1
|
Saadati F, Modarresi Chahardehi A, Jamshidi N, Jamshidi N, Ghasemi D. Coumarin: A natural solution for alleviating inflammatory disorders. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 7:100202. [PMID: 39398983 PMCID: PMC11470182 DOI: 10.1016/j.crphar.2024.100202] [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: 04/28/2024] [Revised: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Coumarin, a naturally occurring compound found in various plants, has a rich history of use in traditional medicine. Recent research has highlighted its anti-inflammatory properties, positioning it as a promising candidate for treating inflammatory disorders such as rheumatoid arthritis, asthma, and inflammatory bowel disease. This narrative review aims to comprehensively summarize the current knowledge regarding coumarin's pharmacological effects in alleviating inflammatory conditions by analyzing preclinical and clinical studies. The review focuses on elucidating the mechanisms through which coumarin exerts its anti-inflammatory effects, including its antioxidant activity, inhibiting pro-inflammatory cytokine production, and modulation of immune cell functions. Additionally, the paper addresses potential limitations of using coumarin, such as concerns about toxicity at high doses or with prolonged use. Before widespread clinical application, further investigation is needed to fully understand coumarin's potential benefits and risks.
Collapse
Affiliation(s)
- Farnoosh Saadati
- Department of Cellular and Molecular Biology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | | | - Negar Jamshidi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| | - Nazanin Jamshidi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| | - Darioush Ghasemi
- Kimia Andisheh Teb Medical and Molecular Research Laboratory Co., Tehran, Iran
| |
Collapse
|
2
|
Ricard-Blum S, Vivès RR, Schaefer L, Götte M, Merline R, Passi A, Heldin P, Magalhães A, Reis CA, Skandalis SS, Karamanos NK, Perez S, Nikitovic D. A biological guide to glycosaminoglycans: current perspectives and pending questions. FEBS J 2024; 291:3331-3366. [PMID: 38500384 DOI: 10.1111/febs.17107] [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: 10/10/2023] [Revised: 01/08/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024]
Abstract
Mammalian glycosaminoglycans (GAGs), except hyaluronan (HA), are sulfated polysaccharides that are covalently attached to core proteins to form proteoglycans (PGs). This article summarizes key biological findings for the most widespread GAGs, namely HA, chondroitin sulfate/dermatan sulfate (CS/DS), keratan sulfate (KS), and heparan sulfate (HS). It focuses on the major processes that remain to be deciphered to get a comprehensive view of the mechanisms mediating GAG biological functions. They include the regulation of GAG biosynthesis and postsynthetic modifications in heparin (HP) and HS, the composition, heterogeneity, and function of the tetrasaccharide linkage region and its role in disease, the functional characterization of the new PGs recently identified by glycoproteomics, the selectivity of interactions mediated by GAG chains, the display of GAG chains and PGs at the cell surface and their impact on the availability and activity of soluble ligands, and on their move through the glycocalyx layer to reach their receptors, the human GAG profile in health and disease, the roles of GAGs and particular PGs (syndecans, decorin, and biglycan) involved in cancer, inflammation, and fibrosis, the possible use of GAGs and PGs as disease biomarkers, and the design of inhibitors targeting GAG biosynthetic enzymes and GAG-protein interactions to develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Univ Lyon 1, ICBMS, UMR 5246 University Lyon 1 - CNRS, Villeurbanne cedex, France
| | | | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Germany
| | - Rosetta Merline
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | | | - Paraskevi Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Sweden
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Portugal
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Serge Perez
- Centre de Recherche sur les Macromolécules Végétales, University of Grenoble-Alpes, CNRS, France
| | - Dragana Nikitovic
- Laboratory of Histology-Embryology, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
3
|
Xu Y, Chen B, Guo Z, Chen C, Wang C, Zhou H, Zhang C, Feng Y. Identification of diagnostic markers for moyamoya disease by combining bulk RNA-sequencing analysis and machine learning. Sci Rep 2024; 14:5931. [PMID: 38467737 PMCID: PMC10928210 DOI: 10.1038/s41598-024-56367-w] [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: 09/27/2023] [Accepted: 03/05/2024] [Indexed: 03/13/2024] Open
Abstract
Moyamoya disease (MMD) remains a chronic progressive cerebrovascular disease with unknown etiology. A growing number of reports describe the development of MMD relevant to infection or autoimmune diseases. Identifying biomarkers of MMD is to understand the pathogenesis and development of novel targeted therapy and may be the key to improving the patient's outcome. Here, we analyzed gene expression from two GEO databases. To identify the MMD biomarkers, the weighted gene co-expression network analysis (WGCNA) and the differential expression analyses were conducted to identify 266 key genes. The KEGG and GO analyses were then performed to construct the protein interaction (PPI) network. The three machine-learning algorithms of support vector machine-recursive feature elimination (SVM-RFE), random forest and least absolute shrinkage and selection operator (LASSO) were used to analyze the key genes and take intersection to construct MMD diagnosis based on the four core genes found (ACAN, FREM1, TOP2A and UCHL1), with highly accurate AUCs of 0.805, 0.903, 0.815, 0.826. Gene enrichment analysis illustrated that the MMD samples revealed quite a few differences in pathways like one carbon pool by folate, aminoacyl-tRNA biosynthesis, fat digestion and absorption and fructose and mannose metabolism. In addition, the immune infiltration profile demonstrated that ACAN expression was associated with mast cells resting, FREM1 expression was associated with T cells CD4 naive, TOP2A expression was associated with B cells memory, UCHL1 expression was associated with mast cells activated. Ultimately, the four key genes were verified by qPCR. Taken together, our study analyzed the diagnostic biomarkers and immune infiltration characteristics of MMD, which may shed light on the potential intervention targets of moyamoya disease patients.
Collapse
Affiliation(s)
- Yifan Xu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiang Su Road, Qingdao City, 266000, China
| | - Bing Chen
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiang Su Road, Qingdao City, 266000, China
| | - Zhongxiang Guo
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiang Su Road, Qingdao City, 266000, China
| | - Cheng Chen
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiang Su Road, Qingdao City, 266000, China
| | - Chao Wang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiang Su Road, Qingdao City, 266000, China
| | - Han Zhou
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiang Su Road, Qingdao City, 266000, China
| | - Chonghui Zhang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiang Su Road, Qingdao City, 266000, China
| | - Yugong Feng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, 16 Jiang Su Road, Qingdao City, 266000, China.
| |
Collapse
|
4
|
Smith MM, Melrose J. Lumican, a Multifunctional Cell Instructive Biomarker Proteoglycan Has Novel Roles as a Marker of the Hypercoagulative State of Long Covid Disease. Int J Mol Sci 2024; 25:2825. [PMID: 38474072 DOI: 10.3390/ijms25052825] [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: 01/01/2024] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 03/14/2024] Open
Abstract
This study has reviewed the many roles of lumican as a biomarker of tissue pathology in health and disease. Lumican is a structure regulatory proteoglycan of collagen-rich tissues, with cell instructive properties through interactions with a number of cell surface receptors in tissue repair, thereby regulating cell proliferation, differentiation, inflammation and the innate and humoral immune systems to combat infection. The exponential increase in publications in the last decade dealing with lumican testify to its role as a pleiotropic biomarker regulatory protein. Recent findings show lumican has novel roles as a biomarker of the hypercoagulative state that occurs in SARS CoV-2 infections; thus, it may also prove useful in the delineation of the complex tissue changes that characterize COVID-19 disease. Lumican may be useful as a prognostic and diagnostic biomarker of long COVID disease and its sequelae.
