1
|
Torres-Machorro AL, Becerril C, Hernández-Plata E, Luis-García ER, Maldonado M, Herrera I, Negreros M, Hernández-Sánchez F, Mendoza-Milla C, Gaxiola M, Ramírez R, Pardo A, Buendía-Roldán I, Selman M, Cisneros J. Altered expression pattern of immune response-related genes and isoforms in hypersensitivity pneumonitis lung fibroblasts. Sci Rep 2024; 14:24002. [PMID: 39402115 PMCID: PMC11473681 DOI: 10.1038/s41598-024-74267-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/24/2024] [Indexed: 10/17/2024] Open
Abstract
Hypersensitivity pneumonitis (HP) is an immune-mediated inflammatory interstitial lung disease that may evolve to pulmonary fibrosis, a progressive disorder with a poor prognosis characterized by fibroblast activation and extracellular matrix accumulation. In HP lung fibroblasts, the gene expression of proteins involved in the interaction with the immune response, their isoforms, and how they influence their phenotype have yet to be elucidated. We analyzed the expression and splicing variants of 16 target genes involved in the interaction between HP fibroblasts and immune signaling and evaluated possible correlations with clinical data. The comparison of HP and control fibroblasts revealed distinct gene expression patterns. HP lung fibroblasts displayed an increased expression of IFI27 and PDFGRA and a downregulation of IL17RC and TGFBR3. IFI27 immunoreactive protein was markedly increased in HP lung tissues and normal fibroblasts treated with TGF-β. Furthermore, IFI27 overexpression in normal fibroblasts increased α-SMA and decreased cell number over time. The isoform analysis showed similar expression patterns for most genes, except for the AGER receptor with increased soluble variants relative to full-length AGER in HP fibroblasts. These findings indicate important differences in the expression of genes related to the immune response by HP fibroblasts, highlighting their unique characteristics and providing further insight into a possible profibrotic role of IFI27 in the disease.
Collapse
Affiliation(s)
- Ana Lilia Torres-Machorro
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Carina Becerril
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Everardo Hernández-Plata
- Investigador Por México, Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCyT), and Instituto Nacional de Medicina Genómica, 14610, Ciudad de México, México
| | - Erika Rubí Luis-García
- Laboratorio de Biología Celular, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Mariel Maldonado
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Iliana Herrera
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Miguel Negreros
- Clínica de Vasculitis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Fernando Hernández-Sánchez
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Criselda Mendoza-Milla
- Laboratorio de Transducción de Señales, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Miguel Gaxiola
- Laboratorio de Morfología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Remedios Ramírez
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Annie Pardo
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Ciudad de México, México
| | - Ivette Buendía-Roldán
- Laboratorio de Investigación Traslacional en Envejecimiento y Enfermedades Fibrosantes, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - Moisés Selman
- Laboratorio de Biopatología Pulmonar INER-Ciencias-UNAM, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México
| | - José Cisneros
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, 14080, Ciudad de México, México.
| |
Collapse
|
2
|
Sharma J, Mudalagiriyappa S, Abdelaal HFM, Kelly TC, Choi W, Ponnuraj N, Vieson MD, Talaat AM, Nanjappa SG. E3 ubiquitin ligase CBLB regulates innate immune responses and bacterial dissemination during nontuberculous mycobacteria infection. J Leukoc Biol 2024; 115:1118-1130. [PMID: 38271280 PMCID: PMC11135617 DOI: 10.1093/jleuko/qiae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/27/2023] [Accepted: 12/20/2023] [Indexed: 01/27/2024] Open
Abstract
Nontuberculous mycobacteria (NTM) are emerging opportunistic pathogens causing pulmonary infection to fatal disseminated disease. NTM infections are steadily increasing in children and adults, and immune-compromised individuals are at a greater risk of fatal infections. The NTM disease's adverse pathology and resistance to antibiotics have further worsened the therapeutic measures. Innate immune regulators are potential targets for therapeutics to NTM, especially in a T cell-suppressed population, and many ubiquitin ligases modulate pathogenesis and innate immunity during infections, including mycobacterial infections. Here, we investigated the role of an E3 ubiquitin ligase, Casitas B-lineage lymphoma proto-oncogene B (CBLB), in immunocompromised mouse models of NTM infection. We found that CBLB is essential to prevent bacterial growth and dissemination. Cblb deficiency debilitated natural killer cells, inflammatory monocytes, and macrophages in vivo. However, Cblb deficiency in macrophages did not wane its ability to inhibit bacterial growth or production of reactive oxygen species or interferon γ production by natural killer cells in vitro. CBLB restricted NTM growth and dissemination by promoting early granuloma formation in vivo. Our study shows that CBLB bolsters innate immune responses and helps prevent the dissemination of NTM during compromised T cell immunity.
Collapse
Affiliation(s)
- Jaishree Sharma
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| | - Srinivasu Mudalagiriyappa
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| | - Hazem F M Abdelaal
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI 53706, United States
| | - Thomas C Kelly
- Integrative Biology Honors Program, University Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Woosuk Choi
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| | - Nagendraprabhu Ponnuraj
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| | - Miranda D Vieson
- Veterinary Diagnostic Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| | - Adel M Talaat
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI 53706, United States
| | - Som Gowda Nanjappa
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL 61802, United States
| |
Collapse
|
3
|
Kim HY, Jeong D, Kim JH, Chung DH. Innate Type-2 Cytokines: From Immune Regulation to Therapeutic Targets. Immune Netw 2024; 24:e6. [PMID: 38455467 PMCID: PMC10917574 DOI: 10.4110/in.2024.24.e6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 03/09/2024] Open
Abstract
The intricate role of innate type-2 cytokines in immune responses is increasingly acknowledged for its dual nature, encompassing both protective and pathogenic dimensions. Ranging from defense against parasitic infections to contributing to inflammatory diseases like asthma, fibrosis, and obesity, these cytokines intricately engage with various innate immune cells. This review meticulously explores the cellular origins of innate type-2 cytokines and their intricate interactions, shedding light on factors that amplify the innate type-2 response, including TSLP, IL-25, and IL-33. Recent advancements in therapeutic strategies, specifically the utilization of biologics targeting pivotal cytokines (IL-4, IL-5, and IL-13), are discussed, offering insights into both challenges and opportunities. Acknowledging the pivotal role of innate type-2 cytokines in orchestrating immune responses positions them as promising therapeutic targets. The evolving landscape of research and development in this field not only propels immunological knowledge forward but also holds the promise of more effective treatments in the future.
Collapse
Affiliation(s)
- Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Korea
| | - Dongjin Jeong
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Ji Hyung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Doo Hyun Chung
- Laboratory of Immune Regulation, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul 03080, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
4
|
Yuan X, Rong Y, Chen Y, Ren C, Meng Y, Mu Y, Chen X. Molecular characterization, expression analysis and cellular location of IL-4/13 receptors in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2022; 120:45-55. [PMID: 34774733 DOI: 10.1016/j.fsi.2021.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/07/2021] [Accepted: 11/07/2021] [Indexed: 06/13/2023]
Abstract
Interleukin (IL)-4 and IL-13 are closely related class I cytokines that play key roles in the T helper (Th)-2 immune response via heterodimeric receptors. IL-4 signals via both the type I (IL-4Rα/γc) and type II (IL-4Rα/IL-13Rα1) receptor complexes, while IL-13 signals only via the type II receptor complex. IL-13Rα2 is traditionally considered a "decoy" receptor for IL-13. However, the IL-4/13 system and its response to pathogenic infection are still not fully understood in fish. In this study, we identified four IL-4/13 receptor subunit genes in the large yellow croaker (Larimichthys crocea): LcIL-4Rα1, LcIL-4Rα2, LcIL-13Rα1, and LcIL-13Rα2. Sequence analysis showed that these receptors possessed typical characteristic domains, including a signal peptide, two fibronectin type III (FN III)-like domains, and a transmembrane domain, but their cytoplasmic regions were not well conserved. The mRNA and protein of the four IL-4/13 receptors were constitutively expressed in all examined tissues of large yellow croaker. Their mRNAs were also detected in primary head kidney macrophages (PKMs), primary head kidney granulocytes (PKGs), and primary head kidney lymphocytes (PKLs). Immunofluorescence assay further showed that LcIL-4Rα and LcIL-13Rα1 were expressed on the membrane of IgM + B cells. After stimulation by Vibrio alginolyticus and poly (I:C) (a viral dsRNA mimic), the mRNA levels of LcIL-4/13 receptors were significantly upregulated in the head kidney and spleen. Their mRNA levels were also upregulated in head kidney leukocytes in response to poly (I:C) and lipopolysaccharide (LPS) treatment. Moreover, both recombinant LcIL-4/13A and LcIL-4/13B upregulated LcIL-4Rα1 and LcIL-4Rα2 in primary leukocytes, but only recombinant LcIL-4/13A upregulated LcIL-13Rα1 and LcIL-13Rα2. These results indicated that LcIL-4/13 receptors, containing conserved functional domains, may be involved in the IL-4/13-mediated immune response to pathogenic infections in the large yellow croaker.
Collapse
Affiliation(s)
- Xiaoqin Yuan
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yi Rong
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - You Chen
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Chaoqun Ren
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yufan Meng
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yinnan Mu
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| | - Xinhua Chen
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, College of Life Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
5
|
Masamba P, Kappo AP. Immunological and Biochemical Interplay between Cytokines, Oxidative Stress and Schistosomiasis. Int J Mol Sci 2021; 22:ijms22137216. [PMID: 34281269 PMCID: PMC8268096 DOI: 10.3390/ijms22137216] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/20/2021] [Accepted: 06/20/2021] [Indexed: 12/17/2022] Open
Abstract
The host–parasite schistosome relationship relies heavily on the interplay between the strategies imposed by the schistosome worm and the defense mechanisms the host uses to counter the line of attack of the parasite. The ultimate goal of the schistosome parasite entails five important steps: evade elimination tactics, survive within the human host, develop into adult forms, propagate in large numbers, and transmit from one host to the next. The aim of the parasitized host on the other hand is either to cure or limit infection. Therefore, it is a battle between two conflicting aspirations. From the host’s standpoint, infection accompanies a plethora of immunological consequences; some are set in place to defend the host, while most end up promoting chronic disease, which ultimately crosses paths with oxidative stress and cancer. Understanding these networks provides attractive opportunities for anti-schistosome therapeutic development. Hence, this review discusses the mechanisms by which schistosomes modulate the human immune response with ultimate links to oxidative stress and genetic instability.
Collapse
|
6
|
Cho MK, Park JG, Iwata H, Kim EY. 2,3,7,8-Tetrachlorodibenzo-p-dioxin prompted differentiation to CD4 +CD8 -CD25 + and CD4 +CD8 +CD25 + Tregs and altered expression of immune-related genes in the thymus of chicken embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 211:111947. [PMID: 33503546 DOI: 10.1016/j.ecoenv.2021.111947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/10/2021] [Accepted: 01/14/2021] [Indexed: 06/12/2023]
Abstract
The chicken (Gallus gallus), which has three aryl hydrocarbon receptor (AHR) isoforms (ckAHR1, ckAHR2, and ckAHR1β) and two AHR nuclear translocator (ARNT) isoforms (ckARNT1 and ckARNT2), is highly sensitive to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and can serve as an avian model to gain an understanding of the mechanism underlying dioxin toxicity. To elucidate the mechanism of TCDD-induced immunotoxicity in avian species, we treated chicken embryos in ovo with graded concentrations of TCDD (1.5, 2.5, 3.0, 3.3, 3.5, and 4.0 μM). Initially, we measured mRNA expression levels of ckAHR and ckARNT isoforms and analyzed the T cell populations and transcriptome in the thymuses of TCDD-treated chicken embryos. Quantitative polymerase chain reaction analysis revealed that mRNA expressions of ckAHR1 and ckARNT2 were dominant in the thymus. Severe weight loss and thymus atrophy were observed in the TCDD-treated embryos. Immunophenotyping analyses demonstrated significant increases in CD4+CD8-CD25+ and CD4+CD8+CD25+ regulatory T cells (Tregs) populations following TCDD exposure, suggesting that TCDD suppresses T cell-mediated immune responses in chicken embryos. In addition, thymic transcriptome analyses intimated that alteration of the signaling pathways related to erb-b2 receptor tyrosine kinase 4 (ERBB4) and wnt family member 5A (WNT5A), and bone morphogenetic protein (BMP) may be associated with the TCDD-induced thymus atrophy. We also observed significantly altered expression levels of genes including interleukine 13 receptor subunit alpha 2 (IL13RA2), transforming growth factor beta 1 (TGFβ1), collagen type III alpha 1 chain (COL3A1), and collagen type IX alpha 3 chain (COL9A3), implying immunosuppression, fibrosis development, and collagen deposition. Collectively, these findings suggest that TCDD exposure activates the ckAHR1-ckARNT2 signaling pathway and suppresses immune responses through the prompted differentiation to CD4+CD8-CD25+ and CD4+CD8+CD25+ Tregs and altered expressions of immune-related genes in the thymus of chicken embryos.
