1
|
Sun Y, Huang S, Zhang B, Peng Y, Lu H, Jia Y, Sun R, Zhang F, Zhou J, Peng L, Li M, Zhang W, Fei Y. Efficacy and safety of anti-CD19 CAR-T in a mouse model of IgG4-related disease. Int Immunopharmacol 2025; 145:113779. [PMID: 39672025 DOI: 10.1016/j.intimp.2024.113779] [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: 09/30/2024] [Revised: 11/30/2024] [Accepted: 12/01/2024] [Indexed: 12/15/2024]
Abstract
Dysregulated B-cell activation plays pivotal roles in IgG4-related disease (IgG4-RD), which makes B-cell depletion a potential strategy for IgG4-RD treatment. In this study, we aimed to investigate the feasibility of applying anti-CD19 chimeric antigen receptor T(CAR-T) cell therapy to IgG4-RD treatment in a mouse disease model based on LatY136F knock-in (Lat) mice. We constructed murine anti-CD19 CARs with either CD28 or 4-1BB as the intracellular costimulatory motif and evaluated the therapeutic function of the corresponding CAR-T cells by infusing them into Lat mice. Next, we assessed the safety of CAR-T infusion by evaluating the risk of cytokine release syndrome (CRS) and the antiviral capabilities in a mouse influenza infection model. Finally, we performed human anti-CD19 CAR-T manufacturing from IgG4-RD patients and evaluated its activation level and functional effects in vitro. Compared with 1D3 antibody treatment, the adoptive transfer of anti-CD19 CAR-T cells with CD28 costimulatory motif showed comparable B-cell-depletion effect in Lat mice. Furthermore, the transfer of syngeneic anti-CD19 CAR-T cells also decreased the percentage of plasma cells as well as IL-4 secreting Th cells, therefore attenuating the inflammation and fibrosis condition. CAR-T cells with CD28 costimulatory motif showed better therapeutic efficiency without the incidence of serious CRS events or increasing the risk of infection. In addition, we validated the feasibility of human CAR-T preparation in vitro from IgG4-RD patients. Taken together, these results show that anti-CD19 CAR-T therapy was effective in the treatment of a murine model of IgG4-RD, indicating its potential for clinical use in patients.
Collapse
Affiliation(s)
- Yeting Sun
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Sicheng Huang
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Bo Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yu Peng
- Department of Rheumatology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang, China
| | - Hui Lu
- Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Yimeng Jia
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Ruijie Sun
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | | | - Jiaxin Zhou
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Linyi Peng
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Mengtao Li
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Wen Zhang
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; The Ministry of Education Key Laboratory, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China.
| | - Yunyun Fei
- Department of Rheumatology and Clinical Immunology, Department of Health Management, Peking Union Medical College and Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, China; Department of Health Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Radosavljevic T, Brankovic M, Djuretić J, Grujic-Milanovic J, Kovacic M, Jevtic J, Stankovic S, Samardzic J, Vucevic D, Jakovljevic V. Alpinetin Exhibits Antioxidant and Anti-Inflammatory Effects in C57BL/6 Mice with Alcoholic Liver Disease Induced by the Lieber-DeCarli Ethanol Liquid Diet. Int J Mol Sci 2024; 26:86. [PMID: 39795945 PMCID: PMC11720451 DOI: 10.3390/ijms26010086] [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: 10/25/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 01/13/2025] Open
Abstract
Alcohol-associated liver disease (ALD) is a common non-communicable chronic liver disease characterized by a spectrum of conditions ranging from steatosis and alcohol-associated steatohepatitis (AH) to fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The pathogenesis of ALD involves a complex interplay of various molecular, biochemical, genetic, epigenetic, and environmental factors. While the mechanisms are well studied, therapeutic options remain limited. Alpinetin, a natural flavonoid with antioxidant and anti-inflammatory properties, has shown potential hepatoprotective effects, though its efficacy in ALD remains unexplored. This study investigated the hepatoprotective effects of alpinetin using a Lieber-DeCarli ethanol liquid diet model of ALD in C57BL/6 mice. Mice were divided into three groups: the control group, the ethanol group, and the ethanol group treated with alpinetin. Serum activity of ALT, AST, γ-GT, and ALP was measured to assess liver function, along with antioxidative and oxidative/nitrosative stress markers in liver tissue. Pro-inflammatory cytokines and endoplasmic reticulum (ER) stress parameters in liver tissue were also evaluated. Histological assessment of disease activity was performed using the SALVE grading and staging system. Treatment with alpinetin significantly reduced serum levels of ALT, AST, γ-GT, and oxidative/nitrosative stress markers while increasing antioxidative markers. The levels of pro-inflammatory cytokines and ER stress parameters were significantly decreased. Histological analysis demonstrated reduced steatosis, hepatocyte ballooning, and inflammation. These findings suggest that alpinetin holds promise as a potential therapeutic agent for managing ALD.
Collapse
Affiliation(s)
- Tatjana Radosavljevic
- Institute of Pathophysiology “Ljubodrag Buba Mihailović”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Milica Brankovic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.B.); (J.S.)
| | - Jasmina Djuretić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia;
| | - Jelica Grujic-Milanovic
- Institute for Medical Research, National Institute of the Republic of Serbia, Department of Cardiovascular Research, University of Belgrade, 11000 Belgrade, Serbia;
| | - Marijana Kovacic
- Group of Immunology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| | - Jovan Jevtic
- Institute of Pathology ‘Dr Đorđe Joannović’, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Sanja Stankovic
- Centre for Medical Biochemistry, University Clinical Centre of Serbia, 11000 Belgrade, Serbia;
- Department of Biochemistry, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Janko Samardzic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.B.); (J.S.)
| | - Danijela Vucevic
- Institute of Pathophysiology “Ljubodrag Buba Mihailović”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Vladimir Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
- Center of Excellence for the Study of Redox Balance in Cardiovascular and Metabolic Disorders, University of Kragujevac, 34000 Kragujevac, Serbia
- Department of Human Pathology, First Moscow State Medical University I.M. Sechenov, Trubetskaya Street 8, Str. 2, 119991 Moscow, Russia
| |
Collapse
|
3
|
Abdoli M, Hoseini SM, Sandoghsaz RS, Javaheri A, Montazeri F, Moshtaghioun SM. Endometriotic lesions and their recurrence: A Study on the mediators of immunoregulatory (TGF-β/miR-20a) and stemness (NANOG/miR-145). J Reprod Immunol 2024; 166:104336. [PMID: 39366216 DOI: 10.1016/j.jri.2024.104336] [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: 04/04/2024] [Revised: 07/21/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024]
Abstract
Endometriosis is a common estrogen-dependent disease that involves various cellular processes. Additionally, miRNAs play a crucial role in the development of the disease as an important component of the microenvironment. In this study, tissue specimens of eutopic and ectopic lesions of 20 women, whose endometriosis was later approved by the pathology laboratory, were biopsied through laparoscopy. As a control group, endometrial tissue specimens were collected from 20 women who underwent curettage for reasons unrelated to endometriosis. The expression levels of miR-20A and miR-145 and their target genes, TGF-β and NANOG, were measured in these samples as markers of stemness and immunomodulatory properties, respectively. The study also aimed to compare the expression levels of target genes and miRNAs in ectopic lesions regarding endometriosis recurrence post-surgery. The study revealed that the expression of TGF-β and NANOG genes was significantly upregulated in endometriotic tissues compared to the control group. There was also a notable increase in miR-20A and miR-145 expression in the endometriotic tissues compared to the control group. While there was no significant correlation between the expression of miR-20a and TGF-β, we observed a negative correlation between the expression level of miR-145 and NANOG. Additionally, the ROC curve analysis emphasized miR-14 as a potential biomarker for endometriosis over miR-20a. However, our findings on disease recurrence underscore the importance of miR-20a in the early detection of endometriosis recurrence.
Collapse
Affiliation(s)
| | - Seyed Mehdi Hoseini
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Reyhaneh Sadat Sandoghsaz
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Atiyeh Javaheri
- Department of Obstetrics and Gynecology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Fateme Montazeri
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | | |
Collapse
|
4
|
Lin M, Lee Y, Liao J, Chou C, Yang Y. PTGES is involved in myofibroblast differentiation via HIF-1α-dependent glycolysis pathway. J Cell Mol Med 2024; 28:e70157. [PMID: 39417702 PMCID: PMC11484478 DOI: 10.1111/jcmm.70157] [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: 01/22/2024] [Revised: 09/09/2024] [Accepted: 10/01/2024] [Indexed: 10/19/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Patients with lung cancer usually exhibit poor prognoses and low 5-year survival rates. Idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD) are both chronic lung dysfunctions resulting in lung fibrosis and increased risk of lung cancer. Myofibroblasts contribute to the progression of asthma, COPD and IPF, leading to fibrosis in the airway and lungs. A growing body of evidence demonstrates that metabolic reprogramming is a major hallmark of fibrosis, being important in the progression of fibrosis. Using gene expression microarray, we identified and validated that the lipid metabolic pathway was upregulated in lung fibroblasts upon interleukin (IL)-4, IL-13 and tumour necrosis factor (TNF)-α treatment. In this study, we described that prostaglandin E synthase (PTGES) was upregulated in lung fibroblasts after IL-4, IL-13 and TNF-α treatments. PTGES increased α-SMA levels and promoted lung fibroblast cell migration and invasion abilities. Furthermore, PTGES was upregulated in a lung fibrosis rat model in vivo. PTGES increased AKT phosphorylation, leading to activation of the HIF-1α-glycolysis pathway in lung fibroblast cells. HIF-1α inhibitor or 2-DG treatments reduced α-SMA expression in recombinant PTGES (rPTGES)-treated lung fibroblast cells. Targeting PGE2 signalling in PTGES-overexpressing cells by a PTGES inhibitor reduced α-SMA expression. In conclusion, the results of this study demonstrate that PTGES increases the expression of myofibroblast marker via HIF-1α-dependent glycolysis and contributes to myofibroblast differentiation.
Collapse
Affiliation(s)
- Min‐Hsi Lin
- Division of Chest MedicineKaohsiung Veterans General HospitalKaohsiungTaiwan
| | - Yi‐Chen Lee
- Department of Anatomy, School of Medicine, College of MedicineKaohsiung Medical UniversityKaohsiungTaiwan
| | - Jia‐Bin Liao
- Department of Pathology and Laboratory MedicineKaohsiung Veterans General HospitalKaohsiungTaiwan
| | - Chih‐Yu Chou
- Department of Medical Education and ResearchKaohsiung Veterans General HospitalKaohsiungTaiwan
| | - Yi‐Fang Yang
- Department of Medical Education and ResearchKaohsiung Veterans General HospitalKaohsiungTaiwan
| |
Collapse
|
5
|
Karaca ZM, Karaca G, Kayhan B, Gül M, Ersan V, Gözükara Bağ H, Yeşilada E. Chronic liver fibrosis induction in aging causes significant ultra-structural deterioration in liver and alteration on immune response gene expressions in liver-spleen axis. Ultrastruct Pathol 2024; 48:261-273. [PMID: 38842161 DOI: 10.1080/01913123.2024.2360447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024]
Abstract
The relationship between damage to the liver and spleen by aging and the immune response status in these two organs, which are anatomically and immunologically interconnected, is unknown. The authors investigated the histopathological, ultrastructural, and immunological effects of aging in young and aged fibrotic mice by using an experimental model. Four groups were planned, with 10 mice in each experimental group. The levels of fibrosis and ultrastructural destruction in the liver were determined by α-SMA staining and TEM analysis. Expression levels of immunity genes (Il2, Il4, Il6, Il10, Il12, Il17, Tnf, Ifng, Tgfb1, Gata3, Rorc, Tbx21, Foxp3, Ccl2, Ccr2, Cxcr3, Pf4, Cxcl10) were carried out by qRT-PCR. While structural disorders were detected in the mitochondria of aged healthy group, cellular destruction in the fibrosis-induced elderly group was at a dramatic level. Fibrosis induction in aged mice caused an elevation in the expression of chemokines (CCl2, CXCL10, CCR2) and cytokine (IL-17a) genes that induce autoinflammatory response in the liver. Unlike the cellular pathology and genes activated in fibrosis in youth and the natural occurrence of fibrosis with aging, induction of fibrosis during aging causes deterioration in the liver and expression of genes responsible for autoimmunity in both the liver and spleen.
