1
|
Dorjay Tamang JS, Banerjee S, Baidya SK, Das S, Ghosh B, Jha T, Adhikari N. An overview of matrix metalloproteinase-12 in multiple disease conditions, potential selective inhibitors, and drug designing strategies. Eur J Med Chem 2025; 283:117154. [PMID: 39709794 DOI: 10.1016/j.ejmech.2024.117154] [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: 07/09/2024] [Revised: 11/08/2024] [Accepted: 12/06/2024] [Indexed: 12/24/2024]
Abstract
Matrix metalloproteases (MMPs) are the proteolytic enzymes accountable for extracellular matrix (ECM) modification through their Zn2+-dependent catalytic activity. Among these, MMP-12 is one of the crucial MMPs that contributes to various disease states including different types of cancers and other major pathophysiological conditions including COPD, asthma, emphysema, skin diseases, arthritis, vascular diseases, and neurological disorders. The majority of the MMP-12 inhibitors should have three constitutional pharmacophoric features (i.e., a hydrophobic group to occupy the S1' pocket, a zinc-binding motif for chelating to the catalytic Zn2+ ion present at the catalytic site, and a flexible and hydrogen bond forming linker region between the S1' pocket substituent and the zinc chelating group for interacting with the catalytic and Ω-loop amino acid residues). This review mainly focuses on the various roles of MMP-12 in different diseases along with the structural comparison with other MMPs as well as promising and MMP-12-selective inhibitors and molecular modeling studies performed on MMP-12 inhibitors. Therefore, this review will provide comprehensive information to the researchers for designing effective and MMP-12-selective inhibitors for therapeutic advancement in the future.
Collapse
Affiliation(s)
- Jigme Sangay Dorjay Tamang
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Suvankar Banerjee
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Sandip Kumar Baidya
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Sanjib Das
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani Hyderabad Campus, Shamirpet, Hyderabad, 500078, India
| | - Tarun Jha
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India.
| | - Nilanjan Adhikari
- Natural Science Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India.
| |
Collapse
|
2
|
Watanabe N, Hara Y, Nishito Y, Kounoe M, Sekiyama K, Takamasu E, Kise T, Chinen N, Shimada K, Sugihara M, Kawaji H. Tissue degrading and remodelling molecules in giant cell arteritis. Rheumatology (Oxford) 2025:keae710. [PMID: 39837478 DOI: 10.1093/rheumatology/keae710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVES GCA is a granulomatous vasculitis affecting large vessels, leading to intimal occlusion accompanied by the accumulation of myofibroblasts. Histopathologically, GCA is characterized by destruction of the tunica media and hypertrophy of the intima with invasion of activated CD4+ T cells, macrophages and multinucleated giant cells (MNGCs). Despite these well-defined histopathological features, the molecular pathology of GCA has largely remained elusive. We aimed to characterize the pathologic features of GCA at the molecular level. METHODS To identify key molecules involved in GCA pathogenesis, we conducted genome-wide gene expression profiling on arterial lesions obtained through temporal artery biopsy of 16 patients who had not received any prior treatment. The resulting data were examined to reveal specific pathways and genes, and some of the molecules were followed up by immunohistochemistry. RESULTS Our analysis revealed a unique gene expression pattern in GCA lesions, including enrichment of immune cells and phagocytic pathways related to microglia and osteoclasts. Subsequent immunohistochemistry analysis identified the presence of MMP12 (macrophage elastase), HLA-DRA, and phagocytosis- and osteoclast-associated molecules in infiltrating macrophages and MNGCs. Additionally, we discovered LRRC15-expressing cells in the tunica intima, suggesting a myofibroblast subpopulation that suppresses cytotoxic CD8+ T cells. These molecules were upregulated in other granulomatous diseases affecting not only arteries but also lymph nodes. CONCLUSION Our study revealed novel molecules associated with the pathological features of GCA, providing a foundation for better understanding of GCA pathogenesis and development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Nobumasa Watanabe
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yuichiro Hara
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yasumasa Nishito
- Center for Basic Technology Research, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mai Kounoe
- Center for Medical Research Cooperation, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazunari Sekiyama
- Center for Medical Research Cooperation, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Eisuke Takamasu
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Tokyo, Japan
| | - Takayasu Kise
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Tokyo, Japan
| | - Naofumi Chinen
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama-Nambu Chiiki Hospital, Tokyo, Japan