Collapse
Affiliation(s)
- Margaret M Smith
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Faculty of Health and Science, University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Arthropharm Pty Ltd., Bondi Junction, NSW 2022, Australia
| | - James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Faculty of Health and Science, University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Graduate School of Biomedical Engineering, Faculty of Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
5
|
Kunnathattil M, Rahul P, Skaria T. Soluble vascular endothelial glycocalyx proteoglycans as potential therapeutic targets in inflammatory diseases. Immunol Cell Biol 2024; 102:97-116. [PMID: 37982607 DOI: 10.1111/imcb.12712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023]
Abstract
Reducing the activity of cytokines and leukocyte extravasation is an emerging therapeutic strategy to limit tissue-damaging inflammatory responses and restore immune homeostasis in inflammatory diseases. Proteoglycans embedded in the vascular endothelial glycocalyx, which regulate the activity of cytokines to restrict the inflammatory response in physiological conditions, are proteolytically cleaved in inflammatory diseases. Here we critically review the potential of proteolytically shed, soluble vascular endothelial glycocalyx proteoglycans to modulate pathological inflammatory responses. Soluble forms of the proteoglycans syndecan-1, syndecan-3 and biglycan exert beneficial anti-inflammatory effects by the removal of chemokines, suppression of proinflammatory cytokine expression and leukocyte migration, and induction of autophagy of proinflammatory M1 macrophages. By contrast, soluble versikine and decorin enhance proinflammatory responses by increasing inflammatory cytokine synthesis and leukocyte migration. Endogenous syndecan-2 and mimecan exert proinflammatory effects, syndecan-4 and perlecan mediate beneficial anti-inflammatory effects and glypican regulates Hh and Wnt signaling pathways involved in systemic inflammatory responses. Taken together, targeting the vascular endothelial glycocalyx-derived, soluble syndecan-1, syndecan-2, syndecan-3, syndecan-4, biglycan, versikine, mimecan, perlecan, glypican and decorin might be a potential therapeutic strategy to suppress overstimulated cytokine and leukocyte responses in inflammatory diseases.
Collapse
Affiliation(s)
- Maneesha Kunnathattil
- Department of Zoology, Government College Madappally, University of Calicut, Calicut, Kerala, India
| | - Pedapudi Rahul
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| | - Tom Skaria
- School of Biotechnology, National Institute of Technology Calicut, Calicut, Kerala, India
| |
Collapse
|
6
|
Miguez PA, Bash E, Musskopf ML, Tuin SA, Rivera-Concepcion A, Chapple ILC, Liu J. Control of tissue homeostasis by the extracellular matrix: Synthetic heparan sulfate as a promising therapeutic for periodontal health and bone regeneration. Periodontol 2000 2024; 94:510-531. [PMID: 37614159 PMCID: PMC10891305 DOI: 10.1111/prd.12515] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/13/2023] [Accepted: 07/22/2023] [Indexed: 08/25/2023]
Abstract
Proteoglycans are core proteins associated with carbohydrate/sugar moieties that are highly variable in disaccharide composition, which dictates their function. These carbohydrates are named glycosaminoglycans, and they can be attached to proteoglycans or found free in tissues or on cell surfaces. Glycosaminoglycans such as hyaluronan, chondroitin sulfate, dermatan sulfate, keratan sulfate, and heparin/heparan sulfate have multiple functions including involvement in inflammation, immunity and connective tissue structure, and integrity. Heparan sulfate is a highly sulfated polysaccharide that is abundant in the periodontium including alveolar bone. Recent evidence supports the contention that heparan sulfate is an important player in modulating interactions between damage associated molecular patterns and inflammatory receptors expressed by various cell types. The structure of heparan sulfate is reported to dictate its function, thus, the utilization of a homogenous and structurally defined heparan sulfate polysaccharide for modulation of cell function offers therapeutic potential. Recently, a chemoenzymatic approach was developed to allow production of many structurally defined heparan sulfate carbohydrates. These oligosaccharides have been studied in various pathological inflammatory conditions to better understand their function and their potential application in promoting tissue homeostasis. We have observed that specific size and sulfation patterns can modulate inflammation and promote tissue maintenance including an anabolic effect in alveolar bone. Thus, new evidence provides a strong impetus to explore heparan sulfate as a potential novel therapeutic agent to treat periodontitis, support alveolar bone maintenance, and promote bone formation.
Collapse
Affiliation(s)
- PA Miguez
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - E Bash
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - ML Musskopf
- Division of Comprehensive Oral Health - Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - SA Tuin
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - A Rivera-Concepcion
- Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC, USA
| | - ILC Chapple
- Periodontal Research Group, School of Dentistry, Institute of Clinical Sciences, College of Medical and Dental Sciences, Birmingham’s NIHR BRC in Inflammation Research, University of Birmingham and Birmingham Community Health Foundation Trust, Birmingham UK Iain Chapple
| | - J Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
7
|
Zhao F, Bai Y, Xiang X, Pang X. The role of fibromodulin in inflammatory responses and diseases associated with inflammation. Front Immunol 2023; 14:1191787. [PMID: 37483637 PMCID: PMC10360182 DOI: 10.3389/fimmu.2023.1191787] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/23/2023] [Indexed: 07/25/2023] Open
Abstract
Inflammation is an immune response that the host organism eliminates threats from foreign objects or endogenous signals. It plays a key role in the progression, prognosis as well as therapy of diseases. Chronic inflammatory diseases have been regarded as the main cause of death worldwide at present, which greatly affect a vast number of individuals, producing economic and social burdens. Thus, developing drugs targeting inflammation has become necessary and attractive in the world. Currently, accumulating evidence suggests that small leucine-rich proteoglycans (SLRPs) exhibit essential roles in various inflammatory responses by acting as an anti-inflammatory or pro-inflammatory role in different scenarios of diseases. Of particular interest was a well-studied member, termed fibromodulin (FMOD), which has been largely explored in the role of inflammatory responses in inflammatory-related diseases. In this review, particular focus is given to the role of FMOD in inflammatory response including the relationship of FMOD with the complement system and immune cells, as well as the role of FMOD in the diseases associated with inflammation, such as skin wounding healing, osteoarthritis (OA), tendinopathy, atherosclerosis, and heart failure (HF). By conducting this review, we intend to gain insight into the role of FMOD in inflammation, which may open the way for the development of new anti-inflammation drugs in the scenarios of different inflammatory-related diseases.