Collapse
Affiliation(s)
- Zeynal Mete Karaca
- Department of Medical Biology and Genetics, Faculty of Medicine, İnönü University, Malatya, Türkiye
- Department of Genetıcs, Faculty of Medıcıne, Kırklarel' Unıversıty, Kırklarelı, Türkıye
| | - Gamze Karaca
- Department of Medical Biology and Genetics, Faculty of Medicine, İnönü University, Malatya, Türkiye
| | - Başak Kayhan
- Liver Transplantation Institute, Transplantation Immunology Laboratory, İnönü University, Malatya, Türkiye
- Department of Microbiology, Faculty of Pharmacy, Anadolu University, Eskişehir, Türkiye
| | - Mehmet Gül
- Department of Histology and Embryology, Faculty of Medicine, İnönü University, Malatya, Türkiye
| | - Veysel Ersan
- Liver Transplantation Institute, Department of General Surgery, İnönü University, Malatya, Türkiye
| | - Harika Gözükara Bağ
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, İnönü University, Malatya, Türkiye
| | - Elif Yeşilada
- Department of Medical Biology and Genetics, Faculty of Medicine, İnönü University, Malatya, Türkiye
| |
Collapse
|
6
|
Xin S, Liu X, He C, Gao H, Wang B, Hua R, Gao L, Shang H, Sun F, Xu J. Inflammation accelerating intestinal fibrosis: from mechanism to clinic. Eur J Med Res 2024; 29:335. [PMID: 38890719 PMCID: PMC11184829 DOI: 10.1186/s40001-024-01932-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024] Open
Abstract
Intestinal fibrosis is a prevalent complication of IBD that that can frequently be triggered by prolonged inflammation. Fibrosis in the gut can cause a number of issues, which continue as an ongoing challenge to healthcare systems worldwide. The primary causes of intestinal fibrosis are soluble molecules, G protein-coupled receptors, epithelial-to-mesenchymal or endothelial-to-mesenchymal transition, and the gut microbiota. Fresh perspectives coming from in vivo and in vitro experimental models demonstrate that fibrogenic pathways might be different, at least to some extent, independent of the ones that influence inflammation. Understanding the distinctive procedures of intestinal fibrogenesis should provide a realistic foundation for targeting and blocking specific fibrogenic pathways, estimating the risk of fibrotic consequences, detecting early fibrotic alterations, and eventually allowing therapy development. Here, we first summarize the inflammatory and non-inflammatory components of fibrosis, and then we elaborate on the underlying mechanism associated with multiple cytokines in fibrosis, providing the framework for future clinical practice. Following that, we discuss the relationship between modernization and disease, as well as the shortcomings of current studies. We outline fibrosis diagnosis and therapy, as well as our recommendations for the future treatment of intestinal fibrosis. We anticipate that the global review will provides a wealth of fresh knowledge and suggestions for future fibrosis clinical practice.
Collapse
Affiliation(s)
- Shuzi Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Xiaohui Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Department of Clinical Laboratory, Aerospace Clinical Medical College, Aerospace Central Hospital, Beijing, 100039, China
| | - Boya Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Lei Gao
- Department of Intelligent Medical Engineering, School of Biomedical Engineering, Capital Medical University, Beijing, 100069, China
| | - Hongwei Shang
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, 100069, China
| | - Fangling Sun
- Department of Laboratory Animal Research, Xuan Wu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Jingdong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
7
|
Rieder F, Mukherjee PK, Massey WJ, Wang Y, Fiocchi C. Fibrosis in IBD: from pathogenesis to therapeutic targets. Gut 2024; 73:854-866. [PMID: 38233198 PMCID: PMC10997492 DOI: 10.1136/gutjnl-2023-329963] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/29/2023] [Indexed: 01/19/2024]
Abstract
BACKGROUND Intestinal fibrosis resulting in stricture formation and obstruction in Crohn's disease (CD) and increased wall stiffness leading to symptoms in ulcerative colitis (UC) is among the largest unmet needs in inflammatory bowel disease (IBD). Fibrosis is caused by a multifactorial and complex process involving immune and non-immune cells, their soluble mediators and exposure to luminal contents, such as microbiota and environmental factors. To date, no antifibrotic therapy is available. Some progress has been made in creating consensus definitions and measurements to quantify stricture morphology for clinical practice and trials, but approaches to determine the degree of fibrosis within a stricture are still lacking. OBJECTIVE We herein describe the current state of stricture pathogenesis, measuring tools and clinical trial endpoints development. DESIGN Data presented and discussed in this review derive from the past and recent literature and the authors' own research and experience. RESULTS AND CONCLUSIONS Significant progress has been made in better understanding the pathogenesis of fibrosis, but additional studies and preclinical developments are needed to define specific therapeutic targets.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Pranab K Mukherjee
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - William J Massey
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Yan Wang
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Mota F. Atopic Dermatitis Patient With Hepatitis C Treated With Dupilumab-A Case Report. ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:T420-T421. [PMID: 38325544 DOI: 10.1016/j.ad.2024.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/06/2022] [Indexed: 02/09/2024] Open
Affiliation(s)
- F Mota
- Photobiology and Cutaneous Immunology Unit, Hospital Senhora da Oliveira, Guimarães, Portugal.
| |
Collapse
|
9
|
Mota F. Atopic Dermatitis Patient With Hepatitis C Treated With Dupilumab-A Case Report. ACTAS DERMO-SIFILIOGRAFICAS 2024; 115:420-421. [PMID: 37290670 DOI: 10.1016/j.ad.2022.07.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/29/2022] [Accepted: 07/06/2022] [Indexed: 06/10/2023] Open
Affiliation(s)
- F Mota
- Photobiology and Cutaneous Immunology Unit, Hospital Senhora da Oliveira, Guimarães, Portugal.
| |
Collapse
|
10
|
Martin KR, Gamell C, Tai TY, Bonelli R, Hansen J, Tatoulis J, Alhamdoosh M, Wilson N, Wicks I. Whole blood transcriptomics reveals granulocyte colony-stimulating factor as a mediator of cardiopulmonary bypass-induced systemic inflammatory response syndrome. Clin Transl Immunology 2024; 13:e1490. [PMID: 38375330 PMCID: PMC10875393 DOI: 10.1002/cti2.1490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 12/20/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Objectives Systemic inflammatory response syndrome (SIRS) is a frequent complication of cardiopulmonary bypass (CPB). SIRS is associated with significant morbidity and mortality, but its pathogenesis remains incompletely understood, and as a result, biomarkers are lacking and treatment remains expectant and supportive. This study aimed to understand the pathophysiological mechanisms driving SIRS induced by CPB and identify novel therapeutic targets that might reduce systemic inflammation and improve patient outcomes. Methods Twenty-one patients undergoing cardiac surgery and CPB were recruited, and blood was sampled before, during and after surgery. SIRS was defined using the American College of Chest Physicians/Society of Critical Care Medicine criteria. We performed immune cell profiling and whole blood transcriptomics and measured individual mediators in plasma/serum to characterise SIRS induced by CPB. Results Nineteen patients fulfilled criteria for SIRS, with a mean duration of 2.7 days. Neutrophil numbers rose rapidly with CPB and remained elevated for at least 48 h afterwards. Transcriptional signatures associated with neutrophil activation and degranulation were enriched during CPB. We identified a network of cytokines governing these transcriptional changes, including granulocyte colony-stimulating factor (G-CSF), a regulator of neutrophil production and function. Conclusions We identified neutrophils and G-CSF as major regulators of CPB-induced systemic inflammation. Short-term targeting of G-CSF could provide a novel therapeutic strategy to limit neutrophil-mediated inflammation and tissue damage in SIRS induced by CPB.
Collapse
Affiliation(s)
- Katherine R Martin
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
| | | | - Tsin Yee Tai
- WEHIParkvilleVICAustralia
- CSL Innovation, Bio21 InstituteParkvilleVICAustralia
| | - Roberto Bonelli
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- CSL Innovation, Bio21 InstituteParkvilleVICAustralia
| | | | - James Tatoulis
- Cardiothoracic SurgeryRoyal Melbourne HospitalParkvilleVICAustralia
- Department of SurgeryUniversity of MelbourneParkvilleVICAustralia
| | | | | | - Ian Wicks
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- Department of RheumatologyRoyal Melbourne HospitalParkvilleVICAustralia
| |
Collapse
|
11
|
Saad EE, Michel R, Borahay MA. Immunosuppressive tumor microenvironment and uterine fibroids: Role in collagen synthesis. Cytokine Growth Factor Rev 2024; 75:93-100. [PMID: 37839993 PMCID: PMC10922281 DOI: 10.1016/j.cytogfr.2023.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 10/17/2023]
Abstract
Uterine fibroids (UF), also called uterine leiomyoma, is one of the most prevalent uterine tumors. UF represents a serious women's health global problem with a significant physical, emotional, and socioeconomic impact. Risk factors for UF include racial disparities, age, race, hormonal factors, obesity, and lifestyle (diet, physical activity, and stress. There are several biological contributors to UF pathogenesis such as cellular proliferation, angiogenesis, and extracellular matrix (ECM) accumulation. This review addresses tumor immune microenvironment as a novel mediator of ECM deposition. Polarization of immune microenvironment towards the immunosuppressive phenotype has been associated with ECM deposition. Immunosuppressive cells include M2 macrophage, myeloid-derived suppressor cells (MDSCs), and Th17 cells, and their secretomes include interleukin 4 (IL-4), IL-10, IL-13, IL-17, IL-22, arginase 1, and transforming growth factor-beta (TGF-β1). The change in the immune microenvironment not only increase tumor growth but also aids in collagen synthesis and ECM disposition, which is one of the main hallmarks of UF pathogenesis. This review invites further investigations on the change in the UF immune microenvironment as well as a novel targeting approach instead of the traditional UF hormonal and supportive treatment.
Collapse
Affiliation(s)
- Eslam E Saad
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rachel Michel
- Department of Population, Family, and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, MD 21205, USA
| | - Mostafa A Borahay
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
12
|
Sadat Sandoghsaz R, Montazeri F, Shafienia H, Mehdi Kalantar S, Javaheri A, Samadi M. Expression of miR-21 &IL-4 in endometriosis. Hum Immunol 2024; 85:110746. [PMID: 38155071 DOI: 10.1016/j.humimm.2023.110746] [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: 12/31/2022] [Revised: 12/14/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Endometriosis characterized with existence of endometrial-like tissue outside the uterus. Fibrosis of ectopic lesions is an important feature of endometriosis. IL-4 induces fibrosis via fibroblast proliferation, collagen production and myofibroblast differentiation. Increasing of miR-21 expression promotes fibroblast activation and fibrosis expansion. The aim of study was to evaluate the expression of miR-21 and its relationship with IL-4 gene expression in endometrial ectopic and eutopic tissues of endometriosis patients. METHODS AND RESULTS Ectopic and eutopic tissue samples were taken from 20 women with endometriosis, and control samples were taken from the endometrium of 20 endometriosis-free women. The relative expression of IL-4 and miR-21 evaluated by Real Time PCR. IL-4 relative gene expression was significantly increased in ectopic tissue compared to eutopic (p = 0.025) and control tissue (p = 0.021). The relative expression of miR-21 gene in ectopic tissue was increased compared to eutopic (p = 0.850) and control tissue (p = 0.978) but these differences were not significant. Also, the correlation between IL-4 and miR-21 relative gene expression was not significant (p = 0.083). CONCLUSION The increased expression of miR-21 in endometrium of women with endometriosis may upregulate the IL-4 gene expression and lead to fibrosis. Further studies may suggest miR-21 and IL-4 as candidates for diagnosis of endometriosis.
Collapse
Affiliation(s)
- Reyhaneh Sadat Sandoghsaz
- International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Montazeri
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hanieh Shafienia
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mehdi Kalantar
- Reproductive & Genetic Unit, Recurrent Abortion Research Center, Yazd Reproductive Science Institute, Yazd University of Medical Sciences, Yazd, Iran
| | - Atiyeh Javaheri
- Department of Obstetrics and Gynecology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Morteza Samadi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
13
|
Sharifinejad N, Mahmoudi E. Dual function of fungi-derived cytokines in inflammatory bowel diseases: protection or inflammation. Gastroenterol Rep (Oxf) 2023; 11:goad068. [PMID: 38058517 PMCID: PMC10697736 DOI: 10.1093/gastro/goad068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/08/2023] [Accepted: 09/27/2023] [Indexed: 12/08/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated inflammatory condition involving both the innate and adaptive immune systems. Recently, the role of intestinal fungal flora and their downstream immune pathways has been highlighted in the pathogenesis of IBD. Cytokines as primary immune mediators require a delicate balance for maintaining intestinal homeostasis. Although most cytokines have a predictable role in either amplifying or attenuating inflammation in IBD, a few cytokines have shown a dual function in the inflammatory state of the intestine. Some of these dual-faced cytokines are also involved in mucosal anti-microbial defense pathways, particularly against intestinal fungal residents. Here, we reviewed the role of these cytokines in IBD pathogenesis to achieve a better understanding of the fungal interactions in the development of IBD.