| | - Kota Shimada
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama Medical Center, Tokyo, Japan
| | - Makoto Sugihara
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Department of Rheumatic Diseases, Tokyo Metropolitan Tama-Hokubu Medical Center, Tokyo, Japan
| | - Hideya Kawaji
- Research Center for Genome & Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
3
|
Habib I, Jawed JJ, Nasrin T, Shaikh S. Briefing of pulmonary sarcoidosis: Reduction-oxidation, misleading and possibilities. Indian J Tuberc 2025; 72:103-111. [PMID: 39890360 DOI: 10.1016/j.ijtb.2024.07.003] [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: 03/15/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 02/03/2025]
Abstract
Sarcoidosis is an inflammatory disease with limited treatment strategies and is characterized by the presence of abnormal lumps (granulomas) of the inflammatory cells. Among the types, pulmonary sarcoidosis most commonly occurs (about 90%), affecting the lungs and intrathoracic lymph nodes. Although the cause of its occurrence is still unknown, perhaps microbes and chemical exposures, as well as genetic history, may trigger the disease occurrence. The updated scenario also depicted the interconnection between oxidative stress and pulmonary sarcoidosis. Thus, the therapeutic value of the genetic consequences, as well as the redox status of pulmonary sarcoidosis, are under consideration. In addition, sarcoidosis complexity has been associated with tumor malignancy and tuberculosis. Therefore, in this review, we summarized the current status of pulmonary sarcoidosis, interference of lung cancer and tuberculosis complications, understanding of the role of reactive species in disease occurrence, and how they are associated with genetic features.
Collapse
Affiliation(s)
- Irfan Habib
- Department of Internal Medicine, College of Medicine and JNM Hospital, WB, India
| | - Junaid Jibran Jawed
- Institute of Health Sciences, Presidency University-2nd Campus, DG/02/02, New Town, Rajarhat, Kolkata, 700156, India
| | - Tina Nasrin
- Dept. of Electrical and Computer Engineering, University of Cyprus, Nicosia, Cyprus
| | - Soni Shaikh
- Laboratory of Histopathology, TATA MEDICAL CENTER 14, MAR (E-W), New Town, Rajarhat, Kolkata, 700160, India.
| |
Collapse
|
4
|
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
|
5
|
Amor M, Bianco V, Buerger M, Lechleitner M, Vujić N, Dobrijević A, Akhmetshina A, Pirchheim A, Schwarz B, Pessentheiner AR, Baumgartner F, Rampitsch K, Schauer S, Klobučar I, Degoricija V, Pregartner G, Kummer D, Svecla M, Sommer G, Kolb D, Holzapfel GA, Hoefler G, Frank S, Norata GD, Kratky D. Genetic deletion of MMP12 ameliorates cardiometabolic disease by improving insulin sensitivity, systemic inflammation, and atherosclerotic features in mice. Cardiovasc Diabetol 2023; 22:327. [PMID: 38017481 PMCID: PMC10685620 DOI: 10.1186/s12933-023-02064-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Matrix metalloproteinase 12 (MMP12) is a macrophage-secreted protein that is massively upregulated as a pro-inflammatory factor in metabolic and vascular tissues of mice and humans suffering from cardiometabolic diseases (CMDs). However, the molecular mechanisms explaining the contributions of MMP12 to CMDs are still unclear. METHODS We investigated the impact of MMP12 deficiency on CMDs in a mouse model that mimics human disease by simultaneously developing adipose tissue inflammation, insulin resistance, and atherosclerosis. To this end, we generated and characterized low-density lipoprotein receptor (Ldlr)/Mmp12-double knockout (DKO) mice fed a high-fat sucrose- and cholesterol-enriched diet for 16-20 weeks. RESULTS DKO mice showed lower cholesterol and plasma glucose concentrations and improved insulin sensitivity compared with LdlrKO mice. Untargeted proteomic analyses of epididymal white adipose tissue revealed that inflammation- and fibrosis-related pathways were downregulated in DKO mice. In addition, genetic deletion of MMP12 led to alterations in immune cell composition and a reduction in plasma monocyte chemoattractant protein-1 in peripheral blood which indicated decreased low-grade systemic inflammation. Aortic en face analyses and staining of aortic valve sections demonstrated reduced atherosclerotic plaque size and collagen content, which was paralleled by an improved relaxation pattern and endothelial function of the aortic rings and more elastic aortic sections in DKO compared to LdlrKO mice. Shotgun proteomics revealed upregulation of anti-inflammatory and atheroprotective markers in the aortas of DKO mice, further supporting our data. In humans, MMP12 serum concentrations were only weakly associated with clinical and laboratory indicators of CMDs. CONCLUSION We conclude that the genetic deletion of MMP12 ameliorates obesity-induced low-grade inflammation, white adipose tissue dysfunction, biomechanical properties of the aorta, and the development of atherosclerosis. Therefore, therapeutic strategies targeting MMP12 may represent a promising approach to combat CMDs.