Collapse
Affiliation(s)
- Feng Zhao
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Bai
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xuerong Xiang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoxiao Pang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
8
|
Liu J, Zhang B, Zhu G, Liu C, Wang S, Zhao Z. Discovering genetic linkage between periodontitis and type 1 diabetes: A bioinformatics study. Front Genet 2023; 14:1147819. [PMID: 37051594 PMCID: PMC10083320 DOI: 10.3389/fgene.2023.1147819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Relationship between periodontitis (PD) and type 1 diabetes (T1D) has been reported, but the detailed pathogenesis requires further elucidation. This study aimed to reveal the genetic linkage between PD and T1D through bioinformatics analysis, thereby providing novel insights into scientific research and clinical treatment of the two diseases.Methods: PD-related datasets (GSE10334, GSE16134, GSE23586) and T1D-related datasets(GSE162689)were downloaded from NCBI Gene Expression Omnibus (GEO). Following batch correction and merging of PD-related datasets as one cohort, differential expression analysis was performed (adjusted p-value <0.05 and ∣log2 fold change| > 0.5), and common differentially expressed genes (DEGs) between PD and T1D were extracted. Functional enrichment analysis was conducted via Metascape website. The protein-protein interaction (PPI) network of common DEGs was generated in The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database. Hub genes were selected by Cytoscape software and validated by receiver operating characteristic (ROC) curve analysis.Results: 59 common DEGs of PD and T1D were identified. Among these DEGs, 23 genes were commonly upregulated, and 36 genes were commonly downregulated in both PD- and T1D-related cohorts. Functional enrichment analysis indicated that common DEGs were mainly enriched in tube morphogenesis, supramolecular fiber organization, 9 + 0 non-motile cilium, plasma membrane bounded cell projection assembly, glomerulus development, enzyme-linked receptor protein signaling pathway, endochondral bone morphogenesis, positive regulation of kinase activity, cell projection membrane and regulation of lipid metabolic process. After PPI construction and modules selection, 6 hub genes (CD34, EGR1, BBS7, FMOD, IGF2, TXN) were screened out and expected to be critical in linking PD and T1D. ROC analysis showed that the AUC values of hub genes were all greater than 70% in PD-related cohort and greater than 60% in T1D-related datasets.Conclusion: Shared molecular mechanisms between PD and T1D were revealed in this study, and 6 hub genes were identified as potential targets in treating PD and T1D.
Collapse
Affiliation(s)
- Junqi Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Guanyin Zhu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chenlu Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuangcheng Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Zhihe Zhao,
| |
Collapse
|
9
|
Vaxevanis CK, Bauer M, Subbarayan K, Friedrich M, Massa C, Biehl K, Al-Ali HK, Wickenhauser C, Seliger B. Biglycan as a mediator of proinflammatory response and target for MDS and sAML therapy. Oncoimmunology 2022; 12:2152998. [PMID: 36531688 PMCID: PMC9757483 DOI: 10.1080/2162402x.2022.2152998] [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] [Indexed: 12/23/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and their progression to secondary acute myeloid leukemia (sAML) are associated with an altered protein expression including extracellular matrix (ECM) components thereby promoting an inflammatory environment. Since the role of the proteoglycan biglycan (BGN) as an inflammatory mediator has not yet been investigated in both diseases and might play a role in disease progression, its expression and/or function was determined in cell lines and bone marrow biopsies (BMBs) of MDS and sAML patients and subpopulations of MDS stem cells by Western blot and immunohistochemistry. The bone marrow (BM) microenvironment was analyzed by multispectral imaging, patients' survival by Cox regression. ROC curves were assessed for diagnostic value of BGN. All cell lines showed a strong BGN surface expression in contrast to only marginal expression levels in mononuclear cells and CD34+ cells from healthy donors. In the MDS-L cell line, CD34-CD33+ and CD34+CD33+ blast subpopulations exhibited a differential BGN surface detection. Increased BGN mediated inflammasome activity of CD34-CD33+TLR4+ cells was observed, which was inhibited by direct targeting of BGN or NLRP3. BGN was heterogeneously expressed in BMBs of MDS and sAML, but was not detected in control biopsies. BGN expression in BMBs positively correlated with MUM1+ and CD8+, but negatively with CD33+TLR4+ cell infiltration and was accompanied by a decreased progression-free survival of MDS patients. BGN-mediated inflammasome activation appears to be a crucial mechanism in MDS pathogenesis implicating its use as suitable biomarker and potential therapeutic target. Abbreviations: Ab, antibody; alloSCT, allogenic stem cell transplant; AML, acute myeloid leukemia; BGN, biglycan; BM, bone marrow; BMB, bone marrow biopsy; casp1, caspase 1; CTLA-4, cytotoxic T lymphocyte-associated protein 4; DAMP, danger-associated molecular pattern; ECM, extracellular matrix; FCS, fetal calf serum; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; HD, healthy donor; HSPC, hematopoietic stem and progenitor cell; HSC, hematopoietic stem cell; IFN, interferon; IHC, immunohistochemistry; IL, interleukin; MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasm; MSI, multispectral imaging; NGS, next-generation sequencing; NLRP3, NLR family pyrin domain containing 3; OS, overall survival; PBMC, peripheral blood mononuclear cell; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1, PFS, progression-free survival; PRR, pattern recognition receptor; SC, stem cell; SLRP, small leucine-rich proteoglycan; TGF, transforming growth factor; TIRAP, toll/interleukin 1 receptor domain-containing adapter protein; TLR, toll-like receptor; Treg, regulatory T cell.
