1
|
Jin Q, Yang H, Jing Z, Hong-hua W, Ben-jing S, Li-ting W, Li-juan Y, Wei X, Xia K, Juan W, Wei Z. IL4/IL4R signaling promotes the osteolysis in metastatic bone of CRC through regulating the proliferation of osteoclast precursors. Mol Med 2021; 27:152. [PMID: 34863091 PMCID: PMC8642926 DOI: 10.1186/s10020-021-00411-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 11/13/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Bone metastasis of colorectal cancer (CRC) often indicates a poor prognosis. Osteolysis can be observed in metastatic sites, implying an aberrant activation of osteoclasts. However, how osteoclastogenesis is regulated in metastatic microenvironment caused by colorectal cancer is still unclear. METHODS In this study, mice bone metastatic model of CRC was established through injection of MC-38 or CT-26 cells. BrdU assays showed primary CD115 ( +) osteoclast precursors (OCPs) proliferated at the first 2 weeks. Transcriptomic profiling was performed to identify differentially expressing genes and pathways in OCPs indirectly co-cultured with CRC cells RESULTS: The expression of IL4Rα was found to be significantly upregulated in OCPs stimulated by tumor conditioned medium (CM). Further investigation indicated that IL-4 signaling regulated proliferation of OPCs through interacting with type I IL4 receptor, and neutrophils were the main source of IL-4 in bone marrow. The proliferation of OCPs can be inhibited in IL4 deficiency mice. In addition, ERK pathway was activated by IL4/IL4R signaling. Ravoxertinib, an ERK antagonists, could significantly prevent bone destruction through inhibiting the proliferation of OCPs. CONCLUSION Our study indicates the essential role of IL4/IL4R signaling for the proliferation of OCPs in early metastasis of CRC predominantly through activating ERK pathway, which remarkedly impacts the number of osteoclasts in later stage and leads to osteolytic lesions. Moreover, Ravoxertinib could be a new therapeutical target for bone metastasis of CRC.
Collapse
Affiliation(s)
- Qian Jin
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
- College of Medicine, Southwest Jiaotong University, North Section 1 No. 111, Second Ring Road, Chengdu, 610000 People’s Republic of China
| | - He Yang
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Zhao Jing
- Biomedical Analysis Center, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Wu Hong-hua
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Song Ben-jing
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Wang Li-ting
- Biomedical Analysis Center, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Ye Li-juan
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Xu Wei
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Kang Xia
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Wu Juan
- Department of Pharmacy, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
| | - Zheng Wei
- Department of Orthopedics, General Hospital of Western Theater Command, Rongdu Avenue No. 270, Chengdu, 610000 People’s Republic of China
- College of Medicine, Southwest Jiaotong University, North Section 1 No. 111, Second Ring Road, Chengdu, 610000 People’s Republic of China
| |
Collapse
|
2
|
Moshkelgosha S, Verhasselt HL, Masetti G, Covelli D, Biscarini F, Horstmann M, Daser A, Westendorf AM, Jesenek C, Philipp S, Diaz-Cano S, Banga JP, Michael D, Plummer S, Marchesi JR, Eckstein A, Ludgate M, Berchner-Pfannschmidt U. Modulating gut microbiota in a mouse model of Graves' orbitopathy and its impact on induced disease. MICROBIOME 2021; 9:45. [PMID: 33593429 PMCID: PMC7888139 DOI: 10.1186/s40168-020-00952-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/06/2020] [Indexed: 05/15/2023]
Abstract
BACKGROUND Graves' disease (GD) is an autoimmune condition in which autoantibodies to the thyrotropin receptor (TSHR) cause hyperthyroidism. About 50% of GD patients also have Graves' orbitopathy (GO), an intractable disease in which expansion of the orbital contents causes diplopia, proptosis and even blindness. Murine models of GD/GO, developed in different centres, demonstrated significant variation in gut microbiota composition which correlated with TSHR-induced disease heterogeneity. To investigate whether correlation indicates causation, we modified the gut microbiota to determine whether it has a role in thyroid autoimmunity. Female BALB/c mice were treated with either vancomycin, probiotic bacteria, human fecal material transfer (hFMT) from patients with severe GO or ddH2O from birth to immunization with TSHR-A subunit or beta-galactosidase (βgal; age ~ 6 weeks). Incidence and severity of GD (TSHR autoantibodies, thyroid histology, thyroxine level) and GO (orbital fat and muscle histology), lymphocyte phenotype, cytokine profile and gut microbiota were analysed at sacrifice (~ 22 weeks). RESULTS In ddH2O-TSHR mice, 84% had pathological autoantibodies, 67% elevated thyroxine, 77% hyperplastic thyroids and 70% orbital pathology. Firmicutes were increased, and Bacteroidetes reduced relative to ddH2O-βgal; CCL5 was increased. The random forest algorithm at the genus level predicted vancomycin treatment with 100% accuracy but 74% and 70% for hFMT and probiotic, respectively. Vancomycin significantly reduced gut microbiota richness and diversity compared with all other groups; the incidence and severity of both GD and GO also decreased; reduced orbital pathology correlated positively with Akkermansia spp. whilst IL-4 levels increased. Mice receiving hFMT initially inherited their GO donors' microbiota, and the severity of induced GD increased, as did the orbital brown adipose tissue volume in TSHR mice. Furthermore, genus Bacteroides, which is reduced in GD patients, was significantly increased by vancomycin but reduced in hFMT-treated mice. Probiotic treatment significantly increased CD25+ Treg cells in orbital draining lymph nodes but exacerbated induced autoimmune hyperthyroidism and GO. CONCLUSIONS These results strongly support a role for the gut microbiota in TSHR-induced disease. Whilst changes to the gut microbiota have a profound effect on quantifiable GD endocrine and immune factors, the impact on GO cellular changes is more nuanced. The findings have translational potential for novel, improved treatments. Video abstract.
Collapse
Affiliation(s)
- Sajad Moshkelgosha
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
- Current address: Latner Thoracic Surgery Laboratories, Toronto General Research Institute, University Health Network and University of Toronto, Toronto, Canada
| | - Hedda Luise Verhasselt
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Cultech Ltd., Baglan, Port Talbot, UK
| | - Giulia Masetti
- Division of Infection & Immunity, School of Medicine, Cardiff University, UHW main building, Heath Park, Cardiff, CF14 4XW, UK
- Department of Bioinformatics, PTP Science Park Srl, Lodi, Italy
- Current address: Computational metagenomics, Department CIBIO, University of Trento, Trento, Italy
| | - Danila Covelli
- Cultech Ltd., Baglan, Port Talbot, UK
- Graves' Orbitopathy Center, Endocrinology, Department of Clinical Sciences and Community Health, Fondazione Ca'Granda IRCCS, University of Milan, Milan, Italy
| | - Filippo Biscarini
- Department of Bioinformatics, PTP Science Park Srl, Lodi, Italy
- Italian National Research Council (CNR), Milano, Italy
| | - Mareike Horstmann
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Anke Daser
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christoph Jesenek
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Svenja Philipp
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | - Salvador Diaz-Cano
- Department of Histopathology, King's College Hospital, King's College, London, UK
| | - J Paul Banga
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany
| | | | | | - Julian R Marchesi
- School of Biosciences, Cardiff University, Cardiff, UK
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Anja Eckstein
- Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
| | - Marian Ludgate
- Division of Infection & Immunity, School of Medicine, Cardiff University, UHW main building, Heath Park, Cardiff, CF14 4XW, UK.
| | - Utta Berchner-Pfannschmidt
- Molecular Ophthalmology, Department of Ophthalmology, University Hospital Essen, University of Duisburg-Essen, 45147, Essen, Germany.
| |
Collapse
|
3
|
Penke LR, Peters-Golden M. Molecular determinants of mesenchymal cell activation in fibroproliferative diseases. Cell Mol Life Sci 2019; 76:4179-4201. [PMID: 31563998 PMCID: PMC6858579 DOI: 10.1007/s00018-019-03212-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/01/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Uncontrolled scarring, or fibrosis, can interfere with the normal function of virtually all tissues of the body, ultimately leading to organ failure and death. Fibrotic diseases represent a major cause of death in industrialized countries. Unfortunately, no curative treatments for these conditions are yet available, highlighting the critical need for a better fundamental understanding of molecular mechanisms that may be therapeutically tractable. The ultimate indispensable effector cells responsible for deposition of extracellular matrix proteins that comprise scars are mesenchymal cells, namely fibroblasts and myofibroblasts. In this review, we focus on the biology of these cells and the molecular mechanisms that regulate their pertinent functions. We discuss key pro-fibrotic mediators, signaling pathways, and transcription factors that dictate their activation and persistence. Because of their possible clinical and therapeutic relevance, we also consider potential brakes on mesenchymal cell activation and cellular processes that may facilitate myofibroblast clearance from fibrotic tissue-topics that have in general been understudied.
Collapse
Affiliation(s)
- Loka R Penke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA.
| |
Collapse
|
4
|
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
|
5
|
Villa O, Brookes SJ, Thiede B, Heijl L, Lyngstadaas SP, Reseland JE. Subfractions of enamel matrix derivative differentially influence cytokine secretion from human oral fibroblasts. J Tissue Eng 2015; 6:2041731415575857. [PMID: 26090085 PMCID: PMC4456328 DOI: 10.1177/2041731415575857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 02/09/2015] [Indexed: 01/09/2023] Open
Abstract
Enamel matrix derivative is used to promote periodontal regeneration during the corrective phase of the treatment of periodontal defects. Our main goal was to analyze the bioactivity of different molecular weight fractions of enamel matrix derivative. Enamel matrix derivative, a complex mixture of proteins, was separated into 13 fractions using size-exclusion chromatography and characterized by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and liquid chromatography-electrospray ionization-tandem mass spectrometry. Human periodontal ligament fibroblasts were treated with either enamel matrix derivative or the different fractions. Proliferation and cytokine secretion to the cell culture medium were measured and compared to untreated cells. The liquid chromatography-electrospray ionization-tandem mass spectrometry analyses revealed that the most abundant peptides were amelogenin and leucine-rich amelogenin peptide related. The fractions containing proteins above 20 kDa induced an increase in vascular endothelial growth factor and interleukin-6 secretion, whereas lower molecular weight fractions enhanced proliferation and secretion of interleukin-8 and monocyte chemoattractant protein-1 and reduced interleukin-4 release. The various molecular components in the enamel matrix derivative formulation might contribute to reported effects on tissue regeneration through their influence on vascularization, the immune response, and chemotaxis.