Collapse
Affiliation(s)
- Melina Amor
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Valentina Bianco
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Martin Buerger
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Margarete Lechleitner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Anja Dobrijević
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
- Institute for Vascular Biology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Alena Akhmetshina
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Birgit Schwarz
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Ariane R Pessentheiner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
- Institute for Molecular Biosciences, University of Graz, Graz, Austria
| | | | | | - Silvia Schauer
- Diagnostics and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Iva Klobučar
- Sisters of Charity, University Hospital Centre, Zagreb, Croatia
| | - Vesna Degoricija
- University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Medicine, Sisters of Charity, University Hospital Centre, Zagreb, Croatia
| | - Gudrun Pregartner
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Daniel Kummer
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Monika Svecla
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Gerhard Sommer
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Dagmar Kolb
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
- Core Facility Ultrastructural Analysis, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Gerhard A Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Department of Structural Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Gerald Hoefler
- Diagnostics and Research Institute of Pathology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Saša Frank
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
- BioTechMed-Graz, Graz, Austria
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
6
|
Ma X, Zhang Y, Jiang J, Ru Y, Luo Y, Luo Y, Fei X, Song J, Ma X, Li B, Tan Y, Kuai L. Metabolism-related biomarkers, molecular classification, and immune infiltration in diabetic ulcers with validation. Int Wound J 2023; 20:3498-3513. [PMID: 37245869 PMCID: PMC10588317 DOI: 10.1111/iwj.14223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 04/21/2023] [Indexed: 05/30/2023] Open
Abstract
Diabetes mellitus (DM) can lead to diabetic ulcers (DUs), which are the most severe complications. Due to the need for more accurate patient classifications and diagnostic models, treatment and management strategies for DU patients still need improvement. The difficulty of diabetic wound healing is caused closely related to biological metabolism and immune chemotaxis reaction dysfunction. Therefore, the purpose of our study is to identify metabolic biomarkers in patients with DU and construct a molecular subtype-specific prognostic model that is highly accurate and robust. RNA-sequencing data for DU samples were obtained from the Gene Expression Omnibus (GEO) database. DU patients and normal individuals were compared regarding the expression of metabolism-related genes (MRGs). Then, a novel diagnostic model based on MRGs was constructed with the random forest algorithm, and classification performance was evaluated utilizing receiver operating characteristic (ROC) analysis. The biological functions of MRGs-based subtypes were investigated using consensus clustering analysis. A principal component analysis (PCA) was conducted to determine whether MRGs could distinguish between subtypes. We also examined the correlation between MRGs and immune infiltration. Lastly, qRT-PCR was utilized to validate the expression of the hub MRGs with clinical validations and animal experimentations. Firstly, 8 metabolism-related hub genes were obtained by random forest algorithm, which could distinguish the DUs from normal samples validated by the ROC curves. Secondly, DU samples could be consensus clustered into three molecular classifications by MRGs, verified by PCA analysis. Thirdly, associations between MRGs and immune infiltration were confirmed, with LYN and Type 1 helper cell significantly positively correlated; RHOH and TGF-β family remarkably negatively correlated. Finally, clinical validations and animal experiments of DU skin tissue samples showed that the expressions of metabolic hub genes in the DU groups were considerably upregulated, including GLDC, GALNT6, RHOH, XDH, MMP12, KLK6, LYN, and CFB. The current study proposed an auxiliary MRGs-based DUs model while proposing MRGs-based molecular clustering and confirmed the association with immune infiltration, facilitating the diagnosis and management of DU patients and designing individualized treatment plans.