Collapse
Affiliation(s)
| | - Marcus Bauer
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale)06112, Germany
| | | | - Michael Friedrich
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale)06112, Germany
| | - Chiara Massa
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale)06112, Germany
| | - Katharina Biehl
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale)06112, Germany
| | - Haifa Kathrin Al-Ali
- Krukenberg Cancer Center Halle, University Hospital Halle, Krukenberg-Krebszentrum, Halle (Saale)06120, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale)06112, Germany
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale)06112, Germany,Department of Good Manufacturing Practice (GMP) Development & Advanced Therapy Medicinal Products (ATMP) Design, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig04103, Germany,Medical School Theodor Fontane, Institute of Translational Medicine, Brandenburg an der Havel14770, Germany,CONTACT Barbara Seliger Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), 06112, Germany
| |
Collapse
|
10
|
Nulali J, Zhan M, Zhang K, Tu P, Liu Y, Song H. Osteoglycin: An ECM Factor Regulating Fibrosis and Tumorigenesis. Biomolecules 2022; 12:1674. [PMID: 36421687 PMCID: PMC9687868 DOI: 10.3390/biom12111674] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 08/27/2023] Open
Abstract
The extracellular matrix (ECM) is made up of noncellular components that have special properties for influencing cell behavior and tissue structure. Small leucine-rich proteoglycans (SLRPs) are nonfibrillar ECM components that serve as structural scaffolds and signaling molecules. osteoglycin (OGN), a class III SLRP, is a ubiquitous ECM component that not only helps to organize the extracellular matrix but also regulates a number of important biological processes. As a glycosylated protein in the ECM, OGN was originally considered to be involved in fiber assembly and was reported to have a connection with fibrosis. In addition to these functions, OGN is found in a variety of cancer tissues and is implicated in cellular processes linked to tumorigenesis, including cell proliferation, invasion, metastasis, and epithelial-mesenchymal transition (EMT). In this review, we summarize the structure and functions of OGN as well as its biological and clinical importance in the context of fibrotic illness and tumorigenesis. This review aims to improve our understanding of OGN and provide some new strategies for the treatment of fibrosis and cancer.
Collapse
Affiliation(s)
- Jiayida Nulali
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Ming Zhan
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Kaiwen Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Pinghui Tu
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yu Liu
- Department of Respiration, Yangpu Hospital, Tongji University School of Medicine, Shanghai 200070, China
| | - Huaidong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics and Endocrinology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
11
|
The Role of Decorin in Autoimmune and Inflammatory Diseases. J Immunol Res 2022; 2022:1283383. [PMID: 36033387 PMCID: PMC9402370 DOI: 10.1155/2022/1283383] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/17/2022] Open
Abstract
Decorin is an extracellular matrix protein that belongs to the family of small leucine-rich proteoglycans. As a matrix protein, the first discovered role of decorin is participating in collagen fibril formation. Many other functions of decorin in various biological processes have been subsequently identified. Decorin is involved in an extensive signaling network and can interact with other extracellular matrix components, growth factors, receptor tyrosine kinases, and various proteases. Decorin has been shown to be involved in wound repair, cell cycle, angiogenesis, tumor metastasis, and autophagy. Recent evidence indicates that it also plays a role in immune regulation and inflammatory diseases. This review summarizes the characteristics of decorin in immune and inflammatory diseases, including inflammatory bowel disease (IBD), Sjögren's syndrome (SS), chronic obstructive pulmonary disease (COPD), IgA nephropathy, rheumatoid arthritis (RA), spondyloarthritis (SpA), osteoarthritis, multiple sclerosis (MS), idiopathic inflammatory myopathies (IIM), and systemic sclerosis (SSc) and discusses the potential role in these disorders.
Collapse
|
12
|
Halasi M, Grinstein M, Adini A, Adini I. Fibromodulin Ablation Exacerbates the Severity of Acute Colitis. J Inflamm Res 2022; 15:4515-4526. [PMID: 35966006 PMCID: PMC9374093 DOI: 10.2147/jir.s366290] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/17/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Epidemiological studies have associated pigment production with protection against certain human diseases. In contrast to African Americans, European descendants are more likely to suffer from angiogenesis-dependent and inflammatory diseases, such as wet age-related macular degeneration (ARMD) and ulcerative colitis (UC), respectively. Methods In a mouse model of dextran sulfate sodium (DSS)-induced acute colitis, the effect of fibromodulin (FMOD) depletion was examined on colitis severity. Results In this study, albino mice that produce high levels of FMOD developed less severe acute colitis compared with mice lacking in FMOD as assessed by clinical symptoms and histopathological changes. FMOD depletion affected the expression of tight junction proteins, contributing to the destruction of the epithelial barrier. Furthermore, this study revealed a stronger inflammatory response after DSS treatment in the absence of FMOD, where FMOD depletion led to an increase in activated T cells, plasmacytoid dendritic cells (pDCs), and type I interferon (IFN) production. Discussion These findings point to FMOD as a potential biomarker of disease severity in UC among light-skinned individuals of European descent.
Collapse
Affiliation(s)
- Marianna Halasi
- Department of Surgery, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mor Grinstein
- Department of Medicine, Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Avner Adini
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Irit Adini
- Department of Surgery, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Szeremeta A, Jura-Półtorak A, Zoń-Giebel A, Olczyk K, Komosińska-Vassev K. TNF-α Inhibitors in Combination with MTX Reduce Circulating Levels of Heparan Sulfate/Heparin and Endothelial Dysfunction Biomarkers (sVCAM-1, MCP-1, MMP-9 and ADMA) in Women with Rheumatoid Arthritis. J Clin Med 2022; 11:jcm11144213. [PMID: 35887981 PMCID: PMC9320287 DOI: 10.3390/jcm11144213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Sulfated glycosaminoglycans (sGAGs) are likely to play an important role in the development and progression of rheumatoid arthritis (RA)-associated atherosclerosis. The present study investigated the effect of anti-tumor necrosis factor-α (anti-TNF-α) therapy in combination with methotrexate on plasma sGAG levels and serum markers of endothelial dysfunction. Among sGAG types, plasma chondroitin/dermatan sulfate (CS/DS) and heparan sulfate/heparin (HS/H) were characterized using electrophoretic fractionation. Serum levels of soluble vascular cell adhesion molecule-1 (sVCAM-1), monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinase-9 (MMP-9) and asymmetric dimethylarginine (ADMA) were measured by immunoassays. The measurements were carried out four times: at baseline and after 3, 9 and 15 months of anti-TNF-α therapy. All analyzed parameters, excluding ADMA, were significantly elevated in patients with RA before the implementation of biological therapy compared to healthy subjects. Performed anti-TNF-α treatment led to a successive decrease in HS/H levels toward normal values, without any effect on CS/DS levels in female RA patients. The treatment was also effective at lowering the serum levels of sVCAM-1, MCP-1, MMP-9 and ADMA. Moreover, a significant positive correlation was found between the circulating HS/H and the 28 joint disease activity score based on the erythrocyte sedimentation rate (DAS28-ESR, r = 0.408; p <0.05), MCP-1 (r = 0.398; p <0.05) and ADMA (r = 0.396; p <0.05) in patients before the first dose of TNF-α inhibitor. In conclusion, a beneficial effect of anti-TNF-α therapy on cell-surface heparan sulfate proteoglycans (HSPGs)/HS turnover and endothelial dysfunction was observed in this study. This was manifested by a decrease in blood HS/H levels and markers of endothelial activation, respectively. Moreover, the decrease in the concentration of HS/H in the blood of patients during treatment, progressing with the decline in disease activity, indicates that the plasma HS/H profile may be useful for monitoring the efficacy of anti-TNF-α treatment in patients with RA.