Collapse
Affiliation(s)
- Niusha Sharifinejad
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Elaheh Mahmoudi
- Department of Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
14
|
Nakamura R, Bing R, Gartling GJ, Garabedian MJ, Branski RC. Dose-Dependent Glucocorticoid Regulation of Transcription Factors in Vocal Fold Fibroblasts and Macrophages. Laryngoscope 2023; 133:2704-2711. [PMID: 36752581 PMCID: PMC10406972 DOI: 10.1002/lary.30594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 02/09/2023]
Abstract
OBJECTIVE Variable outcomes of glucocorticoid (GC) therapy for laryngeal disease are putatively due to diverse interactions of the GC receptor (GR) with cell signaling pathways, limited consideration regarding concentration-dependent effects, and inconsistent selection of GCs. In the current study, we evaluated the concentration-dependent effects of three frequently administered GCs on transcription factors with an emphasis on the phosphorylation of GR at Ser203 and Ser211 regulating the nuclear translocation of GR. This study provides foundational data regarding the diverse functions of GCs to optimize therapeutic approaches. STUDY DESIGN In vitro. METHODS Human vocal fold fibroblasts and THP1-derived macrophages were treated with different concentrations of dexamethasone, methylprednisolone, and triamcinolone in combination with IFN-γ, TNF-α, or IL4. Phosphorylated STAT1, NF-κB family molecules, and phosphorylated STAT6 were analyzed by Western blotting. Ser211-phosphorylated GR (S211-pGR) levels relative to GAPDH and Ser203-phosphorylated GR (S203-pGR) were also analyzed. RESULTS GCs differentially altered phosphorylated STAT1 and NF-κB family molecules in different cell types under IFN-γ and TNF-α stimuli. GCs did not alter phosphorylated STAT6 in IL4-treated macrophages. The three GCs were nearly equivalent. A lower concentration of dexamethasone increased S211-pGR/GAPDH ratios relative to increased S211-pGR/S203-pGR ratios regardless of cell type and treatment. CONCLUSION The three GCs employed in two cell lines had nearly equivalent effects on transcription factor regulation. Relatively high levels of Ser203-phosphorylation at low GC concentrations may be related to concentration-dependent differential effects of GCs in the two cell lines. LEVEL OF EVIDENCE NA Laryngoscope, 133:2704-2711, 2023.
Collapse
Affiliation(s)
- Ryosuke Nakamura
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
| | - Renjie Bing
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
| | - Gary J. Gartling
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
| | | | - Ryan C. Branski
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
- Otolaryngology-Head and Neck Surgery, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
15
|
Moon S, Hong J, Go S, Kim BS. Immunomodulation for Tissue Repair and Regeneration. Tissue Eng Regen Med 2023; 20:389-409. [PMID: 36920675 PMCID: PMC10219918 DOI: 10.1007/s13770-023-00525-0] [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: 10/20/2022] [Revised: 01/11/2023] [Accepted: 01/26/2023] [Indexed: 03/16/2023] Open
Abstract
Various immune cells participate in repair and regeneration following tissue injury or damage, orchestrating tissue inflammation and regeneration processes. A deeper understanding of the immune system's involvement in tissue repair and regeneration is critical for the development of successful reparatory and regenerative strategies. Here we review recent technologies that facilitate cell-based and biomaterial-based modulation of the immune systems for tissue repair and regeneration. First, we summarize the roles of various types of immune cells in tissue repair. Second, we review the principle, examples, and limitations of regulatory T (Treg) cell-based therapy, a representative cell-based immunotherapy. Finally, we discuss biomaterial-based immunotherapy strategies that aim to modulate immune cells using various biomaterials for tissue repair and regeneration.
Collapse
Affiliation(s)
- Sangjun Moon
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jihye Hong
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seokhyeong Go
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Institute of Chemical Processes, Institute of Engineering Research, BioMAX, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
16
|
Nakamura R, Bing R, Gartling GJ, Garabedian MJ, Branski RC. Glucocorticoid Dose Dependency on Gene Expression in Vocal Fold Fibroblasts and Macrophages. Laryngoscope 2023; 133:1169-1175. [PMID: 36779842 PMCID: PMC9925845 DOI: 10.1002/lary.30330] [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: 03/16/2022] [Revised: 06/22/2022] [Accepted: 07/19/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Glucocorticoids (GCs) modulate multiple cellular activities including inflammatory and fibrotic responses. Outcomes of GC treatment for laryngeal disease vary, affording opportunity to optimize treatment. In the current study, three clinically employed GCs were evaluated to identify optimal in vitro concentrations at which GCs mediate favorable anti-inflammatory and fibrotic effects in multiple cell types. We hypothesize a therapeutic window will emerge as a foundation for optimized therapeutic strategies for patients with laryngeal disease. STUDY DESIGN In vitro. METHODS Human vocal fold fibroblasts and human macrophages derived from THP-1 monocytes were treated with 0.03-1000 nM dexamethasone, 0.3-10,000 nM methylprednisolone, and 0.3-10,000 nM triamcinolone in combination with interferon-γ, tumor necrosis factor-α, or interleukin-4. Real-time polymerase chain reaction was performed to analyze inflammatory (CXCL10, CXCl11, PTGS2, TNF, IL1B) and fibrotic (CCN2, LOX, TGM2) genes, and TSC22D3, a target gene of GC signaling. EC50 and IC50 to alter inflammatory and fibrotic gene expression was calculated. RESULTS Interferon-γ and tumor necrosis factor-α increased inflammatory gene expression in both cell types; this response was reduced by GCs. Interleukin-4 increased LOX and TGM2 expression in macrophages; this response was also reduced by GCs. GCs induced TSC22D3 and CCN2 expression independent of cytokine treatment. EC50 for each GC to upregulate CCN2 was higher than the IC50 to downregulate other genes. CONCLUSION Lower concentrations of GCs repressed inflammatory gene expression and only moderately induced genes involved in fibrosis. These data warrant consideration as a foundation for optimized clinical care paradigms to reduce inflammation and mitigate fibrosis. LEVEL OF EVIDENCE NA Laryngoscope, 133:1169-1175, 2023.
Collapse
Affiliation(s)
- Ryosuke Nakamura
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
| | - Renjie Bing
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
| | - Gary J. Gartling
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
| | | | - Ryan C. Branski
- Rehabilitation Medicine, NYU Grossman School of Medicine, New York, NY
- Otolaryngology-Head and Neck Surgery, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
17
|
Jung KI, Lee J, Hong KE, Lee J, Park CK. Cytokines Associated With Onset of a Hypertensive Phase and Surgical Failure After Ahmed Glaucoma Valve Implantation. Am J Ophthalmol 2023; 248:24-34. [PMID: 36410470 DOI: 10.1016/j.ajo.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022]
Abstract
PURPOSE To investigate the relationship between cytokines of the aqueous humor or Tenon capsule, the onset of a hypertensive phase, and surgical failure in patients undergoing Ahmed Glaucoma Valve (AGV) implantation DESIGN: Prospective clinical cohort study. METHODS A total of 36 patients who underwent AGV implantation were included. Samples of aqueous humor and Tenon tissue were collected at the time of surgery. Multiple cytokines were evaluated in the samples using a bead-based multiplex cytokine assay. As for surgical outcomes, a hypertensive phase was defined as an intraocular pressure (IOP) of greater than 21 mm Hg within 3 months after surgery, whereas surgical failure was defined as an IOP greater than 21 mm Hg with maximum tolerable glaucoma medications during 2 consecutive visits. RESULTS Patients who entered a hypertensive phase showed higher transforming growth factor-β2 (TGF-β2) levels in the aqueous humor (P = .012). A longer axial length and higher TGF-β2 concentration of the aqueous humor were associated with higher maximum IOP values during 3 months after surgery per multiple regression analysis (P = .028 and P = .034). In the multiple logistic regression analysis, higher monocyte chemoattractant protein-1 (MCP-1) concentrations in the aqueous humor and higher interleukin-4 concentrations in Tenon tissue were related to surgical failure (P = .022 and P = .040). CONCLUSIONS Greater concentrations of TGF-β2 and MCP-1 were related to surgical outcome after glaucoma drainage device implantation. Further studies are needed to confirm that down-regulation of these cytokines could be helpful in improving surgical outcomes.
Collapse
Affiliation(s)
- Kyoung In Jung
- From the Department of Ophthalmology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jihye Lee
- From the Department of Ophthalmology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyung Euy Hong
- From the Department of Ophthalmology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jiyun Lee
- From the Department of Ophthalmology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chan Kee Park
- From the Department of Ophthalmology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
18
|
The Role of IL-13 and IL-4 in Adipose Tissue Fibrosis. Int J Mol Sci 2023; 24:ijms24065672. [PMID: 36982747 PMCID: PMC10051142 DOI: 10.3390/ijms24065672] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
White adipose tissue (WAT) fibrosis, characterized by an excess of extracellular (ECM) matrix components, is strongly associated with WAT inflammation and dysfunction due to obesity. Interleukin (IL)-13 and IL-4 were recently identified as critical mediators in the pathogenesis of fibrotic diseases. However, their role in WAT fibrosis is still ill-defined. We therefore established an ex vivo WAT organotypic culture system and demonstrated an upregulation of fibrosis-related genes and an increase of α-smooth muscle actin (αSMA) and fibronectin abundance upon dose-dependent stimulation with IL-13/IL-4. These fibrotic effects were lost in WAT lacking il4ra, which encodes for the underlying receptor controlling this process. Adipose tissue macrophages were found to play a key role in mediating IL-13/IL-4 effects in WAT fibrosis as their depletion through clodronate dramatically decreased the fibrotic phenotype. IL-4-induced WAT fibrosis was partly confirmed in mice injected intraperitoneally with IL-4. Furthermore, gene correlation analyses of human WAT samples revealed a strong positive correlation of fibrosis markers with IL-13/IL-4 receptors, whereas IL13 and IL4 correlations failed to confirm this association. In conclusion, IL-13 and IL-4 can induce WAT fibrosis ex vivo and partly in vivo, but their role in human WAT remains to be further elucidated.
Collapse
|
19
|
Secretome from In Vitro Mechanically Loaded Myoblasts Induces Tenocyte Migration, Transition to a Fibroblastic Phenotype and Suppression of Collagen Production. Int J Mol Sci 2021; 22:ijms222313089. [PMID: 34884895 PMCID: PMC8657858 DOI: 10.3390/ijms222313089] [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: 10/27/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
It is known that mechanical loading of muscles increases the strength of healing tendon tissue, but the mechanism involved remains elusive. We hypothesized that the secretome from myoblasts in co-culture with tenocytes affects tenocyte migration, cell phenotype, and collagen (Col) production and that the effect is dependent on different types of mechanical loading of myoblasts. To test this, we used an in vitro indirect transwell co-culture system. Myoblasts were mechanically loaded using the FlexCell® Tension system. Tenocyte cell migration, proliferation, apoptosis, collagen production, and several tenocyte markers were measured. The secretome from myoblasts decreased the Col I/III ratio and increased the expression of tenocyte specific markers as compared with tenocytes cultured alone. The secretome from statically loaded myoblasts significantly enhanced tenocyte migration and Col I/III ratio as compared with dynamic loading and controls. In addition, the secretome from statically loaded myoblasts induced tenocytes towards a myofibroblast-like phenotype. Taken together, these results demonstrate that the secretome from statically loaded myoblasts has a profound influence on tenocytes, affecting parameters that are related to the tendon healing process.
