101
|
Liu Z, Zhou Z, Dang Q, Xu H, Lv J, Li H, Han X. Immunosuppression in tumor immune microenvironment and its optimization from CAR-T cell therapy. Am J Cancer Res 2022; 12:6273-6290. [PMID: 36168626 PMCID: PMC9475465 DOI: 10.7150/thno.76854] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy represents a landmark advance in personalized cancer treatment. CAR-T strategy generally engineers T cells from a specific patient with a new antigen-specificity, which has achieved considerable success in hematological malignancies, but scarce benefits in solid tumors. Recent studies have demonstrated that tumor immune microenvironment (TIME) cast a profound impact on the immunotherapeutic response. The immunosuppressive landscape of TIME is a critical obstacle to the effector activity of CAR-T cells. Nevertheless, every cloud has a silver lining. The immunosuppressive components also shed new inspiration on reshaping a friendly TIME by targeting them with engineered CARs. Herein, we summarize recent advances in disincentives of TIME and discuss approaches and technologies to enhance CAR-T cell efficacy via addressing current hindrances. Simultaneously, we firmly believe that by parsing the immunosuppressive components of TIME, rationally manipulating the complex interactions of immunosuppressive components, and optimizing CAR-T cell therapy for each patient, the CAR-T cell immunotherapy responsiveness for solid malignancies will be substantially enhanced, and novel therapeutic targets will be revealed.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.,Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China.,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| | - Zhaokai Zhou
- Department of Pediatric Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinxiang Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huanyun Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.,Interventional Institute of Zhengzhou University, Zhengzhou, Henan 450052, China.,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan 450052, China
| |
Collapse
|
102
|
Jaén M, Martín-Regalado Á, Bartolomé RA, Robles J, Casal JI. Interleukin 13 receptor alpha 2 (IL13Rα2): Expression, signaling pathways and therapeutic applications in cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188802. [PMID: 36152905 DOI: 10.1016/j.bbcan.2022.188802] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/05/2022] [Accepted: 09/11/2022] [Indexed: 10/14/2022]
Abstract
Interleukin 13 receptor alpha 2 (IL13Rα2) is increasingly recognized as a relevant player in cancer invasion and metastasis. Despite being initially considered a decoy receptor for dampening the levels of interleukin 13 (IL-13) in diverse inflammatory conditions, accumulating evidences in the last decades indicate the capacity of IL13Rα2 for mediating IL-13 signaling in cancer cells. The biological reasons behind the expression of this receptor with such extremely high affinity for IL-13 in cancer cells remain unclear. Elevated expression of IL13Rα2 is commonly associated with invasion, late stage and cancer metastasis that results in poor prognosis for glioblastoma, colorectal or breast cancer, among others. The discovery of new mediators and effectors of IL13Rα2 signaling has been critical for deciphering its underlying molecular mechanisms in cancer progression. Still, many questions about the effects of inflammation, the cancer type and the tumor degree in the expression of IL13Rα2 remain largely uncharacterized. Here, we review and discuss the current status of the IL13Rα2 biology in cancer, with particular emphasis in the role of inflammation-driven expression and the regulation of different signaling pathways. As IL13Rα2 implications in cancer continue to grow exponentially, we highlight new targeted therapies recently developed for glioblastoma, colorectal cancer and other IL13Rα2-positive tumors.
Collapse
Affiliation(s)
- Marta Jaén
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Ángela Martín-Regalado
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Javier Robles
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain; Protein Alternatives SL, Tres Cantos, Madrid, Spain
| | - J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
103
|
Pan S, Li S, Zhan Y, Chen X, Sun M, Liu X, Wu B, Li Z, Liu B. Immune status for monitoring and treatment of bladder cancer. Front Immunol 2022; 13:963877. [PMID: 36159866 PMCID: PMC9492838 DOI: 10.3389/fimmu.2022.963877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022] Open
Abstract
The high recurrence rate of non-muscle invasive bladder cancer (BC) and poor prognosis of advanced BC are therapeutic challenges that need to be solved. Bacillus Calmette-Guerin (BCG) perfusion was the pioneer immunotherapy for early BC, and the discovery of immune checkpoint inhibitors has created a new chapter in the treatment of advanced BC. The benefit of immunotherapy is highly anticipated, but its effectiveness still needs to be improved. In this review, we collated and analysed the currently available information and explored the mechaisms by which the internal immune imbalance of BC leads to tumour progression. The relationship between immunity and progression and the prognosis of BC has been explored through tests using body fluids such as blood and urine. These analytical tests have attempted to identify specific immuyne cells and cytokines to predict treatment outcomes and recurrence. The diversity and proportion of immune and matrix cells in BC determine the heterogeneity and immune status of tumours. The role and classification of immune cells have also been redefined, e.g., CD4 cells having recognised cytotoxicity in BC. Type 2 immunity, including that mediated by M2 macrophages, Th2 cells, and interleukin (IL)-13, plays an important role in the recurrence and progression of BC. Pathological fibrosis, activated by type 2 immunity and cancer cells, enhances the rate of cancer progression and irreversibility. Elucidating the immune status of BC and clarifying the mechanisms of action of different cells in the tumour microenvironment is the research direction to be explored in the future.