Collapse
Affiliation(s)
- Min-Kyung Cho
- Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Jae-Gon Park
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea
| | - Hisato Iwata
- Center for Marine Environmental Studies (CMES), Ehime University, Matsuyama 790-8577, Japan
| | - Eun-Young Kim
- Department of Biology, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea; Department of Life and Nanopharmaceutical Science, Kyung Hee University, Hoegi-Dong, Dongdaemun-Gu, Seoul 130-701, Republic of Korea.
| |
Collapse
|
7
|
Bitton A, Avlas S, Reichman H, Itan M, Karo-Atar D, Azouz NP, Rozenberg P, Diesendruck Y, Nahary L, Rothenberg ME, Benhar I, Munitz A. A key role for IL-13 signaling via the type 2 IL-4 receptor in experimental atopic dermatitis. Sci Immunol 2020; 5:5/44/eaaw2938. [PMID: 32060143 DOI: 10.1126/sciimmunol.aaw2938] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 10/06/2019] [Accepted: 01/09/2020] [Indexed: 12/21/2022]
Abstract
IL-13 and IL-4 are potent mediators of type 2-associated inflammation such as those found in atopic dermatitis (AD). IL-4 shares overlapping biological functions with IL-13, a finding that is mainly explained by their ability to signal via the type 2 IL-4 receptor (R), which is composed of IL-4Rα in association with IL-13Rα1. Nonetheless, the role of the type 2 IL-4R in AD remains to be clearly defined. Induction of two distinct models of experimental AD in Il13ra1 -/- mice, which lack the type 2 IL-4R, revealed that dermatitis, including ear and epidermal thickening, was dependent on type 2 IL-4R signaling. Expression of TNF-α was dependent on the type 2 IL-4R, whereas induction of IL-4, IgE, CCL24, and skin eosinophilia was dependent on the type 1 IL-4R. Neutralization of IL-4, IL-13, and TNF-α as well as studies in bone marrow-chimeric mice revealed that dermatitis, TNF-α, CXCL1, and CCL11 expression were exclusively mediated by IL-13 signaling via the type 2 IL-4R expressed by nonhematopoietic cells. Conversely, induction of IL-4, CCL24, and eosinophilia was dependent on IL-4 signaling via the type 1 IL-4R expressed by hematopoietic cells. Last, we pharmacologically targeted IL-13Rα1 and established a proof of concept for therapeutic targeting of this pathway in AD. Our data provide mechanistic insight into the differential roles of IL-4, IL-13, and their receptor components in allergic skin and highlight type 2 IL-4R as a potential therapeutic target in AD and other allergic diseases such as asthma and eosinophilic esophagitis.
Collapse
Affiliation(s)
- Almog Bitton
- School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel.,Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Shmuel Avlas
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Hadar Reichman
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Michal Itan
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Danielle Karo-Atar
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Nurit P Azouz
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Perri Rozenberg
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Yael Diesendruck
- School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Limor Nahary
- School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel
| | - Marc E Rothenberg
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Itai Benhar
- School of Molecular Cell Biology and Biotechnology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel.
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, 69978 Tel Aviv, Israel.
| |
Collapse
|
8
|
Ayelign B, Akalu Y, Teferi B, Molla MD, Shibabaw T. Helminth Induced Immunoregulation and Novel Therapeutic Avenue of Allergy. J Asthma Allergy 2020; 13:439-451. [PMID: 33116652 PMCID: PMC7548329 DOI: 10.2147/jaa.s273556] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/17/2020] [Indexed: 12/24/2022] Open
Abstract
Allergic diseases are increasing at an alarming rate worldwide, particularly in developed countries. In contrast, there is a decrease in the prevalence of helminthic infections and other neglected diseases. The hygiene hypothesis elaborates parasitic infection, and allergy-associated diseases have an inverse relationship. Acute helminthic infection and allergic reaction stimulate Type 2 helper cells (Th2) immune response with up-regulation of cytokines IL-4-, IL-5-, and IL-13-mediated IgE and mast cell production, as well as eosinophilia. However, people who chronically suffer from helminthic infections are demarcated through polarized Th2 resulting in alternative macrophage activation and T regulatory response. This regulatory system reduces allergy incidence in individuals that are chronically diseased through helminth. As a result, the excretory-secretory (ES) substance derived from parasites and extracellular vesicular components can be used as a novel therapeutic modality of allergy. Therefore, the aim of this review meticulously explored the link between helminth infection and allergy, and utilization of the helminth secretome for therapeutic immunomodulation.
Collapse
Affiliation(s)
- Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Yonas Akalu
- Department of Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Banchamlak Teferi
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
9
|
Furue M, Ulzii D, Nakahara T, Tsuji G, Furue K, Hashimoto-Hachiya A, Kido-Nakahara M. Implications of IL-13Rα2 in atopic skin inflammation. Allergol Int 2020; 69:412-416. [PMID: 32037147 DOI: 10.1016/j.alit.2020.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/16/2022] Open
Abstract
Atopic dermatitis (AD) is a common eczematous skin disorder characterized by skin inflammation, barrier disruption, chronic pruritus and marked scratching. Th2 cytokines, especially IL-13, play a pathogenic role in AD. IL-13 signals via a heterodimeric receptor composed of IL-4Rα and IL-13 Rα1. A second receptor, IL-13 Rα2, binds to IL-13 with high affinity, but it works as a decoy receptor. IL-13 Rα2 is overexpressed in the lesional skin of AD. Notably, mechanical scratching, as well as IL-13 itself, also upregulates IL-13 Rα2 expression. The scratch-induced IL-13 Rα2 upregulation may attenuate the IL-13-mediated epidermal barrier dysfunction and dermal fibrosis. Recent studies stress an importance of another IL-13 Rα2 ligand, chitinase 3-like 1 or YKL-40 in Th2 differentiation. However, the implications of increased IL-13 Rα2 levels remain elusive in AD. In this review, we summarize the recent topics on IL-13 Rα2 in atopic skin inflammation.
Collapse
|
10
|
Lokau J, Garbers C. Biological functions and therapeutic opportunities of soluble cytokine receptors. Cytokine Growth Factor Rev 2020; 55:94-108. [PMID: 32386776 DOI: 10.1016/j.cytogfr.2020.04.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/09/2020] [Indexed: 12/28/2022]
Abstract
Cytokines control the immune system by regulating the proliferation, differentiation and function of immune cells. They activate their target cells through binding to specific receptors, which either are transmembrane proteins or attached to the cell-surface via a GPI-anchor. Different tissues and individual cell types have unique expression profiles of cytokine receptors, and consequently this expression pattern dictates to which cytokines a given cell can respond. Furthermore, soluble variants of several cytokine receptors exist, which are generated by different molecular mechanisms, namely differential mRNA splicing, proteolytic cleavage of the membrane-tethered precursors, and release on extracellular vesicles. These soluble receptors shape the function of cytokines in different ways: they can serve as antagonistic decoy receptors which compete with their membrane-bound counterparts for the ligand, or they can form functional receptor/cytokine complexes which act as agonists and can even activate cells that would usually not respond to the ligand alone. In this review, we focus on the IL-2 and IL-6 families of cytokines and the so-called Th2 cytokines. We summarize for each cytokine which soluble receptors exist, were they originate from, how they are generated, and what their biological functions are. Furthermore, we give an outlook on how these soluble receptors can be exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Juliane Lokau
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany.
| |
Collapse
|
11
|
Sequeida A, Castillo A, Cordero N, Wong V, Montero R, Vergara C, Valenzuela B, Vargas D, Valdés N, Morales J, Tello M, Sandino AM, Maisey K, Imarai M. The Atlantic salmon interleukin 4/13 receptor family: Structure, tissue distribution and modulation of gene expression. FISH & SHELLFISH IMMUNOLOGY 2020; 98:773-787. [PMID: 31734286 DOI: 10.1016/j.fsi.2019.11.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 06/10/2023]
Abstract
Interleukin (IL)-4 and IL-13 play a central role in T helper 2 immune response in mammals. The cell signalling is mediated by the type I heterodimeric receptor containing the IL-4Rα and γC chains, and the type II receptors formed by IL-4Rα and IL-13Rα1. In salmonid species, three paralogues of the IL-4 and IL-13 cytokines have been reported, il-4/13a, il-4/13b1 and il-4/13b2. In regard to receptors, two paralogues of each IL-4/13 receptor chains have been identified in rainbow trout while five genes named γc1, il-4rα, il-13rα1a, il-13rα1b, and il-13rα2 have identified in Atlantic salmon. Since Atlantic salmon is an important farmed fish species, the aim of this work was to get new insights into distribution, structure and expression regulation of the IL-4/13 receptors in salmon. By using qRT-PCR, it was shown that all γc1, il-4rα, il-13rα1a, il-13rα1b, and il-13rα2 receptor chains were expressed in lymphoid and non-lymphoid tissues of healthy salmon, nonetheless γC expression was higher in lymphoid than non-lymphoid tissues. The in silico structural analysis and homology modelling of the predicted receptor proteins showed that domains and most motifs present in the superior vertebrate chains are conserved in salmon suggesting a conserved role for these receptor chains. Only IL-13Rα1B is a receptor chain with a unique structure that seem not to be present in higher vertebrates but in fish species. In order to determine the regulatory role of IL-4/13 on the expression of receptor chains, Atlantic salmon il-4/13A gene was synthetized and cloned in pET15b. The recombinant IL-4/13A was produced in E. coli and the activity of the purified cytokine was confirmed in vitro. The regulatory role of IL-4/13A on the expression of their potential receptors was tested in salmon receiving the recombinant cytokine and effects were compared with those of the control group. The results showed that IL-4/13A induced the expression of its own gene and GATA-3, in the head kidney of fish but not in the spleen, while IL-10 increased in both lymphoid organs indicating a regulatory role of this cytokine on the induction of Th2 responses in salmon. IFN-γ and MHC class II were also later induced in head kidney. In regard to the expression of the receptor chains, IL-4/13A upregulated the expression of γC, IL-13Rα1A and IL-13Rα2A in the spleen but not in the head kidney of salmon, indicating a role on the modulation of cell signalling for the Th2 response. Furthermore, Piscirickettsia salmonis infection of Atlantic salmon occurred with an increase of γC and IL-13Rα1A suggesting a potential role of the IL-4/13 system in bacterial immunity or pathogenesis. This study contributes to a better understanding of the IL-4/13A system in salmon, which as a key axis for Th2 response may be involved not only in pathogen elimination but also in adaptive immune repair that seems critical tolerance to infectious diseases.