Collapse
Affiliation(s)
- Oscar Villa
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Steven J Brookes
- Department of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - Bernd Thiede
- The Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
| | | | - Staale P Lyngstadaas
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Janne E Reseland
- Department of Biomaterials, Institute of Clinical Dentistry, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
6
|
Woidacki K, Zenclussen AC, Siebenhaar F. Mast cell-mediated and associated disorders in pregnancy: a risky game with an uncertain outcome? Front Immunol 2014; 5:231. [PMID: 24904581 PMCID: PMC4033021 DOI: 10.3389/fimmu.2014.00231] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/05/2014] [Indexed: 11/13/2022] Open
Abstract
During pregnancy, the maternal organism is under the influence of tremendous endocrine as well as immunological changes as an adaptation to the implanted and developing fetus. In most cases, the maternal adaptations to pregnancy ensure both, the protection against harmful pathogens and the tolerance toward the growing semi-allogeneic fetus. However, under certain circumstances the unique hormonal milieu during pregnancy is causative of a shift into an unfavorable direction. Of particular importance are cellular disorders previous to pregnancy that involve cell types known for their susceptibility to hormones. One interesting cell type is the mast cell (MC), one of the key figures in allergic disorders. While physiological numbers of MCs were shown to positively influence pregnancy outcome, at least in mouse models, uncontrolled augmentations in quantity, and/or activation can lead to pregnancy complications. Women that have the desire of getting pregnant and been diagnosed with MC mediated disorders such as urticaria and mastocytosis or chronic inflammatory diseases in which MCs are involved, including atopic dermatitis, asthma, or psoriasis, may benefit from specialized medical assistance to ensure a positive pregnancy outcome. In the present review, we address the course of pregnancy in women affected by MC mediated or associated disorders.
Collapse
Affiliation(s)
- Katja Woidacki
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University , Magdeburg , Germany
| | - Ana Claudia Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University , Magdeburg , Germany
| | - Frank Siebenhaar
- Department of Dermatology and Allergy, Allergie-Centrum-Charité, Charité-Universitätsmedizin Berlin , Berlin , Germany
| |
Collapse
|
7
|
Todd NW, Luzina IG, Atamas SP. Molecular and cellular mechanisms of pulmonary fibrosis. FIBROGENESIS & TISSUE REPAIR 2012; 5:11. [PMID: 22824096 PMCID: PMC3443459 DOI: 10.1186/1755-1536-5-11] [Citation(s) in RCA: 309] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 06/28/2012] [Indexed: 12/22/2022]
Abstract
Pulmonary fibrosis is a chronic lung disease characterized by excessive accumulation of extracellular matrix (ECM) and remodeling of the lung architecture. Idiopathic pulmonary fibrosis is considered the most common and severe form of the disease, with a median survival of approximately three years and no proven effective therapy. Despite the fact that effective treatments are absent and the precise mechanisms that drive fibrosis in most patients remain incompletely understood, an extensive body of scientific literature regarding pulmonary fibrosis has accumulated over the past 35 years. In this review, we discuss three broad areas which have been explored that may be responsible for the combination of altered lung fibroblasts, loss of alveolar epithelial cells, and excessive accumulation of ECM: inflammation and immune mechanisms, oxidative stress and oxidative signaling, and procoagulant mechanisms. We discuss each of these processes separately to facilitate clarity, but certainly significant interplay will occur amongst these pathways in patients with this disease.
Collapse
Affiliation(s)
- Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | |
Collapse
|
8
|
Abstract
The pathophysiology of post-PCI restenosis involves neointimal formation that consists of three phases: thrombosis (within 24 h), recruitment (3-8 days), and proliferation, which starts on day 8 of PCI. Various factors suggested to be predictors/risks for restenosis include C-reactive protein (CRP), inflammatory mediators (cytokines and adhesion molecules), oxygen radicals, advanced glycation end products (AGEs) and their receptors (RAGE), and soluble RAGE (sRAGE). The earlier noted factors produce thrombogenesis, vascular smooth muscle cell proliferation, and extracellular matrix formation. Statins have pleiotropic effects. Besides lowering serum cholesterol, they have various other biological effects including antiinflammatory, antithrombotic, CRP-lowering, antioxidant, antimitotic, and inhibition of smooth muscle cell proliferation. They inhibit matrix metalloproteinase and cyclooxygenase-2, lower AGEs, decrease expression of RAGE and increase levels of serum sRAGE. They also increase the synthesis of nitric oxide (NO) by increasing endothelial NO synthase expression and activity. Preprocedural statin therapy is known to reduce peri- and post-PCI myonecrosis and reduce the need for repeat revascularization. There is evidence that statin-eluting stents inhibit in-stent restenosis in animal models. It is concluded that because of the above attributes of statins, they are suitable candidates for reduction of post-PCI restenosis and post-PCI myonecrosis. The future directions for the use of statins in reduction of post-PCI restenosis and myonecrosis have been discussed.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
9
|
Shim JY, Park SW, Kim DS, Shim JW, Jung HL, Park MS. The effect of interleukin-4 and amphiregulin on the proliferation of human airway smooth muscle cells and cytokine release. J Korean Med Sci 2008; 23:857-63. [PMID: 18955794 PMCID: PMC2580012 DOI: 10.3346/jkms.2008.23.5.857] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Airway smooth muscle (ASM) hyperplasia and angiogenesis are important features associated with airway remodeling. We investigated the effect of IL-4 and amphiregulin, an epidermal growth factor family member, on the proliferation of human ASM cells and on the release of vascular endothelial growth factor (VEGF) and monocyte chemotactic protein (MCP)-1 from human ASM cells. Human ASM cells were growth-arrested for 48 hr and incubated with platelet-derived growth factor (PDGF)- BB, interleukin (IL)-4, amphiregulin, and VEGF to evaluate cell proliferation. The cells were treated with PDGF, IL-4 and amphiregulin to evaluate the release of VEGF, MCP-1. IL-4 suppressed unstimulated and PDGF-stimulated ASM cell proliferation. Amphiregulin stimulated ASM cell proliferation in a dose-dependent manner. VEGF did not have any influence on ASM cell proliferation. IL-4 stimulated VEGF secretion by the ASM cells in a dose-dependent manner and showed added stimulatory effects when co-incubated with PDGF. Amphiregulin did not promote VEGF secretion. IL-4 and amphiregulin showed no stimulatory effects on MCP-1 secretion. The results of this study showed that IL-4 had bifunctional effects on airway remodeling, one was the suppression of the proliferation of the ASM cells and the other was the promotion of VEGF release by the ASM cells, and amphiregulin can promote human ASM cell proliferation.
Collapse
Affiliation(s)
- Jung Yeon Shim
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | | | | | | | | | | |
Collapse
|
10
|
Delbridge MS, Shrestha BM, Raftery AT, El Nahas AM, Haylor J. FTY720 reduces extracellular matrix expansion associated with ischemia-reperfusion induced injury. Transplant Proc 2007; 39:2992-2996. [PMID: 18089307 DOI: 10.1016/j.transproceed.2007.04.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2006] [Revised: 02/17/2007] [Accepted: 04/30/2007] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ischemia-reperfusion (IR) is one of the strongest nonimmune factors associated with the development of chronic allograft nephropathy (CAN). This effect is often exacerbated by immunosuppressive medications, most notably cyclosporine. Although traditionally the macrophage was thought to stimulate fibroblast activity in CAN, recent evidence supports a role for lymphocytes. FTY720 is a new immunosuppressant that promotes lymphocyte sequestration into lymph nodes and Peyer's patches. This study investigated the effect of FTY720 on renal fibrosis in the rat following an IR insult (IRI). METHODS A rat model of IRI was used in which male Sprague-Dawley rats (under isoflurane anaesthesia) underwent bilateral flank incision with removal of the right kidney and clamping of the left renal hilum for 45 minutes. Five groups of animals were studied (n=4): nephrectomy only, IRI only, IRI+FTY720 (1 mg/kg/d), IRI+cyclosporine (15 mg/kg/d), and IRI+FTY 720 (1 mg/kg/d) and cyclosporine (15 mg/kg/d). Animals were humanely killed at 30 days. RESULTS Serum creatinine (SCr) level was significantly reduced in the FTY720-treated animals. IRI alone produced a significant increase in SCr level compared with neprectomized animals (138 micromol/L vs 55 micromol/L; P<.05). This effect was potentiated by treatment with cyclosporine (173 micromol/L vs 55 micromol/L; P<.05). Treatment with FTY720 significantly reduced SCr level in rats following IRI alone (81 micromol/L vs 138 micromol/L; P<.01) and in rats following IRI + cyclosporine (98 micromol/L vs 173 micromol/L; P<.014). Parallel changes were seen in the levels of proteinuria. Fibrosis was assessed using Masson's trichrome (MT) staining. IRI alone produced a significant increase in MT staining compared with nephrectomized animals (0.92 vs 0.03; P<.05). This effect was potentiated by treatment with cyclosporine (1.12 vs 0.92; P=.022). Treatment with FTY720 reduced the level of MT staining in rats following IRI alone (0.34 vs 0.92; P<.05) and in rats following IRI+cyclosporine (70.34 vs 1.12; P<.05). Levels of TGF-beta1 were considerably reduced in FTY720-treated animals (compared with cyclosporine+IRI and IRI only), either alone (196+/-31 pg/mL vs 1105+/-59 pg/mL and 611+/-38; P<.05) or in conjunction with cyclosporine (423+/-26 pg/mL vs 1105+/-59 pg/mL and 611+/-38; P<.05). CONCLUSION Our study shows that treatment with FTY720 can reduce renal fibrosis as a result of IRI, both alone and in conjunction with cyclosporine. This provides promising evidence for using FTY720 in a calcineurin-free or reduced-dose immunosuppression protocol in an effort to reduce the incidence of CAN.
Collapse
Affiliation(s)
- M S Delbridge
- Sheffield Kidney Institute, Northern General Hospital, Herries Road, Sheffield, South Yorkshire, United Kingdom.
| | | | | | | | | |
Collapse
|
11
|
Nabeshima Y, Hiragun T, Morita E, Mihara S, Kameyoshi Y, Hide M. IL-4 modulates the histamine content of mast cells in a mast cell/fibroblast co-culture through a Stat6 signaling pathway in fibroblasts. FEBS Lett 2005; 579:6653-8. [PMID: 16298365 DOI: 10.1016/j.febslet.2005.09.104] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2005] [Revised: 08/16/2005] [Accepted: 09/01/2005] [Indexed: 10/25/2022]
Abstract
IL-4 plays a crucial role in the pathogenesis of allergic diseases, such as the induction of IgE synthesis and the development of mast cells. To further understand the effect of IL-4 on mast cells in skin, we utilized a mast cell/fibroblast co-culture system as an in vitro model of dermal mast cells. IL-4 induced mast cell growth in the culture with fibroblasts. Immunoblot analysis revealed that IL-4 activated Stat6 in both mast cells and fibroblasts. The over-expression of dominant-negative Stat6 in fibroblasts in the presence of IL-4 decreased the histamine content per mast cell, but not the number of mast cells. In contrast, the over-expression of constitutively-active Stat6 in fibroblasts increased the histamine content per mast cell, indicating that the activation of Stat6 in fibroblasts supports the maturation of mast cells co-cultured with fibroblasts.