Collapse
Affiliation(s)
- Shen Pan
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shijie Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yunhong Zhan
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaonan Chen
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming Sun
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuefeng Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bin Wu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhenhua Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bitian Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Bitian Liu, ;
| |
Collapse
|
104
|
Spagnolo P, Tonelli R, Samarelli AV, Castelli G, Cocconcelli E, Petrarulo S, Cerri S, Bernardinello N, Clini E, Saetta M, Balestro E. The role of immune response in the pathogenesis of idiopathic pulmonary fibrosis: far beyond the Th1/Th2 imbalance. Expert Opin Ther Targets 2022; 26:617-631. [PMID: 35983984 DOI: 10.1080/14728222.2022.2114897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION . Idiopathic pulmonary fibrosis (IPF) is a chronic disease of unknown origin characterized by progressive scarring of the lung leading to irreversible loss of function. Despite the availability of two drugs that are able to slow down disease progression, IPF remains a deadly disease. The pathogenesis of IPF is poorly understood, but a dysregulated wound healing response following recurrent alveolar epithelial injury is thought to be crucial. Areas covered. In the last few years, the role of the immune system in IPF pathobiology has been reconsidered; indeed, recent data suggest that a dysfunctional immune system may promote and unfavorable interplay with pro-fibrotic pathways thus acting as a cofactor in disease development and progression. In this article, we review and critically discuss the role of T cells in the pathogenesis and progression of IPF in the attempt to highlight ways in which further research in this area may enable the development of targeted immunomodulatory therapies for this dreadful disease. EXPERT OPINION A better understanding of T cells interactions has the potential to facilitate the development of immune modulators targeting multiple T cell-mediated pathways thus halting disease initiation and progression.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Roberto Tonelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy.,Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Gioele Castelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Cocconcelli
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Simone Petrarulo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Stefania Cerri
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicol Bernardinello
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Enrico Clini
- Laboratory of Cell Therapies and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults University Hospital of Modena and Reggio Emilia, Modena, Italy.,University Hospital of Modena, Respiratory Diseases Unit, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Marina Saetta
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Balestro
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| |
Collapse
|
105
|
HSP70 upregulation in nasal mucosa of symptomatic children with allergic rhinitis and potential risk of asthma development. Sci Rep 2022; 12:14104. [PMID: 35982171 PMCID: PMC9388484 DOI: 10.1038/s41598-022-18443-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022] Open
Abstract
Allergic rhinitis and asthma are the most common causes of chronic inflammation of the upper and lower airways in childhood. However, a nasal biomarker that can link to pulmonary inflammation is yet to be found. The present paper aims to investigate the possible role in inflammation of two inducible 70-kDa Heat Shock Proteins (HSP70) members, HSPA1A/B and HSPA6, in nasal mucosa cells of allergic children through their mRNA expression analysis, and their correlation to both spirometric and FeNO values. The relationship between FeNO in lower airways and ∆Cts of HSPA1A/B in nasal mucosa seems to be influenced by clinical symptoms regardless of age, sex, and sensitization patterns. Therefore, HSP70 expression, as well as FeNO levels, could have a predictive capability to identify lower airways inflammation and thus to recognize rhinitic children having a potential risk of asthma development.
Collapse
|
106
|
Kokubo K, Onodera A, Kiuchi M, Tsuji K, Hirahara K, Nakayama T. Conventional and pathogenic Th2 cells in inflammation, tissue repair, and fibrosis. Front Immunol 2022; 13:945063. [PMID: 36016937 PMCID: PMC9395650 DOI: 10.3389/fimmu.2022.945063] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/18/2022] [Indexed: 12/15/2022] Open
Abstract
Type 2 helper T (Th2) cells, a subset of CD4+ T cells, play an important role in the host defense against pathogens and allergens by producing Th2 cytokines, such as interleukin-4 (IL-4), IL-5, and IL-13, to trigger inflammatory responses. Emerging evidence reveals that Th2 cells also contribute to the repair of injured tissues after inflammatory reactions. However, when the tissue repair process becomes chronic, excessive, or uncontrolled, pathological fibrosis is induced, leading to organ failure and death. Thus, proper control of Th2 cells is needed for complete tissue repair without the induction of fibrosis. Recently, the existence of pathogenic Th2 (Tpath2) cells has been revealed. Tpath2 cells produce large amounts of Th2 cytokines and induce type 2 inflammation when activated by antigen exposure or tissue injury. In recent studies, Tpath2 cells are suggested to play a central role in the induction of type 2 inflammation whereas the role of Tpath2 cells in tissue repair and fibrosis has been less reported in comparison to conventional Th2 cells. In this review, we discuss the roles of conventional Th2 cells and pathogenic Th2 cells in the sequence of tissue inflammation, repair, and fibrosis.