Collapse
|
20
|
Wang J, Lin S, Brown JM, van Wagoner D, Fiocchi C, Rieder F. Novel mechanisms and clinical trial endpoints in intestinal fibrosis. Immunol Rev 2021; 302:211-227. [PMID: 33993489 DOI: 10.1111/imr.12974] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022]
Abstract
The incidence of inflammatory bowel diseases (IBD) worldwide has resulted in a global public health challenge. Intestinal fibrosis leading to stricture formation and bowel obstruction is a frequent complication in Crohn's disease (CD), and the lack of anti-fibrotic therapies makes elucidation of fibrosis mechanisms a priority. Progress has shown that mesenchymal cells, cytokines, microbial products, and mesenteric adipocytes are jointly implicated in the pathogenesis of intestinal fibrosis. This recent information puts prevention or reversal of intestinal strictures within reach through innovative therapies validated by reliable clinical trial endpoints. Here, we review the role of immune and non-immune components of the pathogenesis of intestinal fibrosis, including new cell clusters, cytokine networks, host-microbiome interactions, creeping fat, and their translation for endpoint development in anti-fibrotic clinical trials.
Collapse
Affiliation(s)
- Jie Wang
- Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, China.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sinan Lin
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.,Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jonathan Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - David van Wagoner
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Claudio Fiocchi
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| |
Collapse
|
21
|
Mechanosensitive Regulation of Fibrosis. Cells 2021; 10:cells10050994. [PMID: 33922651 PMCID: PMC8145148 DOI: 10.3390/cells10050994] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Cells in the human body experience and integrate a wide variety of environmental cues. A growing interest in tissue mechanics in the past four decades has shown that the mechanical properties of tissue drive key biological processes and facilitate disease development. However, tissue stiffness is not only a potent behavioral cue, but also a product of cellular signaling activity. This review explores both roles of tissue stiffness in the context of inflammation and fibrosis, and the important molecular players driving such processes. During inflammation, proinflammatory cytokines upregulate tissue stiffness by increasing hydrostatic pressure, ECM deposition, and ECM remodeling. As the ECM stiffens, cells involved in the immune response employ intricate molecular sensors to probe and alter their mechanical environment, thereby facilitating immune cell recruitment and potentiating the fibrotic phenotype. This powerful feedforward loop raises numerous possibilities for drug development and warrants further investigation into the mechanisms specific to different fibrotic diseases.
Collapse
|
22
|
Wang H, Xu GB, Chen H, Pan YX. Maternal high-fat diet activates hepatic interleukin-4 in rat male offspring accompanied by increased eosinophil infiltration. Am J Physiol Gastrointest Liver Physiol 2021; 320:G81-G92. [PMID: 33112158 DOI: 10.1152/ajpgi.00153.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Interleukin-4 (IL-4) is activated as an immune response during infection or tissue injury. Epigenetic programming of maternal high-fat (HF) diet has long-term effects in the offspring. In the present study, we investigated the epigenetic regulation of IL-4 in a maternal HF diet model in the liver of adult offspring. Timed-pregnant Sprague-Dawley rats were fed either control (C) or HF diet throughout gestation and lactation. Offspring were placed on a control diet after weaning, generating C/C and HF/C groups. The liver was collected at 12 wk of age, followed by histological and molecular analysis to investigate the maternal programming effects on IL-4 by HF diet. Maternal HF diet significantly induced mRNA expression and protein level of IL-4 and promoted hypomethylation of Il4 compared with the control group. Methylation-selective PCR (MSP) confirmed that maternal HF diet increased RNA polymerase II, acetylation of histone H4, and dimethylation of histone 3 lysine 4 at the +6 kb region of Il4. Moreover, the rat eosinophil marker Siglec-F was increased and colocalized with IL-4 in the liver. In conclusion, our study indicated that IL-4 was increased in liver cells in response to maternal HF diet. This coincides with DNA hypomethylation in combination with chromatin remodeling at the +6 kb region of the 3' downstream region as well as an induced immune cell infiltration, especially eosinophil infiltration, in the liver of offspring.NEW & NOTEWORTHY The present study identifies that maternal high-fat-diet-induced IL-4 upregulation is associated with DNA hypomethylation at the +6 kb region of the 3' downstream region of the gene. Furthermore, our results confirm that the induced Il4 expression in the liver of male offspring corresponds to the induced immune cell, especially eosinophil, infiltration.
Collapse
Affiliation(s)
- Huan Wang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Guanying Bianca Xu
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Hong Chen
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Yuan-Xiang Pan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
23
|
Daseke MJ, Tenkorang-Impraim MAA, Ma Y, Chalise U, Konfrst SR, Garrett MR, DeLeon-Pennell KY, Lindsey ML. Exogenous IL-4 shuts off pro-inflammation in neutrophils while stimulating anti-inflammation in macrophages to induce neutrophil phagocytosis following myocardial infarction. J Mol Cell Cardiol 2020; 145:112-121. [PMID: 32574573 PMCID: PMC7483959 DOI: 10.1016/j.yjmcc.2020.06.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/08/2020] [Accepted: 06/11/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Macrophages and neutrophils are primary leukocytes involved in the inflammatory response to myocardial infarction (MI). While interleukin (IL)-4 is an in vitro anti-inflammatory stimulus, the MI myocardium does not express a considerable amount of IL-4 but does express IL4 receptors. We hypothesized that continuous exogenous IL-4 infusion starting 24 h after MI would promote a polarization switch in inflammatory cells towards a reparative phenotype. METHODS C57BL/6J male mice (3-6 months of age) were subcutaneously infused with either saline (n = 17) or IL-4 (20 ng/g/day; n = 17) beginning 24 h after MI and evaluated at MI day 3. RESULTS Macrophages and neutrophils were isolated ex vivo from the infarct region and examined. Exogenous IL-4 decreased pro-inflammatory Ccl3, Il12a, Tnfa, and Tgfb1 in neutrophils and increased anti-inflammatory Arg1 and Ym1 in macrophages (all p < .05). Tissue clearance by IL-4 treated neutrophils was not different, while selective phagocytosis of neutrophils doubled in IL-4 treated macrophages (p < .05). Of 24,339 genes examined by RNA-sequencing, 2042 genes were differentially expressed in macrophages from IL-4 stimulated infarct (all FDR p < .05). Pdgfc gene expression was ranked first, increasing 3-fold in macrophages stimulated with IL-4 (p = 1 × 10-9). Importantly, changes in macrophage physiology and transcriptome occurred in the absence of global LV effects. Bone marrow derived monocytes stimulated with mouse recombinant PDGF-CC protein (10 μg/ml) or PDGF-CC blocking antibody (200 ng/ml) did not change Arg1 or Ym1 expression, indicating the in vivo effect of IL-4 to stimulate macrophage anti-inflammatory gene expression was independent of PDGF-CC. CONCLUSIONS Our results indicate that exogenous IL-4 promotes inflammation resolution by turning off pro-inflammation in neutrophils while stimulating anti-inflammation in macrophages to mediate removal of apoptotic neutrophils.
Collapse
Affiliation(s)
- Michael J Daseke
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Mavis A A Tenkorang-Impraim
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, USA
| | - Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Michael R Garrett
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristine Y DeLeon-Pennell
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA; Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, USA.
| |
Collapse
|
24
|
Xing XY, Qiang WJ, Bao JL, Yang RC, Hou J, Tao K, Meng ZQ, Zhang JH, Zhang AJ, Sun XB. Jinbei Oral Liquid ameliorates bleomycin-induced idiopathic pulmonary fibrosis in rats via reversion of Th1/Th2 shift. CHINESE HERBAL MEDICINES 2020; 12:273-280. [PMID: 36119009 PMCID: PMC9476682 DOI: 10.1016/j.chmed.2020.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/17/2020] [Accepted: 03/27/2020] [Indexed: 12/28/2022] Open
|
25
|
Yang B, Zhang G, Elias M, Zhu Y, Wang J. The role of cytokine and immune responses in intestinal fibrosis. J Dig Dis 2020; 21:308-314. [PMID: 32410365 DOI: 10.1111/1751-2980.12879] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/11/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022]
Abstract
The rapidly increasing incidence of inflammatory bowel disease (IBD) in South America, eastern Europe, Asia, and Africa has resulted in a global public health challenge. Intestinal fibrosis is a common complication in patients with long-term IBD, which may develop into stenosis and subsequent obstruction. Hitherto, the origin of IBD is unclear and several factors may be involved, including genetic, immune, environmental and microbial influences. Little is known about how the recurrent inflammation in patients with IBD develops into intestinal fibrosis and currently, there is no suitable treatment to reverse intestinal fibrosis in these patients. Here, we review the role of immune components in the pathogenesis of IBD and intestinal fibrosis, including cytokine networks, host-microbiome interactions, and immune cell trafficking.
Collapse
Affiliation(s)
- Bo Yang
- School of Laboratory Medicine, Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Ge Zhang
- School of Laboratory Medicine, Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Michael Elias
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Yijun Zhu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA.,Department of Gastroenterology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jie Wang
- School of Laboratory Medicine, Henan Key Laboratory of Immunology and Targeted Drug, Xinxiang Medical University, Xinxiang, Henan Province, China.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
26
|
Han X, Caron JM, Brooks PC. Cryptic collagen elements as signaling hubs in the regulation of tumor growth and metastasis. J Cell Physiol 2020; 235:9005-9020. [PMID: 32400053 DOI: 10.1002/jcp.29752] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 03/26/2020] [Accepted: 04/17/2020] [Indexed: 12/23/2022]
Abstract
Structural remodeling of the extracellular matrix is a well-established process associated with tumor growth and metastasis. Tumor and stromal cells that compose the tumor mass function cooperatively to promote the malignant phenotype in part by physically interacting with intact and structurally altered matrix proteins. To this end, collagen represents the most abundant component of the extracellular matrix and is known to control the behavior of histologically distinct tumor types as well as a diversity of stromal cells. Although a significant molecular understanding has been established concerning how cellular interactions with intact collagen govern signaling pathways that control tumor progression, considerably less is known concerning how interactions with cryptic or hidden regions within remodeled collagen may selectively alter signaling cascades, or whether inhibition of these cryptic signaling pathways may represent clinically effective therapeutic strategies. Here, we review the emerging evidence concerning the possible mechanisms for the selective generation of cryptic or hidden elements within collagen and their potential cell surface receptors that may facilitate signal transduction. We discuss the concept that cellular communication links between cell surface receptors and these cryptic collagen elements may serve as functional signaling hubs that coordinate multiple signaling pathways operating within both tumor and stromal cells. Finally, we provide examples to help illustrate the possibility that direct targeting of these unique cryptic signaling hubs may lead to the development of more effective therapeutic strategies to control tumor growth and metastasis.
Collapse
Affiliation(s)
- XiangHua Han
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Jennifer M Caron
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| | - Peter C Brooks
- Maine Medical Center Research Institute, Center for Molecular Medicine, Scarborough, Maine
| |
Collapse
|
27
|
El Ayadi A, Jay JW, Prasai A. Current Approaches Targeting the Wound Healing Phases to Attenuate Fibrosis and Scarring. Int J Mol Sci 2020; 21:ijms21031105. [PMID: 32046094 PMCID: PMC7037118 DOI: 10.3390/ijms21031105] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
Cutaneous fibrosis results from suboptimal wound healing following significant tissue injury such as severe burns, trauma, and major surgeries. Pathologic skin fibrosis results in scars that are disfiguring, limit normal movement, and prevent patient recovery and reintegration into society. While various therapeutic strategies have been used to accelerate wound healing and decrease the incidence of scarring, recent studies have targeted the molecular regulators of each phase of wound healing, including the inflammatory, proliferative, and remodeling phases. Here, we reviewed the most recent literature elucidating molecular pathways that can be targeted to reduce fibrosis with a particular focus on post-burn scarring. Current research targeting inflammatory mediators, the epithelial to mesenchymal transition, and regulators of myofibroblast differentiation shows promising results. However, a multimodal approach addressing all three phases of wound healing may provide the best therapeutic outcome.