Collapse
Affiliation(s)
- Anna Szeremeta
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (A.J.-P.); (K.O.); (K.K.-V.)
- Correspondence: ; Tel.: +48-32-364-11-50
| | - Agnieszka Jura-Półtorak
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (A.J.-P.); (K.O.); (K.K.-V.)
| | - Aleksandra Zoń-Giebel
- Department of Rheumatology and Rehabilitation, Specialty Hospital No. 1, Żeromskiego 7, 41-902 Bytom, Poland;
| | - Krystyna Olczyk
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (A.J.-P.); (K.O.); (K.K.-V.)
| | - Katarzyna Komosińska-Vassev
- Department of Clinical Chemistry and Laboratory Diagnostics, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (A.J.-P.); (K.O.); (K.K.-V.)
| |
Collapse
|
14
|
The fibrogenic niche in kidney fibrosis: components and mechanisms. Nat Rev Nephrol 2022; 18:545-557. [PMID: 35788561 DOI: 10.1038/s41581-022-00590-z] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 02/08/2023]
Abstract
Kidney fibrosis, characterized by excessive deposition of extracellular matrix (ECM) that leads to tissue scarring, is the final common outcome of a wide variety of chronic kidney diseases. Rather than being distributed uniformly across the kidney parenchyma, renal fibrotic lesions initiate at certain focal sites in which the fibrogenic niche is formed in a spatially confined fashion. This niche provides a unique tissue microenvironment that is orchestrated by a specialized ECM network consisting of de novo-induced matricellular proteins. Other structural elements of the fibrogenic niche include kidney resident and infiltrated inflammatory cells, extracellular vesicles, soluble factors and metabolites. ECM proteins in the fibrogenic niche recruit soluble factors including WNTs and transforming growth factor-β from the extracellular milieu, creating a distinctive profibrotic microenvironment. Studies using decellularized ECM scaffolds from fibrotic kidneys show that the fibrogenic niche autonomously promotes fibroblast proliferation, tubular injury, macrophage activation and endothelial cell depletion, pathological features that recapitulate key events in the pathogenesis of chronic kidney disease. The concept of the fibrogenic niche represents a paradigm shift in understanding of the mechanism of kidney fibrosis that could lead to the development of non-invasive biomarkers and novel therapies not only for chronic kidney disease, but also for fibrotic diseases of other organs.
Collapse
|
15
|
von Mentzer U, Corciulo C, Stubelius A. Biomaterial Integration in the Joint: Pathological Considerations, Immunomodulation, and the Extracellular Matrix. Macromol Biosci 2022; 22:e2200037. [PMID: 35420256 DOI: 10.1002/mabi.202200037] [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: 02/27/2022] [Revised: 03/30/2022] [Indexed: 11/08/2022]
Abstract
Defects of articular joints are becoming an increasing societal burden due to a persistent increase in obesity and aging. For some patients suffering from cartilage erosion, joint replacement is the final option to regain proper motion and limit pain. Extensive research has been undertaken to identify novel strategies enabling earlier intervention to promote regeneration and cartilage healing. With the introduction of decellularized extracellular matrix (dECM), researchers have tapped into the potential for increased tissue regeneration by designing biomaterials with inherent biochemical and immunomodulatory signals. Compared to conventional and synthetic materials, dECM-based materials invoke a reduced foreign body response. It is therefore highly beneficial to understand the interplay of how these native tissue-based materials initiate a favorable remodeling process by the immune system. Yet, such an understanding also demands increasing considerations of the pathological environment and remodeling processes, especially for materials designed for early disease intervention. This knowledge would avoid rejection and help predict complications in conditions with inflammatory components such as arthritides. This review outlines general issues facing biomaterial integration and emphasizes the importance of tissue-derived macromolecular components in regulating essential homeostatic, immunological, and pathological processes to increase biomaterial integration for patients suffering from joint degenerative diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ula von Mentzer
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| | - Carmen Corciulo
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation, Sahlgrenska Academy at the University of Gothenburg, Guldhedsgatan 10A, Gothenburg, 41296, Sweden
| | - Alexandra Stubelius
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| |
Collapse
|
16
|
Piperigkou Z, Tzaferi K, Makrokanis G, Cheli K, Karamanos NK. The microRNA-cell surface proteoglycan axis in cancer progression. Am J Physiol Cell Physiol 2022; 322:C825-C832. [PMID: 35294845 DOI: 10.1152/ajpcell.00041.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Proteoglycans consist one of the major extracellular matrix class of biomolecules that demonstrate nodal roles in cancer progression. Μodern diagnostic and therapeutic approaches include proteoglycan detection and pharmacological targeting in various cancers. Proteoglycans orchestrate critical signaling pathways for cancer development and progression through dynamic interactions with matrix components. It is well established that the epigenetic signatures of cancer cells play critical role in guiding their functional properties and metastatic potential. Secreted microRNAs (miRNAs) reside in a complex network with matrix proteoglycans, thus affecting cell-cell and cell-matrix communication. This mini-review aims to highlight current knowledge on the proteoglycan-mediated signaling cascades that regulate miRNA biogenesis in cancer. Moreover, the miRNA-mediated proteoglycan regulation during cancer progression and mechanistic aspects on the way that proteoglycans affect miRNA expression are presented. Recent advances on the role of cell surface proteoglycans in exosome biogenesis and miRNA packaging and expression are also discussed.
Collapse
Affiliation(s)
- Zoi Piperigkou
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Kyriaki Tzaferi
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - George Makrokanis
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Konsatntina Cheli
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Nikos K Karamanos
- Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| |
Collapse
|
17
|
Biglycan Promotes Cancer Stem Cell Properties, NFκB Signaling and Metastatic Potential in Breast Cancer Cells. Cancers (Basel) 2022; 14:cancers14020455. [PMID: 35053617 PMCID: PMC8773822 DOI: 10.3390/cancers14020455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Breast cancer stem cells (BCSCs) are a small sub-population of cells within tumors with high metastatic potential. We identified biglycan (BGN) as a prospective molecular target in BCSCs that regulates the aggressive phenotypes of these cells. These findings establish a foundation for the development of therapeutics against BGN to eliminate BCSCs and prevent metastatic breast cancer. Abstract It is a major challenge to treat metastasis due to the presence of heterogenous BCSCs. Therefore, it is important to identify new molecular targets and their underlying molecular mechanisms in various BCSCs to improve treatment of breast cancer metastasis. Here, we performed RNA sequencing on two distinct co-existing BCSC populations, ALDH+ and CD29hi CD61+ from PyMT mammary tumor cells and detected upregulation of biglycan (BGN) in these BCSCs. Genetic depletion of BGN reduced BCSC proportions and tumorsphere formation. Furthermore, BCSC associated aggressive traits such as migration and invasion were significantly reduced by depletion of BGN. Glycolytic and mitochondrial metabolic assays also revealed that BCSCs exhibited decreased metabolism upon loss of BGN. BCSCs showed decreased activation of the NFκB transcription factor, p65, and phospho-IκB levels upon BGN ablation, indicating regulation of NFκB pathway by BGN. To further support our data, we also characterized CD24−/CD44+ BCSCs from human luminal MCF-7 breast cancer cells. These CD24−/CD44+ BCSCs similarly exhibited reduced tumorigenic phenotypes, metabolism and attenuation of NFκB pathway after knockdown of BGN. Finally, loss of BGN in ALDH+ and CD29hi CD61+ BCSCs showed decreased metastatic potential, suggesting BGN serves as an important therapeutic target in BCSCs for treating metastasis of breast cancer.