Collapse
Affiliation(s)
- Kota Kokubo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
| | - Masahiro Kiuchi
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kaori Tsuji
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- *Correspondence: Kiyoshi Hirahara, ; Toshinori Nakayama,
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- AMED-CREST, AMED, Chiba, Japan
- *Correspondence: Kiyoshi Hirahara, ; Toshinori Nakayama,
| |
Collapse
|
107
|
Deng R, Zheng H, Cai H, Li M, Shi Y, Ding S. Effects of helicobacter pylori on tumor microenvironment and immunotherapy responses. Front Immunol 2022; 13:923477. [PMID: 35967444 PMCID: PMC9371381 DOI: 10.3389/fimmu.2022.923477] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/04/2022] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori is closely associated with gastric cancer. During persistent infection, Helicobacter pylori can form a microenvironment in gastric mucosa which facilitates the survival and colony formation of Helicobacter pylori. Tumor stromal cells are involved in this process, including tumor-associated macrophages, mesenchymal stem cells, cancer-associated fibroblasts, and myeloid-derived suppressor cells, and so on. The immune checkpoints are also regulated by Helicobacter pylori infection. Helicobacter pylori virulence factors can also act as immunogens or adjuvants to elicit or enhance immune responses, indicating their potential applications in vaccine development and tumor immunotherapy. This review highlights the effects of Helicobacter pylori on the immune microenvironment and its potential roles in tumor immunotherapy responses.
Collapse
Affiliation(s)
- Ruiyi Deng
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University First Medical School, Beijing, China
| | - Huiling Zheng
- Peking University Third Hospital, Department of Gastroenterology, Beijing, China
| | - Hongzhen Cai
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University First Medical School, Beijing, China
| | - Man Li
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
- Peking University Health Science Center, Peking University Third Medical School, Beijing, China
| | - Yanyan Shi
- Peking University Third Hospital, Research Center of Clinical Epidemiology, Beijing, China
| | - Shigang Ding
- Peking University Third Hospital, Department of Gastroenterology, Beijing, China
| |
Collapse
|
108
|
An In Vitro and In Vivo Translational Research Approach for the Assessment of Sensitization Capacity and Residual Allergenicity of an Extensive Whey Hydrolysate for Cow’s Milk-Allergic Infants. Foods 2022; 11:foods11142005. [PMID: 35885248 PMCID: PMC9320663 DOI: 10.3390/foods11142005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/30/2022] [Accepted: 07/05/2022] [Indexed: 12/10/2022] Open
Abstract
Introduction: Hypoallergenic formulas prepared from hydrolyzed cow’s milk proteins are often used for the management of cow’s milk allergy (CMA) in infants. In this study, both in vitro assays and an in vivo mouse model for CMA were used to assess the sensitizing and allergenic potential of a newly developed, extensive whey hydrolysate (eWH). Methods: Gel permeation chromatography was used to characterize the molecular weight distribution of the peptides. Residual antigenicity was measured using a beta-lactoglobulin ELISA as well as with immunoblotting using anti-beta-lactoglobulin (BLG) and anti-alpha-lactalbumin antibodies. In vitro residual allergenicity was assessed using huFcεRIα-RBL-2H3 cells sensitized with anti-bovine BLG human IgE. In vivo sensitizing and allergenic potential was assessed in a CMA mouse model by measuring the acute allergic skin response, anaphylactic shock score, body temperature, serum mMCP-1, whey-specific IgE, and cytokines. Results: There was no in vitro residual antigenicity and allergenicity observed of the eWH. Mice sensitized with eWH showed no acute allergic skin reaction after challenge with whey, confirmed by an absence of whey-specific IgE and anaphylactic symptoms and decrease in body temperature and mMCP-1 levels. Conclusions: Results from our in vitro and in vivo translational approach to assess sensitization capacity and residual allergenicity indicate that the newly developed eWH is safe for use in CMA infants. This was subsequently confirmed in a clinical study in which this eWH was tolerated by more than 90% (with 95% confidence) of infants or children with confirmed CMA.
Collapse
|
109
|
Haddad EB, Cyr SL, Arima K, McDonald RA, Levit NA, Nestle FO. Current and Emerging Strategies to Inhibit Type 2 Inflammation in Atopic Dermatitis. Dermatol Ther (Heidelb) 2022; 12:1501-1533. [PMID: 35596901 PMCID: PMC9276864 DOI: 10.1007/s13555-022-00737-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 12/30/2022] Open
Abstract
Type 2 immunity evolved to combat helminth infections by orchestrating a combined protective response of innate and adaptive immune cells and promotion of parasitic worm destruction or expulsion, wound repair, and barrier function. Aberrant type 2 immune responses are associated with allergic conditions characterized by chronic tissue inflammation, including atopic dermatitis (AD) and asthma. Signature cytokines of type 2 immunity include interleukin (IL)-4, IL-5, IL-9, IL-13, and IL-31, mainly secreted from immune cells, as well as IL-25, IL-33, and thymic stromal lymphopoietin, mainly secreted from tissue cells, particularly epithelial cells. IL-4 and IL-13 are key players mediating the prototypical type 2 response; IL-4 initiates and promotes differentiation and proliferation of naïve T-helper (Th) cells toward a Th2 cell phenotype, whereas IL-13 has a pleiotropic effect on type 2 inflammation, including, together with IL-4, decreased barrier function. Both cytokines are implicated in B-cell isotype class switching to generate immunoglobulin E, tissue fibrosis, and pruritus. IL-5, a key regulator of eosinophils, is responsible for eosinophil growth, differentiation, survival, and mobilization. In AD, IL-4, IL-13, and IL-31 are associated with sensory nerve sensitization and itch, leading to scratching that further exacerbates inflammation and barrier dysfunction. Various strategies have emerged to suppress type 2 inflammation, including biologics targeting cytokines or their receptors, and Janus kinase inhibitors that block intracellular cytokine signaling pathways. Here we review type 2 inflammation, its role in inflammatory diseases, and current and future therapies targeting type 2 pathways, with a focus on AD. INFOGRAPHIC.