Collapse
Affiliation(s)
- Xiao‐Xuan Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| | - Ying Zhang
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| | - Jing‐Si Jiang
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| | - Yue Luo
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xiao‐Ya Fei
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jian‐Kun Song
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xin Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Bin Li
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yi‐Mei Tan
- Shanghai Skin Disease Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of DermatologyShanghai Academy of Traditional Chinese MedicineshanghaiChina
| |
Collapse
|
7
|
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
|
8
|
Single-cell and spatial transcriptomics reveal aberrant lymphoid developmental programs driving granuloma formation. Immunity 2023; 56:289-306.e7. [PMID: 36750099 PMCID: PMC9942876 DOI: 10.1016/j.immuni.2023.01.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 08/27/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023]
Abstract
Granulomas are lumps of immune cells that can form in various organs. Most granulomas appear unstructured, yet they have some resemblance to lymphoid organs. To better understand granuloma formation, we performed single-cell sequencing and spatial transcriptomics on granulomas from patients with sarcoidosis and bioinformatically reconstructed the underlying gene regulatory networks. We discovered an immune stimulatory environment in granulomas that repurposes transcriptional programs associated with lymphoid organ development. Granuloma formation followed characteristic spatial patterns and involved genes linked to immunometabolism, cytokine and chemokine signaling, and extracellular matrix remodeling. Three cell types emerged as key players in granuloma formation: metabolically reprogrammed macrophages, cytokine-producing Th17.1 cells, and fibroblasts with inflammatory and tissue-remodeling phenotypes. Pharmacological inhibition of one of the identified processes attenuated granuloma formation in a sarcoidosis mouse model. We show that human granulomas adopt characteristic aspects of normal lymphoid organ development in aberrant combinations, indicating that granulomas constitute aberrant lymphoid organs.
Collapse
|
9
|
Different Molecular Features of Epithelioid and Giant Cells in Foreign Body Reaction Identified by Single-Cell RNA Sequencing. J Invest Dermatol 2022; 142:3232-3242.e16. [PMID: 35853485 DOI: 10.1016/j.jid.2022.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/18/2022] [Accepted: 06/21/2022] [Indexed: 01/05/2023]
Abstract
Although macrophage‒epithelioid cell (EPC)‒giant cell (GC) differentiation is acknowledged in foreign body reaction (FBR), the exact molecular features remain elusive. To discover the molecular profiles of EPC and GC, we analyzed mouse sponge and silk FBRs by integrating single-cell RNA sequencing and spatial sequencing, which identified seven cell types, including macrophages and fibroblasts. Macrophages comprised three subsets with a trajectory from M2-like cell to EPC to GC. They were different in many aspects, including cytokine, extracellular matrix organization/degradation, epithelial modules, and glycolysis that were consistent in both sponge and silk FBRs. EPCs exhibited epithelial modules and extracellular matrix organization, and GCs showed glycolysis, extracellular matrix degradation, and cell fusion signatures. Cellular interactions in GCs and M2-like cells were predicted to be higher than that in EPCs. High expression of inflammation or fusion-related (GPNMB, matrix metalloproteinase 12 gene MMP12, DCSTAMP) and glycolysis-related (PGAM1, ALDOA) genes was identified in GCs of human/mouse tissues, suggesting them as GC-specific markers. Our study identified unique signatures of EPCs and GCs in FBR. Importantly, GCs showed strong glycolysis signatures and cellular interactions, suggesting their activation in FBR. Our data on EPC and GC refinement and GC-specific markers enable the understanding of FBR and help to explore preventive and therapeutic management strategies for skin FBRs.
Collapse
|
10
|
Tsunematsu T, Arakaki R, Sato M, Saito M, Otsuka K, Furukawa Y, Taquahashi Y, Kanno J, Ishimaru N. Exposure to Multiwall Carbon Nanotubes Promotes Fibrous Proliferation by Production of Matrix Metalloproteinase-12 via NF-κB Activation in Chronic Peritonitis. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1559-1572. [PMID: 35963465 DOI: 10.1016/j.ajpath.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 07/15/2022] [Accepted: 07/27/2022] [Indexed: 06/15/2023]
Abstract
The toxicologic effects of nanomaterials, such as carbon nanotubes (CNTs), on the immune system are understood well. However, the precise relationship between long-term exposure to CNTs and chronic inflammation remains unclear. In this study, a mouse model of chronic peritonitis was established using i.p. injection of multiwalled CNTs treated by the Taquann method with high dispersion efficiency. Chronic peritonitis with fibrosis was observed in Taquann-treated multiwalled CNT (T-CNT)-injected mice, but not in Taquann-treated titanium dioxide-injected mice. In vivo and in vitro experiments showed that matrix metalloproteinase-12 (MMP-12) of macrophages was up-regulated by T-CNT to enhance fibroblast activation and profibrotic molecule expression in fibroblasts. In addition, T-CNT-induced peritonitis reduced MMP-12 expression in Nfκb1-/- mice, suggesting that MMP-12-producing macrophages play a key role in chronic inflammation due to T-CNT exposure through NF-κB activation. The results of this study could be helpful in understanding the molecular toxicity of nanomaterial and chronic inflammation.