Collapse
Affiliation(s)
- Alvaro Sequeida
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile.
| | - Andrés Castillo
- Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Natalia Cordero
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Valentina Wong
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile.
| | - Ruth Montero
- Laboratory of Comparative Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Laboratory for Comparative Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Claudio Vergara
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Beatriz Valenzuela
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Deborah Vargas
- Laboratory of Virology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Natalia Valdés
- Laboratory of Metagenomics, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile.
| | - Jonathan Morales
- Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Mario Tello
- Laboratory of Metagenomics, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Ana María Sandino
- Laboratory of Virology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| | - Kevin Maisey
- Laboratory of Comparative Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile.
| | - Mónica Imarai
- Laboratory of Immunology, Centre of Aquatic Biotechnology, Department of Biology, Faculty of Chemistry and Biology, University of Santiago of Chile, Av. Bernardo O'Higgins, 3363, Santiago, Chile; Consorcio Tecnológico de Sanidad Acuícola ICTIO Biotechnologies, Federal Research Institute for Animal Health, Institute of Immunology, Südufer 10, 17493, Graeifswald-Insel Riems, Germany.
| |
Collapse
|
12
|
Bhattacharjee S, Mejías-Luque R, Loffredo-Verde E, Toska A, Flossdorf M, Gerhard M, Prazeres da Costa C. Concomitant Infection of S. mansoni and H. pylori Promotes Promiscuity of Antigen-Experienced Cells and Primes the Liver for a Lower Fibrotic Response. Cell Rep 2019; 28:231-244.e5. [DOI: 10.1016/j.celrep.2019.05.108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/29/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
|
13
|
Verstockt B, Perrier C, De Hertogh G, Cremer J, Creyns B, Van Assche G, Ferrante M, Ceuppens JL, Vermeire S, Breynaert C. Effects of Epithelial IL-13Rα2 Expression in Inflammatory Bowel Disease. Front Immunol 2018; 9:2983. [PMID: 30619339 PMCID: PMC6305625 DOI: 10.3389/fimmu.2018.02983] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/04/2018] [Indexed: 12/21/2022] Open
Abstract
Background: Mucosal IL-13 Receptor alpha 2 (IL13RA2) mRNA expression is one of the best predictive markers for primary non-responsiveness to infliximab therapy in patients with inflammatory bowel disease (IBD). The objective of this study was to understand how IL-13Rα2, a negative regulator of IL-13 signaling, can contribute to IBD pathology. Methods:IL13RA2 knockout (KO) and wild type (WT) mice were exposed to dextran sodium sulfate (DSS) in drinking water to induce colitis. Furthermore, mucosal biopsies and resection specimen of healthy individuals and IBD patients before the start of anti-tumor necrosis factor (anti-TNF) therapy were obtained for immunohistochemistry and gene expression analysis. Results: After induction of DSS colitis, IL13RA2 KO mice had similar disease severity, but recovered more rapidly than WT animals. Goblet cell numbers and mucosal architecture were also more rapidly restored in IL13RA2 KO mice. In mucosal biopsies of active IBD patients, immunohistochemistry revealed that IL-13Rα2 protein was highly expressed in epithelial cells, while expression was restricted to goblet cells in healthy controls. Mucosal IL13RA2 mRNA negatively correlated with mRNA of several goblet cell-specific and barrier genes, and with goblet cell numbers. Conclusions: The data suggest that IL-13Rα2 on epithelial cells contributes to IBD pathology by negatively influencing goblet cell recovery, goblet cell function and epithelial restoration after injury. Therefore, blocking IL-13Rα2 could be a promising target for restoration of the epithelial barrier in IBD.
Collapse
Affiliation(s)
- Bram Verstockt
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research for Gastrointestinal Disorders, Leuven, Belgium
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Clémentine Perrier
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research for Gastrointestinal Disorders, Leuven, Belgium
- KU Leuven, Laboratory of Clinical Immunology, Department of Microbiology and Immunology, Leuven, Belgium
| | - Gert De Hertogh
- KU Leuven, Department of Imaging & Pathology, Translational Cell & Tissue Research, Leuven, Belgium
| | - Jonathan Cremer
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research for Gastrointestinal Disorders, Leuven, Belgium
- KU Leuven, Laboratory of Clinical Immunology, Department of Microbiology and Immunology, Leuven, Belgium
| | - Brecht Creyns
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research for Gastrointestinal Disorders, Leuven, Belgium
- KU Leuven, Laboratory of Clinical Immunology, Department of Microbiology and Immunology, Leuven, Belgium
| | - Gert Van Assche
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research for Gastrointestinal Disorders, Leuven, Belgium
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Marc Ferrante
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research for Gastrointestinal Disorders, Leuven, Belgium
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Jan L. Ceuppens
- KU Leuven, Laboratory of Clinical Immunology, Department of Microbiology and Immunology, Leuven, Belgium
| | - Séverine Vermeire
- KU Leuven, Department of Chronic Diseases, Metabolism and Ageing, Translational Research for Gastrointestinal Disorders, Leuven, Belgium
- University Hospitals Leuven, Department of Gastroenterology and Hepatology, Leuven, Belgium
| | - Christine Breynaert
- KU Leuven, Laboratory of Clinical Immunology, Department of Microbiology and Immunology, Leuven, Belgium
| |
Collapse
|
14
|
MicroRNA-96 Promotes Schistosomiasis Hepatic Fibrosis in Mice by Suppressing Smad7. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:73-82. [PMID: 30406154 PMCID: PMC6214875 DOI: 10.1016/j.omtm.2018.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/05/2018] [Indexed: 01/05/2023]
Abstract
Infection with Schistosoma causes aberrant expression of host microRNAs (miRNAs), and normalizing the levels of dysregulated miRNAs can attenuate pathology. Here, we show that the host miRNA, miR-96, is markedly upregulated during the progression of hepatic schistosomiasis. We demonstrate that elevation of miR-96 induces hepatic fibrosis in infected mice by suppressing the expression of its target gene, Smad7. We show that infection with Schistosoma induces the expression of transforming growth factor β1 (TGF-β1), which in turn upregulates the expression of miR-96 through SMAD2/3-DROSHA-mediated post-transcriptional regulation. Furthermore, inhibition of miR-96 with recombinant adeno-associated virus 8 (rAAV8)-mediated delivery of Tough Decoy RNAs in mice attenuated hepatic fibrosis and prevented lethality following schistosome infection. Taken together, our data highlight the potential for rAAV8-mediated inhibition of miR-96 as a therapeutic strategy to treat hepatic schistosomiasis.
Collapse
|
15
|
Serum level of interleukin-13 receptor alpha 2 in infants with biliary atresia - is it of value? Clin Exp Hepatol 2018; 4:91-96. [PMID: 29904725 PMCID: PMC6000744 DOI: 10.5114/ceh.2018.75958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/15/2018] [Indexed: 11/23/2022] Open
Abstract
Aim of the study We aimed to assess the utility of serum level IL-13Rα2 receptors as a non-invasive marker for early diagnosis of biliary atresia (BA) and selection of BA patients indicated for Kasai portoenterostomy. Material and methods The study included 60 infants with neonatal cholestasis in three groups; early BA group (n = 20), delayed BA group (n = 20) and non-BA cholestasis group (n = 20). A fourth group of 20 healthy neonates (n = 20) served as controls. IL-13Rα2 was measured by enzyme-linked immunosorbent assay in all patients and controls. Results The mean value of IL-13Rα2 was significantly higher in delayed BA group (11.05 ± 10.9 ng/ml) compared to early BA (0.34 ± 0.37 ng/ml), non-BA (0.54 ± 0.85 ng/ml) and control (0.24-0.2 ng/ml) groups. The levels of serum IL-13Rα2 increase with the severity of the degree of fibrosis. IL-13Rα2 at a cutoff level > 0.782 ng/ml could predict late fibrosis with accuracy of 77.55% (p < 0.0001). IL-13Rα2 could differentiate between preserved and disturbed liver architecture at a cut off value of more than 0.42 ng/ml with an accuracy of 81.6%. Conclusions Serum IL-13Rα2 not a diagnostic marker for BA however it could be used as a noninvasive marker for detection of advanced liver fibrosis and presence of disturbed liver architecture that helps in patient selection for undergoing Kasai operation. Serum IL-13Rα2 could be a future therapeutic target for management of BA patients and any fibrotic liver disease.
Collapse
|
16
|
Moyo D, Beattie L, Andrews PS, Moore JWJ, Timmis J, Sawtell A, Hoehme S, Sampson AT, Kaye PM. Macrophage Transactivation for Chemokine Production Identified as a Negative Regulator of Granulomatous Inflammation Using Agent-Based Modeling. Front Immunol 2018; 9:637. [PMID: 29636754 PMCID: PMC5880939 DOI: 10.3389/fimmu.2018.00637] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/14/2018] [Indexed: 01/22/2023] Open
Abstract
Cellular activation in trans by interferons, cytokines, and chemokines is a commonly recognized mechanism to amplify immune effector function and limit pathogen spread. However, an optimal host response also requires that collateral damage associated with inflammation is limited. This may be particularly so in the case of granulomatous inflammation, where an excessive number and/or excessively florid granulomas can have significant pathological consequences. Here, we have combined transcriptomics, agent-based modeling, and in vivo experimental approaches to study constraints on hepatic granuloma formation in a murine model of experimental leishmaniasis. We demonstrate that chemokine production by non-infected Kupffer cells in the Leishmania donovani-infected liver promotes competition with infected KCs for available iNKT cells, ultimately inhibiting the extent of granulomatous inflammation. We propose trans-activation for chemokine production as a novel broadly applicable mechanism that may operate early in infection to limit excessive focal inflammation.
Collapse
Affiliation(s)
- Daniel Moyo
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom.,Department of Computer Science, University of York, York, United Kingdom
| | - Lynette Beattie
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Paul S Andrews
- Department of Electronics, University of York, York, United Kingdom.,SimOmics Ltd., York, United Kingdom
| | - John W J Moore
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Jon Timmis
- Department of Electronics, University of York, York, United Kingdom.,SimOmics Ltd., York, United Kingdom
| | - Amy Sawtell
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Stefan Hoehme
- Institute for Computer Science, University of Leipzig, Leipzig, Germany
| | - Adam T Sampson
- Division of Computing and Mathematics, Abertay University, Dundee, United Kingdom
| | - Paul M Kaye
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| |
Collapse
|
17
|
Tricellulin is regulated via interleukin-13-receptor α2, affects macromolecule uptake, and is decreased in ulcerative colitis. Mucosal Immunol 2018; 11:345-356. [PMID: 28612843 PMCID: PMC5730503 DOI: 10.1038/mi.2017.52] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 05/01/2017] [Indexed: 02/04/2023]
Abstract
In the two inflammatory bowel diseases, ulcerative colitis (UC) and Crohn's disease (CD), altered expression of tight junction (TJ) proteins leads to an impaired epithelial barrier including increased uptake of luminal antigens supporting the inflammation. Here, we focused on regulation of tricellulin (Tric), a protein of the tricellular TJ essential for the barrier against macromolecules, and hypothesized a role in paracellular antigen uptake. We report that Tric is downregulated in UC, but not in CD, and that its reduction increases the passage of macromolecules. Using a novel visualization method, passage sites were identified at TJ regions usually sealed by Tric. We show that interleukin-13 (IL-13), beyond its known effect on claudin-2, downregulates Tric expression. These two effects of IL-13 are regulated by different signaling pathways: The IL-13 receptor α1 upregulates claudin-2, whereas IL-13 receptor α2 downregulates Tric. We suggest to target the α2 receptor in future developments of therapeutical IL-13-based biologicals.