Collapse
Affiliation(s)
| | - Sonya L Cyr
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | | | - Noah A Levit
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | |
Collapse
|
110
|
Huan X, Zhao R, Song J, Zhong H, Su M, Yan C, Wang Y, Chen S, Zhou Z, Lu J, Xi J, Luo S, Zhao C. Increased serum IL-2, IL-4, IL-5 and IL-12p70 levels in AChR subtype generalized myasthenia gravis. BMC Immunol 2022; 23:26. [PMID: 35624411 PMCID: PMC9145157 DOI: 10.1186/s12865-022-00501-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 05/20/2022] [Indexed: 12/04/2022] Open
Abstract
Background Myasthenia gravis (MG) is an autoimmune disorder affecting neuromuscular junctions. Cytokines play important roles in facilitating the immune response and augmenting the pathogenic antibody production. The current study aims to sensitively characterize the serum levels of cytokines with very low concentration in generalized MG (gMG). Methods Using ultrasensitive single-molecule arrays (SIMOA), we measured serum IL-2, IL-4, IL-5 and IL-12p70 in 228 participants including 152 immunotherapy-naïve anti-acetylcholine receptor (AChR) subtype gMG from Huashan MG registry and 76 age-matched healthy controls. Subgroup analysis was then performed by stratifying patients according to the onset ages, MGFA classification, disease duration at baseline. Results Serum IL-2, IL-4, IL-5 and IL-12p70 levels were significantly elevated in gMG compared to controls (0.179 pg/mL versus 0.011 pg/mL, P < 0.0001; 0.029 pg/mL versus 0.018 pg/mL, P = 0.0259; 0.215 pg/mL versus 0.143 pg/mL, P = 0.0007; 0.132 pg/mL versus 0.118 pg/mL, P = 0.0401). Subgroup analysis revealed that IL-2 levels were slightly elevated in gMG with MGFA II compared to MGFA III/IV (0.195 pg/mL versus 0.160 pg/mL, P = 0.022), as well as elevated levels of IL-2 (0.220 pg/mL versus 0.159 pg/mL, P = 0.0002) and IL-5 (0.251 pg/mL versus 0.181 pg/mL, P = 0.004) in late-onset gMG compared with the early-onset gMG. gMG patients with a long duration had a significant increased serum IL-12p70 than those with a short duration (0.163 pg/mL versus 0.120 pg/mL, P = 0.011). Conclusion Serum IL-2, IL-4, IL-5 and IL-12p70 levels were increased in AChR subtype gMG using ultrasensitive measurement. Serum cytokines with very low concentrations may provide as potential biomarkers in stratifying gMG patients in future prospective cohort studies.
Collapse
Affiliation(s)
- Xiao Huan
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.,National Center for Neurological Disorders, Shanghai, China
| | - Rui Zhao
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.,National Center for Neurological Disorders, Shanghai, China
| | - Jie Song
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.,National Center for Neurological Disorders, Shanghai, China
| | - Huahua Zhong
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.,National Center for Neurological Disorders, Shanghai, China
| | - Manqiqige Su
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.,National Center for Neurological Disorders, Shanghai, China
| | - Chong Yan
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.,National Center for Neurological Disorders, Shanghai, China
| | - Ying Wang
- Department of Pharmacy, Huashan Hospital Fudan University, Shanghai, China
| | - Sheng Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhirui Zhou
- Radiation Oncology Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiahong Lu
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.,National Center for Neurological Disorders, Shanghai, China
| | - Jianying Xi
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China.,National Center for Neurological Disorders, Shanghai, China
| | - Sushan Luo
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China. .,National Center for Neurological Disorders, Shanghai, China.
| | - Chongbo Zhao
- Huashan Rare Disease Center and Department of Neurology, Huashan Hospital Fudan University, No. 12 Middle Wulumuqi Road, Shanghai, 200040, China. .,National Center for Neurological Disorders, Shanghai, China.
| |
Collapse
|
111
|
Tiwari D, Ahuja N, Kumar S, Kalra R, Nanduri R, Gupta S, Khare AK, Bhagyaraj E, Arora R, Gupta P. Nuclear receptor Nr1d1 alleviates asthma by abating GATA3 gene expression and Th2 cell differentiation. Cell Mol Life Sci 2022; 79:308. [PMID: 35596832 PMCID: PMC11073070 DOI: 10.1007/s00018-022-04323-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 03/30/2022] [Accepted: 04/21/2022] [Indexed: 11/03/2022]
Abstract
Nuclear receptors are a unique family of transcription factors that play cardinal roles in physiology and plethora of human diseases. The adopted orphan nuclear receptor Nr1d1 is a constitutive transcriptional repressor known to modulate several biological processes. In this study, we found that Nr1d1 plays a decisive role in T helper (Th)-cell polarization and transcriptionally impedes the formation of Th2 cells by directly binding to the promoter region of GATA binding protein 3 (GATA3) gene. Nr1d1 interacts with its cellular companion, the nuclear receptor corepressor and histone deacetylase 3 to form a stable repression complex on the GATA3 promoter. The presence of Nr1d1 also imparts protection against associated inflammatory responses in murine model of asthma and its ligand SR9011 eased disease severity by suppressing Th2 responses. Moreover, Chip-seq profiling uncovered Nr1d1 interactions with other gene subsets that impedes Th2-linked pathways and regulates metabolism, immunity and brain functions, therefore, providing empirical evidence regarding the genetic link between asthma and other comorbid conditions. Thus, Nr1d1 emerges as a molecular switch that could be targeted to subdue asthma.
