transcriptome, and miRNA-sequencing in sarcoidosis BAL cells, we detected widespread molecular changes associated with disease, many which are involved in immune response. These molecules may serve as diagnostic/prognostic biomarkers and/or drug targets, although future testing is required for confirmation.
Collapse
Affiliation(s)
- Iain R Konigsberg
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA.
| | - Nancy W Lin
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA.
- Division of Pulmonary and Critical Care Sciences, Department of Medicine, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA.
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Shu-Yi Liao
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA
- Division of Pulmonary and Critical Care Sciences, Department of Medicine, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
- Department of Environmental and Occupational Health, Colorado School of Public Health, Aurora, CO, USA
| | - Cuining Liu
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Kristyn MacPhail
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Margaret M Mroz
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Elizabeth Davidson
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Clara I Restrepo
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Sunita Sharma
- Division of Pulmonary and Critical Care Sciences, Department of Medicine, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Li Li
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA
- Division of Pulmonary and Critical Care Sciences, Department of Medicine, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Lisa A Maier
- Division of Environmental and Occupational Health Sciences, Department of Medicine, National Jewish Health, Denver, CO, USA
- Division of Pulmonary and Critical Care Sciences, Department of Medicine, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
- Department of Environmental and Occupational Health, Colorado School of Public Health, Aurora, CO, USA
| | - Ivana V Yang
- Department of Biomedical Informatics, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
- Division of Pulmonary and Critical Care Sciences, Department of Medicine, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
22
|
Buso H, Discardi C, Bez P, Muscianisi F, Ceccato J, Milito C, Firinu D, Landini N, Jones MG, Felice C, Rattazzi M, Scarpa R, Cinetto F. Sarcoidosis versus Granulomatous and Lymphocytic Interstitial Lung Disease in Common Variable Immunodeficiency: A Comparative Review. Biomedicines 2024; 12:1503. [PMID: 39062076 PMCID: PMC11275071 DOI: 10.3390/biomedicines12071503] [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: 05/08/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Sarcoidosis and Granulomatous and Lymphocytic Interstitial Lung Diseases (GLILD) are two rare entities primarily characterised by the development of Interstitial Lung Disease (ILD) in the context of systemic immune dysregulation. These two conditions partially share the immunological background and pathologic findings, with granuloma as the main common feature. In this narrative review, we performed a careful comparison between sarcoidosis and GLILD, with an overview of their main similarities and differences, starting from a clinical perspective and ending with a deeper look at the immunopathogenesis and possible target therapies. Sarcoidosis occurs in immunocompetent individuals, whereas GLILD occurs in patients affected by common variable immunodeficiency (CVID). Moreover, peculiar extrapulmonary manifestations and radiological and histological features may help distinguish the two diseases. Despite that, common pathogenetic pathways have been suggested and both these disorders can cause progressive impairment of lung function and variable systemic granulomatous and non-granulomatous complications, leading to significant morbidity, reduced quality of life, and survival. Due to the rarity of these conditions and the extreme clinical variability, there are still many open questions concerning their pathogenesis, natural history, and optimal management. However, if studied in parallel, these two entities might benefit from each other, leading to a better understanding of their pathogenesis and to more tailored treatment approaches.
Collapse
Affiliation(s)
- Helena Buso
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Claudia Discardi
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Patrick Bez
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Francesco Muscianisi
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Jessica Ceccato
- Haematology and Clinical Immunology Unit, Department of Medicine (DIMED), University of Padova, 35124 Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), 35131 Padova, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Nicholas Landini
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I Hospital, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Mark G. Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 YD, UK;
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Carla Felice
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Marcello Rattazzi
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Riccardo Scarpa
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| |
Collapse
|
23
|
van Wijck RTA, Sharma HS, Swagemakers SMA, Dik WA, IJspeert H, Dalm VASH, van Daele PLA, van Hagen PM, van der Spek PJ. Bioinformatic meta-analysis reveals novel differentially expressed genes and pathways in sarcoidosis. Front Med (Lausanne) 2024; 11:1381031. [PMID: 38938383 PMCID: PMC11208482 DOI: 10.3389/fmed.2024.1381031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/23/2024] [Indexed: 06/29/2024] Open
Abstract
Introduction Sarcoidosis is a multi-system inflammatory disease of unknown origin with heterogeneous clinical manifestations varying from a single organ non-caseating granuloma site to chronic systemic inflammation and fibrosis. Gene expression studies have suggested several genes and pathways implicated in the pathogenesis of sarcoidosis, however, due to differences in study design and variable statistical approaches, results were frequently not reproducible or concordant. Therefore, meta-analysis of sarcoidosis gene-expression datasets is of great importance to robustly establish differentially expressed genes and signalling pathways. Methods We performed meta-analysis on 22 published gene-expression studies on sarcoidosis. Datasets were analysed systematically using same statistical cut-offs. Differentially expressed genes were identified by pooling of p-values using Edgington's method and analysed for pathways using Ingenuity Pathway Analysis software. Results A consistent and significant signature of novel and well-known genes was identified, those collectively implicated both type I and type II interferon mediated signalling pathways in sarcoidosis. In silico functional analysis showed consistent downregulation of eukaryotic initiation factor 2 signalling, whereas cytokines like interferons and transcription factor STAT1 were upregulated. Furthermore, we analysed affected tissues to detect differentially expressed genes likely to be involved in granuloma biology. This revealed that matrix metallopeptidase 12 was exclusively upregulated in affected tissues, suggesting a crucial role in disease pathogenesis. Discussion Our analysis provides a concise gene signature in sarcoidosis and expands our knowledge about the pathogenesis. Our results are of importance to improve current diagnostic approaches and monitoring strategies as well as in the development of targeted therapeutics.
Collapse
Affiliation(s)
- Rogier T. A. van Wijck
- Department of Pathology & Clinical Bioinformatics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Hari S. Sharma
- Department of Pathology & Clinical Bioinformatics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Sigrid M. A. Swagemakers
- Department of Pathology & Clinical Bioinformatics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Hanna IJspeert
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Virgil A. S. H. Dalm
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Paul L. A. van Daele
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - P. Martin van Hagen
- Laboratory Medical Immunology, Department of Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
- Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus MC University Medical Center, Rotterdam, Netherlands
| | - Peter J. van der Spek
- Department of Pathology & Clinical Bioinformatics, Erasmus MC University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
24
|
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
|
25
|
Robert M, Yatim N, Sacré K, Duffy D. Sarcoidosis immunopathogenesis - a new concept of maladaptive trained immunity. Trends Immunol 2024; 45:406-418. [PMID: 38796404 DOI: 10.1016/j.it.2024.04.013] [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: 03/26/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/28/2024]
Abstract
Sarcoidosis is a chronic immune disease of unknown origin for which we still lack an immunological framework unifying causal agents, host factors, and natural history of disease. Here, we discuss the initial triggers of disease, and how myeloid cells drive granuloma formation and contribute to immunopathogenesis. We highlight recent advances in our understanding of innate immune memory and propose the hypothesis that maladaptive innate immune training connects previous environmental exposure to granuloma maintenance and expansion. Lastly, we consider how this hypothesis may open novel therapeutic avenues, while corticosteroids remain the front-line treatment.