Collapse
|
18
|
Dussoyer M, Page A, Delolme F, Rousselle P, Nyström A, Moali C. Comparison of extracellular matrix enrichment protocols for the improved characterization of the skin matrisome by mass spectrometry. J Proteomics 2022; 251:104397. [PMID: 34678517 DOI: 10.1016/j.jprot.2021.104397] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 09/23/2021] [Accepted: 10/03/2021] [Indexed: 12/14/2022]
Abstract
A striking feature of skin organization is that the extracellular matrix (ECM) occupies a larger volume than the cells. Skin ECM also directly contributes to aging and most cutaneous diseases. In recent years, specific ECM enrichment protocols combined with in silico approaches allowed the proteomic description of the matrisome of various organs and tumor samples. Nevertheless, the skin matrisome remains under-studied and protocols allowing the efficient recovery of the diverse ECM found in skin are still to be described. Here, we compared four protocols allowing the enrichment of ECM proteins from adult mouse back skin and found that all protocols led to a significant enrichment (up to 65%) of matrisome proteins when compared to total skin lysates. The protocols based on decellularization and solubility profiling gave the best results in terms of numbers of proteins identified and confirmed that skin matrisome proteins exhibit very diverse solubility and abundance profiles. We also report the first description of the skin matrisome of healthy adult mice that includes 236 proteins comprising 95 core matrisome proteins and 141 associated matrisome proteins. These results provide a reliable basis for future characterizations of skin ECM proteins and their dysregulations in disease-specific contexts. SIGNIFICANCE: Extracellular matrix proteins are key players in skin physiopathology and have been involved in several diseases such as genetic disorders, wound healing defects, scleroderma and skin carcinoma. However, skin ECM proteins are numerous, diverse and challenging to analyze by mass spectrometry due to the multiplicity of their post-translational modifications and to the heterogeneity of their solubility profiles. Here, we performed the thorough evaluation of four ECM enrichment protocols compatible with the proteomic analysis of mouse back skin and provide the first description of the adult mouse skin matrisome in homeostasis conditions. Our work will greatly facilitate the future characterization of skin ECM alterations in preclinical mouse models and will inspire new optimizations to analyze the skin matrisome of other species and of human clinical samples.
Collapse
Affiliation(s)
- Mélissa Dussoyer
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Adeline Page
- University of Lyon, INSERM, ENS Lyon, CNRS, Protein Science Facility, SFR BioSciences, UAR3444/US8, F-69366 Lyon, France
| | - Frédéric Delolme
- University of Lyon, INSERM, ENS Lyon, CNRS, Protein Science Facility, SFR BioSciences, UAR3444/US8, F-69366 Lyon, France
| | - Patricia Rousselle
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Alexander Nyström
- Department of Clinical Dermatology/Medical Center, University of Freiburg, Freiburg, Germany; Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
| | - Catherine Moali
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France.
| |
Collapse
|
19
|
Colon-Caraballo M, Lee N, Nallasamy S, Myers K, Hudson D, Iozzo RV, Mahendroo M. Novel regulatory roles of small leucine-rich proteoglycans in remodeling of the uterine cervix in pregnancy. Matrix Biol 2022; 105:53-71. [PMID: 34863915 PMCID: PMC9446484 DOI: 10.1016/j.matbio.2021.11.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 01/03/2023]
Abstract
The cervix undergoes rapid and dramatic shifts in collagen and elastic fiber structure to achieve its disparate physiological roles of competence during pregnancy and compliance during birth. An understanding of the structure-function relationships of collagen and elastic fibers to maintain extracellular matrix (ECM) homeostasis requires an understanding of the mechanisms executed by non-structural ECM molecules. Small-leucine rich proteoglycans (SLRPs) play key functions in biology by affecting collagen fibrillogenesis and regulating enzyme and growth factor bioactivities. In the current study, we evaluated collagen and elastic fiber structure-function relationships in mouse cervices using mice with genetic ablation of decorin and/or biglycan genes as representative of Class I SLRPs, and lumican gene representative of Class II SLRP. We identified structural defects in collagen fibril and elastic fiber organization in nonpregnant mice lacking decorin, or biglycan or lumican with variable resolution of defects noted during pregnancy. The severity of collagen and elastic fiber defects was greater in nonpregnant mice lacking both decorin and biglycan and defects were maintained throughout pregnancy. Loss of biglycan alone reduced tissue extensibility in nonpregnant mice while loss of both decorin and biglycan manifested in decreased rupture stretch in late pregnancy. Collagen cross-link density was similar in the Class I SLRP null mice as compared to wild-type nonpregnant and pregnant controls. A broader range in collagen fibril diameter along with an increase in mean fibril spacing was observed in the mutant mice compared to wild-type controls. Collectively, these findings uncover functional redundancy and hierarchical roles of Class I and Class II SLRPs as key regulators of cervical ECM remodeling in pregnancy. These results expand our understating of the critical role SLRPs play to maintain ECM homeostasis in the cervix.