Collapse
Affiliation(s)
- Drishti Tiwari
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India.
| | - Nancy Ahuja
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Sumit Kumar
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Rashi Kalra
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ravikanth Nanduri
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Shalini Gupta
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Asheesh Kumar Khare
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Ella Bhagyaraj
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
- Department of Infectious Disease and Immunology, University of Florida, Gainesville, FL, USA
| | - Rashmi Arora
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Pawan Gupta
- Department of Molecular Immunology, Council of Scientific and Industrial Research, Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India.
| |
Collapse
|
112
|
Williams TC, Loo SL, Nichol KS, Reid AT, Veerati PC, Esneau C, Wark PAB, Grainge CL, Knight DA, Vincent T, Jackson CL, Alton K, Shimkets RA, Girkin JL, Bartlett NW. IL-25 blockade augments antiviral immunity during respiratory virus infection. Commun Biol 2022; 5:415. [PMID: 35508632 PMCID: PMC9068710 DOI: 10.1038/s42003-022-03367-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/13/2022] [Indexed: 12/12/2022] Open
Abstract
IL-25 is implicated in the pathogenesis of viral asthma exacerbations. However, the effect of IL-25 on antiviral immunity has yet to be elucidated. We observed abundant expression and colocalization of IL-25 and IL-25 receptor at the apical surface of uninfected airway epithelial cells and rhinovirus infection increased IL-25 expression. Analysis of immune transcriptome of rhinovirus-infected differentiated asthmatic bronchial epithelial cells (BECs) treated with an anti-IL-25 monoclonal antibody (LNR125) revealed a re-calibrated response defined by increased type I/III IFN and reduced expression of type-2 immune genes CCL26, IL1RL1 and IL-25 receptor. LNR125 treatment also increased type I/III IFN expression by coronavirus infected BECs. Exogenous IL-25 treatment increased viral load with suppressed innate immunity. In vivo LNR125 treatment reduced IL-25/type 2 cytokine expression and increased IFN-β expression and reduced lung viral load. We define a new immune-regulatory role for IL-25 that directly inhibits virus induced airway epithelial cell innate anti-viral immunity.
Collapse
Affiliation(s)
- Teresa C Williams
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Su-Ling Loo
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Kristy S Nichol
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Andrew T Reid
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Punnam C Veerati
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Camille Esneau
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Peter A B Wark
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Christopher L Grainge
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Darryl A Knight
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- UBC Providence Health Care Research Institute, Vancouver, BC, Canada
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Thomas Vincent
- Abeome Corporation/Lanier Biotherapeutics, Athens, GA, USA
| | | | - Kirby Alton
- Abeome Corporation/Lanier Biotherapeutics, Athens, GA, USA
| | | | - Jason L Girkin
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Nathan W Bartlett
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia.
| |
Collapse
|
113
|
Sylvester M, Son A, Schwartz DM. The Interactions Between Autoinflammation and Type 2 Immunity: From Mechanistic Studies to Epidemiologic Associations. Front Immunol 2022; 13:818039. [PMID: 35281022 PMCID: PMC8907424 DOI: 10.3389/fimmu.2022.818039] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/02/2022] [Indexed: 12/30/2022] Open
Abstract
Autoinflammatory diseases are a group of clinical syndromes characterized by constitutive overactivation of innate immune pathways. This results in increased production of or responses to monocyte- and neutrophil-derived cytokines such as interleukin-1β (IL-1β), Tumor Necrosis Factor-α (TNF-α), and Type 1 interferon (IFN). By contrast, clinical allergy is caused by dysregulated type 2 immunity, which is characterized by expansion of T helper 2 (Th2) cells and eosinophils, as well as overproduction of the associated cytokines IL-4, IL-5, IL-9, and IL-13. Traditionally, type 2 immune cells and autoinflammatory effectors were thought to counter-regulate each other. However, an expanding body of evidence suggests that, in some contexts, autoinflammatory pathways and cytokines may potentiate type 2 immune responses. Conversely, type 2 immune cells and cytokines can regulate autoinflammatory responses in complex and context-dependent manners. Here, we introduce the concepts of autoinflammation and type 2 immunity. We proceed to review the mechanisms by which autoinflammatory and type 2 immune responses can modulate each other. Finally, we discuss the epidemiology of type 2 immunity and clinical allergy in several monogenic and complex autoinflammatory diseases. In the future, these interactions between type 2 immunity and autoinflammation may help to expand the spectrum of autoinflammation and to guide the management of patients with various autoinflammatory and allergic diseases.