Collapse
Affiliation(s)
- Takaaki Tsunematsu
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Rieko Arakaki
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mami Sato
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masako Saito
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kunihiro Otsuka
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yusuke Furukawa
- Division of Cellular and Molecular Toxicology, Biological Safety Research Center, National Institute of Health Sciences, Kawasaki, Japan
| | - Yuhji Taquahashi
- Division of Cellular and Molecular Toxicology, Biological Safety Research Center, National Institute of Health Sciences, Kawasaki, Japan
| | - Jun Kanno
- Division of Cellular and Molecular Toxicology, Biological Safety Research Center, National Institute of Health Sciences, Kawasaki, Japan
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.
| |
Collapse
|
11
|
Kandhwal M, Behl T, Singh S, Sharma N, Arora S, Bhatia S, Al-Harrasi A, Sachdeva M, Bungau S. Role of matrix metalloproteinase in wound healing. Am J Transl Res 2022; 14:4391-4405. [PMID: 35958464 PMCID: PMC9360851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 05/27/2022] [Indexed: 06/15/2023]
Abstract
Matrix metalloproteinases (MMPs) are a group of endopeptidases that play a vital role in the restoration of damaged skin. Through mediating various cellular events such as angiogenesis and vasodilation, MMPs are very crucial for the mechanism of wound healing. These enzymes are endopeptidases that are reliant on zinc which are concealed through the extracellular matrix (ECM). MMPs have different targets in different phases of wound healing through which they are capable of promoting timely healing in the body. This review discusses all the possible role of MMPs and their inhibitors that are involved during every step of the wound healing process. This review highlights the latest advances in the respective field about the regulation and mediation of MMPs in human skin and how these studies can be applied to other branches of medical sciences as well. Published papers were searched via MEDLINE, PubMed and MDPI from the available peer reviewed journals. Research done in the past suggests that active MMPs are involved in the healing progression of the wounds or they have a positive effect towards healing of wounds. Present studies in the relative field will further enhance the knowledge about enzymes working along with their inhibitors. These studies will help in a way to resolve some of the parameters that are necessary for modulating them either positively or negatively.
Collapse
Affiliation(s)
- Mimansa Kandhwal
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara UniversityPunjab 140401, India
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of NizwaNizwa 342001, Oman
- School of Health Science, University of Petroleum and Energy StudiesDehradun 248007, Uttarakhand, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of NizwaNizwa 342001, Oman
| | - Monika Sachdeva
- Fatima College of Health SciencesAl Ain 50, United Arab Emirates
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of OradeaOradea 410028, Romania
| |
Collapse
|
12
|
Nienhuis WA, Grutters JC. Potential therapeutic targets to prevent organ damage in chronic pulmonary sarcoidosis. Expert Opin Ther Targets 2021; 26:41-55. [PMID: 34949145 DOI: 10.1080/14728222.2022.2022123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Sarcoidosis is a granulomatous inflammatory disease with high chances of reduced quality of life, irreversible organ damage, and reduced life expectancy when vital organs are involved. Any organ system can be affected, and the lungs are most often affected. There is no preventive strategy as the exact etiology is unknown, and complex immunogenetic and environmental factors determine disease susceptibility and phenotype. Present-day treatment options originated from clinical practice and are effective in many patients. However, a substantial percentage of patients suffer from unacceptable side effects or still develop refractory, threatening pulmonary or extrapulmonary disease. AREAS COVERED As non-caseating granulomas, the pathological hallmark of disease, are assigned to divergent activation and regulation of the immune system, targets in relation to the possible triggers of granuloma formation and their sequelae were searched and reviewed. EXPERT OPINION :The immunopathogenesis underlying sarcoidosis has been a dynamic field of study. Several recent new insights give way to promising new therapeutic targets, such as certain antigenic triggers (e.g. from Aspergillus nidulans), mTOR, JAK-STAT and PPARγ pathways, the NRP2 receptor and MMP-12, which await further exploration. Clinical and trigger related phenotyping, and molecular endotyping in sarcoidosis will likely hold the key for precision medicine in the future.