Collapse
Affiliation(s)
- Yukiko Nabeshima
- Department of Dermatology, Programs for Biomedical Research, Division of Molecular Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | | | | | | | | | | |
Collapse
|
12
|
Hartl D, Griese M, Nicolai T, Zissel G, Prell C, Reinhardt D, Schendel DJ, Krauss-Etschmann S. A role for MCP-1/CCR2 in interstitial lung disease in children. Respir Res 2005; 6:93. [PMID: 16095529 PMCID: PMC1199626 DOI: 10.1186/1465-9921-6-93] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Accepted: 08/11/2005] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Interstitial lung diseases (ILD) are chronic inflammatory disorders leading to pulmonary fibrosis. Monocyte chemotactic protein 1 (MCP-1) promotes collagen synthesis and deletion of the MCP-1 receptor CCR2 protects from pulmonary fibrosis in ILD mouse models. We hypothesized that pulmonary MCP-1 and CCR2+ T cells accumulate in pediatric ILD and are related to disease severity. METHODS Bronchoalveolar lavage fluid was obtained from 25 children with ILD and 10 healthy children. Levels of pulmonary MCP-1 and Th1/Th2-associated cytokines were quantified at the protein and the mRNA levels. Pulmonary CCR2+, CCR4+, CCR3+, CCR5+ and CXCR3+ T cells were quantified by flow-cytometry. RESULTS CCR2+ T cells and MCP-1 levels were significantly elevated in children with ILD and correlated with forced vital capacity, total lung capacity and ILD disease severity scores. Children with lung fibrosis had significantly higher MCP-1 levels and CCR2+ T cells in bronchoalveolar lavage fluid compared to non-fibrotic children. CONCLUSION The results indicate that pulmonary CCR2+ T cells and MCP-1 contribute to the pathogenesis of pediatric ILD and might provide a novel target for therapeutic strategies.
Collapse
Affiliation(s)
- Dominik Hartl
- Childrens' Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Matthias Griese
- Childrens' Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Thomas Nicolai
- Childrens' Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Gernot Zissel
- Department of Pneumology, Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| | - Christine Prell
- Childrens' Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Dietrich Reinhardt
- Childrens' Hospital of the Ludwig-Maximilians-University, Munich, Germany
| | - Dolores J Schendel
- Institute of Molecular Immunology and Immune Monitoring Platform, GSF National Research Center for Environment and Health, Munich, Germany
| | | |
Collapse
|
13
|
Abstract
Pulmonary fibrosis occurs in up to 70% of scleroderma patients and progresses to cause severe restrictive lung disease in about 15% of patients. The mechanisms that cause pulmonary fibrosis in scleroderma remain incompletely understood. Increased amounts of mRNA or protein for multiple profibrotic cytokines and chemokines have been identified in lung tissue or broncholveolar lavage samples from scleroderma patients, when compared to healthy controls. These cytokines include transforming growth factor (TGF)-beta, connective tissue growth factor (CTGF), platelet-derived growth factor (PDGF), oncostatin M (OSM), monocyte chemotactic factor-1 and pulmonary and activation-regulated chemokine (PARC). Potential cellular sources of these profibrotic cytokines and chemokines in scleroderma lung disease include alternatively activated macrophages, activated CD8+ T cells, eosinophils, mast cells, epithelial cells and fibroblasts themselves. This review summarizes the literature on involvement of cytokines and chemokines in the development of pulmonary fibrosis in scleroderma.
Collapse
Affiliation(s)
- Sergei P Atamas
- Baltimore VA Medical Center, University of Maryland School of Medicine, Research Service (151), Room 3C-126, 10 North Greene Street, Baltimore, MD 21201, USA.
| | | |
Collapse
|
14
|
Endo M, Oyadomari S, Terasaki Y, Takeya M, Suga M, Mori M, Gotoh T. Induction of arginase I and II in bleomycin-induced fibrosis of mouse lung. Am J Physiol Lung Cell Mol Physiol 2003; 285:L313-21. [PMID: 12679322 DOI: 10.1152/ajplung.00434.2002] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Arginase, which hydrolyzes arginine to urea and ornithine, is a precursor for the synthesis of polyamines and proline, which is abundant in collagen. The supply of proline can be a crucial factor in the process of lung fibrosis. We investigated the induction of arginine metabolic enzymes in bleomycin-induced mouse lung fibrosis. Histological studies and quantification of lung hydroxyproline showed that lung fibrosis develops in up to 14 days after bleomycin treatment. Under these conditions, collagen I mRNA was induced gradually in up to 15 days, and the content of hydroxyproline reached a maximum at 10 days. Arginase I mRNA was undetectable before bleomycin treatment but was induced 5-10 days after this treatment. Arginase I protein was induced at 7 days and remained little changed for up to 10 days and decreased at 14 days. On the other hand, arginase II mRNA that was detectable before treatment was increased gradually for up to 10 days and decreased at 14 days. Arginase II protein began to increase at day 5, increased for up to 10 days, and was decreased at day 14. mRNAs for cationic amino acid transporter-2 and ornithine decarboxylase were induced in a manner similar to that seen with collagen I mRNA. Immunohistochemical analysis showed that arginase I is induced in macrophages, whereas arginase II is induced in various cell types, including macrophages and myofibroblasts, and roughly colocalizes with the collagen-specific chaperone heat shock protein 47. Our findings suggest that arginine metabolic enzymes play an important role in the development of lung fibrosis, at least in mice.
Collapse
Affiliation(s)
- Motoyoshi Endo
- Department of Molecular Genetics, Kumamoto University School of Medicine, Honjo 2-2-1, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Tissue fibrosis, a serious and even deadly complication of chronic inflammation and environmental exposures, is regulated by a host of factors including interactions with the extracellular matrix, surface of inflammatory cells, hormones, and an extremely complex and redundant network of profibrotic cytokines. The nature of mechanisms by which cytokines regulate fibrosis is dual - indirect, through attraction of inflammatory cells, and direct, through binding to specific receptors on fibroblasts and stimulating proliferation, collagen production and secretion of autocrine factors. This review focuses on systematizing the direct effects of cytokines on fibroblasts. Understanding of the complexity of the cytokine-driven mechanisms of fibrosis is important for identification of potential molecular targets for future pharmacological interventions in prevention and treatment of tissue fibrosis.
Collapse
Affiliation(s)
- Sergei P Atamas
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, MSTF building, Room 8-34, 10 South Pine Street, Baltimore 21201, USA.
| |
Collapse
|
16
|
Izbicki G, Or R, Christensen TG, Segel MJ, Fine A, Goldstein RH, Breuer R. Bleomycin-induced lung fibrosis in IL-4-overexpressing and knockout mice. Am J Physiol Lung Cell Mol Physiol 2002; 283:L1110-6. [PMID: 12376365 DOI: 10.1152/ajplung.00107.2002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The role of IL-4 in the development of lung fibrosis is as yet unclear. Bleomycin (Bleo) or saline (Sal) was injected intratracheally into three groups of C57BL/6J mice: transgenic animals that overexpressed IL-4 (IL-4 TG, n = 14), mice with a targeted knockout mutation of the IL-4 gene (IL-4 KO, n = 11), and wild-type (WT, n = 13) mice. At 14 days, lung fibrosis was evaluated by hydroxyproline measurement and by quantitative image analysis of fibrosis fraction and alveolar wall area fraction. Bronchoalveolar lavage cell counts in all Bleo-treated groups demonstrated an increased percentage of lymphocytes with a corresponding decrease in the percentage of macrophages. Comparing Bleo- to Sal-treated controls within each group of mice showed increases in all lung fibrosis parameters in IL-4 KO and WT, but not in any of the parameters in IL-4 TG mice. The severity of Bleo-induced fibrotic response was decreased in overexpressed IL-4 TG compared with IL-4 KO mice. These data negate a critical profibrotic role for IL-4 in Bleo-induced lung fibrosis.
Collapse
Affiliation(s)
- Gabriel Izbicki
- Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonology, Hadassah University Hospital and The Hebrew University-Hadassah Medical School, 91120 Jerusalem, Israel
| | | | | | | | | | | | | |
Collapse
|
17
|
Kramer G, Steiner GE, Handisurya A, Stix U, Haitel A, Knerer B, Gessl A, Lee C, Marberger M. Increased expression of lymphocyte-derived cytokines in benign hyperplastic prostate tissue, identification of the producing cell types, and effect of differentially expressed cytokines on stromal cell proliferation. Prostate 2002; 52:43-58. [PMID: 11992619 DOI: 10.1002/pros.10084] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) frequently exhibit infiltration of CD4 (+)/CD45RO (+) memory-T-lymphocytes. Expression and impact of lymphocyte-derived growth factors on prostatic stromal cell (PSC) growth were investigated. METHODS; Lymphokine synthesis in normal prostate tissues (n = 3), BPH-tissues (n = 13), BPH-derived T-cells (n = 6), BPH-derived epithelial cells (BPH-EC) (n = 5), normal prostate-derived (n = 3) and BPH-derived stromal cell lines (BPH-SC) (n = 6), and prostate cancer (CaP) lines (n = 3) was analyzed by RT-PCR and Southern-blotting. The effect of interleukin (IL)-2, -4, -7, and interferon-gamma (IFN-gamma) on normal and BPH-SC growth was investigated by (3)H-thymidine incorporation assays. RESULTS All BPH-tissues and, to a lesser degree, normal prostates, expressed significant amounts of IFN-gamma mRNA. However, only BPH-tissues contained IL-2 and IL-4 mRNA (ratio: 10:13). BPH-T-cell lines were heterogeneous in composition and expressed significant amounts of IFN-gamma, IL-2, and IL-4 mRNA. Low level expression of these lymphokines was also observed in BPH-EC, CaP lines, and PSC lines. IL-2, -7 and IFN-gamma stimulated the proliferation of BPH-PSC lines but not that of normal PSC, while IL-4 inhibited BPH-PSC growth. CONCLUSIONS Chronic inflammation may induce an increased growth pattern of fibromuscular tissue in BPH similar to that of wound healing.