Collapse
|
28
|
Farhadihosseinabadi B, Gholipourmalekabadi M, Salimi M, Abdollahifar MA, Bagheri M, Samadikuchaksaraei A, Ghanbarian H, Mozafari M, Kazemi B, Niknejad H. The in vivo effect of Lacto-N-neotetraose (LNnT) on the expression of type 2 immune response involved genes in the wound healing process. Sci Rep 2020; 10:997. [PMID: 31969618 PMCID: PMC6976585 DOI: 10.1038/s41598-020-57860-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/03/2020] [Indexed: 01/08/2023] Open
Abstract
Lacto-n-neotatraose (LNnT) oligosaccharide shows properties such as anti-inflammatory, type 2 immune response induction, induced angiogenesis, and anti-bacterial effects. Here, we hypothesized that the application of LnNT in the skin full-thickness wound can accelerate the healing process through its anti-inflammatory effect as well as induction of type 2 immune responses. In this study, we evaluated the cell viability of fibroblasts in the presence of LNnT. The full-thickness wound model was created by punch biopsy. The mice were treated intradermaly with LNnT at the concentrations of 100 and 200 µg or PBS as a control group. The wounds samples were compared based on the macroscopic and histological evaluations. The amount of collagen deposition and expression of genes involved in type 2 immunity were measured by the hydroxyproline assay and real time PCR method, respectively. Our results showed that LNnT had no negative effect on the cell viability of fibroblasts. LNnT increased the wound closure rate on day 7 post-wounding. H&E stain analysis revealed that mice treated with 200 µg LNnT exhibited better healing score, follicle formation, and lower epidermal thickness index. The mice treated with LNnT exhibited a lower collagen deposition on day 21 and higher collagen content on days 7 and 14 post-treatment. The LNnT groups also exhibited a lower number of neutrophils and a higher number of basal cells and fibroblasts. The expression rate of IL-10, IL-4, and IL-13 was higher in the LNnT groups. These results showed the high potential of LNnT for use in treatment of full-thickness wounds.
Collapse
Affiliation(s)
- Behrouz Farhadihosseinabadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran Universityof Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Salimi
- Department of Biology and Anatomical Sciences, Faculty of Medicine, Shahid Beheshti. University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, Faculty of Medicine, Shahid Beheshti. University of Medical Sciences, Tehran, Iran
| | - Mohammad Bagheri
- Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Samadikuchaksaraei
- Cellular and Molecular Research Centre, Iran Universityof Medical Sciences, Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbarian
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Biology Research center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahram Kazemi
- Cellular and Molecular Biology Research center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
|
30
|
Russo R, Cozzani E, Gasparini G, Parodi A. Targeting interleukin 4 receptor α: A new approach to the treatment of cutaneous autoimmune bullous diseases? Dermatol Ther 2020; 33:e13190. [PMID: 31863534 PMCID: PMC7154653 DOI: 10.1111/dth.13190] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 12/03/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
Bullous pemphigoid, mucous membrane pemphigoid, and pemphigus vulgaris are different cutaneous autoimmune blistering diseases, with complex pathogenic mechanisms. In all of them, a type-2 response is thought to have a central role. Interleukin 4 and Interleukin 13 are crucial cytokines in type-2 response. Treatment of these conditions is often challenging. Dupilumab, a recombinant fully human IgG4 monoclonal antibody with binding specificity to human interleukin-4 receptor IL-4Rα, has the potential to inhibit both IL-4 and IL-13. We propose IL-4Rα as a theoretical drug target for cutaneous autoimmune bullous diseases.
Collapse
Affiliation(s)
- Roberto Russo
- Di.S.SAL Section of DermatologyUniversity of Genoa, Via Pastore 1GenoaItaly
- Section of Dermatology, IRCCS San Martino Policlinic HospitalGenoaItaly
| | - Emanuele Cozzani
- Di.S.SAL Section of DermatologyUniversity of Genoa, Via Pastore 1GenoaItaly
- Section of Dermatology, IRCCS San Martino Policlinic HospitalGenoaItaly
| | - Giulia Gasparini
- Di.S.SAL Section of DermatologyUniversity of Genoa, Via Pastore 1GenoaItaly
- Section of Dermatology, IRCCS San Martino Policlinic HospitalGenoaItaly
| | - Aurora Parodi
- Di.S.SAL Section of DermatologyUniversity of Genoa, Via Pastore 1GenoaItaly
- Section of Dermatology, IRCCS San Martino Policlinic HospitalGenoaItaly
| |
Collapse
|
31
|
Ouaguia L, Moralès O, Aoudjehane L, Wychowski C, Kumar A, Dubuisson J, Calmus Y, Conti F, Delhem N. Hepatitis C Virus Improves Human Tregs Suppressive Function and Promotes Their Recruitment to the Liver. Cells 2019; 8:cells8101296. [PMID: 31652598 PMCID: PMC6829901 DOI: 10.3390/cells8101296] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 12/31/2022] Open
Abstract
Background: The role of regulatory T cells (Tregs) is now well established in the progression of hepatocellular carcinoma (HCC) linked to Hepatitis C virus (HCV) infection. However, nothing is known about the potential interplay between Tregs and HCV. In this pilot study, we have investigated the ability of Tregs to hang HCV on and the subsequent effect on their suppressive function and phenotype. Moreover, we have evaluated how HCV could promote the recruitment of Tregs by infected primary human hepatocytes. Methods: Tregs of healthy donors were incubated with JFH-1/HCVcc. Viral inoculation was assessed using adapted assays (RT-qPCR, Flow Citometry (FACS) and Western Blot (WB). Expression of Tregs phenotypic (CD4, CD25, CD127 and Foxp3) and functional (IL-10, GZMB, TGF-β1 and IL-2) markers was monitored by RT-qPCR, FACS and ELISA. Suppressive activity was validated by suppressive assays. Tregs recruitment by infected primary hepatic cells was evaluated using Boyden Chamber. Results: Tregs express the classical HCV receptors (CD81, CLDN1 and LDLR) and some co-receptors (CD5). HCV inoculation significantly increases the suppressive phenotype and activity of Tregs, and raises their anergy by inducing an unexpected IL-2 production. Moreover, HCV infection induces the expression of chemokines (CCL17, CXCL16, and CCL20) by primary hepatic human hepatocytes and chemokine receptors (CCR4, CXCR6 and CCR6) by Tregs. Finally, infected hepatocytes have a significantly higher potential to recruit Tregs in a seemingly CCL20-dependent manner. Conclusions: Direct interaction between HCV and Tregs represents a newly defined mechanism that could potentiate HCV immune evasion and favor intratumoral recruitment contributing to HCC progression.
Collapse
Affiliation(s)
- Laurissa Ouaguia
- Université Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France.
- CNRS-UMR 8161, F-59000 Lille, France.
- Institut Pasteur de Lille, F-59000 Lille, France.
| | - Olivier Moralès
- Université Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France.
- CNRS-UMR 8161, F-59000 Lille, France.
- Institut Pasteur de Lille, F-59000 Lille, France.
| | - Lynda Aoudjehane
- Sorbonne Université, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), F-75013 Paris, France.
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.
| | - Czeslaw Wychowski
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France.
| | - Abhishek Kumar
- Université Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France.
- CNRS-UMR 8161, F-59000 Lille, France.
- Institut Pasteur de Lille, F-59000 Lille, France.
| | - Jean Dubuisson
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019-UMR 8204-CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France.
| | - Yvon Calmus
- Sorbonne Université, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), F-75013 Paris, France.
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.
- Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Department of Medical Liver Transplantation, F-75013 Paris, France.
| | - Filomena Conti
- Sorbonne Université, INSERM, Institute of Cardiometabolism and Nutrition (ICAN), F-75013 Paris, France.
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), F-75012 Paris, France.
- Assistance Publique-Hôpitaux de Paris (AP-HP), Pitié-Salpêtrière Hospital, Department of Medical Liver Transplantation, F-75013 Paris, France.
| | - Nadira Delhem
- Université Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, F-59000 Lille, France.
- CNRS-UMR 8161, F-59000 Lille, France.
- Institut Pasteur de Lille, F-59000 Lille, France.
| |
Collapse
|
32
|
Antonopoulos I, Daoussis D, Lalioti ME, Markatseli TE, Drosos AA, Taraviras S, Andonopoulos AP, Liossis SNC. B cell depletion treatment decreases CD4+IL4+ and CD4+CD40L+ T cells in patients with systemic sclerosis. Rheumatol Int 2019; 39:1889-1898. [PMID: 31227855 DOI: 10.1007/s00296-019-04350-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/15/2019] [Indexed: 01/29/2023]
Abstract
Recent data suggests that rituximab may favorably affect skin fibrosis and lung function in patients with systemic sclerosis. Based on experimental data suggesting a key role of B and T cells in scleroderma we aimed to explore the effect(s) of rituximab treatment on T cell subpopulations. Fifteen patients with scleroderma who received rituximab treatment and six who received standard treatment alone were recruited. Peripheral CD4+IL4+, CD4+INFγ+, CD4+IL17+ and CD4+CD40L+ T cells were assessed using flow cytometry. Using ELISA, serum levels of IL4 were assessed. Skin CD4+IL4+ T cells were assessed with confocal microscopy from skin biopsies. Following rituximab treatment skin CD4+IL4+ T cells obviously decreased as seen with confocal microscopy. Moreover, peripheral CD4+IL4+ T cells decreased significantly compared to those from patients who received standard treatment alone: median (IQR): 14.9 (22.63-12.88) vs 7.87 (12.81-4.9)%, p = 0.005 and 9.43 (19.53-7.50)% vs 14.86 (21.96-6.75)%, p = NS at baseline and 6 months later respectively, whereas there was no difference in serum IL4 levels. Peripheral CD4+CD40L+ T cells also decreased significantly following rituximab treatment compared to those from patients who received standard treatment alone: median (IQR): 17.78 (25.64-14.44)% vs 8.15 (22.85-3.08)%, p = 0.04 and 22.13 (58.77-8.20)% vs 72.11 (73.05-20.45)%, p = NS at baseline and 6 months later respectively. Furthermore, peripheral CD4+INFγ+ and CD4+IL17+ T cells revealed no differences following rituximab treatment. Our study demonstrates a link between rituximab treatment and CD4+IL4+ T cell decrease both in the skin and peripheral blood of patients with SSc.
Collapse
Affiliation(s)
- Ioannis Antonopoulos
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, University of Patras Medical School, Rion, 26504, Patras, Greece
| | - Dimitrios Daoussis
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, University of Patras Medical School, Rion, 26504, Patras, Greece
| | - Maria-Eleni Lalioti
- Department of Physiology, School of Medicine, University of Patras, Rion, 26504, Patras, Greece
| | - Theodora E Markatseli
- Department of Rheumatology, Ioannina University Hospital, University of Ioannina Medical School, Ioannina, Greece
| | - Alexandros A Drosos
- Department of Rheumatology, Ioannina University Hospital, University of Ioannina Medical School, Ioannina, Greece
| | - Stavros Taraviras
- Department of Physiology, School of Medicine, University of Patras, Rion, 26504, Patras, Greece
| | - Andrew P Andonopoulos
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, University of Patras Medical School, Rion, 26504, Patras, Greece
| | - Stamatis-Nick C Liossis
- Division of Rheumatology, Department of Internal Medicine, Patras University Hospital, University of Patras Medical School, Rion, 26504, Patras, Greece.
| |
Collapse
|
33
|
Bhattacharjee O, Ayyangar U, Kurbet AS, Ashok D, Raghavan S. Unraveling the ECM-Immune Cell Crosstalk in Skin Diseases. Front Cell Dev Biol 2019; 7:68. [PMID: 31134198 PMCID: PMC6514232 DOI: 10.3389/fcell.2019.00068] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 04/09/2019] [Indexed: 01/06/2023] Open
Abstract
The extracellular matrix (ECM) is a complex network of proteins and proteoglycans secreted by keratinocytes, fibroblasts and immune cells. The function of the skin ECM has expanded from being a scaffold that provides structural integrity, to a more dynamic entity that is constantly remodeled to maintain tissue homeostasis. The ECM functions as ligands for cell surface receptors such as integrins, dystroglycans, and toll-like receptors (TLRs) and regulate cellular signaling and immune cell dynamics. The ECM also acts as a sink for growth factors and cytokines, providing critical cues during epithelial morphogenesis. Dysregulation in the organization and deposition of ECMs lead to a plethora of pathophysiological conditions that are exacerbated by aberrant ECM-immune cell interactions. In this review, we focus on the interplay between ECM and immune cells in the context of skin diseases and also discuss state of the art therapies that target the key molecular players involved.