Collapse
|
18
|
IL-13 -1112 polymorphism and periodontitis susceptibility: a meta-analysis. BMC Oral Health 2018; 18:21. [PMID: 29415708 PMCID: PMC5803991 DOI: 10.1186/s12903-018-0481-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 01/29/2018] [Indexed: 01/13/2023] Open
Abstract
Background Several studies have examined the association between the IL-13 -1112C/T polymorphism and the risk of periodontitis. However, these studies have reached different conclusions. The aim of the current study was to investigate the link between this IL-13 -1112 polymorphism and susceptibility to periodontitis. Methods We utilized electronic databases, including the CNKI (China National Knowledge Infrastructure), Wanfang, PubMed, Embase, and Cochrane Library databases, to manually search for relevant research published through November 30, 2016. The Chinese and English terms used to search the literature included “periodontitis”, “periodontal disease”, “IL 13”, “IL-13”, and “interleukin-13”. In accordance with our inclusion criteria, we selected studies that involved case-control trials. All of these case-control trials described their objectives, design and specific statistical methods. For all included studies, odds ratios (ORs) and 95% confidence intervals (95% CIs) were provided or could be calculated from the study data. The quality of the included literature was evaluated using the Newcastle-Ottawa scale (NOS). STATA 12.0 was used to calculate the sizes of the combined effects and conduct a sensitivity analysis of the results. Results Our meta-analysis included 4 articles representing 5 case-control studies with a total of 710 cases and 671 control subjects. The meta-analysis results indicated that the CC vs TT model, CT vs TT model and TT vs CT + CC model (CC VS TT: OR = 0.615, 95% CI = 0.395–0.957; CT vs TT: OR = 0.518, 95% CI = 0.323–0.830; and TT vs CT + CC: OR = 1.739, 95% CI = 1.130–2.676) were significant in five IL-13 -1112 gene polymorphism and periodontitis susceptibility models. Subgroup analysis indicated that the CC vs TT, CT vs TT and TT vs CT + CC models were significant in the chronic periodontitis (CP) group, whereas no significant differences were found in the five aggressive periodontitis (AgP) group models. The sensitivity analysis showed that dropping any single study did not affect the pooled analysis results. Conclusion The IL-13 -1112 polymorphism may be associated with susceptibility to periodontitis. The IL-13 -1112 gene polymorphism may be associated with susceptibility to CP but not to AgP. Thus, large-scale, multi-ethnic case-control trials are still warranted.
Collapse
|
19
|
Yang Y, He JJ, Hu S, Chang H, Xiang X, Yang JF, Zou FC. Comparative Study of Transcriptome Profiles of Mouse Livers and Skins Infected by Fork-Tailed or Non-Fork-Tailed Schistosoma japonicum. Front Microbiol 2017; 8:1648. [PMID: 28912762 PMCID: PMC5582157 DOI: 10.3389/fmicb.2017.01648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 08/15/2017] [Indexed: 12/29/2022] Open
Abstract
Schistosoma japonicum (S. japonicum) is a worldwide spread pathogen which penetrates host skin and then induces several diseases in infected host, such as fibrosis, formation of granulomas, hepatocirrhosis, and hepatomegaly. In present study, for the first time, transcriptomic profiles of mouse livers and skins infected by fork-tailed S. japonicum cercaria or non-fork-tailed S. japonicum cercaria were analyzed by using RNA-seq. The present findings demonstrated that transcriptomic landscapes of livers and skins infected by fork-tailed S. japonicum cercaria or non-fork-tailed S. japonicum cercaria were different. S. japonicum has great influence on hepatic metabolic processes. Fork-tailed S. japonicum cercaria upregulated hepatic metabolic processes, while non-fork-tailed S. japonicum cercaria downregulated hepatic metabolic processes. For the metabolism process or the metabolism enzyme expressional change, the pharmacokinetics of host could be changed during S. japonicum infection, regardless the biotypes of S. japonicum cercariae. The changes of infected skins focused on upregulation of immune response. During the S. japonicum skin infection period, fork-tailed S. japonicum cercaria infection induced stronger immune response comparing with that immune response triggered by non-fork-tailed S. japonicum cercaria. The transcription factor enrichment analysis showed that Irf7, Stat1 and Stat2 could play important roles in gene regulation during fork-tailed S. japonicum cercaria infection.
Collapse
Affiliation(s)
- Yan Yang
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural UniversityKunming, China
| | - Jun-Jun He
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural SciencesLanzhou, China
| | - Shuang Hu
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural UniversityKunming, China
| | - Hua Chang
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural UniversityKunming, China
| | - Xun Xiang
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural UniversityKunming, China
| | - Jian-Fa Yang
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural UniversityKunming, China
| | - Feng-Cai Zou
- Key Laboratory of Veterinary Public Health of Yunnan Province, College of Veterinary Medicine, Yunnan Agricultural UniversityKunming, China
| |
Collapse
|
20
|
Abstract
Type 2 immunity is characterized by the production of IL-4, IL-5, IL-9 and IL-13, and this immune response is commonly observed in tissues during allergic inflammation or infection with helminth parasites. However, many of the key cell types associated with type 2 immune responses - including T helper 2 cells, eosinophils, mast cells, basophils, type 2 innate lymphoid cells and IL-4- and IL-13-activated macrophages - also regulate tissue repair following injury. Indeed, these cell populations engage in crucial protective activity by reducing tissue inflammation and activating important tissue-regenerative mechanisms. Nevertheless, when type 2 cytokine-mediated repair processes become chronic, over-exuberant or dysregulated, they can also contribute to the development of pathological fibrosis in many different organ systems. In this Review, we discuss the mechanisms by which type 2 immunity contributes to tissue regeneration and fibrosis following injury.
Collapse
Affiliation(s)
- Richard L Gieseck
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20852, USA
| | - Mark S Wilson
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, USA
| | - Thomas A Wynn
- Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20852, USA
| |
Collapse
|
21
|
Singh B, Kasam RK, Sontake V, Wynn TA, Madala SK. Repetitive intradermal bleomycin injections evoke T-helper cell 2 cytokine-driven pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2017; 313:L796-L806. [PMID: 28775096 DOI: 10.1152/ajplung.00184.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/11/2017] [Accepted: 07/28/2017] [Indexed: 02/06/2023] Open
Abstract
IL-4 and IL-13 are major T-helper cell (Th) 2 cytokines implicated in the pathogenesis of several lung diseases, including pulmonary fibrosis. In this study, using a novel repetitive intradermal bleomycin model in which mice develop extensive lung fibrosis and a progressive decline in lung function compared with saline-treated control mice, we investigated profibrotic functions of Th2 cytokines. To determine the role of IL-13 signaling in the pathogenesis of bleomycin-induced pulmonary fibrosis, wild-type, IL-13, and IL-4Rα-deficient mice were treated with bleomycin, and lungs were assessed for changes in lung function and pulmonary fibrosis. Histological staining and lung function measurements demonstrated that collagen deposition and lung function decline were attenuated in mice deficient in either IL-13 or IL-4Rα-driven signaling compared with wild-type mice treated with bleomycin. Furthermore, our results demonstrated that IL-13 and IL-4Rα-driven signaling are involved in excessive migration of macrophages and fibroblasts. Notably, our findings demonstrated that IL-13-driven migration involves increased phospho-focal adhesion kinase signaling and F-actin polymerization. Importantly, in vivo findings demonstrated that IL-13 augments matrix metalloproteinase (MMP)-2 and MMP9 activity that has also been shown to increase migration and invasiveness of fibroblasts in the lungs during bleomycin-induced pulmonary fibrosis. Together, our findings demonstrate a pathogenic role for Th2-cytokine signaling that includes excessive migration and protease activity involved in severe fibrotic lung disease.
Collapse
Affiliation(s)
- Brijendra Singh
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Rajesh K Kasam
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Biochemistry, National Institute of Nutrition, Hyderabad, Telangana, India; and
| | - Vishwaraj Sontake
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.,Department of Biochemistry, National Institute of Nutrition, Hyderabad, Telangana, India; and
| | - Thomas A Wynn
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland
| | - Satish K Madala
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio;
| |
Collapse
|
22
|
Citrin DE, Prasanna PGS, Walker AJ, Freeman ML, Eke I, Barcellos-Hoff MH, Arankalayil MJ, Cohen EP, Wilkins RC, Ahmed MM, Anscher MS, Movsas B, Buchsbaum JC, Mendonca MS, Wynn TA, Coleman CN. Radiation-Induced Fibrosis: Mechanisms and Opportunities to Mitigate. Report of an NCI Workshop, September 19, 2016. Radiat Res 2017; 188:1-20. [PMID: 28489488 PMCID: PMC5558616 DOI: 10.1667/rr14784.1] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A workshop entitled "Radiation-Induced Fibrosis: Mechanisms and Opportunities to Mitigate" (held in Rockville, MD, September 19, 2016) was organized by the Radiation Research Program and Radiation Oncology Branch of the Center for Cancer Research (CCR) of the National Cancer Institute (NCI), to identify critical research areas and directions that will advance the understanding of radiation-induced fibrosis (RIF) and accelerate the development of strategies to mitigate or treat it. Experts in radiation biology, radiation oncology and related fields met to identify and prioritize the key areas for future research and clinical translation. The consensus was that several known and newly identified targets can prevent or mitigate RIF in pre-clinical models. Further, basic and translational research and focused clinical trials are needed to identify optimal agents and strategies for therapeutic use. It was felt that optimally designed preclinical models are needed to better study biomarkers that predict for development of RIF, as well as to understand when effective therapies need to be initiated in relationship to manifestation of injury. Integrating appropriate endpoints and defining efficacy in clinical trials testing treatment of RIF were felt to be critical to demonstrating efficacy. The objective of this meeting report is to (a) highlight the significance of RIF in a global context, (b) summarize recent advances in our understanding of mechanisms of RIF,
Collapse
Affiliation(s)
- Deborah E. Citrin
- Radiation Oncology Branch, Center for Cancer Research, Bethesda, Maryland
| | - Pataje G. S. Prasanna
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Amanda J. Walker
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Michael L. Freeman
- Department of Radiation Oncology, Vanderbilt School of Medicine, Nashville, Tennessee
| | - Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, Bethesda, Maryland
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | | | - Eric P. Cohen
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ruth C. Wilkins
- Radiobiology Division, Consumer and Clinical Radiation Protection Bureau, Health Canada, Ottawa, Ontario
| | - Mansoor M. Ahmed
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Mitchell S. Anscher
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia
| | - Benjamin Movsas
- Department of Radiation Oncology, Henry Ford Hospital, Detroit, Michigan
| | - Jeffrey C. Buchsbaum
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| | - Marc S. Mendonca
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, Virginia
| | - Thomas A. Wynn
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - C. Norman Coleman
- Radiation Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
23
|
Figueiredo ALC, Domingues ALC, Melo WG, Tashiro T, de Lorena VMB, Montenegro SML, Morais CNL. Receptor Antagonist of IL-13 Exerts a Potential Negative Regulation During Early Infection of Human Schistosomiasis. Scand J Immunol 2017; 84:284-290. [PMID: 27507682 DOI: 10.1111/sji.12472] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 08/08/2016] [Indexed: 11/30/2022]
Abstract
The pathology of schistosomiasis is associated with the formation of granulomas, and this process is associated with liver fibrosis. Studies indicate that Th1 cytokines reduce fibrosis in schistosomiasis, while Th2 cytokines play a part in the progression of fibrosis, and IL-13 has a critical role in this process. The IL-13Rα2 receptor, known as a 'receptor antagonist' binds with high affinity to IL-13, and studies have identified that this plays a part in reducing fibrosis and the size of granulomas. The objective of this study was to evaluate the function of IL-13Rα2 and cellular immune response in hepatic fibrosis. A negative correlation between IL-13Rα2 and IL-13 was found, suggesting an increase in cytokine in early fibrosis. Initially, a negative correlation between IFN-γ and IL-13 was found in patients without fibrosis, and subsequently, this correlation was found to be positive in patients with severe fibrosis, thereby highlighting a new mechanism for regulating the progress of periportal fibrosis. There was a positive correlation between the profiles of Th1 and Th2 cytokines, suggesting the presence of both responses, thus regulating the disease. The results contribute to a better understanding of the immune mechanisms that control the process of hepatic fibrogenesis in schistosomiasis in humans.