Collapse
Affiliation(s)
- McKella Sylvester
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Aran Son
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniella M Schwartz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
114
|
Zeng Z, Xu X, Zhu Y, Wang Q, Zhang Y, Huo X. Pb and Cd exposure linked with Il-10 and Il-13 gene polymorphisms in asthma risk relevant immunomodulation in children. CHEMOSPHERE 2022; 294:133656. [PMID: 35051511 DOI: 10.1016/j.chemosphere.2022.133656] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/05/2023]
Abstract
Genetic variants and environmental factor of heavy metal exposure accelerate the risk of immune-mediated respiratory diseases, including asthma in children. This study aims to investigate the effects and interaction of Pb, Cd exposure from e-waste and interleukin (IL)-10, IL-13 gene polymorphisms on interleukin expressions in children. Pb, Cd levels, Il-10 (rs1800871, rs1800872, rs1800896) and Il-13 (rs20541, rs1800925) polymorphisms were determined in blood or urine of 155 children (75 from e-waste-exposed area, Guiyu and 80 from reference area, Haojiang). Blood Pb and urine Cd levels of e-waste-exposed children were both higher (mean: 5.89 vs. 3.35 μg/dL; 6.04 vs. 1.82 μg/g, both P < 0.05). Exposed children had a larger proportion of high blood Pb level (> 5.00 μg/dL) (66.67% vs. 4.11%, P < 0.001), but they had no statistically different proportion of high urine Cd level (> 1.00 μg/g creatinine). Median levels of IL-13 decreased (3.674 vs. 4.410 ng/L, P < 0.01), but of IL-10 did not vary in serum of exposed children. The analyses revealed no significant associations of Pb or Cd with Il-10 or Il-13 gene polymorphisms. The high blood Pb and urine Cd level were respectively associated with the low IL-13 ( < 3.696 ng/L) and low IL-10 ( < 0.361 ng/L) level, after adjustment for children's age, gender, and BMI (both P ≤ 0.05). Children homozygous carrying major allele for Il-13 (rs20541 and rs1800925) had additive interactions with high blood Pb level on low IL-13 expression (OR = 5.37, 95% CI: 1.96, 14.73 and OR = 8.45, 95% CI: 2.61, 27.32; both P ≤ 0.001). In contrast, no interaction was observed amongst Pb or Cd with Il-10 gene polymorphisms on its expression. Our findings suggest that Pb exposure interacting with Il-13 gene polymorphisms negatively regulates IL-13 expression, which may pose a risk to abnormal asthma-relevant immunomodulation in preschool children.
Collapse
Affiliation(s)
- Zhijun Zeng
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China; Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Yongzhan Zhu
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Qihua Wang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China
| | - Yuling Zhang
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, Guangzhou, 511443, Guangdong, China.
| |
Collapse
|
115
|
Yang YQ, Chen H, Qiu LR, Zhu RF. Case Report: The Application of Dupilumab in Atopic Dermatitis Children Complicated With Nephrotic Syndrome. Front Med (Lausanne) 2022; 9:813313. [PMID: 35573014 PMCID: PMC9099359 DOI: 10.3389/fmed.2022.813313] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/13/2022] [Indexed: 12/01/2022] Open
Abstract
Nephrotic syndrome (NS) tends to be more common in patients with history of allergies. Atopic dermatitis (AD) is one of the most common allergic diseases in children. Dupilumab, a dual IL-4 and IL-13 inhibitor, has been widely used to treat AD patients. However, the efficacy and safety of Dupilumab in NS is unclear. We reported two AD patients with NS comorbidities treated with Dupilumab. The outcomes showed the good control of NS and less systemic steroids and/or immunosuppressive agents use during the Dupilumab treatment period, accompanied by significant relief of AD symptoms. We suggest prospective pilot studies and randomized controlled trials could be carried out to validate the efficacy and safety of Dupilumab in the treatment of NS patients.
Collapse
Affiliation(s)
- Ya-Qi Yang
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Chen
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Ru Qiu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Li-Ru Qiu
| | - Rong-Fei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Rong-Fei Zhu
| |
Collapse
|
116
|
Oyesola OO, Souza COS, Loke P. The Influence of Genetic and Environmental Factors and Their Interactions on Immune Response to Helminth Infections. Front Immunol 2022; 13:869163. [PMID: 35572520 PMCID: PMC9103684 DOI: 10.3389/fimmu.2022.869163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/04/2022] [Indexed: 12/20/2022] Open
Abstract
Helminth infection currently affect over 2 billion people worldwide, with those with the most pathologies and morbidities, living in regions with unequal and disproportionate access to effective healthcare solutions. Host genetics and environmental factors play critical roles in modulating and regulating immune responses following exposure to various pathogens and insults. However, the interplay of environment and genetic factors in influencing who gets infected and the establishment, persistence, and clearance of helminth parasites remains unclear. Inbred strains of mice have long been used to investigate the role of host genetic factors on pathogenesis and resistance to helminth infection in a laboratory setting. This review will discuss the use of ecological and environmental mouse models to study helminth infections and how this could be used in combination with host genetic variation to explore the relative contribution of these factors in influencing immune response to helminth infections. Improved understanding of interactions between genetics and the environment to helminth immune responses would be important for efforts to identify and develop new prophylactic and therapeutic options for the management of helminth infections and their pathogenesis.