Collapse
Affiliation(s)
- Oindrila Bhattacharjee
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Uttkarsh Ayyangar
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Ambika S. Kurbet
- School of Chemical and Biotechnology, Sastra University, Thanjavur, India
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Driti Ashok
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| | - Srikala Raghavan
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bangalore, India
| |
Collapse
|
34
|
Batsaikhan B, Lu MY, Yeh ML, Huang CI, Huang CF, Lin ZY, Chen SC, Huang JF, Hsieh PH, Chuang WL, Lee JC, Yu ML, Dai CY. Elevated interleukin-4 levels predicted advanced fibrosis in chronic hepatitis C. J Chin Med Assoc 2019; 82:277-281. [PMID: 30946707 DOI: 10.1097/jcma.0000000000000064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cytokine imbalance has been associated with chronic hepatitis C virus (HCV) infection. We hypothesized that cytokines have an important role in fibrosis development in HCV infection. METHODS Data of 92 patients were analyzed retrospectively. Fluorescent Bead immunoassay was used to measure the following serum cytokine levels: Interferon γ, tumor necrosis factor α, interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, and IL-12. Various statistical analyses were used as appropriate. RESULTS Of the 92 HCV-infected patients, 49 (53.3%) were male, 23 (25%) patients had advanced (fibrosis grades 3-4) fibrosis, and the mean age of the study population was 51.9 ± 9.4 years. Elevation of baseline IL-4 level (>490 pg/mL) was associated with liver fibrosis grade by χ test (odds ratio [OR] = 2.99; 95%, CI = 1.02-8.78; p = 0.042) and multivariate logistic regression (OR = 4.26; 95% CI = 1.13-16.02; p = 0.032). Also, IL-4 had strong diagnostic value in advanced liver fibrosis by using area under receiver operating characteristics curve analysis. Assessment of fibrosis score was consequently developed from our findings and compared with other noninvasive serum markers to assess liver fibrosis. CONCLUSION This study provides evidence that increased IL-4 expression predicted advanced liver fibrosis in treatment of naive HCV-infected patients. The newly developed "FIL4" score had good predictive value for advanced fibrosis before treatment and this value was even strong in HCV-genotype 1b patients.
Collapse
Affiliation(s)
- Batbold Batsaikhan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Internal Medicine, Institute of Medical Sciences, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Ming-Ying Lu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Ming-Lun Yeh
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Ching-I Huang
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Chung-Feng Huang
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Chi Mei Hospital, Liouying, Tainan, Taiwan, ROC
| | - Zu-Yau Lin
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Shinn-Cherng Chen
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Jee-Fu Huang
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | | | - Wan-Long Chuang
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Jin-Ching Lee
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Ming-Lung Yu
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
| | - Chia-Yen Dai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Occupational and Environmental Medicine, Health Management Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan, ROC
- Department of Biological Science and Technology, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan, ROC
| |
Collapse
|
35
|
Kotsiou OS, Gourgoulianis KI, Zarogiannis SG. IL-33/ST2 Axis in Organ Fibrosis. Front Immunol 2018; 9:2432. [PMID: 30405626 PMCID: PMC6207585 DOI: 10.3389/fimmu.2018.02432] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/02/2018] [Indexed: 12/19/2022] Open
Abstract
Interleukin 33 (IL-33) is highly expressed in barrier sites, acting via the suppression of tumorigenicity 2 receptor (ST2). IL-33/ST2 axis has long been known to play a pivotal role in immunity and cell homeostasis by promoting wound healing and tissue repair. However, it is also involved in the loss of balance between extensive inflammation and tissue regeneration lead to remodeling, the hallmark of fibrosis. The aim of the current review is to critically evaluate the available evidence regarding the role of the IL-33/ST2 axis in organ fibrosis. The role of the axis in tissue remodeling is better understood considering its crucial role reported in organ development and regeneration. Generally, the IL-33/ST2 signaling pathway has mainly anti-inflammatory/anti-proliferative effects; however, chronic tissue injury is responsible for pro-fibrogenetic responses. Regarding pulmonary fibrosis mature IL-33 enhances pro-fibrogenic type 2 cytokine production in an ST2- and macrophage-dependent manner, while full-length IL-33 is also implicated in the pulmonary fibrotic process in an ST2-independent, Th2-independent fashion. In liver fibrosis, evidence indicate that when acute and massive liver damage occurs, the release of IL-33 might act as an activator of tissue-protective mechanisms, while in cases of chronic injury IL-33 plays the role of a hepatic fibrotic factor. IL-33 signaling has also been involved in the pathogenesis of acute and chronic pancreatitis. Moreover, IL-33 could be used as an early marker for ulcer-associated activated fibroblasts and myofibroblast trans-differentiation; thus one cannot rule out its potential role in inflammatory bowel disease-associated fibrosis. Similarly, the upregulation of the IL-33/ST2 axismay contribute to tubular cell injury and fibrosis via epithelial to mesenchymal transition (EMT) of various cell types in the kidneys. Of note, IL-33 exerts a cardioprotective role via ST2 signaling, while soluble ST2 has been demonstrated as a marker of myocardial fibrosis. Finally, IL-33 is a crucial cytokine in skin pathology responsible for abnormal fibroblast proliferation, leukocyte infiltration and morphologic differentiation of human endothelial cells. Overall, emerging data support a novel contribution of the IL-33/ST2 pathway in tissue fibrosis and highlight the significant role of the Th2 pattern of immune response in the pathophysiology of organ fibrosis.
Collapse
Affiliation(s)
- Ourania S. Kotsiou
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
| | - Konstantinos I. Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
| | - Sotirios G. Zarogiannis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
| |
Collapse
|
36
|
Sheng J, Yang Y, Cui Y, He S, Wang L, Liu L, He Q, Lv T, Han W, Yu W, Hu S, Jin J. M2 macrophage-mediated interleukin-4 signalling induces myofibroblast phenotype during the progression of benign prostatic hyperplasia. Cell Death Dis 2018; 9:755. [PMID: 29988032 PMCID: PMC6037751 DOI: 10.1038/s41419-018-0744-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/23/2018] [Accepted: 04/26/2018] [Indexed: 12/24/2022]
Abstract
Benign prostatic hyperplasia (BPH) is a progressive disease in elderly men, but potential factors accelerating its progression remain largely unknown. The aim of this study was to elucidate the factors affecting BPH progression by understanding the complex mechanisms causing early- progressed BPH, which progresses rapidly and requires surgical intervention before the age of 50. Three groups of human prostate tissue samples, from patients with early-progressed BPH, age-matched prostate and elderly BPH tissues, were collected (n = 25 each). We compared these tissues to determine the histologic features and molecular mechanisms underlying BPH progression. We found that early-progressed BPH samples were characterised by aberrant stromal hyper-proliferation, collagen deposition and increased M2 macrophage infiltration, compared to those from age-matched prostate and elderly BPH tissues. The M2 macrophage–fibroblast co-culture system demonstrated that the myofibroblast phenotypes were strongly induced only in fibroblasts from the early-progressed BPH samples, while the co-cultured M2 macrophages expressed high levels of pro-fibrotic cytokines, such as IL4 and TGFβ1. M2 macrophage-derived IL4, but not TGFβ1, selectively induced the myofibroblast phenotype through the JAK/STAT6, PI3K/AKT and MAPK/ERK signalling pathways in the early-progressed BPH prostate fibroblasts. Taken together, our results indicate that induction of the myofibroblast phenotype may lead to BPH progression through M2 macrophage-mediated IL4 signalling, and that IL4 may represent a potential therapeutic target, allowing the prevention of M2 macrophage activation and fibroblast-to-myofibroblast differentiation.
Collapse
Affiliation(s)
- Jindong Sheng
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Yang Yang
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Yun Cui
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China.,Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Gongti South Road, Chaoyang District, Beijing 100020, China
| | - Shiming He
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Lu Wang
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Libo Liu
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Qun He
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Tianjing Lv
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Wenke Han
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Wei Yu
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China
| | - Shuai Hu
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China.
| | - Jie Jin
- Department of Urology, National Research Center for Genitourinary Oncology, Peking University First Hospital, 8 Xishiku Street, Xicheng District, Beijing 100034, China.
| |
Collapse
|
37
|
McCoy SS, Reed TJ, Berthier CC, Tsou PS, Liu J, Gudjonsson JE, Khanna D, Kahlenberg JM. Scleroderma keratinocytes promote fibroblast activation independent of transforming growth factor beta. Rheumatology (Oxford) 2017; 56:1970-1981. [PMID: 28968684 DOI: 10.1093/rheumatology/kex280] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Indexed: 01/06/2023] Open
Abstract
Objectives SSc is a devastating disease that results in fibrosis of the skin and other organs. Fibroblasts are a key driver of the fibrotic process through deposition of extracellular matrix. The mechanisms by which fibroblasts are induced to become pro-fibrotic remain unclear. Thus, we examined the ability of SSc keratinocytes to promote fibroblast activation and the source of this effect. Methods Keratinocytes were isolated from skin biopsies of 9 lcSSc, 10 dcSSc and 13 control patients. Conditioned media was saved from the cultures. Normal fresh primary fibroblasts were exposed to healthy control and SSc keratinocyte conditioned media in the presence or absence of neutralizing antibodies for TGF-β. Gene expression was assessed by microarrays and real-time PCR. Immunocytochemistry was performed for α-smooth muscle actin (α-SMA), collagen type 1 (COL1A1) and CCL5 expression. Results SSc keratinocyte conditioned media promoted fibroblast activation, characterized by increased α-SMA and COL1A1 mRNA and protein expression. This effect was independent of TGF-β. Microarray analysis identified upregulation of nuclear factor κB (NF-κB) and downregulation of peroxisome proliferator-activated receptor γ (PPAR-γ) pathways in both SSc subtypes. Scleroderma keratinocytes exhibited increased expression of NF-κB-regulated cytokines and chemokines and lesional skin staining confirmed upregulation of CCL5 in basal keratinocytes. Conclusion Scleroderma keratinocytes promote the activation of fibroblasts in a TGF-β-independent manner and demonstrate an imbalance in NF-κB1 and PPAR-γ expression leading to increased cytokine and CCL5 production. Further study of keratinocyte mediators of fibrosis, including CCL5, may provide novel targets for skin fibrosis therapy.
Collapse
Affiliation(s)
- Sara S McCoy
- Department of Internal Medicine, Division of Rheumatology, University of Wisconsin, Madison, WI
| | - Tamra J Reed
- Department of Internal Medicine, Division of Rheumatology
| | | | - Pei-Suen Tsou
- Department of Internal Medicine, Division of Rheumatology
| | - Jianhua Liu
- Department of Internal Medicine, Division of Rheumatology
| | | | - Dinesh Khanna
- Department of Internal Medicine, Division of Rheumatology
| | | |
Collapse
|
38
|
|
39
|
Mattyasovszky SG, Mausbach S, Ritz U, Langendorf E, Wollstädter J, Baranowski A, Drees P, Rommens PM, Hofmann A. Influence of the anti-inflammatory cytokine interleukin-4 on human joint capsule myofibroblasts. J Orthop Res 2017; 35:1290-1298. [PMID: 27504740 DOI: 10.1002/jor.23386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 08/05/2016] [Indexed: 02/04/2023]
Abstract
Post-traumatic joint contracture was reported to be associated with elevated numbers of contractile myofibroblasts (MFs) in the healing capsule. During the physiological healing process, the number of MFs declines; however, in fibroconnective disorders, MFs persist. The manifold interaction of the cytokines regulating the appearance and persistence of MFs in the pathogenesis of joint contracture remains to be elucidated. The objective of our current study was to analyze the impact of the anti-inflammatory cytokine interleukin (IL)-4 on functional behavior of MFs. Cells were isolated from human joint capsule specimens and challenged with three different concentrations of IL-4 with or without its neutralizing antibody. MF viability, contractile properties, and the gene expression of both alpha-smooth muscle actin (α-SMA) and collagen type I were examined. Immunofluorescence staining revealed the presence of IL-4 receptor (R)-alpha (α) on the membrane of cultured MFs. The cytokine IL-4 promoted MF viability and enhanced MF modulated contraction of collagen gels. Moreover, IL-4 intervened in gene expression by up-regulation of α-SMA and collagen type I mRNA. These effects could be specifically lowered by the neutralizing IL-4 antibody. On the basis of our findings we conclude that the anti-inflammatory cytokine IL-4 specifically regulates viability and the contractile properties of MFs via up-regulating the gene expression of α-SMA and collagen type I. IL-4 may be a helpful target in developing anti-fibrotic therapeutics for post-traumatic joint contracture in human. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1290-1298, 2017.