Collapse
Affiliation(s)
- W A Nienhuis
- ILD Center of Excellence, Department of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - J C Grutters
- ILD Center of Excellence, Department of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Hearth and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
13
|
Mouse innate-like B-1 lymphocytes promote inhaled particle-induced in vitro granuloma formation and inflammation in conjunction with macrophages. Arch Toxicol 2021; 96:585-599. [PMID: 34935064 PMCID: PMC8837577 DOI: 10.1007/s00204-021-03200-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022]
Abstract
The current paradigm for explaining lung granulomatous diseases induced by inhaled particles is mainly based on macrophages. This mechanism is now challenging because B lymphocytes also infiltrate injured tissue, and the deficiency in B lymphocytes is associated with limited lung granulomas in silica-treated mice. Here, we investigated how B lymphocytes respond to micro- and nanoparticles by combining in vivo and in vitro mouse models. We first demonstrated that innate-like B-1 lymphocytes (not conventional B-2 lymphocytes or plasma cells) specifically accumulated during granuloma formation in mice instilled with crystalline silica (DQ12, 2.5 mg/mouse) and carbon nanotubes (CNT Mitsui, 0.2 mg/mouse). In comparison to macrophages, peritoneal B-1 lymphocytes purified from naïve mice were resistant to the pyroptotic activity of reactive particles (up to 1 mg/mL) but clustered to establish in vitro cell/particle aggregates. Mouse B-1 lymphocytes (not B-2 lymphocytes) in coculture with macrophages and CNT (0.1 µg/mL) organized three-dimensional spheroid structures in Matrigel and stimulated the release of TIMP-1. Furthermore, purified B-1 lymphocytes are sensitive to nanosilica toxicity through radical generation in culture. Nanosilica-exposed B-1 lymphocytes released proinflammatory cytokines and alarmins. In conclusion, our data indicate that in addition to macrophages, B-1 lymphocytes participate in micrometric particle-induced granuloma formation and display inflammatory functions in response to nanoparticles.
Collapse
|
14
|
The M2a Macrophage Phenotype Accompanies Pulmonary Granuloma Resolution in Mmp12 Knock-Out Mice Instilled with Multiwall Carbon Nanotubes. Int J Mol Sci 2021; 22:ijms222011019. [PMID: 34681679 PMCID: PMC8537143 DOI: 10.3390/ijms222011019] [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: 09/09/2021] [Revised: 09/30/2021] [Accepted: 10/09/2021] [Indexed: 11/17/2022] Open
Abstract
Sarcoidosis is a chronic disease with unknown etiology and pathophysiology, characterized by granuloma formation. Matrix Metalloproteinase-12 (MMP12) is an elastase implicated in active granulomatous sarcoidosis. Previously, we reported that oropharyngeal instillation of multiwall carbon nanotubes (MWCNT) into C57Bl/6 mice induced sarcoid-like granulomas and upregulation of MMP12. When Mmp12 knock-out (KO) mice were instilled with MWCNT, granuloma formation occurred 10 days post-instillation but subsequently resolved at 60 days. Thus, we concluded that MMP12 was essential to granuloma persistence. The aim of the current study was to identify potential mechanisms of granuloma resolution in Mmp12KO mice. Strikingly, an M2 macrophage phenotype was present in Mmp12KO but not in C57Bl/6 mice. Between 10 and 60 days, macrophage populations in MWCNT-instilled Mmp12KO mice demonstrated an M2c to M2a phenotypic shift, with elevations in levels of IL-13, an M2 subtype-regulating factor. Furthermore, the M2 inducer, Apolipoprotein E (ApoE), and Matrix Metalloproteinase-14 (MMP14), a promoter of collagen degradation, were upregulated in 60-day MWCNT-instilled Mmp12KO mice. In conclusion, alveolar macrophages express two M2 phenotypes in Mmp12KO mice: M2c at 10 days when granulomas form, and M2a at 60 days when granulomas are resolving. Findings suggest that granuloma resolution in 60-day Mmp12KO mice requires an M2a macrophage phenotype.