Collapse
Affiliation(s)
- Marie Robert
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, Paris, France; Department of Internal Medicine, Hôpital Bichat, Paris, France; Université Paris-Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, Paris, France
| | - Nader Yatim
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, Paris, France; Department of Internal Medicine, Hôpital Bichat, Paris, France
| | - Karim Sacré
- Department of Internal Medicine, Hôpital Bichat, Paris, France; Université Paris-Cité, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Laboratoire d'Excellence Inflamex, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris-Cité, Paris, France; CBUtechS, Institut Pasteur, Université Paris-Cité, Paris, France.
| |
Collapse
|
26
|
Hutton A, Deshane JS. The Fats and the Furious: Unraveling the Role of Macrophage Lipid Metabolism in Sarcoidosis. Am J Respir Crit Care Med 2024; 209:1064-1066. [PMID: 38690977 PMCID: PMC11092947 DOI: 10.1164/rccm.202402-0287ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024] Open
Affiliation(s)
- Anneka Hutton
- Division of Pulmonary Allergy and Critical Care Medicine Department of Medicine University of Alabama at Birmingham Birmingham, Alabama
| | - Jessy S Deshane
- Division of Pulmonary Allergy and Critical Care Medicine Department of Medicine University of Alabama at Birmingham Birmingham, Alabama
| |
Collapse
|
27
|
Lim CX, Redl A, Kleissl L, Pandey RV, Mayerhofer C, El Jammal T, Mazic M, Gonzales K, Sukhbaatar N, Krausgruber T, Bock C, Hengstschläger M, Calender A, Pacheco Y, Stary G, Weichhart T. Aberrant Lipid Metabolism in Macrophages Is Associated with Granuloma Formation in Sarcoidosis. Am J Respir Crit Care Med 2024; 209:1152-1164. [PMID: 38353578 DOI: 10.1164/rccm.202307-1273oc] [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: 08/01/2023] [Accepted: 02/14/2024] [Indexed: 05/02/2024] Open
Abstract
Rationale: Chronic sarcoidosis is a complex granulomatous disease with limited treatment options that can progress over time. Understanding the molecular pathways contributing to disease would aid in new therapeutic development. Objectives: To understand whether macrophages from patients with nonresolving chronic sarcoidosis are predisposed to macrophage aggregation and granuloma formation and whether modulation of the underlying molecular pathways influence sarcoidosis granuloma formation. Methods: Macrophages were cultivated in vitro from isolated peripheral blood CD14+ monocytes and evaluated for spontaneous aggregation. Transcriptomics analyses and phenotypic and drug inhibitory experiments were performed on these monocyte-derived macrophages. Human skin biopsies from patients with sarcoidosis and a myeloid Tsc2-specific sarcoidosis mouse model were analyzed for validatory experiments. Measurements and Main Results: Monocyte-derived macrophages from patients with chronic sarcoidosis spontaneously formed extensive granulomas in vitro compared with healthy control participants. Transcriptomic analyses separated healthy and sarcoidosis macrophages and identified an enrichment in lipid metabolic processes. In vitro patient granulomas, sarcoidosis mouse model granulomas, and those directly analyzed from lesional patient skin expressed an aberrant lipid metabolism profile and contained increased neutral lipids. Conversely, a combination of statins and cholesterol-reducing agents reduced granuloma formation both in vitro and in vivo in a sarcoidosis mouse model. Conclusions: Together, our findings show that altered lipid metabolism in sarcoidosis macrophages is associated with its predisposition to granuloma formation and suggest cholesterol-reducing therapies as a treatment option in patients.
Collapse
Affiliation(s)
- Clarice X Lim
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics
| | - Anna Redl
- Department of Dermatology, and
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Lisa Kleissl
- Department of Dermatology, and
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | | | - Thomas El Jammal
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR5305, University Claude Bernard Lyon 1, IBCP, Lyon, France; and
| | - Mario Mazic
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics
| | - Karine Gonzales
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics
| | | | - Thomas Krausgruber
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Bock
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Alain Calender
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR5305, University Claude Bernard Lyon 1, IBCP, Lyon, France; and
- Department of Genetics, Hospices Civils de Lyon, University Claude Bernard Lyon 1, Bron, France
| | - Yves Pacheco
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR5305, University Claude Bernard Lyon 1, IBCP, Lyon, France; and
| | - Georg Stary
- Department of Dermatology, and
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Weichhart
- Institute of Medical Genetics, Center of Pathobiochemistry and Genetics
| |
Collapse
|
28
|
Chen C, Luo N, Dai F, Zhou W, Wu X, Zhang J. Advance in pathogenesis of sarcoidosis: Triggers and progression. Heliyon 2024; 10:e27612. [PMID: 38486783 PMCID: PMC10938127 DOI: 10.1016/j.heliyon.2024.e27612] [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: 11/15/2023] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024] Open
Abstract
Sarcoidosis, a multisystemic immune disease, significantly impacts patients' quality of life. The complexity and diversity of its pathogenesis, coupled with limited comprehensive research, had hampered both diagnosis and treatment, resulting in an unsatisfactory prognosis for many patients. In recent years, the research had made surprising progress in the triggers of sarcoidosis (genetic inheritance, infection and environmental factors) and the abnormal regulations on immunity during the formation of granuloma. This review consolidated the latest findings on sarcoidosis research, providing a systematic exploration of advanced studies on triggers, immune-related regulatory mechanisms, and clinical applications. By synthesizing previous discoveries, we aimed to offer valuable insights for future research directions and the development of clinical diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Cong Chen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| | - Nanzhi Luo
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| | - Fuqiang Dai
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Wenjing Zhou
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| | - Xiaoqing Wu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| | - Jian Zhang
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| |
Collapse
|
29
|
Abstract
At present, no biomarker exists which is truly specific for sarcoidosis and the ones available have modest sensitivity and specificity. The clinical context should dictate the choice of biomarker(s) used to address different clinical questions such as diagnosis, monitoring disease activity or monitoring response to treatment. In the future, in addition to known serum biomarkers, it seems fruitful to further explore a possible role of imaging, exhaled air and even biopsy-related biomarkers in sarcoidosis to guide clinical management.
Collapse
Affiliation(s)
- Sophie C van der Mark
- Department of Pulmonology, ILD Center of Excellence, St. Antonius Hospital, Koekoekslaan 1, 3435 CM, Nieuwegein, The Netherlands; Division of Heart and Lungs, University Medical Center, Utrecht, The Netherlands
| | - Vikaash W S Bajnath
- Department of Pulmonology, ILD Center of Excellence, St. Antonius Hospital, Koekoekslaan 1, 3435 CM, Nieuwegein, The Netherlands
| | - Marcel Veltkamp
- Department of Pulmonology, ILD Center of Excellence, St. Antonius Hospital, Koekoekslaan 1, 3435 CM, Nieuwegein, The Netherlands; Division of Heart and Lungs, University Medical Center, Utrecht, The Netherlands.
| |
Collapse
|
30
|
Nunes H, Brillet PY, Bernaudin JF, Gille T, Valeyre D, Jeny F. Fibrotic Pulmonary Sarcoidosis. Clin Chest Med 2024; 45:199-212. [PMID: 38245367 DOI: 10.1016/j.ccm.2023.08.011] [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] [Indexed: 01/22/2024]
Abstract
Fibrotic pulmonary sarcoidosis (fPS) affects about 20% of patients. fPS carries a significant morbidity and mortality. However, its prognosis is highly variable, depending mainly on fibrosis extent, functional impairment severity, and the development of pulmonary hypertension. Moreover, fPS outcomes are also influenced by several other complications, including acute exacerbations, and infections. fPS natural history is unknown, in particular regarding the risk of progressive self-sustaining fibrosis. The management of fPS is challenging, including anti-inflammatory treatment if granulomatous activity persists, rehabilitation, and in highly selected patients antifibrotic treatment and lung transplantation.