Collapse
Affiliation(s)
- Gero Kramer
- Department of Urology, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Atamas SP, Luzina IG, Dai H, Wilt SG, White B. Synergy between CD40 ligation and IL-4 on fibroblast proliferation involves IL-4 receptor signaling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:1139-45. [PMID: 11801648 DOI: 10.4049/jimmunol.168.3.1139] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Fibrosis can be an undesired consequence of activated cellular immune responses. The purpose of this work was to determine whether CD40 ligation and the pro-fibrotic cytokine IL-4 interact in regulating fibroblast proliferation and collagen production, and, if so, the mechanisms used. This study found that the combination of IL-4 and ligation of CD40 on the fibroblast cell surface had synergistic effects in stimulating fibroblast proliferation. In contrast, CD40 ligation negated the inhibitory effects of IFN-gamma on fibroblast proliferation. Western blotting analyses of fibroblast crude lysates revealed that a potential mechanism of the synergy between CD40 ligation and IL-4 was the phosphorylation of proteins at 130 kDa and, to a lesser degree, at 95, 85, and 75 kDa. Immunoprecipitation-Western blotting experiments showed that phosphorylation levels of IL-4Ralpha, Janus kinase 1, insulin receptor substrate 1, and insulin receptor substrate 2, factors with molecular mass close to the observed 130 kDa major phosphorylation band, increased in response to the combined CD40 ligation and IL-4 action. In contrast, there was no evidence that synergy was mediated by an increased expression of IL-4Ralpha chain, CD40, or the autocrine profibrotic cytokines IL-6 and TGF-beta. These findings suggest that CD40-CD40 ligand contacts between fibroblasts and cells secreting IL-4 may promote the profibrotic effects of IL-4 by affecting signal transduction and reducing the anti-fibrotic effects of IFN-gamma.
Collapse
Affiliation(s)
- Sergei P Atamas
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine, MSTF 8-34, 10 South Pine Street, Baltimore, MD 21201, USA.
| | | | | | | | | |
Collapse
|
19
|
Gharaee-Kermani M, Nozaki Y, Hatano K, Phan SH. Lung interleukin-4 gene expression in a murine model of bleomycin-induced pulmonary fibrosis. Cytokine 2001; 15:138-47. [PMID: 11554783 DOI: 10.1006/cyto.2001.0903] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interleukin-4 (IL-4) is known to activate mononuclear cells as well as fibroblasts, all of which are important in the pathogenesis of pulmonary fibrosis. To investigate the potential role of this cytokine, lung IL-4 expression was examined in a murine model of bleomycin-induced pulmonary fibrosis. Lung fibrosis was induced in CBA/J mice by endotracheal injection of bleomycin on day 0. On selected days after treatment, lungs were harvested for reverse transcriptase polymerase chain reaction (RT-PCR), Northern, in-situ hybridization and immunohistochemical analyses. RT-PCR and Northern analyses revealed significant increases in lung IL-4 mRNA content between days 3 and 14 after induction of lung injury, which decreased toward control level after day 21. Both in-situ hybridization and immunohistochemistry showed low or undetectable IL-4 expression in control lungs and in injured lungs before day 3 after bleomycin injection. There was however elevated expression in mononuclear cells and macrophages between days 3 and 14, localized to areas of active fibrosis. These results demonstrate that IL-4 is upregulated significantly in this model. They suggest a potential role for this cytokine in pulmonary fibrosis, perhaps via its ability to stimulate and amplify the inflammatory response, stimulate collagen synthesis in fibroblasts, and thus promote the progression to fibrosis and end stage lung disease.
Collapse
Affiliation(s)
- M Gharaee-Kermani
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0602, USA
| | | | | | | |
Collapse
|
20
|
Madison JM, Ethier MF. Interleukin-4 rapidly inhibits calcium transients in response to carbachol in bovine airway smooth muscle cells. Am J Respir Cell Mol Biol 2001; 25:239-44. [PMID: 11509335 PMCID: PMC2043474 DOI: 10.1165/ajrcmb.25.2.4286] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
To assess interleukin (IL)-4 effects on calcium signaling, bovine airway smooth-muscle (ASM) cells were loaded with fura-2 and cytosolic calcium ([Ca(2+)](i)) was measured in single cells by digital microscopy. Human recombinant IL-4 (50 ng/ml) caused small increases in [Ca(2+)](i). For single cells, carbachol-stimulated calcium transients were compared before (S1) and after (S2) exposure to IL-4 or IL-13. When cells were treated with IL-4 (50 ng/ml) for 20 min, the S2/S1 ratio was 0.17 +/- 0.04 (n = 7) even though IL-4 had been washed from the chamber for 10 min before the S2 response. In contrast, controls not treated with IL-4 had S2/S1 of 0.70 +/- 0.04 (n = 13, P < 0.01). Lower concentrations of IL-4 variably decreased transients and IL-13 had no effect. In other experiments, 5 min of IL-4 did not immediately decrease transients but did after a 25-min delay. Goat antihuman IL-4 antibody abolished the effect of IL-4. IL-4 (50 ng/ml) also inhibited responses to caffeine (S2/S1: 0.30 +/- 0.04 and 0.54 +/- 0.06 for IL-4-treated versus control). We conclude that IL-4 rapidly inhibited calcium transients. Because caffeine-stimulated transients were inhibited, IL-4 may act, at least in part, by depleting calcium stores. IL-4 inhibition of cholinergic signaling may be important for modulating ASM responses during inflammation.
Collapse
|
21
|
Wei LH, Jacobs AT, Morris SM, Ignarro LJ. IL-4 and IL-13 upregulate arginase I expression by cAMP and JAK/STAT6 pathways in vascular smooth muscle cells. Am J Physiol Cell Physiol 2000; 279:C248-56. [PMID: 10898736 DOI: 10.1152/ajpcell.2000.279.1.c248] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The objectives of this study were to determine whether rat aortic smooth muscle cells (RASMC) express arginase and to elucidate the possible mechanisms involved in the regulation of arginase expression. The results show that RASMC contain basal arginase I (AI) activity, which is significantly enhanced by stimulating the cells with either interleukin (IL)-4 or IL-13, but arginase II (AII) expression was not detected under any condition studied here. We further investigated the signal transduction pathways responsible for AI induction. AI mRNA and protein levels were enhanced by addition of forskolin (1 microM) and inhibited by H-89 (30 microM), suggesting positive regulation of AI by a protein kinase A pathway. Genistein (10 microgramg/ml) and sodium orthovanadate (Na(3)VO(4); 10 microM) were used to investigate the role of tyrosine phosphorylation in the control of AI expression. Genistein inhibited, whereas Na(3)VO(4) enhanced the induction of AI by IL-4 or IL-13. Along with immunoprecipitation and immunoblot analyses, these data implicate the JAK/STAT6 pathway in AI regulation. Dexamethasone (Dex) and interferon (IFN)-gamma were investigated for their effects on AI induction. Dex (1 microM) and IFN-gamma (100 U/ml) alone had no effect on basal AI expression in RASMC, but both reduced AI induction by IL-4 and IL-13. In combination, Dex and IFN-gamma abolished AI induction by IL-4 and IL-13. Finally, both IL-4 and IL-13 significantly increased RASMC DNA synthesis as monitored by [(3)H]thymidine incorporation, demonstrating that upregulation of AI is correlated with an increase in cell proliferation. Blockade of AI induction by IFN-gamma, H-89, or genistein also blocked the increase in cell proliferation. These observations are consistent with the possibility that upregulation of AI might play an important role in the pathophysiology of vascular disorders characterized by excessive smooth muscle growth.
Collapse
Affiliation(s)
- L H Wei
- Department of Molecular and Medical Pharmacology, UCLA School of Medicine, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
22
|
Müller-Ladner U, Judex M, Ballhorn W, Kullmann F, Distler O, Schlottmann K, Gay RE, Schölmerich J, Gay S. Activation of the IL-4 STAT pathway in rheumatoid synovium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:3894-901. [PMID: 10725752 DOI: 10.4049/jimmunol.164.7.3894] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
STATs act as second messenger after binding of a signaling molecule to its receptor. IL-4 STAT is directly involved in the IL-4-dependent gene transcription in the nucleus. We examined the expression and activation of IL-4 STAT and its related kinase Jak-1 in rheumatoid synovium. Rheumatoid arthritis (RA) synovial frozen sections of patients with short-term (<1 year) and long-term disease (>2 years) were examined using in situ hybridization and immunohistochemistry. IL-4 STAT mRNA could be detected in synovium of patients with short-term and long-term RA. The most intensive expression of IL-4 STAT mRNA could be seen in follicular inflammatory infiltrates. In the synovial lining, both fibroblasts and macrophages expressed IL-4 STAT mRNA. IL-4 STAT and Jak-1 protein was expressed by synoviocytes, and up-regulation could be induced after stimulation with IL-4. Activation of IL-4 STAT was reflected by phosphorylation of IL-4 STAT. The results indicate that IL-4 STAT is involved in key pathomechanisms in RA synovium and that IL-4 STAT-dependent pathways operate in early and late stages of the disease and presumably contribute to inhibitory immune mechanisms in RA synovium.
Collapse
Affiliation(s)
- U Müller-Ladner
- Department of Internal Medicine I, University of Regensburg, Regensburg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lubberts E, Joosten LAB, van den Bersselaar L, Helsen MMA, Bakker AC, van Meurs JBJ, Graham FL, Richards CD, van den Berg WB. Adenoviral Vector-Mediated Overexpression of IL-4 in the Knee Joint of Mice with Collagen-Induced Arthritis Prevents Cartilage Destruction. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.8.4546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Rheumatoid arthritis is a chronic inflammatory joint disease, leading to cartilage and bone destruction. In this study, we investigated the effects of local IL-4 application, introduced by a recombinant human type 5 adenovirus vector, in the knee joint of mice with collagen-induced arthritis. One intraarticular injection with an IL-4-expressing virus caused overexpression of IL-4 in the mouse knee joint. Enhanced onset and aggravation of the synovial inflammation were found in the IL-4 group. However, despite ongoing inflammation, histologic analysis showed impressive prevention of chondrocyte death and cartilage erosion. In line with this, chondrocyte proteoglycan synthesis was enhanced in the articular cartilage. This was quantified with ex vivo 35S-sulfate incorporation in patellar cartilage and confirmed by autoradiography on whole knee joint sections. Reduction of cartilage erosion was further substantiated by lack of expression of the stromelysin-dependent cartilage proteoglycan breakdown neoepitope VDIPEN in the Ad5E1 mIL-4-treated knee joint. Reduced metalloproteinase activity was also supported by markedly diminished mRNA expression of stromelysin-3 in the synovial tissue. Histologic analysis revealed marked reduction of polymorphonuclear cells in the synovial joint space in the IL-4-treated joints. This was confirmed by immunolocalization studies on knee joint sections using NIMP-R14 staining and diminished mRNA expression of macrophage-inflammatory protein-2 in the synovium tissue. mRNA levels of TNF-α and IL-1β were suppressed as well, and IL-1β and nitric oxide production by arthritic synovial tissue were strongly reduced. Our data show an impressive cartilage-protective effect of local IL-4 and underline the feasibility of local gene therapy with this cytokine in arthritis.