Collapse
Affiliation(s)
| | - A L C Domingues
- Gastroenterology Outpatients Clinic, Hospital das Clínicas, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - W G Melo
- Aggeu Magalhães Research Center, Recife, Pernambuco, Brazil
| | - T Tashiro
- Department of Physical Education, Health Sciences Center, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | | | - C N L Morais
- Aggeu Magalhães Research Center, Recife, Pernambuco, Brazil.
| |
Collapse
|
24
|
Nady S, Shata MTM, Mohey MA, El-Shorbagy A. Protective role of IL-22 against Schistosoma mansoni soluble egg antigen-induced granuloma in Vitro. Parasite Immunol 2017; 39. [PMID: 27741351 DOI: 10.1111/pim.12392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 10/07/2016] [Indexed: 12/14/2022]
Abstract
The role of T helper-17 (Th17) lymphocytes in the regulation of Schistosoma mansoni soluble egg antigen (SEA)-induced granuloma is unknown. This study examined the effect of Th17 cytokines (IL-17 and IL-22) on granulocyte recruitment and functions during SEA-induced granuloma formation in vitro in Schistosoma-infected and noninfected individuals. Granulocytes were isolated from 27 Schistosoma-infected patients and 13 controls and were used for granuloma induction using SEA-conjugated polyacrylamide beads in the presence of Th17 cytokines. Granuloma index was assessed, and granulocyte mediators such as tumour necrosis factor (TNF-α), hydrogen peroxide (H2 O2 ) and nitric oxide (NO) were measured in the culture supernatant at the 7th day using enzyme-linked immunosorbent assay (ELISA). Schistosoma-infected patients had significant larger SEA-induced granuloma than controls. IL-17 (125 pg/mL) induced the optimum size for granuloma within 3-7 days. However, IL-22 at different concentrations up to 300 pg/mL had no effect on granuloma formation. Using both cytokines simultaneously, IL-22 suppressed the effect of IL-17 and prevented granuloma formation. IL-17 significantly decreased TNF-α, H2 O2 and NO levels in Schistosoma-infected individuals. In contrast, IL-22 increased TNF-α and H2 O2 levels. In conclusion, IL-17 accelerates SEA-induced granuloma formation and inhibits granulocytes functions in Schistosoma-infected patients, while IL-22 inhibited the granuloma formation, but enhanced granulocyte functions.
Collapse
Affiliation(s)
- S Nady
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
| | - M T M Shata
- Division of Digestive Diseases, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - M A Mohey
- Department of Endemic Medicine and Hepatology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - A El-Shorbagy
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
| |
Collapse
|
25
|
Bioanalytical challenges and improved detection of circulating levels of IL-13. Bioanalysis 2016; 8:323-32. [PMID: 26847873 DOI: 10.4155/bio.15.254] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND IL-13 is a key mediator of type 2 inflammation-driven diseases. Circulating IL-13 levels are very low and challenging to detect reliably. We assessed the ability of immunoassays on the Erenna(®) and IMPACT platforms to measure serum IL-13 in asthma, idiopathic pulmonary fibrosis (IPF) and atopic dermatitis (AD) patients and in healthy controls (HC). RESULTS The Erenna IL-13 assay exhibited significant specificity issues and had limited ability to detect IL-13 in serum samples. The IMPACT IL-13 assay had excellent specificity and detected IL-13 in 100% of serum samples tested from asthma, IPF and AD patients and HC. Serum IL-13 levels were significantly elevated in asthma, IPF and AD patients, relative to HC. CONCLUSION The IMPACT IL-13 assay had fg/ml sensitivity and excellent specificity, enabling reliable detection of circulating levels of IL-13.
Collapse
|
26
|
Crosby A, Soon E, Jones FM, Southwood MR, Haghighat L, Toshner MR, Raine T, Horan I, Yang P, Moore S, Ferrer E, Wright P, Ormiston ML, White RJ, Haight DA, Dunne DW, Morrell NW. Hepatic Shunting of Eggs and Pulmonary Vascular Remodeling in Bmpr2(+/-) Mice with Schistosomiasis. Am J Respir Crit Care Med 2015; 192:1355-65. [PMID: 26308618 PMCID: PMC4731697 DOI: 10.1164/rccm.201412-2262oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 08/09/2015] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Schistosomiasis is a major cause of pulmonary arterial hypertension (PAH). Mutations in the bone morphogenetic protein type-II receptor (BMPR-II) are the commonest genetic cause of PAH. OBJECTIVES To determine whether Bmpr2(+/-) mice are more susceptible to schistosomiasis-induced pulmonary vascular remodeling. METHODS Wild-type (WT) and Bmpr2(+/-) mice were infected percutaneously with Schistosoma mansoni. At 17 weeks postinfection, right ventricular systolic pressure and liver and lung egg counts were measured. Serum, lung and liver cytokine, pulmonary vascular remodeling, and liver histology were assessed. MEASUREMENTS AND MAIN RESULTS By 17 weeks postinfection, there was a significant increase in pulmonary vascular remodeling in infected mice. This was greater in Bmpr2(+/-) mice and was associated with an increase in egg deposition and cytokine expression, which induced pulmonary arterial smooth muscle cell proliferation, in the lungs of these mice. Interestingly, Bmpr2(+/-) mice demonstrated dilatation of the hepatic central vein at baseline and postinfection, compared with WT. Bmpr2(+/-) mice also showed significant dilatation of the liver sinusoids and an increase in inflammatory cells surrounding the central hepatic vein, compared with WT. This is consistent with an increase in the transhepatic passage of eggs. CONCLUSIONS This study has shown that levels of BMPR-II expression modify the pulmonary vascular response to chronic schistosomiasis. The likely mechanism involves the increased passage of eggs to the lungs, caused by altered diameter of the hepatic veins and sinusoids in Bmpr2(+/-) mice. Genetically determined differences in the remodeling of hepatic vessels may represent a new risk factor for PAH associated with schistosomiasis.
Collapse
Affiliation(s)
- Alexi Crosby
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Elaine Soon
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Frances M. Jones
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Mark R. Southwood
- Department of Pathology, Papworth Hospital, Cambridge, United Kingdom
| | - Leila Haghighat
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Mark R. Toshner
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Tim Raine
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Ian Horan
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Peiran Yang
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Stephen Moore
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Elisabet Ferrer
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Penny Wright
- Addenbrooke’s Hospital, Cambridge, United Kingdom; and
| | - Mark L. Ormiston
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | | | | | - David W. Dunne
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s Hospital, Cambridge, United Kingdom
| |
Collapse
|
27
|
Russell SB, Smith JC, Huang M, Trupin JS, Williams SM. Pleiotropic Effects of Immune Responses Explain Variation in the Prevalence of Fibroproliferative Diseases. PLoS Genet 2015; 11:e1005568. [PMID: 26540410 PMCID: PMC4634921 DOI: 10.1371/journal.pgen.1005568] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many diseases are differentially distributed among human populations. Differential selection on genetic variants in ancestral environments that coincidentally predispose to disease can be an underlying cause of these unequal prevalence patterns. Selected genes may be pleiotropic, affecting multiple phenotypes and resulting in more than one disease or trait. Patterns of pleiotropy may be helpful in understanding the underlying causes of an array of conditions in a population. For example, several fibroproliferative diseases are more prevalent and severe in populations of sub-Saharan ancestry. We propose that this disparity is due to selection for an enhanced Th2 response that confers resistance to helminthic infections, and concurrently increases susceptibility to fibrosis due to the profibrotic action of Th2 cytokines. Many studies on selection of Th2-related genes for host resistance to helminths have been reported, but the pleiotropic impact of this selection on the distribution of fibrotic disorders has not been explicitly investigated. We discuss the disproportionate occurrence of fibroproliferative diseases in individuals of African ancestry and provide evidence that adaptation of the immune system has shaped the genetic structure of these human populations in ways that alter the distribution of multiple fibroproliferative diseases.
Collapse
Affiliation(s)
- Shirley B. Russell
- Vanderbilt Genetics Institute, Division of Dermatology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- * E-mail: (SBR); , (SMW)
| | - Joan C. Smith
- Meharry Medical College, Nashville, Tennessee, United States of America
| | - Minjun Huang
- Department of Genetics, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States of America
| | - Joel S. Trupin
- Meharry Medical College, Nashville, Tennessee, United States of America
| | - Scott M. Williams
- Department of Genetics, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, United States of America
- * E-mail: (SBR); , (SMW)
| |
Collapse
|
28
|
Borthwick LA, Wynn TA. IL-13 and TGF-β1: Core Mediators of Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0091-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
29
|
Zheng S, Lu Q, Xu Y, Wang X, Shen J, Wang W. GdCl3 Attenuates Schistosomiasis japonicum Egg-Induced Granulomatosis Accompanied by Decreased Macrophage Infiltration in Murine Liver. PLoS One 2015; 10:e0132222. [PMID: 26317423 PMCID: PMC4552789 DOI: 10.1371/journal.pone.0132222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 06/12/2015] [Indexed: 11/19/2022] Open
Abstract
Early-stage hepatic granuloma and advanced-stage fibrosis are important characteristics of schistosomiasis. The direct consequences of gadolinium chloride (GdCl3) in egg-induced granuloma formation have not been reported, although GdCl3 is known to block the macrophages. In present study, mice were infected with 15 Schistosoma japonicum (S. japonicum) cercariae and treated with GdCl3 (10 mg/kg body weight) twice weekly from day 21 to day 42 post-infection during the onset of egg-laying towards early granuloma formation. Histochemical staining showed that repeated injection of GdCl3 decreased macrophages infiltration in liver of mice infected with S. japonicum. Macrophage depletion by GdCl3 during the initial phase attenuated liver pathological injury characterized by smaller granuloma size and decreased immune inflammation as well as less fibrogenesis. In addition, IL-13Rα2 expression was reduced by GdCl3 in liver of mice infected with S. japonicum. The results suggest that GdCl3 depleted macrophages, which attenuated helminth infected immune responses involving with IL-13Rα2 signal. These findings would highlight a therapeutic potential via manipulating IL-13Rα2+ macrophage in schistosomiasis.
Collapse
Affiliation(s)
- Shengsheng Zheng
- Department of Pathobiology, Key Laboratories of Zoonoses of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Qiang Lu
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230032, China
| | - Yuanhong Xu
- Department of Laboratory Diagnostics, First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xiaonan Wang
- Department of Pathobiology, Key Laboratories of Zoonoses of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Jilong Shen
- Department of Pathobiology, Key Laboratories of Zoonoses of Anhui Province, Anhui Medical University, Hefei, 230032, China
| | - Wei Wang
- Department of Pathobiology, Key Laboratories of Zoonoses of Anhui Province, Anhui Medical University, Hefei, 230032, China
- * E-mail:
| |
Collapse
|
30
|
McCormick SM, Heller NM. Commentary: IL-4 and IL-13 receptors and signaling. Cytokine 2015; 75:38-50. [PMID: 26187331 PMCID: PMC4546937 DOI: 10.1016/j.cyto.2015.05.023] [Citation(s) in RCA: 230] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 05/16/2015] [Accepted: 05/21/2015] [Indexed: 12/21/2022]
Abstract
Interleukin (IL)-4 and IL-13 were discovered approximately 30years ago and were immediately linked to allergy and atopic diseases. Since then, new roles for IL-4 and IL-13 and their receptors in normal gestation, fetal development and neurological function and in the pathogenesis of cancer and fibrosis have been appreciated. Studying IL-4/-13 and their receptors has revealed important clues about cytokine biology and led to the development of numerous experimental therapeutics. Here we aim to highlight new discoveries and consolidate concepts in the field of IL-4 and IL-13 structure, receptor regulation, signaling and experimental therapeutics.