Collapse
Affiliation(s)
- Hilario Nunes
- AP-HP, Pulmonology Department, Avicenne Hospital, Bobigny, 93009, France; INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, 93009, France.
| | - Pierre-Yves Brillet
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, 93009, France; AP-HP, Radiology Department, Avicenne Hospital, Bobigny, 93009, France
| | | | - Thomas Gille
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, 93009, France; AP-HP, Physiology Department, Avicenne Hospital, Bobigny, 93009, France
| | - Dominique Valeyre
- INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, 93009, France; Groupe Hospitalier Paris Saint-Joseph, Pulmonology Department, Paris, 75014 France
| | - Florence Jeny
- AP-HP, Pulmonology Department, Avicenne Hospital, Bobigny, 93009, France; INSERM UMR 1272, Sorbonne Paris-Nord University, Bobigny, 93009, France
| |
Collapse
|
31
|
Redl A, Doberer K, Unterluggauer L, Kleissl L, Krall C, Mayerhofer C, Reininger B, Stary V, Zila N, Weninger W, Weichhart T, Bock C, Krausgruber T, Stary G. Efficacy and safety of mTOR inhibition in cutaneous sarcoidosis: a single-centre trial. THE LANCET. RHEUMATOLOGY 2024; 6:e81-e91. [PMID: 38267106 DOI: 10.1016/s2665-9913(23)00302-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Sarcoidosis is an inflammatory condition that can affect various organs and tissues, causing the formation of granulomas and subsequent functional impairment. The origin of sarcoidosis remains unknown and there are few treatment options. Mechanistic target of rapamycin (mTOR) activation is commonly seen in granulomas of patients across different tissues and has been shown to induce sarcoidosis-like granulomas in a mouse model. This study aimed to examine the efficacy and safety of the mTOR inhibitor sirolimus as a treatment for cutaneous sarcoidosis. METHODS We did a single-centre, randomised study treating patients with persistent and glucocorticoid-refractory cutaneous sarcoidosis with sirolimus at the Vienna General Hospital, Medical University of Vienna (Vienna, Austria). We recruited participants who had persistent, active, and histologically proven cutaneous sarcoidosis. We used an n-of-1 crossover design in a placebo-controlled, double-blind topical treatment period and a subsequent single-arm systemic treatment phase for 4 months in the same participants. Participants initially received either 0·1% topical sirolimus in Vaseline or placebo (Vaseline alone), twice daily. After a washout period, all participants were subsequently administered a 6 mg loading dose followed by 2 mg sirolimus solution orally once daily, aiming to achieve serum concentrations of 6 ng/mL. The primary endpoint was change in the Cutaneous Sarcoidosis Activity and Morphology Index (CSAMI) after topical or systemic treatment. All participants were included in the safety analyses, and patients having completed the respective treatment period (topical treatment or systemic treatment) were included in the primary analyses. Adverse events were assessed at each study visit by clinicians and were categorised according to their correlation with the study drug, severity, seriousness, and expectedness. This study is registered with EudraCT (2017-004930-27) and is now closed. FINDINGS 16 participants with persistent cutaneous sarcoidosis were enrolled in the study between Sept 3, 2019, and June 15, 2021. Six (37%) of 16 participants were men, ten (63%) were women, and 15 (94%) were White. The median age of participants was 54 years (IQR 48-58). 14 participants were randomly assigned in the topical phase and 2 entered the systemic treatment phase directly. Daily topical treatment did not improve cutaneous lesions (effect estimate -1·213 [95% CI -2·505 to 0·079], p=0·066). Systemic treatment targeting trough serum concentrations of 6 ng/mL resulted in clinical and histological improvement of skin lesions in seven (70%) of ten participants (median -7·0 [95% CI -16·5 to -3·0], p=0·018). Various morphologies of cutaneous sarcoidosis, including papular, nodular, plaque, scar, and tattoo-associated sarcoidosis, responded to systemic sirolimus therapy with a long-lasting effect for more than 1 year after treatment had been stopped. There were no serious adverse events and no deaths. INTERPRETATION Short-term treatment with systemic sirolimus might be an effective and safe treatment option for patients with persistent glucocorticoid-refractory sarcoidosis with a long-lasting disease-modulating effect. The effect of sirolimus in granulomatous inflammation should be investigated further in large, multi-centre, randomised clinical trials. FUNDING Vienna Science and Technology Fund, Austrian Science Fund.
Collapse
Affiliation(s)
- Anna Redl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | | | - Lisa Kleissl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Christoph Krall
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | | | - Bärbel Reininger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Victoria Stary
- Department of General Surgery, Medical University of Vienna, Vienna, Austria.
| | - Nina Zila
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Division of Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Thomas Weichhart
- Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Christoph Bock
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Thomas Krausgruber
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|
32
|
Padovani CM, Yin K. Immunosuppression in Sepsis: Biomarkers and Specialized Pro-Resolving Mediators. Biomedicines 2024; 12:175. [PMID: 38255280 PMCID: PMC10813323 DOI: 10.3390/biomedicines12010175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Severe infection can lead to sepsis. In sepsis, the host mounts an inappropriately large inflammatory response in an attempt to clear the invading pathogen. This sustained high level of inflammation may cause tissue injury and organ failure. Later in sepsis, a paradoxical immunosuppression occurs, where the host is unable to clear the preexisting infection and is susceptible to secondary infections. A major issue with sepsis treatment is that it is difficult for physicians to ascertain which stage of sepsis the patient is in. Sepsis treatment will depend on the patient's immune status across the spectrum of the disease, and these immune statuses are nearly polar opposites in the early and late stages of sepsis. Furthermore, there is no approved treatment that can resolve inflammation without contributing to immunosuppression within the host. Here, we review the major mechanisms of sepsis-induced immunosuppression and the biomarkers of the immunosuppressive phase of sepsis. We focused on reviewing three main mechanisms of immunosuppression in sepsis. These are lymphocyte apoptosis, monocyte/macrophage exhaustion, and increased migration of myeloid-derived suppressor cells (MDSCs). The biomarkers of septic immunosuppression that we discuss include increased MDSC production/migration and IL-10 levels, decreased lymphocyte counts and HLA-DR expression, and increased GPR18 expression. We also review the literature on the use of specialized pro-resolving mediators (SPMs) in different models of infection and/or sepsis, as these compounds have been reported to resolve inflammation without being immunosuppressive. To obtain the necessary information, we searched the PubMed database using the keywords sepsis, lymphocyte apoptosis, macrophage exhaustion, MDSCs, biomarkers, and SPMs.