Collapse
Affiliation(s)
- Mariano Colon-Caraballo
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biological Science, The University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Nicole Lee
- Department of Mechanical Engineering, Columbia University New York, New York 10027
| | - Shanmugasundaram Nallasamy
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biological Science, The University of Texas Southwestern Medical Center, Dallas, Texas 75390,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont Burlington, Vermont 05405
| | - Kristin Myers
- Department of Mechanical Engineering, Columbia University New York, New York 10027
| | - David Hudson
- Department of Orthopaedics and Sports Medicine, University of Washington Seattle, Washington 98165
| | - Renato V. Iozzo
- Department of Pathology, Anatomy, and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Mala Mahendroo
- Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biological Science, The University of Texas Southwestern Medical Center, Dallas, Texas 75390,Correspondence to: Mala Mahendroo, Ph.D, Department of Ob/Gyn and Cecil H. and Ida Green Center for Reproductive Biological Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas 75390.
| |
Collapse
|
20
|
Diehl V, Huber LS, Trebicka J, Wygrecka M, Iozzo RV, Schaefer L. The Role of Decorin and Biglycan Signaling in Tumorigenesis. Front Oncol 2021; 11:801801. [PMID: 34917515 PMCID: PMC8668865 DOI: 10.3389/fonc.2021.801801] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
The complex and adaptive nature of malignant neoplasm constitute a major challenge for the development of effective anti-oncogenic therapies. Emerging evidence has uncovered the pivotal functions exerted by the small leucine-rich proteoglycans, decorin and biglycan, in affecting tumor growth and progression. In their soluble forms, decorin and biglycan act as powerful signaling molecules. By receptor-mediated signal transduction, both proteoglycans modulate key processes vital for tumor initiation and progression, such as autophagy, inflammation, cell-cycle, apoptosis, and angiogenesis. Despite of their structural homology, these two proteoglycans interact with distinct cell surface receptors and thus modulate distinct signaling pathways that ultimately affect cancer development. In this review, we summarize growing evidence for the complex roles of decorin and biglycan signaling in tumor biology and address potential novel therapeutic implications.
Collapse
Affiliation(s)
- Valentina Diehl
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Lisa Sophie Huber
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Jonel Trebicka
- Department of Internal Medicine I, Goethe University, Frankfurt, Germany
| | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung, Member of the German Center for Lung Research, University of Giessen and Marburg Lung Center, Giessen, Germany
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, United States
| | - Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| |
Collapse
|
21
|
Zheng Y, Huang C, Zhao L, Chen Y, Liu F. Regulation of decorin by ursolic acid protects against non-alcoholic steatohepatitis. Biomed Pharmacother 2021; 143:112166. [PMID: 34560554 DOI: 10.1016/j.biopha.2021.112166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 11/21/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) has become a global health issue, which poses additional financial burden to public health care. However, no specific pharmacological therapy is recommended in current guidelines. Ursolic acid (UA) has been proven to perform multiple biological activities, thereby having a broad application prospect in healthcare field. Thus, this current research was conducted to investigate the protective mechanisms of UA on NASH. Integrative genomic analyses were performed to identify characteristic genes for NASH, and human proteomics chip was applied to seek out differentially binding proteins for UA. The combining bioinformatic analyses revealed 529 and 502 differentially expressed genes for NASH and UA, respectively. And further enrichment analyses indicated that IGF-IR signaling pathway was intimately involved in the therapeutic effects of UA on NASH. Experimental studies displayed that UA up-regulated the decorin expression to activate IGF-IR signaling as well as to inhibit HIF-1 signaling, resulting in alleviation on metabolic dysfunction, liver steatosis, inflammation and hypoxia in high-fat-fed mice. And additionally, these results were confirmed by lipotoxic and decorin-interference cell model. Taken together, we found that UA could regulate IGF-IR and HIF-1 signaling pathways via decorin to provide dual protective functions on metabolic dysfunction and liver hypoxia, and therefore turned to be an effective option for the treatment of NASH.
Collapse
Affiliation(s)
- Yiyuan Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chaoyuan Huang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China; The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lina Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Youlan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Fengbin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
22
|
Kiripolsky J, Kasperek EM, Zhu C, Li QZ, Wang J, Yu G, Kramer JM. Immune-Intrinsic Myd88 Directs the Production of Antibodies With Specificity for Extracellular Matrix Components in Primary Sjögren's Syndrome. Front Immunol 2021; 12:692216. [PMID: 34381449 PMCID: PMC8350326 DOI: 10.3389/fimmu.2021.692216] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/12/2021] [Indexed: 02/05/2023] Open
Abstract
Primary Sjögren's syndrome is an autoimmune disease that is predominantly seen in women. The disease is characterized by exocrine gland dysfunction in combination with serious systemic manifestations. At present, the causes of pSS are poorly understood. Pulmonary and renal inflammation are observed in pSS mice, reminiscent of a subset of pSS patients. A growing body of evidence indicates that inflammation mediated by Damage-Associated Molecular Patterns (DAMPs) contributes to autoimmunity, although this is not well-studied in pSS. Degraded extracellular matrix (ECM) constituents can serve as DAMPs by binding pattern-recognition receptors and activating Myd88-dependent signaling cascades, thereby exacerbating and perpetuating inflammatory cascades. The ECM components biglycan (Bgn) and decorin (Dcn) mediate sterile inflammation and both are implicated in autoimmunity. The objective of this study was to determine whether these ECM components and anti-ECM antibodies are altered in a pSS mouse model, and whether this is dependent on Myd88 activation in immune cells. Circulating levels of Bgn and Dcn were similar among pSS mice and controls and tissue expression studies revealed pSS mice had robust expression of both Bgn and Dcn in the salivary tissue, saliva, lung and kidney. Sera from pSS mice displayed increased levels of autoantibodies directed against ECM components when compared to healthy controls. Further studies using sera derived from conditional knockout pSS mice demonstrated that generation of these autoantibodies relies, at least in part, on Myd88 expression in the hematopoietic compartment. Thus, this study demonstrates that ECM degradation may represent a novel source of chronic B cell activation in the context of pSS.
Collapse
Affiliation(s)
- Jeremy Kiripolsky
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Eileen M. Kasperek
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Chengsong Zhu
- Department of Immunology, Microarray & Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Quan-Zhen Li
- Department of Immunology, Microarray & Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jia Wang
- Department of Biostatistics, School of Public Health and Health Professions, The University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Guan Yu
- Department of Biostatistics, School of Public Health and Health Professions, The University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jill M. Kramer
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, United States
- Department of Oral Diagnostics Sciences, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, United States
| |
Collapse
|
23
|
Schaefer L, Dikic I. Autophagy: Instructions from the extracellular matrix. Matrix Biol 2021; 100-101:1-8. [PMID: 34217800 DOI: 10.1016/j.matbio.2021.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 06/27/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023]
Abstract
In recent years, extensive research has uncovered crucial regulatory roles for the extracellular matrix (ECM) in regulating autophagy. Autophagy is a ubiquitous and highly conserved catabolic process that allows the selective removal and recycling of cytosolic components via lysosomal or vacuolar degradation. Due to its pivotal role in cellular homeostasis, the impairment of autophagy is involved in the pathophysiology of numerous diseases, comprising infectious diseases, immune and neurodegenerative disorders, renal and hepatic diseases, intervertebral and cartilage disorders, as well as fibrosis and cancer. Several ECM-derived proteoglycans and proteins, including decorin, biglycan, endorepellin, endostatin, collagen VI, and plasminogen kringle 5, have been identified as strong inducers of autophagy. In contrast, laminin α2, perlecan, and lumican exert opposite function by suppressing autophagy. Importantly, by direct interaction with various receptors, which interplay with their co-receptors and adhesion molecules, the ECM is able to direct autophagy in a molecular and cell context-specific manner. Thus, vast pharmacological potential resides in translating this knowledge into the development of ECM-derived therapeutics selectively regulating autophagy.