Collapse
Affiliation(s)
- Sarah M McCormick
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Nicola M Heller
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Division of Allergy and Clinical Immunology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| |
Collapse
|
31
|
An IL-13 promoter polymorphism associated with liver fibrosis in patients with Schistosoma japonicum. PLoS One 2015; 10:e0135360. [PMID: 26258681 PMCID: PMC4530950 DOI: 10.1371/journal.pone.0135360] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/21/2015] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to determine whether two polymorphisms in the gene encoding IL13 previously associated with Schistosoma hematobium (S. hematobium) and S. mansoni infection are associated with S. japonicum infection. Single nucleotide polymorphisms (SNPs) rs1800925 (IL13/-1112C>T) and rs20541 (IL13R130Q) were genotyped in 947 unrelated individuals (307 chronically infected, 339 late-stage with liver fibrosis, 301 uninfected controls) from a schistosomiasis-endemic area of Hubei province in China. Regression models were used to evaluate allelic and haplotypic associations with chronic and late-stage schistosomiasis adjusted for non-genetic covariates. Expression of IL-13 was measured in S. japonicun-infected liver fibrosis tissue and normal liver tissue from uninfected controls by immunohistochemistry (IHC). The role of rs1800925 in IL-13 transcription was further determined by Luciferase report assay using the recombinant PGL4.17-rs180092 plasmid. We found SNP rs1800925T was associated with late-stage schistosomiasis caused by S. japonicum but not chronic schistosomiasis (OR = 1.39, 95%CI = 1.02-1.91, p = 0.03) and uninfected controls (OR = 1.49, 95%CI = 1.03-2.13, p = 0.03). Moreover, the haplotype rs1800925T-rs20541C increased the risk of disease progression to late-stage schistosomiasis (OR = 1.46, p = 0.035), whereas haplotype rs1800925C-rs20541A showed a protective role against development of late-stage schistosomiasis (F = 0.188, OR = 0.61, p = 0.002). Furthermore, S. japonicum-induced fibrotic liver tissue had higher IL13 expression than normal liver tissue. Plasmid PGL4.17-rs1800925T showed a stronger relative luciferase activity than Plasmid PGL4.17-rs1800925C in 293FT, QSG-7701 and HL-7702 cell lines. In conclusion, the functional IL13 polymorphism, rs1800925T, previously associated with risk of schistosomiasis, also contributes to risk of late-stage schistosomiasis caused by S. japonicum.
Collapse
|
32
|
Abstract
Type 2 immune responses are defined by the cytokines interleukin-4 (IL-4), IL-5, IL-9 and IL-13, which can either be host protective or have pathogenic activity. Type 2 immunity promotes antihelminth immunity, suppresses type 1-driven autoimmune disease, neutralizes toxins, maintains metabolic homeostasis, and regulates wound repair and tissue regeneration pathways following infection or injury. Nevertheless, when type 2 responses are dysregulated, they can become important drivers of disease. Type 2 immunity induces a complex inflammatory response characterized by eosinophils, mast cells, basophils, type 2 innate lymphoid cells, IL-4-and/or IL-13-conditioned macrophages and T helper 2 (TH2) cells, which are crucial to the pathogenesis of many allergic and fibrotic disorders. As chronic type 2 immune responses promote disease, the mechanisms that regulate their maintenance are thought to function as crucial disease modifiers. This Review discusses the many endogenous negative regulatory mechanisms that antagonize type 2 immunity and highlights how therapies that target some of these pathways are being developed to treat type 2-mediated disease.
Collapse
Affiliation(s)
- Thomas A Wynn
- Immunopathogenesis Section, Program in Barrier Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland 20892-0425, USA
| |
Collapse
|
33
|
Abstract
Type 2 immune responses are defined by the cytokines interleukin-4 (IL-4), IL-5, IL-9 and IL-13, which can either be host protective or have pathogenic activity. Type 2 immunity promotes antihelminth immunity, suppresses type 1-driven autoimmune disease, neutralizes toxins, maintains metabolic homeostasis, and regulates wound repair and tissue regeneration pathways following infection or injury. Nevertheless, when type 2 responses are dysregulated, they can become important drivers of disease. Type 2 immunity induces a complex inflammatory response characterized by eosinophils, mast cells, basophils, type 2 innate lymphoid cells, IL-4-and/or IL-13-conditioned macrophages and T helper 2 (TH2) cells, which are crucial to the pathogenesis of many allergic and fibrotic disorders. As chronic type 2 immune responses promote disease, the mechanisms that regulate their maintenance are thought to function as crucial disease modifiers. This Review discusses the many endogenous negative regulatory mechanisms that antagonize type 2 immunity and highlights how therapies that target some of these pathways are being developed to treat type 2-mediated disease.
Collapse
|
34
|
Lumsden RV, Worrell JC, Boylan D, Walsh SM, Cramton J, Counihan I, O'Beirne S, Medina MF, Gauldie J, Fabre A, Donnelly SC, Kane R, Keane MP. Modulation of pulmonary fibrosis by IL-13Rα2. Am J Physiol Lung Cell Mol Physiol 2015; 308:L710-8. [DOI: 10.1152/ajplung.00120.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 02/01/2015] [Indexed: 11/22/2022] Open
Abstract
Pulmonary fibrosis is a progressive and fatal disease that involves the remodeling of the distal airspace and the lung parenchyma, which results in compromised gas exchange. The median survival time once diagnosed is less than three years. Interleukin (IL)-13 has been shown to play a role in a number of inflammatory and fibrotic diseases. IL-13 modulates its effector functions via a complex receptor system that includes the IL-4 receptor (R) α, IL-13Rα1, and the IL-13Rα2. IL-13Rα1 binds IL-13 with low affinity, yet, when it forms a complex with IL-4α, it binds with much higher affinity, inducing the effector functions of IL-13. IL-13Rα2 binds IL-13 with high affinity but has a short cytoplasmic tail and has been shown to act as a nonsignaling decoy receptor. Transfection of fibroblasts and epithelial cells with IL-13Rα2 inhibited the IL-13 induction of soluble collagen, TGF-β, and CCL17. Adenoviral overexpression of IL-13Rα2 in the lung reduced bleomycin-induced fibrosis. Our work shows that overexpression of IL-13Rα2 inhibits the IL-13 induction of fibrotic markers in vitro and inhibits bleomycin-induced pulmonary fibrosis. In summary our study highlights the antifibrotic nature of IL-13Ra2.
Collapse
Affiliation(s)
- Robert V. Lumsden
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
| | - Julie C. Worrell
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
| | - Denise Boylan
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
| | - Sinead M. Walsh
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
| | - Jennifer Cramton
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
| | - Ian Counihan
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
| | - Sarah O'Beirne
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
| | - Maria Fe Medina
- Fitzhenry Laboratory, Department of Pathology, McMaster University, Hamilton, Ontario, Canada; and
| | - Jack Gauldie
- Fitzhenry Laboratory, Department of Pathology, McMaster University, Hamilton, Ontario, Canada; and
| | - Aurelie Fabre
- Department of Pathology, St. Vincent's University Hospital, Dublin, Ireland
| | - Seamas C. Donnelly
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
| | - Rosemary Kane
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
| | - Michael P. Keane
- Conway Institute of Biomedical and Biomolecular Science, University College Dublin, Dublin, Ireland
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
35
|
Papamatheakis DG, Mocumbi AOH, Kim NH, Mandel J. Schistosomiasis-associated pulmonary hypertension. Pulm Circ 2015; 4:596-611. [PMID: 25610596 DOI: 10.1086/678507] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Accepted: 07/13/2014] [Indexed: 01/26/2023] Open
Abstract
Schistosomiasis, a parasite-borne disease, is highly prevalent in Africa and Asia; it is estimated that close to 20 million people worldwide have a severe form of the disease. The chronic form can affect the gastrointestinal system and lead to hepatosplenic disease, and it may cause cardiopulmonary complications, including pulmonary hypertension. The exact pathogenesis of schistosomiasis-associated pulmonary hypertension (Sch-PH) remains unclear, although several mechanisms, including parasitic arterial embolization, pulmonary arteriopathy, and portopulmonary hypertension-like pathophysiology, have been suggested. The immunopathology of the disease is also unclear, although there are similarities with the immunology of idiopathic pulmonary arterial hypertension (PAH). Finally, the treatment of Sch-PH has not been well studied. There is some evidence on treating the underlying infection, with unclear effect on Sch-PH, and advanced PAH therapies are now being suggested, but more studies are needed to confirm their efficacy.
Collapse
Affiliation(s)
- Demosthenes G Papamatheakis
- Division of Pulmonary and Critical Care Medicine, University of California San Diego Health System, La Jolla, California, USA
| | - Ana Olga H Mocumbi
- Instituto Nacional de Saúde and Universidade Eduardo Mondlane, Maputo, Mozambique
| | - Nick H Kim
- Division of Pulmonary and Critical Care Medicine, University of California San Diego Health System, La Jolla, California, USA
| | - Jess Mandel
- Division of Pulmonary and Critical Care Medicine, University of California San Diego Health System, La Jolla, California, USA
| |
Collapse
|
36
|
Gidwani S, Nair A. The burden of pulmonary hypertension in resource-limited settings. Glob Heart 2014; 9:297-310. [PMID: 25667181 DOI: 10.1016/j.gheart.2014.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 08/11/2014] [Accepted: 08/18/2014] [Indexed: 12/11/2022] Open
Abstract
Pulmonary vascular disease (PVD) is a significant global health problem and accounts for a substantial portion of cardiovascular disease in the developing world. Although there have been considerable advances in therapeutics for pulmonary arterial hypertension, over 97% of the disease burden lies within the developing world where there is limited access to health care and pharmaceuticals. The causes of pulmonary arterial hypertension differ between industrialized and developing nations. Infectious diseases-including schistosomiasis human immunodeficiency virus, and rheumatic fever-are common causes of PVD, as are hemoglobinopathies, and untreated congenital heart disease. High altitude and exposure to household air pollutants also contribute to a significant portion of PVD cases. Although diagnosis of pulmonary arterial hypertension requires the use of imaging and invasive hemodynamics, access to equipment may be limited. PVD therapies may be prohibitively expensive and limited to a select few. Prevention is therefore important in limiting the global PVD burden.
Collapse
Affiliation(s)
| | - Ajith Nair
- Cardiovascular Institute, Mount Sinai Hospital, New York, NY, USA.
| |
Collapse
|
37
|
Vannella KM, Barron L, Borthwick LA, Kindrachuk KN, Narasimhan PB, Hart KM, Thompson RW, White S, Cheever AW, Ramalingam TR, Wynn TA. Incomplete deletion of IL-4Rα by LysM(Cre) reveals distinct subsets of M2 macrophages controlling inflammation and fibrosis in chronic schistosomiasis. PLoS Pathog 2014; 10:e1004372. [PMID: 25211233 PMCID: PMC4161449 DOI: 10.1371/journal.ppat.1004372] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 08/01/2014] [Indexed: 01/09/2023] Open
Abstract
Mice expressing a Cre recombinase from the lysozyme M-encoding locus (Lyz2) have been widely used to dissect gene function in macrophages and neutrophils. Here, we show that while naïve resident tissue macrophages from IL-4Rαflox/deltaLysMCre mice almost completely lose IL-4Rα function, a large fraction of macrophages elicited by sterile inflammatory stimuli, Schistosoma mansoni eggs, or S. mansoni infection, fail to excise Il4rα. These F4/80hiCD11bhi macrophages, in contrast to resident tissue macrophages, express lower levels of Lyz2 explaining why this population resists LysMCre-mediated deletion. We show that in response to IL-4 and IL-13, Lyz2loIL-4Rα+ macrophages differentiate into an arginase 1-expressing alternatively-activated macrophage (AAM) population, which slows the development of lethal fibrosis in schistosomiasis. In contrast, we identified Lyz2hiIL-4Rα+ macrophages as the key subset of AAMs mediating the downmodulation of granulomatous inflammation in chronic schistosomiasis. Our observations reveal a limitation on using a LysMCre mouse model to study gene function in inflammatory settings, but we utilize this limitation as a means to demonstrate that distinct populations of alternatively activated macrophages control inflammation and fibrosis in chronic schistosomiasis. Chronic injury and inflammation lead to irreversible fibrosis in a range of diseases and infections. Macrophages alternatively activated by the immune system are capable of regulating inflammation and fibrosis, but our understanding of the source and function of these cells is incomplete. Mice genetically engineered to specifically prevent macrophages from becoming alternatively activated have been used to study the cells' role following infection with the parasite, Schistosoma mansoni. To our surprise, we found these mice prevent alternative activation only in macrophages that have had time to mature and some, perhaps more nascent, macrophages can become alternatively activated following exposure to S. mansoni eggs. We detected lower expression of Lyz2 gene in these cells, leading to less expression of the enzyme excising the receptor gene necessary for alternative activation. Following S. mansoni infection, the livers of these mice have similar levels of fibrosis but significantly more inflammation compared to controls. We conclude that during schistosomiasis, distinct populations of alternatively activated macrophages control inflammation and fibrosis: macrophages expressing low levels of Lyz2 express Arg1 and thus are sufficient to control fibrosis, while more mature Lyz2-expressing macrophages are required for downmodulation of egg-induced inflammation in chronic schistosomiasis.