Collapse
Affiliation(s)
- Cristina M. Padovani
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Virtua Health College of Life Sciences of Rowan University, Stratford, NJ 08084, USA;
| | | |
Collapse
|
33
|
Baratta S, Cazzato G, Foti C, Ingravallo G, Lospalluti L, Laface C, Filotico R, Ambrogio F. Tattoo-Associated Skin Reaction in a Melanoma Patient Receiving B-RAF and MEK Inhibitors: A Case Report with an Emphasis on Etiopathogenic and Histological Features. J Clin Med 2024; 13:321. [PMID: 38256455 PMCID: PMC10816304 DOI: 10.3390/jcm13020321] [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: 10/13/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Tattoo-associated cutaneous reactions have become quite frequent given the increasing percentage of tattooed subjects globally and also in Italy. On the other hand, the increasing use of target therapy is showing the ability of these drugs to affect the immune system and also cause adverse tattoo-related reactions. In this paper, we report a case of a 42-year-old patient with stage-IIID melanoma undergoing treatment with Dabrafenib and Trametinib. The patient reported erythema, oedema and scaling in areas of the body containing a black tattoo, and, conversely, no signs and/or symptoms in areas with tattoos of a different color. Histopathological and immunohistochemical features indicated a lympho-histiocytic reaction with a granulomatous morphology, mainly distributed around the vessels and hair adnexa. By discussing the cases reported in the literature prior to ours, we concluded and provided the possible indications of the pathogenesis.
Collapse
Affiliation(s)
- Silvia Baratta
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.B.); (C.F.); (L.L.); (R.F.); (F.A.)
| | - Gerardo Cazzato
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Caterina Foti
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.B.); (C.F.); (L.L.); (R.F.); (F.A.)
| | - Giuseppe Ingravallo
- Section of Molecular Pathology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Lucia Lospalluti
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.B.); (C.F.); (L.L.); (R.F.); (F.A.)
| | - Carmelo Laface
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Raffaele Filotico
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.B.); (C.F.); (L.L.); (R.F.); (F.A.)
| | - Francesca Ambrogio
- Section of Dermatology and Venereology, Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari “Aldo Moro”, 70124 Bari, Italy; (S.B.); (C.F.); (L.L.); (R.F.); (F.A.)
| |
Collapse
|
34
|
Weeratunga P, Moller DR, Ho LP. Immune mechanisms of granuloma formation in sarcoidosis and tuberculosis. J Clin Invest 2024; 134:e175264. [PMID: 38165044 PMCID: PMC10760966 DOI: 10.1172/jci175264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
Sarcoidosis is a complex immune-mediated disease characterized by clusters of immune cells called granulomas. Despite major steps in understanding the cause of this disease, many questions remain. In this Review, we perform a mechanistic interrogation of the immune activities that contribute to granuloma formation in sarcoidosis and compare these processes with its closest mimic, tuberculosis, highlighting shared and divergent immune activities. We examine how Mycobacterium tuberculosis is sensed by the immune system; how the granuloma is initiated, formed, and perpetuated in tuberculosis compared with sarcoidosis; and the role of major innate and adaptive immune cells in shaping these processes. Finally, we draw these findings together around several recent high-resolution studies of the granuloma in situ that utilized the latest advances in single-cell technology combined with spatial methods to analyze plausible disease mechanisms. We conclude with an overall view of granuloma formation in sarcoidosis.
Collapse
Affiliation(s)
- Praveen Weeratunga
- MRC Translational Immunology Discovery Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Ling-Pei Ho
- MRC Translational Immunology Discovery Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
35
|
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
|
36
|
Mangione MC, Wen J, Cao DJ. Mechanistic target of rapamycin in regulating macrophage function in inflammatory cardiovascular diseases. J Mol Cell Cardiol 2024; 186:111-124. [PMID: 38039845 PMCID: PMC10843805 DOI: 10.1016/j.yjmcc.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/14/2023] [Accepted: 10/18/2023] [Indexed: 12/03/2023]
Abstract
The mechanistic target of rapamycin (mTOR) is evolutionarily conserved from yeast to humans and is one of the most fundamental pathways of living organisms. Since its discovery three decades ago, mTOR has been recognized as the center of nutrient sensing and growth, homeostasis, metabolism, life span, and aging. The role of dysregulated mTOR in common diseases, especially cancer, has been extensively studied and reported. Emerging evidence supports that mTOR critically regulates innate immune responses that govern the pathogenesis of various cardiovascular diseases. This review discusses the regulatory role of mTOR in macrophage functions in acute inflammation triggered by ischemia and in atherosclerotic cardiovascular disease (ASCVD) and heart failure with preserved ejection fraction (HFpEF), in which chronic inflammation plays critical roles. Specifically, we discuss the role of mTOR in trained immunity, immune senescence, and clonal hematopoiesis. In addition, this review includes a discussion on the architecture of mTOR, the function of its regulatory complexes, and the dual-arm signals required for mTOR activation to reflect the current knowledge state. We emphasize future research directions necessary to understand better the powerful pathway to take advantage of the mTOR inhibitors for innovative applications in patients with cardiovascular diseases associated with aging and inflammation.
Collapse
Affiliation(s)
- MariaSanta C Mangione
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jinhua Wen
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dian J Cao
- Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; VA North Texas Health Care System, Dallas TX 75216, USA.
| |
Collapse
|
37
|
Huang X, Yang X, Xiang L, Chen Y. Serine metabolism in macrophage polarization. Inflamm Res 2024; 73:83-98. [PMID: 38070057 DOI: 10.1007/s00011-023-01815-y] [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: 07/20/2023] [Revised: 10/09/2023] [Accepted: 11/06/2023] [Indexed: 01/10/2024] Open
Abstract
OBJECTIVE Emerging studies have revealed that macrophages possess different dependences on the uptake, synthesis, and metabolism of serine for their activation and functionalization, necessitating our insight into how serine availability and utilization impact macrophage activation and inflammatory responses. METHODS This article summarizes the reports published domestically and internationally about the serine uptake, synthesis, and metabolic flux by the macrophages polarizing with distinct stimuli and under different pathologic conditions, and particularly analyzes how altered serine metabolism rewires the metabolic behaviors of polarizing macrophages and their genetic and epigenetic reprogramming. RESULTS Macrophages dynamically change serine metabolism to orchestrate their anabolism, redox balance, mitochondrial function, epigenetics, and post-translation modification, and thus match the distinct needs for both classical and alternative activation. CONCLUSION Serine metabolism coordinates multiple metabolic pathways to tailor macrophage polarization and their responses to different pathogenic attacks and thus holds the potential as therapeutic target for types of acute and chronic inflammatory diseases.
Collapse
Affiliation(s)
| | - Xue Yang
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, 28 West Changsheng Road, Hengyang, 421001, Hunan, China
| | - Li Xiang
- Hengyang Medical School, Hengyang, China
| | - Yuping Chen
- Hengyang Medical School, Hengyang, China.
- Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, 28 West Changsheng Road, Hengyang, 421001, Hunan, China.
| |
Collapse
|
38
|
Kim JS, Gupta R. Clinical Manifestations and Management of Fibrotic Pulmonary Sarcoidosis. J Clin Med 2023; 13:241. [PMID: 38202248 PMCID: PMC10780222 DOI: 10.3390/jcm13010241] [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: 11/13/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Fibrotic pulmonary sarcoidosis represents a distinct and relatively uncommon manifestation within the spectrum of sarcoidosis and has substantial morbidity and mortality. Due to the scarcity of research focused on this specific disease subtype, our current understanding of pathogenesis and optimal management remains constrained. This knowledge gap underscores the need for further investigation into areas such as targeted therapies, lung transplantation, and quality of life of patients with fibrotic pulmonary sarcoidosis. The primary aim of this review is to discuss recent developments within the realm of fibrotic pulmonary sarcoidosis to foster a more comprehensive understanding of the underlying mechanisms, prognosis, and potential treatment modalities.
Collapse
Affiliation(s)
- Jin Sun Kim
- Department of Thoracic Medicine and Surgery, Temple University Hospital, Philadelphia, PA 19140, USA
| | - Rohit Gupta
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University Hospital, Philadelphia, PA 19140, USA;
| |
Collapse
|
39
|
Nelson NC, Kogan R, Condos R, Hena KM. Emerging Therapeutic Options for Refractory Pulmonary Sarcoidosis: The Evidence and Proposed Mechanisms of Action. J Clin Med 2023; 13:15. [PMID: 38202021 PMCID: PMC10779381 DOI: 10.3390/jcm13010015] [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: 11/14/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 01/12/2024] Open
Abstract
Sarcoidosis is a systemic disease with heterogenous clinical phenotypes characterized by non-necrotizing granuloma formation in affected organs. Most disease either remits spontaneously or responds to corticosteroids and second-line disease-modifying therapies. These medications are associated with numerous toxicities that can significantly impact patient quality-of-life and often limit their long-term use. Additionally, a minority of patients experience chronic, progressive disease that proves refractory to standard treatments. To date, there are limited data to guide the selection of alternative third-line medications for these patients. This review will outline the pathobiological rationale behind current and emerging therapeutic agents for refractory or drug-intolerant sarcoidosis and summarize the existing clinical evidence in support of their use.
Collapse
Affiliation(s)
| | | | | | - Kerry M. Hena
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University, 301 E 17th St Suite 550, New York, NY 10003, USA
| |
Collapse
|
40
|
Do MH, Shi W, Ji L, Ladewig E, Zhang X, Srivastava RM, Capistrano KJ, Edwards C, Malik I, Nixon BG, Stamatiades EG, Liu M, Li S, Li P, Chou C, Xu K, Hsu TW, Wang X, Chan TA, Leslie CS, Li MO. Reprogramming tumor-associated macrophages to outcompete endovascular endothelial progenitor cells and suppress tumor neoangiogenesis. Immunity 2023; 56:2555-2569.e5. [PMID: 37967531 PMCID: PMC11284818 DOI: 10.1016/j.immuni.2023.10.010] [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: 03/11/2023] [Revised: 07/03/2023] [Accepted: 10/18/2023] [Indexed: 11/17/2023]
Abstract
Tumors develop by invoking a supportive environment characterized by aberrant angiogenesis and infiltration of tumor-associated macrophages (TAMs). In a transgenic model of breast cancer, we found that TAMs localized to the tumor parenchyma and were smaller than mammary tissue macrophages. TAMs had low activity of the metabolic regulator mammalian/mechanistic target of rapamycin complex 1 (mTORC1), and depletion of negative regulator of mTORC1 signaling, tuberous sclerosis complex 1 (TSC1), in TAMs inhibited tumor growth in a manner independent of adaptive lymphocytes. Whereas wild-type TAMs exhibited inflammatory and angiogenic gene expression profiles, TSC1-deficient TAMs had a pro-resolving phenotype. TSC1-deficient TAMs relocated to a perivascular niche, depleted protein C receptor (PROCR)-expressing endovascular endothelial progenitor cells, and rectified the hyperpermeable blood vasculature, causing tumor tissue hypoxia and cancer cell death. TSC1-deficient TAMs were metabolically active and effectively eliminated PROCR-expressing endothelial cells in cell competition experiments. Thus, TAMs exhibit a TSC1-dependent mTORC1-low state, and increasing mTORC1 signaling promotes a pro-resolving state that suppresses tumor growth, defining an innate immune tumor suppression pathway that may be exploited for cancer immunotherapy.
Collapse
Affiliation(s)
- Mytrang H Do
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Wei Shi
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Liangliang Ji
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Erik Ladewig
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Xian Zhang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Raghvendra M Srivastava
- Immunogenomics & Precision Oncology Platform (IPOP), Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kristelle J Capistrano
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chaucie Edwards
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Isha Malik
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Briana G Nixon
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Efstathios G Stamatiades
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ming Liu
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Shun Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Peng Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chun Chou
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ke Xu
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Ting-Wei Hsu
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Graduate Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Xinxin Wang
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA
| | - Timothy A Chan
- Immunogenomics & Precision Oncology Platform (IPOP), Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christina S Leslie
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ming O Li
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
41
|
Fritsch SD, Sukhbaatar N, Gonzales K, Sahu A, Tran L, Vogel A, Mazic M, Wilson JL, Forisch S, Mayr H, Oberle R, Weiszmann J, Brenner M, Vanhoutte R, Hofmann M, Pirnes-Karhu S, Magnes C, Kühnast T, Weckwerth W, Bock C, Klavins K, Hengstschläger M, Moissl-Eichinger C, Schabbauer G, Egger G, Pirinen E, Verhelst SHL, Weichhart T. Metabolic support by macrophages sustains colonic epithelial homeostasis. Cell Metab 2023; 35:1931-1943.e8. [PMID: 37804836 DOI: 10.1016/j.cmet.2023.09.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 06/23/2023] [Accepted: 09/14/2023] [Indexed: 10/09/2023]
Abstract
The intestinal epithelium has a high turnover rate and constantly renews itself through proliferation of intestinal crypt cells, which depends on insufficiently characterized signals from the microenvironment. Here, we showed that colonic macrophages were located directly adjacent to epithelial crypt cells in mice, where they metabolically supported epithelial cell proliferation in an mTORC1-dependent manner. Specifically, deletion of tuberous sclerosis complex 2 (Tsc2) in macrophages activated mTORC1 signaling that protected against colitis-induced intestinal damage and induced the synthesis of the polyamines spermidine and spermine. Epithelial cells ingested these polyamines and rewired their cellular metabolism to optimize proliferation and defense. Notably, spermine directly stimulated proliferation of colon epithelial cells and colon organoids. Genetic interference with polyamine production in macrophages altered global polyamine levels in the colon and modified epithelial cell proliferation. Our results suggest that macrophages act as "commensals" that provide metabolic support to promote efficient self-renewal of the colon epithelium.