Collapse
Affiliation(s)
- Erik Lubberts
- *Rheumatology Research Lab, Department of Rheumatology, University Hospital Nijmegen, Nijmegen, The Netherlands; and
| | - Leo A. B. Joosten
- *Rheumatology Research Lab, Department of Rheumatology, University Hospital Nijmegen, Nijmegen, The Netherlands; and
| | - Liduine van den Bersselaar
- *Rheumatology Research Lab, Department of Rheumatology, University Hospital Nijmegen, Nijmegen, The Netherlands; and
| | - Monique M. A. Helsen
- *Rheumatology Research Lab, Department of Rheumatology, University Hospital Nijmegen, Nijmegen, The Netherlands; and
| | - Andrew C. Bakker
- *Rheumatology Research Lab, Department of Rheumatology, University Hospital Nijmegen, Nijmegen, The Netherlands; and
| | - Joyce B. J. van Meurs
- *Rheumatology Research Lab, Department of Rheumatology, University Hospital Nijmegen, Nijmegen, The Netherlands; and
| | - Frank L. Graham
- †Department of Pathology, McMaster University, Hamilton, Ontario, Canada
| | - Carl D. Richards
- †Department of Pathology, McMaster University, Hamilton, Ontario, Canada
| | - Wim B. van den Berg
- *Rheumatology Research Lab, Department of Rheumatology, University Hospital Nijmegen, Nijmegen, The Netherlands; and
| |
Collapse
|
24
|
Atamas SP, White B. Interleukin 4 in systemic sclerosis: not just an increase. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 1999; 6:658-9. [PMID: 10473512 PMCID: PMC95749 DOI: 10.1128/cdli.6.5.658-659.1999] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- S P Atamas
- University of Maryland School of Medicine and Veterans Affairs Maryland Health Care Center, Baltimore, Maryland 21201, USA.
| | | |
Collapse
|
25
|
Ong CJ, Ip S, Teh SJ, Wong C, Jirik FR, Grusby MJ, Teh HS. A role for T helper 2 cells in mediating skin fibrosis in tight-skin mice. Cell Immunol 1999; 196:60-8. [PMID: 10486156 DOI: 10.1006/cimm.1999.1537] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mice heterozygous for the tight-skin (Tsk) mutation develop skin fibrosis. Previous studies have implicated a role for the immune system and, specifically, CD4(+) T cells, in the etiology of skin fibrosis in Tsk/+ mice. We have recently shown that the administration of neutralizing anti-IL-4 antibodies to Tsk/+ mice prevented the development of skin fibrosis in these mice. Since IL-4 is a major cytokine produced by T helper 2 (Th2) cells, we investigated the role of Th2 cells in mediating skin fibrosis in Tsk/+ mice. Previous studies have shown that the development of Th2 cells in non-Tsk mice is abrogated in mice with null mutation for either the IL-4 or the Stat6 gene. In this study we showed that the polarization of CD4(+) T cells from Tsk/+ mice toward the Th2 lineage is also dependent on a functioning IL-4 or Stat6 gene. More importantly, the development of skin fibrosis in Tsk/+ mice was abrogated by the IL4(-/-) or the Stat6(-/-) mutation. We also determined whether alteration of the TCR repertoire in Tsk/+ mice, achieved by the introduction of TCR transgenes, was able to prevent the development of skin fibrosis in Tsk/+ mice. We found that the exclusive usage of the Vbeta8.2 gene segment by T cells was sufficient to prevent skin fibrosis in Tsk/+ mice. This result suggests that the exclusive use of this Vbeta gene segment by T cells may have prevented the development of fibrosis-causing Th2 cells.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Disease Models, Animal
- Female
- Fibrosis/immunology
- Fibrosis/pathology
- H-2 Antigens/immunology
- Interferon-gamma/biosynthesis
- Interleukin-4/biosynthesis
- Interleukin-4/genetics
- Interleukin-4/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Mutant Strains
- Mice, Transgenic
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- STAT6 Transcription Factor
- Scleroderma, Systemic/immunology
- Scleroderma, Systemic/pathology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/immunology
- Trans-Activators/genetics
- Trans-Activators/immunology
Collapse
Affiliation(s)
- C J Ong
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | | | | | | | | | | | | |
Collapse
|
26
|
Sugihara A, Tsujimura T, Fujita Y, Nakata Y, Terada N. Evaluation of role of mast cells in the development of liver fibrosis using mast cell-deficient rats and mice. J Hepatol 1999; 30:859-67. [PMID: 10365813 DOI: 10.1016/s0168-8278(99)80140-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND/AIMS Several studies have suggested that mast cells participate in the development of liver fibrosis in rodent models. In this study mast cell-deficient mutant Ws/Ws rats and W/Wv mice were used to examine whether mast cells are involved in the development of liver fibrosis. METHODS Liver fibrosis was induced in rats by bile duct resection (BDR), and by intraperitoneal injections of carbon tetrachloride (CCl4) or porcine serum, and in mice by intragastric administrations of CCl4, and BDR. The degree of fibrosis was evaluated by measuring the hydroxyproline content (microg/mg tissue) of the liver as an index of the collagen content. The density of mast cells (number/cm2 liver section) was determined by counting mast cells in liver sections stained with alcian blue. RESULTS In the liver of control non-mutant (+/+) rats, mast cells were found principally in portal areas, and their average density was 200-300/cm2 liver section. BDR, and treatments with CCl4 and porcine serum increased the density of mast cells in the liver of +/+ rats several-fold, and induced liver fibrosis, increasing the liver hydroxyproline content markedly. BDR, and treatments with CCl4 and porcine serum also induced liver fibrosis in Ws/Ws rats, increasing the liver hydroxyproline content to a similar or higher level than that in +/+ rats. However, the average densities of mast cells in the liver of Ws/Ws rats after BDR and treatment with CCl4 and porcine serum were at most 10.2/cm2 liver section. The density of mast cells in the liver of control +/+ mice was extremely low (average, less than 2), and neither BDR nor treatment with CCl4 caused any significant increase in their density, whereas these treatments induced liver fibrosis and markedly increased the liver hydroxyproline content. Furthermore, treatment with CCl4 induced fibrosis in the liver of W/Wv mice similarly to that in +/+ mice, but the density of mast cells in the liver of W/Wv mice was very low (average, less than 1), and was not increased by treatment with CCl4. CONCLUSIONS The present results indicate that mast cells play no role in the development of liver fibrosis in rats and mice.
Collapse
Affiliation(s)
- A Sugihara
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Japan
| | | | | | | | | |
Collapse
|
27
|
Yamamoto T, Takagawa S, Katayama I, Yamazaki K, Hamazaki Y, Shinkai H, Nishioka K. Animal model of sclerotic skin. I: Local injections of bleomycin induce sclerotic skin mimicking scleroderma. J Invest Dermatol 1999; 112:456-62. [PMID: 10201529 DOI: 10.1046/j.1523-1747.1999.00528.x] [Citation(s) in RCA: 309] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We have established a mouse model for scleroderma induced by repeated local injections of bleomycin (BLM). Daily injection of BLM at a dose of >10 microg per ml for 4 wk induced histologic changes of dermal sclerosis, but not fibrosis, with thickened and homogenous collagen bundles and cellular infiltrates in BALB/C mice, whereas clinical signs of scleroderma were not apparent. In addition, lung fibrosis was also induced preceding the cutaneous changes. Sclerotic changes were not found in other sites of the skin distant from the injection site. Dermal sclerosis could also be induced by injecting BLM only every other day. The sclerotic changes of the dermis were sustained after ceasing BLM applications for at least 6 wk. Mast cells gradually increased in number as the sclerotic changes developed. Marked degranulation of mast cells was observed with elevated histamine release. The amount of hydroxyproline in skin was significantly increased at 4 wk of BLM treatment as compared with that in untreated or phosphate-buffered saline-treated mice. Anti-nuclear antibody was detected in serum of BLM-treated mice. Transforming growth factor-beta1 mRNA was detected at an early phase, while transforming growth factor-beta2 mRNA was strongly expressed at 4 wk when the sclerotic features were prominent. These results suggest that dermal sclerosis induced by BLM closely resembles systemic sclerosis both histologically and biochemically. Our mouse model can provide a powerful tool of inducing dermal sclerosis to examine the pathogenesis and the therapeutic approach of scleroderma.
Collapse
Affiliation(s)
- T Yamamoto
- Department of Dermatology, Tokyo Medical and Dental University, School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Hogaboam CM, Steinhauser ML, Chensue SW, Kunkel SL. Novel roles for chemokines and fibroblasts in interstitial fibrosis. Kidney Int 1998; 54:2152-9. [PMID: 9853282 DOI: 10.1046/j.1523-1755.1998.00176.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Regardless of its involvement in either wound healing or excessive fibrosis, the interstitial fibroblast can now be considered an important early participant in inflammatory responses. Although it is recognized that certain immune cells and proinflammatory mediators are intricately linked to fibrotic disease, little is presently known about the manner in which these mediators and cells are orchestrated to a fibrotic finale. Experimental studies have shown that interstitial fibroblasts are capable of participating in an inflammatory response by promoting direct fibroblast-to-immune cell communication and/or modulating the release of soluble mediators that are mutually recognized by both types of cells. METHODS Primary cultures of murine fibroblasts were recovered from either normal tissue or tissue undergoing a cell-mediated inflammatory response. These stromal cells were assessed for the expression of various cytokines and chemokines indicative of a type 1 or type 2 response. In addition, the fibroblasts were co-cultured with mononuclear cells to assess the cell-to-cell communication. RESULTS Fibroblasts recovered from different cell-mediated inflammatory responses demonstrated a dramatic alteration in their cytokine profile. Fibroblasts recovered from the type 2 immune response produced high levels of monocyte chemotactic protein-1 (MCP-1), as compared to the normal fibroblasts and fibroblasts recovered from the type 1 lesion. Mononuclear cells co-cultured with fibroblasts induced a contact-dependent expression of elevated levels of chemokines, especially the macrophage-derived MIP-1 alpha. Thus, both fibroblasts themselves and fibroblasts co-cultured with immune-inflammatory cells have the ability to participate in the maintenance of an inflammatory response via the expression of chemokines. CONCLUSIONS Our laboratory and others have addressed the role of chemotactic cytokines or chemokines in the fibrotic process, and have demonstrated that fibroblasts are capable of modulating the activation of various immune cells that have been implicated in fibrotic disease. In addition, the interstitial fibroblast is capable of regulating its own behavior within the interstitial environment via the expression of chemokines and chemokine receptors. Thus, novel strategies aimed at preventing fibrotic disease will likely need to address the early engagement of inflammatory cells by fibroblasts, and possibly modulate the ability of fibroblasts to generate and/or recognize profibrotic signals supplied by chemokines.