Collapse
Affiliation(s)
- Kevin M. Vannella
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Luke Barron
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lee A. Borthwick
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Tissue Fibrosis and Repair Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kristen N. Kindrachuk
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Prakash Babu Narasimhan
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kevin M. Hart
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert W. Thompson
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sandra White
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Allen W. Cheever
- Biomedical Research Institute, Rockville, Maryland, United States of America
| | - Thirumalai R. Ramalingam
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas A. Wynn
- Program in Tissue Immunity and Repair, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
38
|
Kamel MM, Fouad SA, Basyoni MMA. P selectins and immunological profiles in HCV and Schistosoma mansoni induced chronic liver disease. BMC Gastroenterol 2014; 14:132. [PMID: 25066324 PMCID: PMC4119237 DOI: 10.1186/1471-230x-14-132] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 07/17/2014] [Indexed: 01/01/2023] Open
Abstract
Background Hepatitis C virus (HCV) and Schistosoma mansoni are major causes of chronic liver disease (CLD) in which immune alteration is common. Recent studies suggested that certain platelets and lymphocytes activation markers may have an impact on progression of CLD. This study aimed to evaluate the potential of platelets and lymphocytes activation molecules expression on the pathogenesis of CLD in distinct or concomitant chronic HCV and schistosomiasis mansoni infections. Methods The study populations were divided into group-I: patients with chronic schistosomiasis mansoni, group-II: HCV patients without cirrhosis, group-III: patients with combined liver diseases without cirrhosis, group-IV: patients with chronic HCV and liver cirrhosis and group-V: Age and sex matched healthy individuals as normal controls. All groups were subjected to full clinical evaluation, ELISA anti-HCV antibodies screening, parasitological examination for diagnosing S. mansoni and flow cytometry for lymphocyte (CD3, CD4, CD8, CD19, CD22, & CD56) and platelets activation (CD41, CD42 & CD62P (P- selectins)) markers. Results The platelet count was significantly decreased in HCV and/or S. mansoni patients. The total T-lymphocytes and T-helper cells were significantly reduced, while T-cytotoxics were increased. The patients possessed a significantly higher platelets activation marker; CD62P (P-selectins) and higher mean fluorescent intensity (MFI) positivity. There were considerable correlations between platelets count and both of CD62P and MFI. Conclusion Our Findings suggest an increased expression of certain platelets and lymphocytes activation markers in chronic HCV and S. mansoni induced CLD that may have a role in disease progression.
Collapse
Affiliation(s)
- Mahmoud M Kamel
- Department of Clinical Pathology, National Cancer Institute Cairo University, Cairo, Egypt.
| | | | | |
Collapse
|
39
|
Chandriani S, DePianto DJ, N’Diaye EN, Abbas AR, Jackman J, Bevers J, Ramirez-Carrozzi V, Pappu R, Kauder SE, Toy K, Ha C, Modrusan Z, Wu LC, Collard HR, Wolters PJ, Egen JG, Arron JR. Endogenously Expressed IL-13Rα2 Attenuates IL-13–Mediated Responses but Does Not Activate Signaling in Human Lung Fibroblasts. THE JOURNAL OF IMMUNOLOGY 2014; 193:111-9. [DOI: 10.4049/jimmunol.1301761] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
40
|
CD14 influences host immune responses and alternative activation of macrophages during Schistosoma mansoni infection. Infect Immun 2014; 82:3240-51. [PMID: 24866794 DOI: 10.1128/iai.01780-14] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Antigen-presenting cell (APC) plasticity is critical for controlling inflammation in metabolic diseases and infections. The roles that pattern recognition receptors (PRRs) play in regulating APC phenotypes are just now being defined. We evaluated the expression of PRRs on APCs in mice infected with the helminth parasite Schistosoma mansoni and observed an upregulation of CD14 expression on macrophages. Schistosome-infected Cd14(-/-) mice showed significantly increased alternative activation of (M2) macrophages in the livers compared to infected wild-type (wt) mice. In addition, splenocytes from infected Cd14(-/-) mice exhibited increased production of CD4(+)-specific interleukin-4 (IL-4), IL-5, and IL-13 and CD4(+)Foxp3(+)IL-10(+) regulatory T cells compared to cells from infected wt mice. S. mansoni-infected Cd14(-/-) mice also presented with smaller liver egg granulomas associated with increased collagen deposition compared to granulomas in infected wt mice. The highest expression of CD14 was found on liver macrophages in infected mice. To determine if the Cd14(-/-) phenotype was in part due to increased M2 macrophages, we adoptively transferred wt macrophages into Cd14(-/-) mice and normalized the M2 and CD4(+) Th cell balance close to that observed in infected wt mice. Finally, we demonstrated that CD14 regulates STAT6 activation, as Cd14(-/-) mice had increased STAT6 activation in vivo, suggesting that lack of CD14 impacts the IL-4Rα-STAT6 pathway, altering macrophage polarization during parasite infection. Collectively, these data identify a previously unrecognized role for CD14 in regulating macrophage plasticity and CD4(+) T cell biasing during helminth infection.
Collapse
|
41
|
The effect of recombinant sTGFβ1RII and sIL13Rα2 receptor proteins on schistosomiasis japonica, hepatic fibrosis and signal transduction in a mouse model of schistosome disease. Exp Parasitol 2014; 142:17-26. [PMID: 24746639 DOI: 10.1016/j.exppara.2014.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 03/11/2014] [Accepted: 04/06/2014] [Indexed: 11/20/2022]
Abstract
This study was designed to investigate the effect of recombinant sTGFβ1RII and sIL13Rα2 receptor proteins on schistosomiasis japonica, hepatic fibrosis and the expression of SMAD3 and STAT6. The proteins sTGFβ1RII and sIL13Rα2 were expressed in Escherichiacoli, purified using affinity chromatography and characterized by Western blotting. Female BALB/C mice (48) were randomly divided into eight groups and infected with Schistosoma japonicum. Five weeks after infection, test groups were injected with the recombinant proteins at different doses. Eight weeks after infection, lung and hepatic tissue samples were obtained and stained with hematoxylin and eosin (HE) and Masson's trichrome. Immunohistochemical staining was used to detect the expression of SMAD3 and STAT6. The recombinant proteins sTGFβ1RII and sIL13Rα2 were successfully expressed, purified, and characterized. The granuloma area, hepatic hydroxyproline (HYP) level and hepatic fibrosis of the protein therapeutic groups were significantly smaller than those of the positive control group (P<0.01). Treatment with sTGFβ1RII was more effective when the protein was administered for 4weeks rather than 2 (P<0.01). Hepatic fibrosis in the groups using a low dose of protein sTGFβ1 was lower that of the combination group (P<0.05). The expression level of STAT6 was significantly lower in groups treated with sIL13Rα2 than in groups not treated with the protein (P<0.01). The recombinant proteins TGFβ1RII and sIL13Rα2 were able to decrease granuloma area and hepatic fibrosis in schistosomiasis japonica, and also reduced the expression of the signal transduction proteins SMAD3 and STAT6. The proteins were more effective when used in combination than when applied singly.
Collapse
|
42
|
Chuah C, Jones MK, Burke ML, McManus DP, Gobert GN. Cellular and chemokine-mediated regulation in schistosome-induced hepatic pathology. Trends Parasitol 2014; 30:141-50. [PMID: 24433721 DOI: 10.1016/j.pt.2013.12.009] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 12/17/2013] [Accepted: 12/18/2013] [Indexed: 02/06/2023]
Abstract
In hepatic schistosomiasis, pathology arises when schistosome eggs become lodged in the host liver, evoking an interleukin 4 (IL-4)- and IL-13-mediated dominant CD4(+) Th2 immune response. This response leads to the development of granulomas and fibrosis, with eosinophils, neutrophils, macrophages, hepatic stellate cells, and lymphocytes all identified as major cellular contributors to these events. This review outlines the cellular and molecular mechanisms of hepatic schistosomiasis, with an emphasis on the major cellular components and their release of chemokines. The differences between Schistosoma mansoni- and Schistosoma japonicum-induced hepatic granuloma are also discussed. This comprehensive overview of the processes associated with hepatic schistosomiasis may provide new insights into improved treatment for both schistosomiasis and other granulofibrotic diseases.
Collapse
Affiliation(s)
- Candy Chuah
- Parasite Cell Biology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia; School of Veterinary Sciences, The University of Queensland, Gatton, 4343, Australia; School of Medical Sciences, Universiti Sains Malaysia, 16150, Kelantan, Malaysia
| | - Malcolm K Jones
- Parasite Cell Biology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia; School of Veterinary Sciences, The University of Queensland, Gatton, 4343, Australia
| | - Melissa L Burke
- Division of Mycobacterial Research, National Institute for Medical Research, London, NW7 1AA, UK
| | - Donald P McManus
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia
| | - Geoffrey N Gobert
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, 4006, Australia.
| |
Collapse
|
43
|
Wick G, Grundtman C, Mayerl C, Wimpissinger TF, Feichtinger J, Zelger B, Sgonc R, Wolfram D. The immunology of fibrosis. Annu Rev Immunol 2013; 31:107-35. [PMID: 23516981 DOI: 10.1146/annurev-immunol-032712-095937] [Citation(s) in RCA: 233] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fibrosis is the production of excessive amounts of connective tissue, i.e., scar formation, in the course of reactive and reparative processes. Fibrosis develops as a consequence of various underlying diseases and presents a major diagnostically and therapeutically unsolved problem. In this review, we postulate that fibrosis is always a sequela of inflammatory processes and that the many different causes of fibrosis all channel into the same final stereotypical pathways. During the inflammatory phase, both innate and adaptive immune mechanisms are operative. This concept is exemplified by fibrotic diseases that develop as a consequence of tissue damage, primary inflammatory diseases, fibrotic alterations induced by foreign body implants, "spontaneous" fibrosis, and tumor-associated fibrotic changes.