Collapse
Affiliation(s)
| | - Nyamdelger Sukhbaatar
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Karine Gonzales
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Alishan Sahu
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Department of Pathology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| | - Andrea Vogel
- Institute for Vascular Biology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Mario Mazic
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Jayne Louise Wilson
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Stephan Forisch
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Hannah Mayr
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Raimund Oberle
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Jakob Weiszmann
- Department of Ecogenomics and Systems Biology, University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Martin Brenner
- Department of Ecogenomics and Systems Biology, University of Vienna, Vienna, Austria; Department of Pharmaceutical Sciences/ Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Roeland Vanhoutte
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Melanie Hofmann
- Institute for Vascular Biology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Sini Pirnes-Karhu
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Christoph Magnes
- HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Graz, Austria
| | - Torben Kühnast
- Diagnostic and Research Department of Microbiology, Hygiene and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Wolfram Weckwerth
- Department of Ecogenomics and Systems Biology, University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Institute of Artificial Intelligence, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Kristaps Klavins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Markus Hengstschläger
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Christine Moissl-Eichinger
- Diagnostic and Research Department of Microbiology, Hygiene and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Gernot Schabbauer
- Institute for Vascular Biology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Steven H L Verhelst
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Thomas Weichhart
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
42
|
Wu W, Xia X, Tang L, Luo J, Xiong S, Ma G, Lei H. Phosphoinositide 3-kinase as a therapeutic target in angiogenic disease. Exp Eye Res 2023; 236:109646. [PMID: 37716399 DOI: 10.1016/j.exer.2023.109646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023]
Abstract
Phosphoinositide 3-kinases (PI3Ks) generate lipids that control multitudinous intracellular cell signaling events which participate in cell survival and proliferation. In addition, PI3K signaling also contributes to metabolism, immunity, angiogenesis and cardiovascular homeostasis, and many diseases. The diverse actions of PI3K stem from the existence of their various isoforms and a variety of protein effectors. Hence, PI3K isoform-specific inhibitors have already achieved a wonderful effect on treating cancer. Herein, we summarize the molecular mechanism of PI3K inhibitors in preventing the permeability of vessels and neovascularization. Additionally, we briefly illustrate how PI3K signaling modulates blood vessel growth and discuss the different roles that PI3K isoforms play in angiogenesis.
Collapse
Affiliation(s)
- Wenyi Wu
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Ophthalmology, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Xiaobo Xia
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Ophthalmology, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Luosheng Tang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Luo
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siqi Xiong
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Ophthalmology, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Gaoen Ma
- Department of Ophthalmology, The First Affiliated Hospital of Hainan Medical University, Haikou, 571199, China.
| | - Hetian Lei
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China.
| |
Collapse
|
43
|
Bueno‐Beti C, Lim CX, Protonotarios A, Szabo PL, Westaby J, Mazic M, Sheppard MN, Behr E, Hamza O, Kiss A, Podesser BK, Hengstschläger M, Weichhart T, Asimaki A. An mTORC1-Dependent Mouse Model for Cardiac Sarcoidosis. J Am Heart Assoc 2023; 12:e030478. [PMID: 37750561 PMCID: PMC10727264 DOI: 10.1161/jaha.123.030478] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 08/15/2023] [Indexed: 09/27/2023]
Abstract
Background Sarcoidosis is an inflammatory, granulomatous disease of unknown cause affecting multiple organs, including the heart. Untreated, unresolved granulomatous inflammation can lead to cardiac fibrosis, arrhythmias, and eventually heart failure. Here we characterize the cardiac phenotype of mice with chronic activation of mammalian target of rapamycin (mTOR) complex 1 signaling in myeloid cells known to cause spontaneous pulmonary sarcoid-like granulomas. Methods and Results The cardiac phenotype of mice with conditional deletion of the tuberous sclerosis 2 (TSC2) gene in CD11c+ cells (TSC2fl/flCD11c-Cre; termed TSC2KO) and controls (TSC2fl/fl) was determined by histological and immunological stains. Transthoracic echocardiography and invasive hemodynamic measurements were performed to assess myocardial function. TSC2KO animals were treated with either everolimus, an mTOR inhibitor, or Bay11-7082, a nuclear factor-kB inhibitor. Activation of mTOR signaling was evaluated on myocardial samples from sudden cardiac death victims with a postmortem diagnosis of cardiac sarcoidosis. Chronic activation of mTORC1 signaling in CD11c+ cells was sufficient to initiate progressive accumulation of granulomatous infiltrates in the heart, which was associated with increased fibrosis, impaired cardiac function, decreased plakoglobin expression, and abnormal connexin 43 distribution, a substrate for life-threatening arrhythmias. Mice treated with the mTOR inhibitor everolimus resolved granulomatous infiltrates, prevented fibrosis, and improved cardiac dysfunction. In line, activation of mTOR signaling in CD68+ macrophages was detected in the hearts of sudden cardiac death victims who suffered from cardiac sarcoidosis. Conclusions To our best knowledge this is the first animal model of cardiac sarcoidosis that recapitulates major pathological hallmarks of human disease. mTOR inhibition may be a therapeutic option for patients with cardiac sarcoidosis.
Collapse
Affiliation(s)
- Carlos Bueno‐Beti
- Clinical Cardiology Academic Group, Molecular and Clinical Research Science InstituteSt George’s University of LondonLondonUnited Kingdom
| | - Clarice X. Lim
- Center for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Alexandros Protonotarios
- Institute of Cardiovascular Science, Clinical Science Research GroupUniversity College LondonLondonUnited Kingdom
| | - Petra Lujza Szabo
- Center for Biomedical ResearchMedical University of ViennaViennaAustria
| | - Joseph Westaby
- Clinical Cardiology Academic Group, Molecular and Clinical Research Science InstituteSt George’s University of LondonLondonUnited Kingdom
| | - Mario Mazic
- Center for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Mary N. Sheppard
- Clinical Cardiology Academic Group, Molecular and Clinical Research Science InstituteSt George’s University of LondonLondonUnited Kingdom
| | - Elijah Behr
- Clinical Cardiology Academic Group, Molecular and Clinical Research Science InstituteSt George’s University of LondonLondonUnited Kingdom
| | - Ouafa Hamza
- Center for Biomedical ResearchMedical University of ViennaViennaAustria
| | - Attila Kiss
- Center for Biomedical ResearchMedical University of ViennaViennaAustria
| | - Bruno K. Podesser
- Center for Biomedical ResearchMedical University of ViennaViennaAustria
| | | | - Thomas Weichhart
- Center for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Angeliki Asimaki
- Clinical Cardiology Academic Group, Molecular and Clinical Research Science InstituteSt George’s University of LondonLondonUnited Kingdom
| |
Collapse
|
44
|
Bandyopadhyay D, Mirsaeidi MS. Sarcoidosis-associated pulmonary fibrosis: joining the dots. Eur Respir Rev 2023; 32:230085. [PMID: 37758275 PMCID: PMC10523156 DOI: 10.1183/16000617.0085-2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/10/2023] [Indexed: 09/30/2023] Open
Abstract
Sarcoidosis is a multisystem granulomatous disorder of unknown aetiology. A minority of patients with sarcoidosis develop sarcoidosis-associated pulmonary fibrosis (SAPF), which may become progressive. Genetic profiles differ between patients with progressive and self-limiting disease. The mechanisms of fibrosis in SAPF are not fully understood, but SAPF is likely a distinct clinicopathological entity, rather than a continuum of acute inflammatory sarcoidosis. Risk factors for the development of SAPF have been identified; however, at present, it is not possible to make a robust prediction of risk for an individual patient. The bulk of fibrotic abnormalities in SAPF are located in the upper and middle zones of the lungs. A greater extent of SAPF on imaging is associated with a worse prognosis. Patients with SAPF are typically treated with corticosteroids, second-line agents such as methotrexate or azathioprine, or third-line agents such as tumour necrosis factor inhibitors. The antifibrotic drug nintedanib is an approved treatment for slowing the decline in lung function in patients with progressive fibrosing interstitial lung diseases, but more evidence is needed to assess its efficacy in SAPF. The management of patients with SAPF should include the identification and treatment of complications such as bronchiectasis and pulmonary hypertension. Further research is needed into the mechanisms underlying SAPF and biomarkers that predict its clinical course.