Collapse
Affiliation(s)
- C M Hogaboam
- Department of Pathology, University of Michigan Medical School, Ann Arbor, USA
| | | | | | | |
Collapse
|
29
|
|
30
|
Hawker KM, Johnson PR, Hughes JM, Black JL. Interleukin-4 inhibits mitogen-induced proliferation of human airway smooth muscle cells in culture. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:L469-77. [PMID: 9728041 DOI: 10.1152/ajplung.1998.275.3.l469] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The increase in the amount of airway smooth muscle in the bronchial wall associated with asthma is partly due to hyperplasia. It is therefore important to determine which factors regulate growth and especially proliferation. In this study, we describe the effect of interleukin-4 (IL-4), a mast cell- and T lymphocyte-derived cytokine, on human airway smooth muscle proliferation as determined by [3H]thymidine uptake in the presence of fetal bovine serum (FBS), platelet-derived growth factor, basic fibroblast growth factor, and thrombin. IL-4 (5, 15, 50, and 150 ng/ml) significantly decreased 10% FBS-induced proliferation by 50, 73, 43, and 46%, respectively. The proliferative responses to platelet-derived growth factor (20 and 40 ng/ml), basic fibroblast growth factor (30 ng/ml), and thrombin (1 and 10 U/ml) were significantly reduced by 19, 21, 37, 36, and 57% respectively in the presence of 50 ng/ml of IL-4. We investigated the effect of IL-4 and other known inhibitors of smooth muscle proliferation, namely PGE2, heparin, and forskolin, on intracellular cAMP concentrations. IL-4 (50 ng/ml) and heparin (100 U/ml) did not alter intracellular cAMP levels when cells were treated with 1 or 10% FBS. PGE2 (1 microM) and forskolin (10 microM) significantly increased cAMP concentration above the control value in nonproliferating cells (1% FBS treated) by 7- and 37-fold, respectively. The effect of IL-4 (50 ng/ml), PGE2 (1 microM), and forskolin (10 microM) on cyclin D1 protein expression in 10% FBS-stimulated human airway smooth muscle cells was also examined. PGE2 and forskolin did not significantly inhibit cyclin D1 expression. However, IL-4 decreased cyclin D1 expression by 21%. These results provide evidence that IL-4 decreases human airway smooth muscle cell proliferation via a mechanism that is cAMP independent and mediated, in part, by a decrease in cyclin D1 protein expression.
Collapse
Affiliation(s)
- K M Hawker
- Department of Pharmacology, University of Sydney, New South Wales 2006, Australia
| | | | | | | |
Collapse
|
31
|
Ong C, Wong C, Roberts CR, Teh HS, Jirik FR. Anti-IL-4 treatment prevents dermal collagen deposition in the tight-skin mouse model of scleroderma. Eur J Immunol 1998; 28:2619-29. [PMID: 9754550 DOI: 10.1002/(sici)1521-4141(199809)28:09<2619::aid-immu2619>3.0.co;2-m] [Citation(s) in RCA: 125] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The tight-skin (Tsk/+) mutant mouse, a putative murine model of scleroderma, is characterized primarily by the excessive deposition of collagen and other extracellular matrix molecules in the dermis, and also by a developmentally acquired defect in pulmonary architecture. Passive transfer experiments have suggested an etiologic role for the immune system in Tsk/+ dermal pathology. In addition, CD4+ T lymphocytes have been shown to be required for the excessive accumulation of dermal collagen in these mice. As IL-4, a product of differentiated CD4+ T cells, is capable of regulating the synthesis of various matrix molecules (including type I collagen) by fibroblasts in vitro, we investigated the potential role of IL-4 in mediating Tsk/+ dermal fibrosis. Confirming that Tsk/+ cells are capable of responding to IL-4, we found receptors for this cytokine on Tsk/+ embryonic fibroblasts and a dermal fibroblast cell line derived from these mice. Furthermore, IL-4 receptors on Tsk/+ fibroblasts were functional since IL-4 stimulation in vitro increased type I collagen secretion from these cells. These results demonstrated the potential for IL-4 to be directly involved in the excessive deposition of dermal collagen in Tsk/+ mice. Critical insight into the role played by IL-4 in mediating the dermal phenotype, however, was obtained following the administration of neutralizing anti-lL-4 antibodies to Tsk/+ mice. This treatment prevented the development of dermal fibrosis, leading to normalization of dermal collagen content. Given the requirement for CD4+ T cells in Tsk/+ dermal fibrosis, our results suggest that Th2 cells and/or factors elaborated by this T cell subset may play a key role in regulating dermal collagen content in this strain.
Collapse
Affiliation(s)
- C Ong
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | | | | | | | | |
Collapse
|
32
|
Trautmann A, Krohne G, Bröcker EB, Klein CE. Human Mast Cells Augment Fibroblast Proliferation by Heterotypic Cell-Cell Adhesion and Action of IL-4. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.10.5053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Mast cells have been implicated in the pathogenesis of fibrosis because of their increased number in chronic inflammatory reactions. In a previous study, we had shown that human mast cells readily attach and form heterotypic cell-cell contacts when seeded on top of fibroblast monolayers. Here, we report that human mast cells stimulate fibroblast proliferation after cell-cell contact. Proliferation was measured by 5-bromo-2′-deoxyuridine or [3H]thymidine uptake of subconfluent fibroblast monolayers after attachment of mast cells that had been preincubated with mitomycin C. An 18-h coculture of the human mast cell line HMC-1 doubled proliferation of normal skin fibroblasts. Moreover, normal mast cells prepared from neonatal foreskin doubled fibroblast proliferation. The stimulatory effect was dependent on heterotypic cell-cell contact since it was not transferred by tissue culture supernatants from mast cells. We hypothesized that mast cell cytokines secreted after heterotypic cell-cell contact stimulate fibroblast proliferation. Several mast cell-derived cytokines were tested for effects on fibroblast proliferation. Only IL-4 was able to double fibroblast proliferation. Additional experiments revealed that: 1) the stimulatory effect of IL-4 as well as of the mast cell coculture could be completely abrogated by preincubation of fibroblasts with an anti-IL-4R mAb blocking ligand binding; 2) mast cell-derived IL-4 acts as a second signal for fibroblasts since it amplifies the action of low doses of obligatory fibroblast growth factors such as fibroblast growth factor or platelet-derived growth factor.
Collapse
Affiliation(s)
| | - Georg Krohne
- †Theodor-Boveri-Institute, University of Würzburg, Würzburg, Germany
| | | | | |
Collapse
|
33
|
Affiliation(s)
- A E Postlethwaite
- Department of Medicine, University of Tennessee, Memphis 38163-0001, USA
| |
Collapse
|
34
|
Elbadawi A. Interstitial cystitis: a critique of current concepts with a new proposal for pathologic diagnosis and pathogenesis. Urology 1997; 49:14-40. [PMID: 9145999 DOI: 10.1016/s0090-4295(99)80329-x] [Citation(s) in RCA: 160] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Interstitial cystitis (IC) has continued to be an unresolved problem in clinical urology despite intense investigation over the past 16 or more years. Its etiology and pathogenesis are still undetermined, and its pathologic diagnosis is essentially one of exclusion, with no specific or clear criteria. In this review, current concepts of the etiology/pathogenesis and pathology are critically analyzed, new pathologic observations summarized, and a proposal of neurogenic inflammation as the primary pathogenetic factor is presented in the context of all currently available information. The popular postulate attributing IC to a deficient or defective glycosaminoglycan urothelial surface layer is not substantiated by morphologic, experimental, clinical, or therapeutic observations. Although the consensus seems to discount an infectious etiology, there is sufficient evidence that a microbial factor-short of a bona fide clinical infection-may have a role. Both autoimmunity and mast cell infiltration also appear to have a role, despite the lack of evidence that either is involved as the primary etiologic factor. Claims that the so-called feline urologic syndrome may represent a natural animal model of IC are shaky. As it now stands, there is no natural or induced animal model that duplicates IC as it occurs in humans. No specific or diagnostic light microscopic pathologic features are provided by either routine histopathology or immunohistochemistry. Increasingly, it has been recognized that detrusor mast cell count has little or no diagnostic value. On the other hand, electron microscopy has provided important new observations: (a) presence of mast cells, activated by piecemeal degranulation, in close proximity to intrinsic nerves-particularly in suburothelium: (b) distinctive pathologic changes in urothelium, suburothelium, and muscularis in biopsy samples obtained after diagnostic bladder hydrodistension; (c) constant associated changes in venules, capillaries, and neural elements in the same biopsy samples; and (d) diffuse involvement of bladder wall, with the most evident and profound pathologic changes in posthydrodistension biopsy samples obtained from cystoscopically obvious lesions (glomerulations). These features are sufficiently distinctive to allow definitive pathologic diagnosis of IC, and provide a firm basis for primary involvement of neurogenic inflammation in its pathogenesis. A proposal is presented regarding the mechanisms invoked by neurogenic inflammation. This proposal revolves around sensory nerve excitation, the release of neuropeptides, and activated differential secretion of potent mast cell mediators. This proposal can account for the heterogeneity and variability of observed pathologic features, and upholds the tacit acceptance of IC as a disease of pluricausal etiology and multifactorial pathogenesis.