Collapse
Affiliation(s)
- Stefan G Mattyasovszky
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Stefan Mausbach
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Eva Langendorf
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Jochen Wollstädter
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Andreas Baranowski
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Phillipp Drees
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Alexander Hofmann
- Department of Orthopaedics and Traumatology, University Medical Centre of the Johannes Gutenberg-University of Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
40
|
Valatas V, Filidou E, Drygiannakis I, Kolios G. Stromal and immune cells in gut fibrosis: the myofibroblast and the scarface. Ann Gastroenterol 2017; 30:393-404. [PMID: 28655975 PMCID: PMC5479991 DOI: 10.20524/aog.2017.0146] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/19/2017] [Indexed: 02/07/2023] Open
Abstract
Post-inflammatory scarring is the end-result of excessive extracellular matrix (ECM) accumulation and tissue architectural destruction. It represents a failure to effectively remodel ECM and achieve proper reinstitution and healing during chronic relapsing inflammatory processes. Scarring may affect the functionality of any organ, and in the case of inflammatory bowel disease (IBD)-associated fibrosis leads to stricture formation and often surgery to remove the affected bowel. The activated myofibroblast is the final effector cell that overproduces ECM under the influence of various mediators generated by an intense interplay of classic and non-classic immune cells. This review focuses on how proinflammatory mediators from various sources produced in different stages of intestinal inflammation can form profibrotic pathways that eventually lead to tissue scarring through sustained activation of myofibroblasts.
Collapse
Affiliation(s)
- Vassilis Valatas
- Laboratory of Gastroenterology, Faculty of Medicine, University of Crete, Heraklion (Vassilis Valatas, Ioannis Drygiannakis)
| | - Eirini Filidou
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Dragana, Alexandroupolis (Eirini Filidou, George Kolios), Greece
| | - Ioannis Drygiannakis
- Laboratory of Gastroenterology, Faculty of Medicine, University of Crete, Heraklion (Vassilis Valatas, Ioannis Drygiannakis)
| | - George Kolios
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Dragana, Alexandroupolis (Eirini Filidou, George Kolios), Greece
| |
Collapse
|
41
|
Wang W, Cheng ZS, Chen YF, Lin YH. Increased circulating IL-9-producing CD8 + T cells are associated with eosinophilia and high FeNO in allergic asthmatics. Exp Ther Med 2016; 12:4055-4060. [PMID: 28105134 DOI: 10.3892/etm.2016.3870] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 10/07/2016] [Indexed: 12/11/2022] Open
Abstract
Allergic asthma is a chronic airway disorder mediated by Th2 cells. It has been shown that IL-9-producing CD8+ cytotoxic T (Tc9) cells promote the subsequent onset of allergic airway inflammation in mice mediated by abnormal Th2 immunity. Whether Tc9 cells are associated with the immunopathogenesis of asthmatic patients remains unknown. In the present study, peripheral blood mononuclear cells (PBMCs) were separated by Ficoll-Hypaque gradient centrifugation from all subjects. The frequency of Tc9 cells was measured by flow cytometry. Serum IL-9 levels were assessed by enzyme-linked immunosorbent assay (ELISA). mRNA expression levels of IL-9, STAT6, and IRF4 in PBMCs from healthy controls and asthmatic patients were detected by reverse transcription-quantitative polymerase chain reaction. The results showed that the numbers of Tc9 cells in allergic asthmatics were significantly increased, compared with healthy controls (P<0.0001). Notably, IL-9 protein and mRNA levels were increased in allergic asthmatics and STAT6 and IRF4 mRNA levels were elevated, as compared with healthy controls. In addition, circulating numbers of Tc9 cells were positively correlated with blood eosinophil counts and fractioned exhaled nitric oxide (FeNO) levels in asthmatic patients. Moreover, the number of Tc9 cells and serum IL-9 levels in asthmatic patients were significantly decreased after treatment with glucocorticoids (P<0.05). These findings suggest that increased circulating Tc9 cells are associated with eosinophilia and high FeNO of allergic asthma, and that abnormal Tc9 immunity may contribute to the pathogenesis of allergic asthmatics.
Collapse
Affiliation(s)
- Wei Wang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zhen-Shun Cheng
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yi-Fei Chen
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yu-Hui Lin
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
42
|
Aoudjehane L, Boelle PY, Bisch G, Delelo R, Paye F, Scatton O, Housset C, Becquart J, Calmus Y, Conti F. Development of an in vitro model to test antifibrotic drugs on primary human liver myofibroblasts. J Transl Med 2016; 96:672-9. [PMID: 26950484 DOI: 10.1038/labinvest.2016.36] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/28/2015] [Accepted: 01/11/2016] [Indexed: 02/08/2023] Open
Abstract
We have developed a culture model to assess antifibrotic drugs using normal human liver myofibroblasts (HLMFs) obtained from 31 subjects. Activation was evaluated in terms of α-smooth muscle actin (α-SMA) and collagen 1 (Coll1) expression using RT-PCR, and proliferation as the uptake of 5-ethynil-2'-deoxyuridine. Under analysis of variance, between-subject differences accounted for 70% of all variability and inter-experiment differences for 30%. The sensitivity of the model was determined by quantifying the effects in terms of relative expression, which were 0.74±0.03 for cyclosporine A (CsA) and 2.4±0.10 for transforming growth factor-beta (TGF-β) (P<0.0001 vs no treatment) for α-SMA expression. Inter-subject variations in α-SMA and Coll1 expression enabled the classification of subjects as potentially low or high fibrosers. Finally, we observed that pirfenidone (which has beneficial effects in vivo) significantly reduced the expressions of α-SMA and Coll1, whereas the angiotensin-converting enzyme inhibitor losartan (which has no effect in vivo) had no significant effect. Our model may thus detect the antifibrotic properties of drugs. Antifibrotic drugs with promising clinical relevance could possibly be selected using a bank of HLMFs from high fibrosers.
Collapse
Affiliation(s)
- Lynda Aoudjehane
- Human HepCell, Faculté de Médecine Pierre et Marie Curie, Site Saint-Antoine, Paris, France.,Sorbonne Universités, UPMC University Paris 06, Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France
| | | | - Grégoire Bisch
- Human HepCell, Faculté de Médecine Pierre et Marie Curie, Site Saint-Antoine, Paris, France
| | - Rolland Delelo
- Sorbonne Universités, UPMC University Paris 06, Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France
| | - François Paye
- Sorbonne Universités, UPMC University Paris 06, Paris, France.,AP-HP, Hôpital Saint Antoine, Service de Chirurgie Digestive, Paris, France
| | - Olivier Scatton
- Sorbonne Universités, UPMC University Paris 06, Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Servie de Chirurgie Digestive et Transplantation Hépatique, Paris, France
| | - Chantal Housset
- Sorbonne Universités, UPMC University Paris 06, Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France
| | - Jérôme Becquart
- Human HepCell, Faculté de Médecine Pierre et Marie Curie, Site Saint-Antoine, Paris, France
| | - Yvon Calmus
- Sorbonne Universités, UPMC University Paris 06, Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Unité de Transplantation Hépatique, Paris, France
| | - Filomena Conti
- Sorbonne Universités, UPMC University Paris 06, Paris, France.,INSERM, UMR_S 938, CDR Saint-Antoine, Paris, France.,AP-HP, Hôpital Pitié-Salpêtrière, Unité de Transplantation Hépatique, Paris, France
| |
Collapse
|
43
|
Jacob N, Targan SR, Shih DQ. Cytokine and anti-cytokine therapies in prevention or treatment of fibrosis in IBD. United European Gastroenterol J 2016; 4:531-40. [PMID: 27536363 DOI: 10.1177/2050640616649356] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/19/2016] [Indexed: 12/18/2022] Open
Abstract
The frequency of fibrosing Crohn's disease (CD) is significant, with approximately 40% of CD patients with ileal disease developing clinically apparent strictures throughout their lifetime. Although strictures may be subdivided into fibrotic, inflammatory, or mixed forms, despite immunosuppressive therapy in CD patients in the form of steroids or immunomodulators, the frequency of fibrostenosing complications has still remained significant. A vast number of genetic and epigenetic variables are thought to contribute to fibrostenosing disease, including those that affect cytokine biology, and therefore highlight the complexity of disease, but also shed light on targetable pathways. Exclusively targeting fibrosis may be difficult, however, because of the relatively slow evolution of fibrosis in CD, and the potential adverse effects of inhibiting pathways involved in tissue repair and mucosal healing. Acknowledging these caveats, cytokine-targeted therapy has become the mainstay of treatment for many inflammatory conditions and is being evaluated for fibrotic disorders. The question of whether anti-cytokine therapy will prove useful for intestinal fibrosis is, therefore, acutely relevant. This review will highlight some of the current therapeutics targeting cytokines involved in fibrosis.
Collapse
Affiliation(s)
- Noam Jacob
- F. Widjaja Foundation, Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Digestive Diseases, Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Stephan R Targan
- F. Widjaja Foundation, Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David Q Shih
- F. Widjaja Foundation, Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
44
|
RAGE and TGF-β1 Cross-Talk Regulate Extracellular Matrix Turnover and Cytokine Synthesis in AGEs Exposed Fibroblast Cells. PLoS One 2016; 11:e0152376. [PMID: 27015414 PMCID: PMC4807770 DOI: 10.1371/journal.pone.0152376] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/14/2016] [Indexed: 01/11/2023] Open
Abstract
AGEs accumulation in the skin affects extracellular matrix (ECM) turnover and triggers diabetes associated skin conditions and accelerated skin aging. The receptor of AGEs (RAGE) has an essential contribution to cellular dysfunction driven by chronic inflammatory responses while TGF-β1 is critical in both dermal homeostasis and inflammation. We investigated the contribution of RAGE and TGF-β1 to the modulation of inflammatory response and ECM turnover in AGEs milieu, using a normal fibroblast cell line. RAGE, TGF-β1, collagen I and III gene and protein expression were upregulated after exposure to AGEs-BSA, and MMP-2 was activated. AGEs-RAGE was pivotal in NF-κB dependent collagen I expression and joined with TGF-β1 to stimulate collagen III expression, probably via ERK1/2 signaling. AGEs-RAGE axis induced upregulation of TGF-β1, TNF-α and IL-8 cytokines. TNF-α and IL-8 were subjected to TGF-β1 negative regulation. RAGE’s proinflammatory signaling also antagonized AGEs-TGF-β1 induced fibroblast contraction, suggesting the existence of an inhibitory cross-talk mechanism between TGF-β1 and RAGE signaling. RAGE and TGF-β1 stimulated anti-inflammatory cytokines IL-2 and IL-4 expression. GM-CSF and IL-6 expression appeared to be dependent only on TGF-β1 signaling. Our data also indicated that IFN-γ upregulated in AGEs-BSA milieu in a RAGE and TGF-β1 independent mechanism. Our findings raise the possibility that RAGE and TGF-β1 are both involved in fibrosis development in a complex cross-talk mechanism, while also acting on their own individual targets. This study contributes to the understanding of impaired wound healing associated with diabetes complications.
Collapse
|
45
|
Aoudjehane L, Bisch G, Scatton O, Granier C, Gaston J, Housset C, Roingeard P, Cosset FL, Perdigao F, Balladur P, Wakita T, Calmus Y, Conti F. Infection of Human Liver Myofibroblasts by Hepatitis C Virus: A Direct Mechanism of Liver Fibrosis in Hepatitis C. PLoS One 2015. [PMID: 26214688 PMCID: PMC4516308 DOI: 10.1371/journal.pone.0134141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background Chronic hepatitis C is a major cause of liver fibrosis and cirrhosis. It is generally accepted that inflammation that occurs in response to hepatocyte infection by the hepatitis C virus (HCV) is the main mechanism that triggers myofibroblast differentiation and stimulation in chronic hepatitis C. The aim of this study was to determine if HCV might infect human liver myofibroblasts (HLMF) and directly stimulate their fibrogenic activities. Methods We evaluated the expression of the viral entry receptors, levels of HCV-RNA and HCV-protein and the expression of fibrosis markers in HLMF by using quantitative PCR, western blot and immunofluorescence analyses. Pseudoparticles (HCVpp) and cell culture–derived HCV (HCVcc) were used to study the ability of HLMF to support viral entry, replication and fibrosis induction. Results We showed that HLMF expressed all known molecules of the HCV receptor complex, i.e. CD81, LDL-R, scavenger receptor-BI, claudin-1 and occludin. These cells were also permissive to HCVpp entry. Inoculation with HCVcc caused short-term infection of these cells, as shown by their content in positive- and negative-strand HCV RNA, in core and NS3 viral proteins, and by their release of core protein levels in the culture supernatants. HCV infection stimulated myofibroblastic differentiation, proliferation and collagen production in these cells. In addition, evidence of in vivo infection was provided by the detection of positive- and negative-strand HCV RNA in preparations of HLMF obtained from HCV-infected patients. Conclusion These findings indicate that HCV infection of HLMF can occur and trigger extracellular matrix overproduction, thereby contributing to the development of HCV-related liver fibrosis.