Collapse
Affiliation(s)
| | - Mehdi S Mirsaeidi
- Division of Pulmonary and Critical Care, University of Florida, Jacksonville, FL, USA
| |
Collapse
|
45
|
Celada SI, Lim CX, Carisey AF, Ochsner SA, Arce Deza CF, Rexie P, Poli De Frias F, Cardenas-Castillo R, Polverino F, Hengstschläger M, Tsoyi K, McKenna NJ, Kheradmand F, Weichhart T, Rosas IO, Van Kaer L, Celada LJ. SHP2 promotes sarcoidosis severity by inhibiting SKP2-targeted ubiquitination of TBET in CD8 + T cells. Sci Transl Med 2023; 15:eade2581. [PMID: 37703351 PMCID: PMC11126869 DOI: 10.1126/scitranslmed.ade2581] [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: 08/04/2022] [Accepted: 08/24/2023] [Indexed: 09/15/2023]
Abstract
Sarcoidosis is an interstitial lung disease (ILD) characterized by interferon-γ (IFN-γ) and T-box expressed in T cells (TBET) dysregulation. Although one-third of patients progress from granulomatous inflammation to severe lung damage, the molecular mechanisms underlying this process remain unclear. Here, we found that pharmacological inhibition of phosphorylated SH2-containing protein tyrosine phosphatase-2 (pSHP2), a facilitator of aberrant IFN-γ abundance, decreased large granuloma formation and macrophage infiltration in the lungs of mice with sarcoidosis-like disease. Positive treatment outcomes were dependent on the effective enhancement of TBET ubiquitination within CD8+ T cells. Mechanistically, we identified a posttranslational modification pathway in which the E3 F-box protein S-phase kinase-associated protein 2 (SKP2) targets TBET for ubiquitination in T cells under normal conditions. However, this pathway was disrupted by aberrant pSHP2 signaling in CD8+ T cells from patients with progressive pulmonary sarcoidosis and end-stage disease. Ex vivo inhibition of pSHP2 in CD8+ T cells from patients with end-stage sarcoidosis enhanced TBET ubiquitination and suppressed IFN-γ and collagen synthesis. Therefore, these studies provided new mechanistic insights into the SHP2-dependent posttranslational regulation of TBET and identified SHP2 inhibition as a potential therapeutic intervention against severe sarcoidosis. Furthermore, these studies also suggest that the small-molecule SHP2 inhibitor SHP099 might be used as a therapeutic measure against human diseases linked to TBET or ubiquitination.
Collapse
Affiliation(s)
- Sherly I. Celada
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Clarice X. Lim
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Alexandre F. Carisey
- William T. Shearer Center for Human Immunobiology, Texas Children’s Hospital, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Cell and Molecular Biology, St. Jude Children’s Hospital, Memphis, TN 38105, USA
| | - Scott A. Ochsner
- Department of Molecular and Cellular Biology, Houston, TX 77030, USA
| | - Carlos F. Arce Deza
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Praveen Rexie
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Fernando Poli De Frias
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Mout Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Rafael Cardenas-Castillo
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Francesca Polverino
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Markus Hengstschläger
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Konstantin Tsoyi
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Neil J. McKenna
- Department of Molecular and Cellular Biology, Houston, TX 77030, USA
| | - Farrah Kheradmand
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey, Houston, TX 77030, USA
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| | - Ivan O. Rosas
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| | - Lindsay J. Celada
- Department of Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| |
Collapse
|
46
|
Hwang E, Abdelghaffar M, Shields BE, Damsky W. Molecularly Targeted Therapies for Inflammatory Cutaneous Granulomatous Disorders: A Review of the Evidence and Implications for Understanding Disease Pathogenesis. JID INNOVATIONS 2023; 3:100220. [PMID: 37719661 PMCID: PMC10500476 DOI: 10.1016/j.xjidi.2023.100220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 09/19/2023] Open
Abstract
Inflammatory cutaneous granulomatous diseases, including granuloma annulare, cutaneous sarcoidosis, and necrobiosis lipoidica, are distinct diseases unified by the hallmark of macrophage accumulation and activation in the skin. There are currently no Food and Drug Administration-approved therapies for these conditions except prednisone and repository corticotropin injection for pulmonary sarcoidosis. Treatment of these diseases has generally been guided by low-quality evidence and may involve broadly immunomodulatory medications. Development of new treatments has in part been limited by an incomplete understanding of disease pathogenesis. Recently, there has been substantial progress in better understanding the molecular pathogenesis of these disorders, opening the door for therapeutic innovation. Likewise, reported outcomes of treatment with immunologically targeted therapies may offer insights into disease pathogenesis. In this systematic review, we summarize progress in deciphering the pathomechanisms of these disorders and discuss this in the context of emerging evidence on the use of molecularly targeted therapies in treatment of these diseases.
Collapse
Affiliation(s)
- Erica Hwang
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Mariam Abdelghaffar
- School of Medicine, Royal College of Surgeons in Ireland, Busaiteen, Bahrain
| | - Bridget E. Shields
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
47
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [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
|
48
|
Daoud A, Lema DA, Won T, Čiháková D. Integrative single-cell analysis of cardiac and pulmonary sarcoidosis using publicly available cardiac and bronchoalveolar lavage fluid sequencing datasets. Front Cardiovasc Med 2023; 10:1227818. [PMID: 37576111 PMCID: PMC10419306 DOI: 10.3389/fcvm.2023.1227818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/03/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Cardiac presentation of autoimmune sarcoidosis, known as cardiac sarcoidosis (CS), is a poorly understood disease with high mortality and low diagnosis rate. While CS is an immunological syndrome, little is known about how cardiac parenchymal and stromal cells mediate its pathogenesis. Moreover, while most current sarcoidosis research is based on research in pulmonary sarcoidosis (PS), it remains unclear how much both presentations of sarcoidosis overlap. To tackle these concerns, we leveraged publicly available sarcoidosis transcriptomic datasets. Methods Two publicly available bronchoalveolar lavage single-cell RNA sequencing datasets were integrated to analyze PS relative to control. Additionally, two publicly available cardiac single-nucleus RNA sequencing datasets were integrated to analyze CS relative to control. Following integration, we ran cell-cell communication, transcription factor, and differential expression analyses on parenchymal, stromal, and immune subsets identified in our analysis. Results Our analysis revealed that there was an expansion of stromal and immune cells in PS and CS. We also observed upregulation of Th17.1 and attenuated activation transcriptional profiles in the immune cells of CS and PS relative to control. Additionally, we found upregulation of pro-inflammatory and pro-fibrotic transcriptional profiles in the cardiac stromal cells of CS relative to control. We also found that cardiomyocytes exhibited upregulated cardiac stress and proliferation transcriptional profiles in CS relative to control. Conclusions Our integrative transcriptomic analysis shows that despite tissue-specific differences, there are shared transcriptional trends between CS and PS. It also shows that stromal and parenchymal populations exhibit transcriptional trends that could explain their pathogenic role in CS.