Collapse
|
15
|
Qian W, Zhao M, Wang R, Li H. Fibrinogen-like protein 1 (FGL1): the next immune checkpoint target. J Hematol Oncol 2021; 14:147. [PMID: 34526102 PMCID: PMC8444356 DOI: 10.1186/s13045-021-01161-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint therapy has achieved significant efficacy by blocking inhibitory pathways to release the function of T lymphocytes. In the clinic, anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) monoclonal antibodies (mAbs) have progressed to first-line monotherapies in certain tumor types. However, the efficacy of anti-PD-1/PD-L1 mAbs is still limited due to toxic side effects and de novo or adaptive resistance. Moreover, other immune checkpoint target and biomarkers for therapeutic response prediction are still lacking; as a biomarker, the PD-L1 (CD274, B7-H1) expression level is not as accurate as required. Hence, it is necessary to seek more representative predictive molecules and potential target molecules for immune checkpoint therapy. Fibrinogen-like protein 1 (FGL1) is a proliferation- and metabolism-related protein secreted by the liver. Multiple studies have confirmed that FGL1 is a newly emerging checkpoint ligand of lymphocyte activation gene 3 (LAG3), emphasizing the potential of targeting FGL1/LAG3 as the next generation of immune checkpoint therapy. In this review, we summarize the substantial regulation mechanisms of FGL1 in physiological and pathological conditions, especially tumor epithelial to mesenchymal transition, immune escape and immune checkpoint blockade resistance, to provide insights for targeting FGL1 in cancer treatment.
Collapse
Affiliation(s)
- Wenjing Qian
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, Liaoning, 110006, People's Republic of China.,The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, 116001, People's Republic of China
| | - Mingfang Zhao
- Department of Medical Oncology, the First Hospital of China Medical University, No.155 Nanjingbei Road, Shenyang, Liaoning, 110001, People's Republic of China
| | - Ruoyu Wang
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, No. 6 Jiefang Street, Dalian, Liaoning, 110006, People's Republic of China. .,The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, 116001, People's Republic of China.
| | - Heming Li
- Department of Medical Oncology, the First Hospital of China Medical University, No.155 Nanjingbei Road, Shenyang, Liaoning, 110001, People's Republic of China.
| |
Collapse
|
16
|
Barna BP, Judson MA, Thomassen MJ. Inflammatory Pathways in Sarcoidosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:39-52. [PMID: 34019262 DOI: 10.1007/978-3-030-68748-9_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Concepts regarding etiology and pathophysiology of sarcoidosis have changed remarkably within the past 5 years. Sarcoidosis is now viewed as a complex multi-causation disease related to a diverse collection of external environmental or infectious signals. It is generally accepted that the cause of sarcoidosis is unknown. Moreover, concepts of the inflammatory pathway have been modified by the realization that intrinsic genetic factors and innate immunity may modify adaptive immune responses to external triggers. With those potential regulatory pathways in mind, we will attempt to discuss the current understanding of the inflammatory response in sarcoidosis with emphasis on development of pulmonary granulomatous pathology. In that context, we will emphasize that both macrophages and T lymphocytes play key roles, with sometimes overlapping cytokine production (i.e., TNFα and IFN-γ) but also with unique mediators that influence the pathologic picture. Numerous studies have shown that in a sizable number of sarcoidosis patients, granulomas spontaneously resolve, usually within 3 years. Other sarcoidosis patients, however, may develop a chronic granulomatous disease which may subsequently lead to fibrosis. This chapter will outline our current understanding of inflammatory pathways in sarcoidosis which initiate and mediate granulomatous changes or onset of pulmonary fibrosis.