Collapse
Affiliation(s)
- Liliana Schaefer
- Institute of Pharmacology and Toxicology, Goethe University, Frankfurt, Germany.
| | - Ivan Dikic
- Institute of Biochemistry II, School of Medicine, Goethe University, Frankfurt, Germany; Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt, Germany.
| |
Collapse
|
24
|
Biglycan: A regulator of hepatorenal inflammation and autophagy. Matrix Biol 2021; 100-101:150-161. [DOI: 10.1016/j.matbio.2021.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 02/07/2023]
|
25
|
Neill T, Kapoor A, Xie C, Buraschi S, Iozzo RV. A functional outside-in signaling network of proteoglycans and matrix molecules regulating autophagy. Matrix Biol 2021; 100-101:118-149. [PMID: 33838253 PMCID: PMC8355044 DOI: 10.1016/j.matbio.2021.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Proteoglycans and selected extracellular matrix constituents are emerging as intrinsic and critical regulators of evolutionarily conversed, intracellular catabolic pathways. Often, these secreted molecules evoke sustained autophagy in a variety of cell types, tissues, and model systems. The unique properties of proteoglycans have ushered in a paradigmatic shift to broaden our understanding of matrix-mediated signaling cascades. The dynamic cellular pathway controlling autophagy is now linked to an equally dynamic and fluid signaling network embedded in a complex meshwork of matrix molecules. A rapidly emerging field of research encompasses multiple matrix-derived candidates, representing a menagerie of soluble matrix constituents including decorin, biglycan, endorepellin, endostatin, collagen VI and plasminogen kringle 5. These matrix constituents are pro-autophagic and simultaneously anti-angiogenic. In contrast, perlecan, laminin α2 chain, and lumican have anti-autophagic functions. Mechanistically, each matrix constituent linked to intracellular catabolic events engages a specific cell surface receptor that often converges on a common core of the autophagic machinery including AMPK, Peg3 and Beclin 1. We consider this matrix-evoked autophagy as non-canonical given that it occurs in an allosteric manner and is independent of nutrient availability or prevailing bioenergetics control. We propose that matrix-regulated autophagy is an important outside-in signaling mechanism for proper tissue homeostasis that could be therapeutically leveraged to combat a variety of diseases.
Collapse
Affiliation(s)
- Thomas Neill
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | - Aastha Kapoor
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Christopher Xie
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Simone Buraschi
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Renato V Iozzo
- Department of Pathology, Anatomy, and Cell Biology, and the Translational Cellular Oncology Program, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
26
|
Matsushima N, Miyashita H, Kretsinger RH. Sequence features, structure, ligand interaction, and diseases in small leucine rich repeat proteoglycans. J Cell Commun Signal 2021; 15:519-531. [PMID: 33860400 DOI: 10.1007/s12079-021-00616-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/25/2021] [Indexed: 12/26/2022] Open
Abstract
Small leucine rich repeat proteoglycans (SLRPs) are a group of active components of the extracellular matrix in all tissues. SLRPs bind to collagens and regulate collagen fibril growth and fibril organization. SLRPs also interact with various cytokines and extracellular compounds, which lead to various biological functions such cell adhesion and signaling, proliferation, and differentiation. Mutations in SLRP genes are associated with human diseases. Now crystal structures of five SLRPs are available. We describe some features of amino acid sequence and structures of SLRPs. We also review ligand interactions and then discuss the interaction surfaces. Furthermore, we map mutations associated with human diseases and discuss possible effects on structures by the mutations.
Collapse
Affiliation(s)
- Norio Matsushima
- Division of Bioinformatics, Institute of Tandem Repeats, Noboribetsu, 059-0464, Japan.
- Center for Medical Education, Sapporo Medical University, Sapporo, 060-8556, Japan.
| | - Hiroki Miyashita
- Division of Bioinformatics, Institute of Tandem Repeats, Noboribetsu, 059-0464, Japan
- Hokubu Rinsho Co., Ltd, Sapporo, 060⎼0061, Japan
| | - Robert H Kretsinger
- Department of Biology, University of Virginia, Charlottesville, VA, 22904, USA
| |
Collapse
|
27
|
Kiripolsky J, Kasperek EM, Zhu C, Li QZ, Wang J, Yu G, Kramer JM. Tissue-specific activation of Myd88-dependent pathways governs disease severity in primary Sjögren's syndrome. J Autoimmun 2021; 118:102608. [PMID: 33596533 PMCID: PMC8299268 DOI: 10.1016/j.jaut.2021.102608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 12/17/2022]
Abstract
Myd88 activation is an important driver of autoimmunity. Primary Sjögren's syndrome (pSS) is an autoimmune disease characterized by exocrine gland dysfunction in combination with serious systemic disease manifestations. Myd88-dependent signaling networks remain incompletely understood in the context of pSS. The objective of this study was to establish the contribution of tissue-specific Myd88 activation to local (exocrine) and systemic pSS manifestations. To this end, we generated two novel conditional knockout pSS mouse models; one lacking Myd88 in hematopoietic cells and a second strain in which Myd88 was deleted in the stromal compartment. Spontaneous production of inflammatory mediators was altered in salivary tissue, and nephritis was diminished in both conditional knockout strains. In contrast, pulmonary inflammation was increased in mice lacking Myd88 in hematopoietic cells and was reduced when Myd88 was ablated in stromal cells. Finally, anti-nuclear autoantibodies (ANAs) were attenuated in pSS mice lacking Myd88 in immune cells. Additionally, the ANA-specific B cell repertoire was skewed in the Myd88-deficient strains. Collectively, these data demonstrate that Myd88 activation in specific cell types is essential for distinct aspects of pSS pathology.
Collapse
Affiliation(s)
- Jeremy Kiripolsky
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Eileen M Kasperek
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, 14214, USA
| | - Chengsong Zhu
- Department of Immunology, Microarray & Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Quan-Zhen Li
- Department of Immunology, Microarray & Immune Phenotyping Core Facility, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Jia Wang
- Department of Biostatistics, School of Public Health and Health Professions, The University at Buffalo, State University of New York, 3435 Main Street, 718 Kimball Tower, Buffalo, NY, 14214, USA
| | - Guan Yu
- Department of Biostatistics, School of Public Health and Health Professions, The University at Buffalo, State University of New York, 3435 Main Street, 718 Kimball Tower, Buffalo, NY, 14214, USA
| | - Jill M Kramer
- Department of Oral Biology, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, 14214, USA; Department of Oral Diagnostics Sciences, School of Dental Medicine, The University at Buffalo, State University of New York, Buffalo, NY, 14214, USA.
| |
Collapse
|