Collapse
Affiliation(s)
- Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Diego A. Lema
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
49
|
He J, Liu S, Guo X, Zhang F, Fan Y, Wu L, Takiff HE, Zhao Y. Association of PI3K/AKT/mTOR pathway autophagy-related gene polymorphisms with pulmonary tuberculosis susceptibility in a Chinese population. Rev Soc Bras Med Trop 2023; 56:e01042023. [PMID: 37493735 PMCID: PMC10367219 DOI: 10.1590/0037-8682-0104-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Autophagy can inhibit the survival of intracellular microorganisms including Mycobacterium tuberculosis (Mtb), and the PI3K/AKT/mTOR pathway plays a crucial role. This study investigated the association between PI3K/AKT/mTOR pathway autophagy-related gene polymorphisms and pulmonary tuberculosis (PTB) susceptibility. METHODS KEGG pathway and gene ontology (GO) databases were searched for genes belonging to the PI3K/AKT/mTOR and autophagy pathways. Thirty SNPs in nine genes were identified and tested for their associations with tuberculosis in 130 patients with PTB and 271 controls. We constructed genetic risk scores (GRSs) and divided the participants into 3 subgroups based on their GRSs:0-5, 6-10, and 11-16. RESULTS This analysis revealed that the AKT1 (rs12432802), RPTOR (rs11654508, rs12602885, rs2090204, rs2589144, and rs2672897), and TSC2 (rs2074969) polymorphisms were significantly associated with PTB risk. A decreasing trend was observed (P trend 0.020), in which a lower GRS was associated with a higher risk of PTB ([6-10] vs. [0-5]: OR (95%CI) 0.590 (0.374-0.931); [11-16] vs. [0-5]: OR (95%CI) 0.381 (0.160-0.906)). CONCLUSIONS Polymorphisms in AKT1, RPTOR, and TSC2 may influence susceptibility to PTB.
Collapse
Affiliation(s)
- Juan He
- Harbin Medical University, School of Public Health, Department of Epidemiology, Harbin, China
- Shenzhen Nanshan Center for Chronic Disease Control, Department of Tuberculosis Control and Prevention, Shenzhen, China
| | - Shengyuan Liu
- Shenzhen Nanshan Center for Chronic Disease Control, Department of Tuberculosis Control and Prevention, Shenzhen, China
| | - Xujun Guo
- Shenzhen Nanshan Center for Chronic Disease Control, Department of Tuberculosis Control and Prevention, Shenzhen, China
| | - Fan Zhang
- Shenzhen Nanshan Center for Chronic Disease Control, Department of Tuberculosis Control and Prevention, Shenzhen, China
| | - Yuzheng Fan
- Shenzhen Nanshan Center for Chronic Disease Control, Department of Tuberculosis Control and Prevention, Shenzhen, China
| | - Lijuan Wu
- Shenzhen Nanshan Center for Chronic Disease Control, Department of Tuberculosis Control and Prevention, Shenzhen, China
| | | | - Yashuang Zhao
- Harbin Medical University, School of Public Health, Department of Epidemiology, Harbin, China
| |
Collapse
|
50
|
Linkova N, Diatlova A, Zinchenko Y, Kornilova A, Snetkov P, Morozkina S, Medvedev D, Krasichkov A, Polyakova V, Yablonskiy P. Pulmonary Sarcoidosis: Experimental Models and Perspectives of Molecular Diagnostics Using Quantum Dots. Int J Mol Sci 2023; 24:11267. [PMID: 37511027 PMCID: PMC10379333 DOI: 10.3390/ijms241411267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Sarcoidosis is a complex inflammatory multisystem disease of unknown etiology that is characterised by epithelioid cell granulomatous lesions affecting various organs, mainly the lungs. In general, sarcoidosis is asymptomatic, but some cases result in severe complications and organ failure. So far, no accurate and validated modelling for clinical and pathohistological manifestations of sarcoidosis is suggested. Moreover, knowledge about disease-specific diagnostic markers for sarcoidosis is scarce. For instance, pulmonary granulomatosis is associated with the upregulated production of proinflammatory molecules: TNF-α, IL-6, CXCL1, CCL2, CCL18, CD163, serum angiotensin-converting enzyme (sACE), lysozyme, neopterin, and serum amyloid A (SAA). Quantum dots (QDs) are widely applied for molecular diagnostics of various diseases. QDs are semiconductor nanoparticles of a few nanometres in size, made from ZnS, CdS, ZnSe, etc., with unique physical and chemical properties that are useful for the labelling and detection in biological experiments. QDs can conjugate with various antibodies or oligonucleotides, allowing for high-sensitivity detection of various targets in organs and cells. Our review describes existing experimental models for sarcoidosis (in vitro, in vivo, and in silico), their advantages and restrictions, as well as the physical properties of quantum dots and their potential applications in the molecular diagnostics of sarcoidosis. The most promising experimental models include mice with TSC2 deletion and an in silico multiscale computational model of sarcoidosis (SarcoidSim), developed using transcriptomics and flow cytometry of human sarcoid biopsies. Both models are most efficient to test different candidate drugs for sarcoidosis.
Collapse
Affiliation(s)
- Natalia Linkova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2-4, 191036 Saint Petersburg, Russia
- St. Petersburg Institute of Bioregulation and Gerontology, Dynamo pr., 3, 197110 Saint Petersburg, Russia
| | - Anastasiia Diatlova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2-4, 191036 Saint Petersburg, Russia
| | - Yulia Zinchenko
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2-4, 191036 Saint Petersburg, Russia
| | - Anastasiia Kornilova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2-4, 191036 Saint Petersburg, Russia
| | - Petr Snetkov
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2-4, 191036 Saint Petersburg, Russia
- Chemical Bioengineering Center, ITMO University, Kronverksky Pr, 49A, 197101 Saint Petersburg, Russia
| | - Svetlana Morozkina
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2-4, 191036 Saint Petersburg, Russia
- Chemical Bioengineering Center, ITMO University, Kronverksky Pr, 49A, 197101 Saint Petersburg, Russia
| | - Dmitrii Medvedev
- St. Petersburg Institute of Bioregulation and Gerontology, Dynamo pr., 3, 197110 Saint Petersburg, Russia
| | - Alexandr Krasichkov
- Department of Radio Engineering Systems, Electrotechnical University "LETI", Prof. Popova Street 5F, 197022 Saint Petersburg, Russia
| | - Victoria Polyakova
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2-4, 191036 Saint Petersburg, Russia
| | - Piotr Yablonskiy
- St. Petersburg Research Institute of Phthisiopulmonology, Ligovskii Prospect, 2-4, 191036 Saint Petersburg, Russia
- Department of Hospital Surgery of the Faculty of Medicine, St. Petersburg State University, University Embankment, 7-9, 199034 Saint Petersburg, Russia
| |
Collapse
|