Collapse
Affiliation(s)
- Oyebola O. Oyesola
- Laboratory of Parasitic Disease, National Institute of Allergy and Infectious Disease (NIAID), National Institute of Health, Bethesda, MD, United States
| | | | | |
Collapse
|
117
|
Guo B, Dong W, Huo J, Sun G, Qin Z, Liu X, Zhang B, Wang W. Integrated Metabolomics and Network Pharmacology Analysis Immunomodulatory Mechanisms of Qifenggubiao Granules. Front Pharmacol 2022; 13:828175. [PMID: 35479328 PMCID: PMC9037242 DOI: 10.3389/fphar.2022.828175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/07/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Qifenggubiao granules (QFGBG) is a new Chinese medicine independently developed by Heilongjiang Academy of Traditional Chinese Medicine, which combines the essence of Yupingfeng powder and Shengmai yin (invention patent number: CN1325098C, approval number: Sinopharm Zhunzi B20020410), and has been included in the 2020 edition of Chinese Pharmacopoeia. It has remarkable pharmacodynamic results and conclusive clinical effects in the treatment of allergic rhinitis, chronic cough and other diseases. Previous pharmacological studies have shown that it has immunomodulatory effect, but its immunomodulatory mechanism is still unclear. Methods: In this study, cyclophosphamide (CTX) was used to establish the immune hypofunction model in mice, and the weight change, index of immune organs in spleen and thymus, pathological sections of immune organs and inflammatory factors were used to evaluate the model. Based on the metabolic biomarkers obtained by metabonomics technology, the potential targets of Qifeng Gubiao Granule immunomodulation were obtained by integrating the targets of blood components, metabolites and diseases through network pharmacology. Meanwhile, GO enrichment analysis and KEGG pathway analysis were carried out on the potential targets. Results: QFGBG can increase body weight and organ index, and recover immune organ damage caused by CP. Metabonomics identified 13 metabolites with significant changes, among which the level of phospholipid (PC) metabolites decreased significantly in the model group. Sphingosine -1- phosphate, 1- palmitoyl phosphatidylcholine [LysoPC (16:0/0:0)] and other metabolites were significantly increased in the model group, and 98 targets of Qifeng’s external immune regulation were obtained by intersecting 629 component targets, 202 metabolite targets and 1916 disease targets. KEGG pathway analysis obtained 233 related metabolic pathways, and the top 20 metabolic pathways mainly involved IL-17 signaling pathway, TNF signaling pathway, Sphingolipid signaling pathway, and so on. Conclusion: QFGBG may act on AKT1, IL6, MAPK3, PTGS2, CASP3, MAPK1, ESR1, PPARG, HSP90AA1, PPARA and other targets, acting through Sphingolipid signaling pathway and signaling pathway. Combined with pharmacodynamic evaluation, the immunomodulatory effect of QFGBG was confirmed, and the immunomodulatory mechanism of QFGBG with multiple targets and multiple pathways was preliminarily clarified.
Collapse
Affiliation(s)
- Bindan Guo
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Wenting Dong
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Jinhai Huo
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Guodong Sun
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Zhiwei Qin
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Xiaodong Liu
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Bihai Zhang
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| | - Weiming Wang
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin, China
| |
Collapse
|
118
|
Zhao M, Ren K, Xiong X, Xin Y, Zou Y, Maynard JC, Kim A, Battist AP, Koneripalli N, Wang Y, Chen Q, Xin R, Yang C, Huang R, Yu J, Huang Z, Zhang Z, Wang H, Wang D, Xiao Y, Salgado OC, Jarjour NN, Hogquist KA, Revelo XS, Burlingame AL, Gao X, von Moltke J, Lin Z, Ruan HB. Epithelial STAT6 O-GlcNAcylation drives a concerted anti-helminth alarmin response dependent on tuft cell hyperplasia and Gasdermin C. Immunity 2022; 55:623-638.e5. [PMID: 35385697 PMCID: PMC9109499 DOI: 10.1016/j.immuni.2022.03.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/25/2022] [Accepted: 03/14/2022] [Indexed: 12/14/2022]
Abstract
The epithelium is an integral component of mucosal barrier and host immunity. Following helminth infection, the intestinal epithelial cells secrete "alarmin" cytokines, such as interleukin-25 (IL-25) and IL-33, to initiate the type 2 immune responses for helminth expulsion and tolerance. However, it is unknown how helminth infection and the resulting cytokine milieu drive epithelial remodeling and orchestrate alarmin secretion. Here, we report that epithelial O-linked N-Acetylglucosamine (O-GlcNAc) protein modification was induced upon helminth infections. By modifying and activating the transcription factor STAT6, O-GlcNAc transferase promoted the transcription of lineage-defining Pou2f3 in tuft cell differentiation and IL-25 production. Meanwhile, STAT6 O-GlcNAcylation activated the expression of Gsdmc family genes. The membrane pore formed by GSDMC facilitated the unconventional secretion of IL-33. GSDMC-mediated IL-33 secretion was indispensable for effective anti-helminth immunity and contributed to induced intestinal inflammation. Protein O-GlcNAcylation can be harnessed for future treatment of type 2 inflammation-associated human diseases.