Collapse
Affiliation(s)
- A Elbadawi
- Department of Pathology, State University of New York, Health Science Center, Syracuse 13210, USA
| |
Collapse
|
35
|
FcεRI-induced Cytokine Production and Gene Expression. IGE RECEPTOR (FCΕRI) FUNCTION IN MAST CELLS AND BASOPHILS 1997. [DOI: 10.1007/978-3-662-22022-1_11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
36
|
Kendall JC, Li XH, Galli SJ, Gordon JR. Promotion of mouse fibroblast proliferation by IgE-dependent activation of mouse mast cells: role for mast cell tumor necrosis factor-alpha and transforming growth factor-beta 1. J Allergy Clin Immunol 1997; 99:113-23. [PMID: 9003219 DOI: 10.1016/s0091-6749(97)70308-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Pathologic fibroblast proliferation or tissue fibrosis develops in certain chronic allergic diseases and in a wide array of other inflammatory disorders in which mast cell activation is also a prominent feature. In this study we investigated a number of potential mechanisms by which IgE-dependent activation of mouse mast cells might influence the proliferation of mouse fibroblasts in vitro. We found that supernatants from in vitro-derived mast cells that had been activated by IgE and specific antigen (but not those from quiescent mast cells) promoted the proliferation of mouse embryonic skin or 3T3 fibroblasts, and we showed that this effect was detectable in the absence of fetal calf serum. We analyzed the kinetics with which the fibroblast-proliferative activity was secreted from bone marrow-derived cultured mast cells and found that it was released both rapidly (i.e., in 30 minutes or less) and for a more prolonged period (i.e., for more than 2 hours) after IgE-dependent mast cell activation. We then measured the levels at which the mast cells produce a number of cytokines that are known to affect fibroblasts (IL-1, IL-6, transforming growth factor-beta 1 [TGF-beta 1], and tumor necrosis factor-alpha [TNF-alpha]) and assessed their relative effects, as recombinant cytokines, on fibroblast proliferation. Our mast cells secreted high levels of TGF-beta 1 and TNF-alpha, intermediate amounts of IL-6, and low levels of IL-1. We titrated the fibroproliferative effects of each of these cytokines and determined that at a dose of 50 pg/ml their rank order of activity was TGF-beta 1 > TNF-alpha > IL-1 > IL-6, with all but IL-6 having significant effects. The ability of supernatants from activated bone marrow-derived cultured mast cells to promote fibroblast proliferation was partially diminished by absorption with neutralizing antibodies against either TNF-alpha or TGF-beta 1, and absorption of the supernatants with a combination of antibodies against TNF-alpha and TGF-beta 1 reduced their ability to induce fibroblast proliferation by approximately 50% (p < or = 0.001, n = 5). These findings show that IgE-dependent activation of mouse mast cells can result in the release of mediators that promote fibroblast proliferation in the absence of any other cell type and suggest that mast cell-derived TNF-alpha and TGF-beta 1 contribute substantially to this effect. They also suggest that these cytokines exert their effects through synergistic interactions with other mast cell mediators.
Collapse
Affiliation(s)
- J C Kendall
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | | | | | | |
Collapse
|
37
|
Kaklamanis L, Koukourakis MI, Leek R, Giatromanolaki A, Ritter M, Whitehouse R, Gatter KC, Harris AL. Loss of interleukin 4 receptor-associated molecule gp200-MR6 in human breast cancer: prognostic significance. Br J Cancer 1996; 74:1627-31. [PMID: 8932345 PMCID: PMC2074844 DOI: 10.1038/bjc.1996.599] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Several in vitro studies stress a potentially important role of interleukin 4 (IL-4) and the related gp200-MR6 molecule in the immunological response to cancer and in tumour proliferation. In the present study, we assessed the expression of gp200-MR6 in primary breast cacrinomas using the MR6 monoclonal antibody. Results were correlated with tumour parameters (T-,N-stage, histology, grade, oestrogen and epidermal growth factor (EGF) receptors), and the impact on survival was assessed. Twenty-four out of 110 cases (22%) were positive for gp200-MR6, 62 out of 110 (56%) expressed weak staining and 24 out of 114 (22%) did not stain. The normal breast epithelia were invariably stained for gp200-MR6 showing that down-regulation or loss of this molecule occurred during the evolution of breast cancer. Gp200-MR6 loss was independent from differentiation, nodal positivity and oestrogen receptor levels as well as patients' age. Loss of the gp200-MR6 molecule was more frequent in lobular cases (P=0.03). The overall survival was better, although not reaching statistical significance, in patients with positive gp200-MR6 expression (92% alive at 5 years compared with 70% for those with weak or no expression, P=0.1). The local relapse-free survival was independent of gp200-MR6 status. It is concluded that loss of gp200-MR6 may be one of the mechanisms through which breast cancer cells escape immune surveillance, resulting in an increased metastatic potential and poorer outcome. Evidence of down-regulation of the gp200-MR6 molecule has implications for IL-4-linked toxin therapy and, as IL-4 is an inhibitor of breast epithelial growth, may represent loss of a tumour-suppression mechanism.
Collapse
Affiliation(s)
- L Kaklamanis
- Department of Cellular Science, John Radcliffe Hospital, Oxford, UK
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Rankin JA, Picarella DE, Geba GP, Temann UA, Prasad B, DiCosmo B, Tarallo A, Stripp B, Whitsett J, Flavell RA. Phenotypic and physiologic characterization of transgenic mice expressing interleukin 4 in the lung: lymphocytic and eosinophilic inflammation without airway hyperreactivity. Proc Natl Acad Sci U S A 1996; 93:7821-5. [PMID: 8755560 PMCID: PMC38832 DOI: 10.1073/pnas.93.15.7821] [Citation(s) in RCA: 289] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
To investigate the contribution of interleukin-4 (IL-4) to airway inflammation in vivo and to explore directly its relationship to airway reactivity, we created transgenic mice in which the murine cDNA for IL-4 was regulated by the rat Clara cell 10 protein promoter. Expression was detected only in the lung and not in thymus, heart, liver, spleen, kidney, or uterus. The expression of IL-4 elicited hypertrophy of epithelial cells of the trachea, bronchi, and bronchioles. Hypertrophy is due, at least in part, to the accumulation of mucus glycoprotein. Histologic examination of parenchyma revealed multinucleated macrophages and occasional islands of cells consisting largely of eosinophils or lymphocytes. Analysis of lung lavage fluid revealed the presence of a leukocytic infiltrate consisting of lymphocytes, neutrophils and eosinophils. Mice expressing IL-4 had greater baseline airway resistance but did not demonstrate hyperreactivity to methacholine. Thus, the expression of IL-4 selectively within the lung elicits an inflammatory response characterized by epithelial cell hypertrophy, and the accumulation of macrophages, lymphocytes, eosinophils, and neutrophils without resulting in an alteration in airway reactivity to inhaled methacholine.
Collapse
Affiliation(s)
- J A Rankin
- Pulmonary and Critical Care Medicine, VA Connecticut Health Care System, West Haven 06516, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sempowski GD, Derdak S, Phipps RP. Interleukin-4 and interferon-gamma discordantly regulate collagen biosynthesis by functionally distinct lung fibroblast subsets. J Cell Physiol 1996; 167:290-6. [PMID: 8613470 DOI: 10.1002/(sici)1097-4652(199605)167:2<290::aid-jcp13>3.0.co;2-c] [Citation(s) in RCA: 110] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Pulmonary fibrosis is a potentially fatal consequence of treatments for malignancy and is an increasing problem in bone marrow transplant patients and in cases of allogenic lung transplant. The fibrotic response is characterized by increases in lung fibroblast number and collagen synthesis. This laboratory previously isolated stable, functionally distinct, murine lung fibroblast subsets (Thy-1+ and Thy-1-) to study the contribution of fibroblast subpopulations in lung fibrosis. The fibroblast fibrotic response may be induced by cytokines secreted by infiltrating cells such as T lymphocytes and mast cells. In the current study two key regulatory cytokines, interferon-gamma (IFN-gamma) and interleukin-4 (IL-4), were investigated for their effects on the collagen synthesis of murine lung fibroblast subsets. IL-4 and IFN-gamma are putatively characterized as fibrogenic and anti-fibrogenic cytokines, respectively, and are found in repairing lung tissue. Stimulation with recombinant IL-4 induced a100% increase in total collagen production only by Thy-1+ fibroblasts. Types I and III collagen mRNA were increased in the Thy-1+ fibroblasts, unlike the Thy-1- subset. In contrast, IFN-gamma decreased constitutive collagen production by more than 50% in Thy-1+ and Thy-1- fibroblasts. Interestingly, the two subsets utilized their collagen production machinery (collagenase, tissue inhibitors of metalloproteinases) differently to further regulate collagen turnover in response to IL-4 and IFN-gamma. Overall, our data support the hypothesis that IL-4 is fibrogenic and IFN-gamma is anti-fibrogenic. Moreover, selective expansion of IL-4 responsive fibroblasts (e.g., Thy-1+) may be important in the transition from repair to chronic fibrosis. In addition, these data suggest that an inflammatory response dominated by IL-4-producing Th2 lymphocytes and/or mast cells will promote fibrosis development.
Collapse
Affiliation(s)
- G D Sempowski
- University of Rochester Cancer Center, New York 14642, USA
| | | | | |
Collapse
|
40
|
Kotanides H, Moczygemba M, White MF, Reich NC. Characterization of the interleukin-4 nuclear activated factor/STAT and its activation independent of the insulin receptor substrate proteins. J Biol Chem 1995; 270:19481-6. [PMID: 7642632 DOI: 10.1074/jbc.270.33.19481] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The activation of a latent DNA binding factor by interleukin-4 (IL-4), the IL-4 nuclear activated factor (IL-4 NAF), occurs within minutes of IL-4 binding to its receptor. Molecular characterization of IL-4NAF by ultraviolet light cross-linking experiments revealed a single protein of 120-130 kDa in contact with the DNA target site. Glycerol gradient sedimentation analysis indicated a molecular mass of IL-4 NAF consistent with a monomer that is capable of binding DNA. The IL-4 NAF target site is a palindromic sequence that is also recognized by the interferon-induced transcription factor, p91/STAT1 alpha. However, IL-4 NAF and p91/STAT1 alpha display distinguishable DNA binding specificities that may generate one level of specificity in the expression of target genes. Previous studies suggested the involvement of the insulin receptor substrate-1 (IRS-1) in the IL-4 signal transduction pathway. Although IRS-1 is involved in the stimulation of mitogenesis, our results demonstrate that activation of IL-4 NAF is independent of IRS-signaling proteins. The results of this study indicate that IL-4 stimulates bifurcating signal pathways that can direct mitogenesis via the IRS-signaling proteins and specific gene expression via the IL-4 NAF.
Collapse
Affiliation(s)
- H Kotanides
- Graduate Program in Molecular and Cellular Biology, State University of New York, Stony Brook 11794-8691, USA
| | | | | | | |
Collapse
|
41
|
D'Amico G, Ferrario F, Rastaldi MP. Tubulointerstitial damage in glomerular diseases: its role in the progression of renal damage. Am J Kidney Dis 1995; 26:124-32. [PMID: 7611242 DOI: 10.1016/0272-6386(95)90165-5] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The evidence that tubular damage, interstitial infiltration, and interstitial fibrosis occur in all glomerular diseases, either immunologically or non-immunologically mediated, is reviewed on the basis of personal data and data from the literature. The proposed mechanisms linking glomerular and tubular damage to the interstitial recruitment of mononuclear leukocytes and fibroblast proliferation, with abnormal extracellular matrix production leading to interstitial fibrosis, also are analyzed. The role of persistent heavy proteinuria and exposure to proinflammatory cytokines in inducing the damage of the tubular epithelial cells, with consequent acquisition by these cells of the ability to interact as antigen-presenting cells with T lymphocytes, is especially emphasized. Finally, the importance of the tubulointerstitial damage as a marker of unfavorable prognosis in glomerular diseases is documented.