Collapse
Affiliation(s)
- Lynda Aoudjehane
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75005, Paris, France
- INSERM, UMR_S 938, CDR Saint-Antoine, F-75012, Paris, France
- Human HepCell, Hôpital Saint-Antoine, F-75012 Paris, France
- * E-mail:
| | - Grégoire Bisch
- Human HepCell, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Olivier Scatton
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75005, Paris, France
- INSERM, UMR_S 938, CDR Saint-Antoine, F-75012, Paris, France
- AP-HP, Hôpital Pitié-Salpêtrière, Unité de Transplantation Hépatique, F-75013, Paris, France
| | - Christelle Granier
- CIRI–International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France
- Inserm, U1111, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France
- CNRS, UMR5308, Lyon, France
- LabEx Ecofect, Université de Lyon, Lyon, France
| | - Jesintha Gaston
- Institut Cochin, Université Paris Descartes, CNRS (UMR 8104), INSERM U1016, Paris, France
| | - Chantal Housset
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75005, Paris, France
- INSERM, UMR_S 938, CDR Saint-Antoine, F-75012, Paris, France
| | | | - François-Loïc Cosset
- CIRI–International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France
- Inserm, U1111, Lyon, France
- Ecole Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, Centre International de Recherche en Infectiologie, Lyon, France
- CNRS, UMR5308, Lyon, France
- LabEx Ecofect, Université de Lyon, Lyon, France
| | - Fabiano Perdigao
- AP-HP, Hôpital Pitié-Salpêtrière, Unité de Transplantation Hépatique, F-75013, Paris, France
| | - Pierre Balladur
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75005, Paris, France
- INSERM, UMR_S 938, CDR Saint-Antoine, F-75012, Paris, France
- AP-HP, Hôpital Saint Antoine, Département de la chirurgie digestive, F-75012, Paris, France
| | - Takaji Wakita
- National Institute of Infectious Diseases, Department of Virology II, Tokyo, Japan
| | - Yvon Calmus
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75005, Paris, France
- INSERM, UMR_S 938, CDR Saint-Antoine, F-75012, Paris, France
- AP-HP, Hôpital Pitié-Salpêtrière, Unité de Transplantation Hépatique, F-75013, Paris, France
| | - Filomena Conti
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S 938, CDR Saint-Antoine, F-75005, Paris, France
- INSERM, UMR_S 938, CDR Saint-Antoine, F-75012, Paris, France
- AP-HP, Hôpital Pitié-Salpêtrière, Unité de Transplantation Hépatique, F-75013, Paris, France
| |
Collapse
|
46
|
Peng H, Sarwar Z, Yang XP, Peterson EL, Xu J, Janic B, Rhaleb N, Carretero OA, Rhaleb NE. Profibrotic Role for Interleukin-4 in Cardiac Remodeling and Dysfunction. Hypertension 2015. [PMID: 26195478 DOI: 10.1161/hypertensionaha.115.05627] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Elevated interleukin-4 (IL-4) levels are associated with cardiac fibrosis in hypertension and heart failure in both patients and experimental animals. We hypothesized that chronically elevated IL-4 induces cardiac fibrosis, resulting in a predisposition of the heart to angiotensin II-induced damage. Wild-type Balb/c (WT, high circulating IL-4) and IL-4-deficient Balb/c mice (IL-4(-/-)) were used. WT mice exhibited cardiac fibrosis (evidenced by an increase in expression of procollagen genes/interstitial collagen fraction), enlarged left ventricle chamber, and declined cardiac function associated with a greater number of mast cells and macrophages in the heart compared with IL-4(-/-). In contrast, IL-4(-/-) mice had normal cardiac architecture/function while showing a 57.9% reduction in heart interstitial collagen compared with WT, despite elevated proinflammatory cytokines in heart tissue. In response to angiotensin II administration, IL-4(-/-) had reduced interstitial myocardial fibrosis and were protected from developing dilated cardiomyopathy, which was seen in WT mice. This was associated with increased macrophage infiltration into the hearts of WT mice, despite a similar degree of hypertension and increased cardiac transforming growth factor-β1 in both groups. In vitro data demonstrated that IL-4 upregulates procollagen genes and stimulates collagen production in mouse cardiac fibroblasts. This process is mediated by signal transducer and activator of transcription 6 signaling pathway via IL-4 receptor alpha. This study not only establishes a causal relationship between IL-4 and cardiac fibrosis/dysfunction, but also reveals a critical role for IL-4 in angiotensin II-induced cardiac damage. IL-4 could serve as an additional target for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Hongmei Peng
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI.
| | - Zeyd Sarwar
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI
| | - Xiao-Ping Yang
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI
| | - Edward L Peterson
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI
| | - Jiang Xu
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI
| | - Branislava Janic
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI
| | - Nadia Rhaleb
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI
| | - Oscar A Carretero
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI
| | - Nour-Eddine Rhaleb
- From the Hypertension and Vascular Research Division, Department of Internal Medicine (H.P., Z.S., X.-P.Y., J.X., B.J., N.R., O.A.C., N.-E.R.) and Department of Public Health Sciences (E.L.P.), Henry Ford Hospital, Detroit, MI.
| |
Collapse
|
47
|
Huang XL, Wang YJ, Yan JW, Wan YN, Chen B, Li BZ, Yang GJ, Wang J. Role of anti-inflammatory cytokines IL-4 and IL-13 in systemic sclerosis. Inflamm Res 2015; 64:151-9. [PMID: 25725697 DOI: 10.1007/s00011-015-0806-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 02/16/2015] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The aim of this paper is to review the anti-inflammatory cytokines IL-4 and IL-13 and their receptor signals; we discuss new insight into their possible roles in systemic sclerosis (SSc) and their overlapping function in SSc. INTRODUCTION SSc is a connective tissue disease characterized by fibrosis. The exact etiology of SSc is unknown, and no therapy has been proved effective in modifying its course. Recently the roles of IL-4 and IL-13 in the development of SSc have been extensively considered. The possible roles of IL-4 and IL-13, especially their overlapping function, in SSc are not well documented. METHODS A literature survey was performed using a PubMed database search to gather complete information regarding IL-4 and IL-13 and their role in inflammation. RESULTS AND CONCLUSIONS The participation of complex pathways of IL-4 and IL-13 in the process of inflammation and fibrosis action in SSc is still not very clear, and some pathogenesis of regulation found in vitro needs to be further proved. There is still more work which could be done to achieve useful developments with therapeutic benefit in SSc.
Collapse
Affiliation(s)
- Xiao-Lei Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, No. 81, Meishan Road, 230032, Hefei, Anhui, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Barber T, Esteban-Pretel G, Marín MP, Timoneda J. Vitamin a deficiency and alterations in the extracellular matrix. Nutrients 2014; 6:4984-5017. [PMID: 25389900 PMCID: PMC4245576 DOI: 10.3390/nu6114984] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 09/26/2014] [Accepted: 10/20/2014] [Indexed: 12/13/2022] Open
Abstract
Vitamin A or retinol which is the natural precursor of several biologically active metabolites can be considered the most multifunctional vitamin in mammals. Its deficiency is currently, along with protein malnutrition, the most serious and common nutritional disorder worldwide. It is necessary for normal embryonic development and postnatal tissue homeostasis, and exerts important effects on cell proliferation, differentiation and apoptosis. These actions are produced mainly by regulating the expression of a variety of proteins through transcriptional and non-transcriptional mechanisms. Extracellular matrix proteins are among those whose synthesis is known to be modulated by vitamin A. Retinoic acid, the main biologically active form of vitamin A, influences the expression of collagens, laminins, entactin, fibronectin, elastin and proteoglycans, which are the major components of the extracellular matrix. Consequently, the structure and macromolecular composition of this extracellular compartment is profoundly altered as a result of vitamin A deficiency. As cell behavior, differentiation and apoptosis, and tissue mechanics are influenced by the extracellular matrix, its modifications potentially compromise organ function and may lead to disease. This review focuses on the effects of lack of vitamin A in the extracellular matrix of several organs and discusses possible molecular mechanisms and pathologic implications.
Collapse
Affiliation(s)
- Teresa Barber
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Avda V. Andrés Estellés s/n, 46100-Burjassot, Spain.
| | - Guillermo Esteban-Pretel
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Avda V. Andrés Estellés s/n, 46100-Burjassot, Spain.
| | - María Pilar Marín
- Unidad de Microscopía IIS La Fe Valencia, Avda Campanar, 21, 46009-Valencia, Spain.
| | - Joaquín Timoneda
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Avda V. Andrés Estellés s/n, 46100-Burjassot, Spain.
| |
Collapse
|
49
|
Sun C, Zhu M, Yang Z, Pan X, Zhang Y, Wang Q, Xiao W. LL-37 secreted by epithelium promotes fibroblast collagen production: a potential mechanism of small airway remodeling in chronic obstructive pulmonary disease. J Transl Med 2014; 94:991-1002. [PMID: 24955895 DOI: 10.1038/labinvest.2014.86] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 05/27/2014] [Accepted: 05/30/2014] [Indexed: 12/30/2022] Open
Abstract
Emerging evidence suggests that the process of small airway remodeling is mediated by profibrotic growth factors produced by epithelium, which are capable of activating the underlying mesenchymal cells with excessive collagen production. It has been demonstrated that human cathelicidin antimicrobial protein LL-37 is highly expressed in small airway epithelium from COPD patients. However, it is unknown whether the increased levels of LL-37 in epithelium are involved in the pathogenesis of small airway remodeling in COPD. In this study, we examined the expression of LL-37 in small airways from smokers with COPD and controls (non-smokers and smokers without COPD) by immunohistochemistry, and then the association between LL-37 expression in epithelium and the structural changes of small airway remodeling was analyzed. In vitro, the effect of CSE-induced epithelial secretion of LL-37 on collagen production in human lung fibroblasts (HFL-1 cell line) was studied in a co-culture system. Finally, the signaling pathways involved in the effect of LL-37 on fibroblast collagen production were evaluated. The results showed that LL-37 immunoreactivity in airway epithelium was significantly elevated in smokers with COPD compared with controls. In addition, the magnitude of LL-37 expression in epithelium was positively correlated with airway wall thickness and collagen deposition. In vitro, CSE-induced epithelial secretion of LL-37 promoted fibroblast collagen production. Finally, we showed that formyl peptide receptor-like 1 (FPRL1)-dependent extracellular signal-regulated kinase (ERK) signaling pathway was essential for LL-37-induced collagen production in HFL-1 cells. These results suggest that after cigarette smoke exposure, the increased levels of LL-37 in airway epithelium could stimulate collagen production in the underlying lung fibroblasts and may contribute to small airway remodeling in COPD.
Collapse
Affiliation(s)
- Congcong Sun
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Maoxiang Zhu
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhihua Yang
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiujie Pan
- Department of Radiation Toxicology and Oncology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yuke Zhang
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Qin Wang
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Wei Xiao
- Department of Respiratory Medicine, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
50
|
Abstract
Hepatic fibrosis is a response to repair of chronic hepatic injury caused by different diseases. Its pathological features are inflammatory cell aggregation and associated cascade reaction. There are myofibroblast formation (MFB) and excessive deposition of extracellular matrix (ECM) in the liver, and the structure of the liver is destroyed. The recent research finds that Janus kinase/signal transducer and activator of transcription (JAK-STAT) plays an important role in the development of hepatic fibrosis. In general, STAT1 and STAT5 act as anti-fibrotic signaling molecules preventing injury-driven liver fibrosis. Although the antiviral effect of STAT2 is well documented, it is not clear whether or not STAT2 also plays a role in contributing to the interferon-α/β-mediated anti-fibrotic effects in the liver. What's more, the biological functions of STAT3, STAT4 and STAT6 in hepatic fibrosis remains largely unknown. Therefore, this review focuses on the role of STAT family members and cytokines in hepatic fibrosis.
Collapse
|