Collapse
Affiliation(s)
- Georg Wick
- Division of Experimental Pathophysiology and Immunology, 6020 Innsbruck, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Chen W, Sivaprasad U, Gibson AM, Ericksen MB, Cunningham CM, Bass SA, Kinker KG, Finkelman FD, Wills-Karp M, Khurana Hershey GK. IL-13 receptor α2 contributes to development of experimental allergic asthma. J Allergy Clin Immunol 2013; 132:951-8.e1-6. [PMID: 23763980 DOI: 10.1016/j.jaci.2013.04.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 03/21/2013] [Accepted: 04/05/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND IL-13 receptor α2 (IL-13Rα2) binds IL-13 with high affinity and modulates IL-13 responses. There are soluble and membrane forms of IL-13Rα2 generated by alternative splicing in mice, but human subjects express only the membrane form of IL-13Rα2 (memIL-13Rα2). OBJECTIVE We determined the role of memIL-13Rα2 in the development of allergic inflammation in mouse models of asthma. METHODS IL-13Rα2-deficient and memIL-13Rα2 lung epithelium-specific transgenic mice were challenged with house dust mite (HDM). Airway hyperresponsiveness (AHR) and inflammation were assessed based on the airway pressure-time index, bronchoalveolar lavage (BAL) cell counts, and lung histology. Mucus production was determined by means of periodic acid-Schiff staining of lung sections, Western blot analysis of chloride channel calcium activated 3 (CLCA3) expression in lung homogenates, and ELISA of Muc5ac in BAL fluid. The expression of cytokines and chemokines was determined by using RT-quantitative PCR. RESULTS In IL-13Rα2-deficient mice AHR and airway inflammation were attenuated compared with levels seen in wild-type mice after HDM challenge. Lung epithelial overexpression of memIL-13Rα2 in the IL-13Rα2-deficient mice reconstituted AHR and inflammation to levels similar to those observed in HDM-challenged wild-type mice. Mucus production was attenuated in lungs from HDM-treated IL-13Rα2-deficient mice, whereas lung epithelial overexpression of memIL-13Rα2 increased mucus production. Lung epithelial overexpression of memIL-13Rα2 had no effect on levels of the soluble form of IL-13Rα2 in serum or BAL fluid and did not affect IL-13-dependent signal transducer and activator of transcription 6 activation in the lungs. CONCLUSION These data collectively support a distinct role for memIL-13Rα2 in the lung and suggest that memIL-13Rα2 might contribute to allergic inflammation.
Collapse
Affiliation(s)
- Weiguo Chen
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Osburn WO, Levine JS, Chattergoon MA, Thomas DL, Cox AL. Anti-inflammatory cytokines, pro-fibrogenic chemokines and persistence of acute HCV infection. J Viral Hepat 2013; 20:404-13. [PMID: 23647957 PMCID: PMC3793396 DOI: 10.1111/jvh.12052] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2012] [Accepted: 12/01/2012] [Indexed: 12/12/2022]
Abstract
Chemokines and cytokines play a vital role in directing and regulating immune responses to viral infections. Persistent hepatitis C virus (HCV) infection is characterized by the loss of anti-HCV cellular immune responses, while control of HCV infection is associated with maintenance of anti-HCV cellular immune responses. To determine whether plasma concentrations of 19 chemokines and cytokines controlling T-cell trafficking and function differed based on infection outcome, we compared them in at-risk subjects followed prospectively for HCV infection. Levels were compared over time in subjects who controlled HCV infection (Clearance) and subjects who developed persistent HCV infection (Persistence) at two time points during acute infection: (i) first viraemic sample (initial viraemia) and (ii) last viraemic sample in Clearance subjects and time-matched samples in Persistence subjects. At initial viraemia, increased pro-inflammatory tumour necrosis factor α (TNFα) plasma concentrations were observed in the Clearance group, while the plasma levels of anti-inflammatory interleukin (IL)-2, IL-10 and IL-13 were higher in the Persistence group. IL-13 was positively correlated with IL-2 and IL-10 at initial viraemia in the Persistence group. At the time of last viraemia, plasma levels of eotaxin, macrophage chemoattractant protein-4 (MCP-4), IL-5 and IL-10 were higher in the Persistence group and IL-10 and IL-5 levels were positively correlated. Collectively, these results suggest that the development of persistent infection is associated with an anti-inflammatory and pro-fibrogenic chemokine and cytokine profile that is evident at the onset of infection and maintained throughout acute infection.
Collapse
Affiliation(s)
- W O Osburn
- Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
46
|
Ray D, Nelson TA, Fu CL, Patel S, Gong DN, Odegaard JI, Hsieh MH. Transcriptional profiling of the bladder in urogenital schistosomiasis reveals pathways of inflammatory fibrosis and urothelial compromise. PLoS Negl Trop Dis 2012; 6:e1912. [PMID: 23209855 PMCID: PMC3510078 DOI: 10.1371/journal.pntd.0001912] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 10/03/2012] [Indexed: 11/19/2022] Open
Abstract
Urogenital schistosomiasis, chronic infection by Schistosoma haematobium, affects 112 million people worldwide. S. haematobium worm oviposition in the bladder wall leads to granulomatous inflammation, fibrosis, and egg expulsion into the urine. Despite the global impact of urogenital schistosomiasis, basic understanding of the associated pathologic mechanisms has been incomplete due to the lack of suitable animal models. We leveraged our recently developed mouse model of urogenital schistosomiasis to perform the first-ever profiling of the early molecular events that occur in the bladder in response to the introduction of S. haematobium eggs. Microarray analysis of bladders revealed rapid, differential transcription of large numbers of genes, peaking three weeks post-egg administration. Many differentially transcribed genes were related to the canonical Type 2 anti-schistosomal immune response, as reflected by the development of egg-based bladder granulomata. Numerous collagen and metalloproteinase genes were differentially transcribed over time, revealing complex remodeling and fibrosis of the bladder that was confirmed by Masson's Trichrome staining. Multiple genes implicated in carcinogenesis pathways, including vascular endothelial growth factor-, oncogene-, and mammary tumor-related genes, were differentially transcribed in egg-injected bladders. Surprisingly, junctional adhesion molecule, claudin and uroplakin genes, key components for maintaining the urothelial barrier, were globally suppressed after bladder exposure to eggs. This occurred in the setting of urothelial hyperplasia and egg shedding in urine. Thus, S. haematobium egg expulsion is associated with intricate modulation of the urothelial barrier on the cellular and molecular level. Taken together, our findings have important implications for understanding host-parasite interactions and carcinogenesis in urogenital schistosomiasis, and may provide clues for novel therapeutic strategies.
Collapse
Affiliation(s)
- Debalina Ray
- Department of Urology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Tyrrell A. Nelson
- Department of Urology, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Chi-Ling Fu
- Department of Urology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Shailja Patel
- Department of Urology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Diana N. Gong
- Department of Urology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Justin I. Odegaard
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Michael H. Hsieh
- Department of Urology, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
47
|
Beschin A, De Baetselier P, Van Ginderachter JA. Contribution of myeloid cell subsets to liver fibrosis in parasite infection. J Pathol 2012; 229:186-97. [DOI: 10.1002/path.4112] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 08/24/2012] [Accepted: 09/13/2012] [Indexed: 12/23/2022]
Affiliation(s)
- Alain Beschin
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| | - Patrick De Baetselier
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| | - Jo A Van Ginderachter
- Myeloid Cell Immunology Laboratory; VIB Brussels Belgium
- Cellular and Molecular Immunology Unit; Vrije Universiteit Brussel; Brussels Belgium
| |
Collapse
|
48
|
Role of resident liver cells in the pathogenesis of schistosomiasis. Trends Parasitol 2012; 28:572-9. [PMID: 23099112 DOI: 10.1016/j.pt.2012.09.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 09/14/2012] [Accepted: 09/21/2012] [Indexed: 12/12/2022]
Abstract
Pathology in schistosomiasis occurs as a result of eggs deposited in the liver by the schistosome parasite. A granulomatous reaction occurs, resulting in portal hypertension and hepatic fibrosis. Resident non-parenchymal cells within the liver take part in this process, including hepatic stellate cells, which are responsible for collagen production, and Kupffer cells, the liver macrophages involved in both host protection and in pathology. Other cells such as liver sinusoidal endothelial cells or portal fibroblasts may also be involved in this process. This review discusses the possible role of these resident liver cells in the pathology associated with schistosomiasis and provides information which may assist our understanding of the mechanisms associated with chronic liver disease in general.
Collapse
|
49
|
Wu J, Zhou M, Wan Y, Xu A. CD8+ T cells from vitiligo perilesional margins induce autologous melanocyte apoptosis. Mol Med Rep 2012; 7:237-41. [PMID: 23042234 PMCID: PMC3572717 DOI: 10.3892/mmr.2012.1117] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 08/08/2012] [Indexed: 11/16/2022] Open
Abstract
Cell-mediated autoimmunity has been suggested to be involved in the melanocyte apoptosis that occurs in vitiligo. We investigated the cytotoxicity to autologous melanocytes of CD8+ T cells from the perilesional margins and peripheral blood samples of vitiligo patients. CD8+ T cells isolated from skin biopsied from the edges of depigmented skin patches of vitiligo patients or from peripheral blood samples of the same donors were proliferated in culture medium. The primary cultures of CD8+ T cells and autologous melanocytes were mixed at ratios of 1:1, 1:2 or 1:5 and incubated for 3 days. The apoptosis of the melanocytes was analyzed by flow cytometry. Secreted cytokines in selected samples were measured by cytokine arrays. The results show that the CD8+ T cells were successfully isolated from the vitiligo perilesional margins. This cell population showed a significantly higher percentage of CD69 expression (56.13±3.55 versus 29.93±2.35%, p<0.01) and CD137 expression (41.74±1.06 versus 25.97±1.63%, p<0.01) compared with CD8+ T cells in peripheral blood from the same donors. The co-culturing of CD8+ T cells from lesional skin with autologous melanocytes induced apoptosis in the melanocytes (16.63±1.21, 16.71±0.63 and 18.32±1.60% for CD8+ T cells and autologous melanocytes at ratios of 1:1, 1:2 and 1:5, respectively). IL-6 levels were much higher in the co-culture (3.01-fold higher than in a melanocyte monoculture and 17.32-fold higher than in a CD8+ T-cell monoculture). The CD8+ T cells were also demonstrated to secrete more IL-13. Taken together, our data demonstrate that the infiltration of active CD8+ T cells takes place in the vitiligo perilesional margins. Those CD8+ T cells present significantly higher activation levels and higher cytotoxicity to autologous melanocytes than their counterparts from peripheral blood samples. These data suggest that CD8+ T cells are likely to be involved in the pathogenesis of vitiligo.
Collapse
Affiliation(s)
- Jilong Wu
- Department of Dermatology, The Third People's Hospital of Hangzhou, Hangzhou 310009, PR China
| | | | | | | |
Collapse
|
50
|
Raso P, Raso LAM, Melo FDA, Tafuri WL. Schistosoma mansoni granuloma in late evolutive phase, in a case of tumoral form in man. Rev Soc Bras Med Trop 2012; 45:627-32. [DOI: 10.1590/s0037-86822012000500016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 08/31/2012] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION: Authors describe human schistosomal granuloma in late chronic phase, from the morphological and evolutionary viewpoints. METHODS: The study was based on a histological analysis of two fragments obtained from a surgical biopsy of peritoneum and large intestine of a 42-year-old patient, with a pseudotumoral form mimicking a peritoneal carcinomatosis associated to the schistosomiasis hepatointestinal form. RESULTS: Two hundred and three granulomas were identified in the pseudotumor and 27 in the intestinal biopsy, with similar morphological features, most in the late chronic phase, in fibrotic healing. A new structural classification was suggested for granulomas: zone 1 (internal), 2 (intermediate) and 3 (external). CONCLUSIONS: Regarding granuloma as a whole, we may conclude that fibrosis is likely to be controlled by different and independent mechanisms in the three zones of the granuloma. Lamellar fibrosis in zone 3 seems to be controlled by matrix mesenchymal cells (fibroblasts and myoepithelial cells) and by inflammatory exudate cells (lymphocytes, plasmocytes, neutrophils, eosinophils). Annular fibrosis in zone 2, comprising a dense fibrous connective tissue, with few cells in the advanced phase, would be controlled by epithelioid cells involving zone 1 in recent granulomas. In zone 1, replacing periovular necrosis, an initialy loose and tracery connective neoformation, housing stellate cells or with fusiform nuclei, a dense paucicellular nodular connctive tissue emerges, probably induced by fibroblasts. In several granulomas, one of the zones is missing and granuloma is represented by two of them: Z3 and Z2, Z3 and Z1 or Z2 and Z1 and, ultimately, by a scar.
Collapse
|