Collapse
Affiliation(s)
- G D'Amico
- Division of Nephrology, San Carlo Borromeo Hospital, Milano, Italy
| | | | | |
Collapse
|
42
|
Wegrowski Y, Paltot V, Gillery P, Kalis B, Randoux A, Maquart FX. Stimulation of sulphated glycosaminoglycan and decorin production in adult dermal fibroblasts by recombinant human interleukin-4. Biochem J 1995; 307 ( Pt 3):673-8. [PMID: 7741696 PMCID: PMC1136704 DOI: 10.1042/bj3070673] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Interleukin-4 (IL-4) is a pleiotropic cytokine expressed by inflammatory cells. Previous work from our laboratory has shown that it stimulates collagen synthesis in fibroblasts. Here we report the effects of recombinant human IL-4 on glycosaminoglycan (GAG) and proteoglycan synthesis in normal dermal fibroblasts from adult donors. IL-4 (10 and 100 units/ml) induced a dose-dependent increase of [3H]glucosamine and [35S]sulphate incorporation into total GAGs. The analysis of the different GAG fractions indicated the enhanced synthesis of dermatan/chondroitin sulphates. IL-4 had no effect on hyaluronan synthesis. The increase of sulphated GAG synthesis was correlated with an increase of proteoglycans in the culture medium. Decorin was identified as the major chondroitin/dermatan sulphate-containing proteoglycan in the culture medium of fibroblasts. Its synthesis was strongly stimulated by IL-4. Both the core-protein synthesis and mRNA expression were enhanced, indicating that the cytokine acted, at least in part, at the pre-translational level. These results indicate that IL-4 is able to modulate not only collagen, but also proteoglycan, production by human fibroblasts. Their implications in physiopathological processes such as wound healing or fibrosis is suggested.
Collapse
Affiliation(s)
- Y Wegrowski
- Laboratoire de Biochimie (CNRS ER 084), Faculté de Médecine, France
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
A number of humoral and cellular immune abnormalities are present in patients with early scleroderma (systemic sclerosis). Most of these abnormalities reflect ongoing autoimmune reactions of the cellular and humoral types, resulting in a variety of autoantibodies to cellular and tissue constituents. Evidence exists for a defect(s) in immunoregulation favoring excessive helper T cell activity. The presence of circulating cytokines and shed interleukin-2 receptors suggest ongoing cellular immune reactions are occurring, generating cytokines and lymphokines that are capable of effecting the vascular and fibrotic lesions that are hallmarks of the disease. Future directions for research are suggested that would focus on determining if, and at what point, fibroblasts might function autonomously to generate excessive matrix components and on determining the nature of the original antigenic stimulus that starts the scleroderma process.
Collapse
|
44
|
Gordon JR, Galli SJ. Promotion of mouse fibroblast collagen gene expression by mast cells stimulated via the Fc epsilon RI. Role for mast cell-derived transforming growth factor beta and tumor necrosis factor alpha. J Exp Med 1994; 180:2027-37. [PMID: 7964480 PMCID: PMC2191776 DOI: 10.1084/jem.180.6.2027] [Citation(s) in RCA: 153] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Chronic allergic diseases and other disorders associated with mast cell activation can also be associated with tissue fibrosis, but a direct link between mast cell mediator release and fibroblast collagen gene expression has not been established. Using in situ hybridization, we show that the elicitation of an IgE-dependent passive cutaneous anaphylaxis (PCA) reaction in mice results in a transient, but marked augmentation of steady state levels of type alpha-1 (I) collagen mRNA in the dermis. While peak levels of collagen mRNA expression in the skin are observed 16-24 h after mast cell activation, substantial numbers of dermal cells are strongly positive for collagen mRNA at 1 and 2 h after antigen challenge, before circulating inflammatory cells are recruited into the tissues. Furthermore, experiments in mast cell-reconstituted or genetically mast cell-deficient WBB6F1-W/Wv mice demonstrate that the increased expression of collagen mRNA at sites of PCA reactions is entirely mast cell dependent. In vitro studies show that the supernatants of mouse serosal mast cells activated via the Fc epsilon RI markedly increase type alpha-1 (I) collagen mRNA levels in mouse embryonic skin fibroblasts, and also upregulate collagen secretion by these cells. The ability of mast cell supernatants to induce increased steady state levels of collagen mRNA in mouse skin fibroblasts is markedly diminished by absorption with antibodies specific for either of two mast cell-derived cytokines, transforming growth factor beta (TGF-beta 1) or tumor necrosis factor alpha (TNF-alpha), and is eliminated entirely by absorption with antibodies against both cytokines. Taken together, these findings demonstrate that IgE-dependent mouse mast cell activation can induce a transient and marked increase in steady state levels of type alpha-1 (I) collagen mRNA in dermal fibroblasts and that mast cell-derived TGF-beta 1 and TNF-alpha importantly contribute to this effect.
Collapse
Affiliation(s)
- J R Gordon
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | | |
Collapse
|
45
|
Affiliation(s)
- F Levi-Schaffer
- Department of Pharmacology, Hebrew University--Hadassah Medical School, Jerusalem, Israel
| | | |
Collapse
|
46
|
Horsmanheimo L, Harvima IT, Järvikallio A, Harvima RJ, Naukkarinen A, Horsmanheimo M. Mast cells are one major source of interleukin-4 in atopic dermatitis. Br J Dermatol 1994; 131:348-53. [PMID: 7918008 DOI: 10.1111/j.1365-2133.1994.tb08522.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Several reports have shown the presence of T-helper lymphocytes with Th2 characteristics in the skin lesions of atopic dermatitis (AD). However, Th2 cells themselves require an exogenous pulse of IL-4 to initiate their differentiation and synthesis of IL-4. As mast cells have recently been shown to contain IL-4, this finding prompted us to investigate IL-4 in mast cells of AD lesions, to determine if they might provide the IL-4 pulse needed by the Th2 cells. In this study, we measured IL-4 immunoreactivity in mast cells of non-lesional and lesional skin sections from 20 patients with AD. Ten patients with nummular eczema (NE) without any atopic features or background, and five healthy subjects, served as the control groups. Mast cells were first identified using an enzyme--histochemical staining method for tryptase. Subsequently, the sections were photographed, the tryptase stain was removed, and IL-4 was demonstrated with a polyclonal antibody. The sections were photographed again, and the percentage of IL-4 positive mast cells was calculated. The percentage of mast cells exhibiting IL-4 immunoreactivity in the upper dermis in lesional vs. non-lesional skin was 66 +/- 18% vs. 37 +/- 18% in AD (P < 0.0001, paired t-test), but only 46 +/- 19% vs. 31 +/- 22% in NE. In the skin of healthy controls, only 23 +/- 25% of the mast cells were positive for IL-4. In addition, a significant difference was found between lesional skin of AD vs. NE patients (P < 0.008, unpaired t-test).(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- L Horsmanheimo
- Department of Dermatology, Kuopio University Hospital, Finland
| | | | | | | | | | | |
Collapse
|
47
|
Shimada H, Eguchi K, Ueki Y, Nakashima M, Yamashita I, Kawabe Y, Sakai M, Ida H, Aoyagi T, Nagataki S. Interleukin 4 increases human synovial cell expression of VCAM-1 and T cell binding. Ann Rheum Dis 1994; 53:601-7. [PMID: 7526808 PMCID: PMC1005412 DOI: 10.1136/ard.53.9.601] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE The effects were studied of interleukin 4 (IL-4) on T cell-synovial cell adhesion and on the expression of adhesion molecules on the surface of synovial fibroblast-like cells. METHODS The adhesion of T cells toward the synovial cells were measured by 51chromium-labelled adhesion assay. The expression of adhesion molecules on synovial cells were analysed by flowcytometry. RESULTS Stimulation of synovial cells with IL-4 increased T cell-synovial cells adhesion in a time- and dose-dependent manner. IL-4 considerably enhanced the expression of VCAM-1 on the surface of synovial cells, but not the expression of ICAM-1 and ELAM-1. The combination of IL-1 beta and IL-4 had no effect on the expression of ICAM-1 or VCAM-1 on the surface of synovial cells. The increased adhesion of T cells to IL-4 stimulated synovial cells was inhibited significantly by adding anti-VCAM-1 or anti-CD29 monoclonal antibody. Furthermore, anti-VLA-4 alpha or the combination of anti-VLA-4 alpha and anti-VCAM-1 antibodies blocked completely T-cell binding to IL-4 stimulated synovial cells. CONCLUSIONS These results suggest that the increased adhesion of T cells to IL-4-stimulated synovial cells is mediated by VLA-4/VCAM-1 pathway.
Collapse
Affiliation(s)
- H Shimada
- First Department of Internal Medicine, Nagasaki University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- T H Piela-Smith
- Section of Rheumatology, Boston University School of Medicine, Massachusetts
| | | |
Collapse
|
49
|
Affiliation(s)
- D M Fagundus
- Department of Medicine, Medical University of South Carolina, Charleston 29425-2229
| | | |
Collapse
|
50
|
Saita N, Sakata KM, Matsumoto M, Iyonaga K, Ando M, Adachi M, Hirashima M. Production of fibroblast proliferative cytokines from T lymphocytes stimulated by a B cell lymphoma line and their functional heterogeneity. Immunol Lett 1994; 41:279-86. [PMID: 8002051 DOI: 10.1016/0165-2478(94)90146-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Human mononuclear leukocytes (MNL) produced several factors with fibroblast proliferation activity (FPA) for HFL-1, a human lung fibroblast cell line, when MNL were cocultured with irradiated BALL-1, a B cell lymphoma line (BCLL), but not with other BCLL. The cellular source of BALL-1-induced FPA seemed to be CD4-positive T lymphocytes. On isoelectric electrophoresis, major activity of BALL-1-induced FPA was detected in the fractions around pH 4-5, and minor activity was present in the fractions around pH 6-7. Major BALL-1-induced FPA consisted of at least 4 different fibroblast proliferation factors (FPFs) according to their molecular weight; 320-600 kDa (P-I), 50-110 kDa (P-II), 22-38 kDa (P-III) and 4.6-11 kDa (P-IV). P-I had affinity to heparin though the rest had little or no affinity. FPA of P-I was suppressed by an antibody against acidic FGF, and FPA of P-III was suppressed by an antibody against IL-6. On the other hand, FPA of P-II and P-IV was suppressed by none of the antibodies against cytokines with FPA, such as FGF, IL-4, IL-6, IFN-gamma, TGF-beta and TNF-alpha. It was thus suggested that P-I was acidic FGF, that P-III was IL-6, and that P-II and P-IV were different cytokines from those described above. Furthermore, it was found that P-II and P-IV failed to exhibit proliferation activity for human umbilical vein endothelial cells (HUVEC).(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- N Saita
- Department of Internal Medicine, Kumamoto University School of Medicine, Takasaki, Japan
| | | | | | | | | | | | | |
Collapse
|