Collapse
Affiliation(s)
- Barbara P Barna
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary and Critical Care Medicine, East Carolina University, Greenville, NC, USA
| | - Marc A Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Mary Jane Thomassen
- Program in Lung Cell Biology and Translational Research, Division of Pulmonary and Critical Care Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
17
|
Barna BP, Malur A, Thomassen MJ. Studies in a Murine Granuloma Model of Instilled Carbon Nanotubes: Relevance to Sarcoidosis. Int J Mol Sci 2021; 22:ijms22073705. [PMID: 33918196 PMCID: PMC8038141 DOI: 10.3390/ijms22073705] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/25/2021] [Accepted: 03/31/2021] [Indexed: 12/23/2022] Open
Abstract
Poorly soluble environmental antigens, including carbon pollutants, are thought to play a role in the incidence of human sarcoidosis, a chronic inflammatory granulomatous disease of unknown causation. Currently, engineered carbon products such as multiwall carbon nanotubes (MWCNT) are manufactured commercially and have been shown to elicit acute and chronic inflammatory responses in experimental animals, including the production of granulomas or fibrosis. Several years ago, we hypothesized that constructing an experimental model of chronic granulomatosis resembling that associated with sarcoidosis might be achieved by oropharyngeal instillation of MWCNT into mice. This review summarizes the results of our efforts to define mechanisms of granuloma formation and identify potential therapeutic targets for sarcoidosis. Evidence is presented linking findings from the murine MWCNT granuloma model to sarcoidosis pathophysiology. As our goal was to determine what pulmonary inflammatory pathways might be involved, we utilized mice of knock-out (KO) backgrounds which corresponded to deficiencies noted in sarcoidosis patients. A primary example of this approach was to study mice with a myeloid-specific knock-out of the lipid-regulated transcription factor, peroxisome proliferator-activated receptor gamma (PPARγ) which is strikingly depressed in sarcoidosis. Among the major findings associated with PPARγ KO mice compared to wild-type were: (1) exacerbation of granulomatous and fibrotic histopathology in response to MWCNT; (2) elevation of inflammatory mediators; and (3) pulmonary retention of a potentially antigenic ESAT-6 peptide co-instilled with MWCNT. In line with these data, we also observed that activation of PPARγ in wild-type mice by the PPARγ-specific ligand, rosiglitazone, significantly reduced both pulmonary granuloma and inflammatory mediator production. Similarly, recognition of a deficiency of ATP-binding cassette (ABC) lipid transporter ABCG1 in sarcoidosis led us to study MWCNT instillation in myeloid-specific ABCG1 KO mice. As anticipated, ABCG1 deficiency was associated with larger granulomas and increased levels of inflammatory mediators. Finally, a transcriptional survey of alveolar macrophages from MWCNT-instilled wild-type mice and human sarcoidosis patients revealed several common themes. One of the most prominent mediators identified in both human and mouse transcriptomic analyses was MMP12. Studies with MMP12 KO mice revealed similar acute reactions to those in wild-type but at chronic time points where wild-type maintained granulomatous disease, resolution occurred with MMP12 KO mice suggesting MMP12 is necessary for granuloma progression. In conclusion, these studies suggest that the MWCNT granuloma model has relevance to human sarcoidosis study, particularly with respect to immune-specific pathways.
Collapse
|
18
|
Mousapasandi A, Herbert C, Thomas P. Potential use of biomarkers for the clinical evaluation of sarcoidosis. J Investig Med 2021; 69:jim-2020-001659. [PMID: 33452128 DOI: 10.1136/jim-2020-001659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2020] [Indexed: 12/20/2022]
Abstract
Sarcoidosis is a systemic granulomatous disease of unknown etiology and pathogenesis with a heterogeneous clinical presentation. In the appropriate clinical and radiological context and with the exclusion of other diagnoses, the disease is characterized by the pathological presence of non-caseating epithelioid cell granulomas. Sarcoidosis is postulated to be a multifactorial disease caused by chronic antigenic stimulation. The immunopathogenesis of sarcoidosis encompasses a complex interaction between the host, genetic factors and postulated environmental and infectious triggers, which result in granuloma development.The exact pathogenesis of the disease has yet to be elucidated, but some of the inflammatory pathways that play a key role in disease progression and outcomes are becoming apparent, and these may form the logical basis for selecting potential biomarkers.Biomarkers are biological molecules that are altered pathologically. To date, there exists no single reliable biomarker for the evaluation of sarcoidosis, either diagnostically or prognostically but new candidates are emerging. A diagnosis of sarcoidosis ideally requires a biopsy confirming non-caseating granulomas, but the likelihood of progression that requires intervention remains unpredictable. These challenging aspects could be potentially resolved by incorporating biomarkers into clinical practice for both diagnosis and monitoring disease activity.This review outlines the current knowledge on sarcoidosis with an emphasis on pulmonary sarcoidosis, and delineates the understanding surrounding the implication of biomarkers for the clinical evaluation of sarcoidosis.
Collapse
Affiliation(s)
- Amir Mousapasandi
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Cristan Herbert
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Paul Thomas
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Department of Respiratory Medicine, Prince of Wales' Hospital and Prince of Wales' Clinical School, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|