Collapse
Affiliation(s)
- Ming Zhao
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kaiqun Ren
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; College of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Xiwen Xiong
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yue Xin
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yujie Zou
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jason C Maynard
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Angela Kim
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Alexander P Battist
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Navya Koneripalli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Yusu Wang
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Qianyue Chen
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Ruyue Xin
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Chenyan Yang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Rong Huang
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jiahui Yu
- School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zan Huang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Zengdi Zhang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Haiguang Wang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Daoyuan Wang
- College of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Yihui Xiao
- College of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Oscar C Salgado
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Nicholas N Jarjour
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kristin A Hogquist
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xavier S Revelo
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Alma L Burlingame
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California, USA
| | - Xiang Gao
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Zhaoyu Lin
- MOE Key Laboratory of Model Animals for Disease Study, State Key Laboratory of Pharmaceutical Biotechnology, Model Animal Research Center, National Resource Center for Mutant Mice of China, Nanjing Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China.
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA.
| |
Collapse
|
119
|
West Nile Virus Neuroinfection in Humans: Peripheral Biomarkers of Neuroinflammation and Neuronal Damage. Viruses 2022; 14:v14040756. [PMID: 35458486 PMCID: PMC9027124 DOI: 10.3390/v14040756] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 01/27/2023] Open
Abstract
Among emerging arthropod-borne viruses (arbovirus), West Nile virus (WNV) is a flavivirus that can be associated with severe neuroinvasive infections in humans. In 2018, the European WNV epidemic resulted in over 2000 cases, representing the most important arboviral epidemic in the European continent. Characterization of inflammation and neuronal biomarkers released during WNV infection, especially in the context of neuronal impairments, could provide insight into the development of predictive tools that could be beneficial for patient outcomes. We first analyzed the inflammatory signature in the serum of WNV-infected mice and found increased concentrations of several inflammatory cytokines. We next analyzed serum and cerebrospinal-fluid (CSF) samples from a cohort of patients infected by WNV between 2018 and 2019 in Hungary to quantify a large panel of inflammatory cytokines and neurological factors. We found higher levels of inflammatory cytokines (e.g., IL4, IL6, and IL10) and neuronal factors (e.g., BDNF, GFAP, MIF, TDP-43) in the sera of WNV-infected patients with neuroinvasive disease. Furthermore, the serum inflammatory profile of these patients persisted for several weeks after initial infection, potentially leading to long-term sequelae and having a deleterious effect on brain neurovasculature. This work suggests that early signs of increased serum concentrations of inflammatory cytokines and neuronal factors could be a signature underlying the development of severe neurological impairments. Biomarkers could play an important role in patient monitoring to improve care and prevent undesirable outcomes.
Collapse
|
120
|
Czaja AJ. Immune Inhibitory Properties and Therapeutic Prospects of Transforming Growth Factor-Beta and Interleukin 10 in Autoimmune Hepatitis. Dig Dis Sci 2022; 67:1163-1186. [PMID: 33835375 DOI: 10.1007/s10620-021-06968-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
Transforming growth factor-beta and interleukin 10 have diverse immune inhibitory properties that have restored homeostatic defense mechanisms in experimental models of autoimmune disease. The goals of this review are to describe the actions of each cytokine, review their investigational use in animal models and patients, and indicate their prospects as interventions in autoimmune hepatitis. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Transforming growth factor-beta expands the natural and inducible populations of regulatory T cells, limits the proliferation of natural killer cells, suppresses the activation of naïve CD8+ T cells, decreases the production of interferon-gamma, and stimulates fibrotic repair. Interleukin 10 selectively inhibits the CD28 co-stimulatory signal for antigen recognition and impairs antigen-specific activation of uncommitted CD4+ and CD8+ T cells. It also inhibits maturation of dendritic cells, suppresses Th17 cells, supports regulatory T cells, and limits production of diverse pro-inflammatory cytokines. Contradictory immune stimulatory effects have been associated with each cytokine and may relate to the dose and accompanying cytokine milieu. Experimental findings have not translated into successful early clinical trials. The recombinant preparation of each agent in low dosage has been safe in human studies. In conclusion, transforming growth factor-beta and interleukin 10 have powerful immune inhibitory actions of potential therapeutic value in autoimmune hepatitis. The keys to their therapeutic application will be to match their predominant non-redundant function with the pivotal pathogenic mechanism or cytokine deficiency and to avoid contradictory immune stimulatory actions.
Collapse
Affiliation(s)
- Albert J Czaja
- Professor Emeritus of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
121
|
Yuan X, Chinnaswamy K, Stuckey JA, Yang CY. Computational Cosolvent Mapping Analysis Leads to Identify Salicylic Acid Analogs as Weak Inhibitors of ST2 and IL33 Binding. J Phys Chem B 2022; 126:2394-2406. [PMID: 35294837 PMCID: PMC9354565 DOI: 10.1021/acs.jpcb.2c00341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cytokine signaling initiated by the binding of the cytokine receptors to cytokines plays important roles in immune regulation and diseases. Structurally, cytokine receptors interact with cytokines via an extensive, rugged interface that represents a challenge in inhibitor development. Our computational analysis has previously indicated that butyric acid, mimicking acidic residues, preferentially binds to sites in ST2 (Stimulation-2) that interact with acidic residues of IL33, the endogenous cytokine for ST2. To investigate if a charged group in small molecules facilitates ligand binding to ST2, we developed a biochemical homogeneous time resolved fluorescence assay to determine the inhibition of ST2/IL33 binding by five molecules containing an aromatic ring and a charged group. Three molecules, including niacin, salicylic acid, and benzamidine, exhibit inhibition activities at millimolar concentrations. We further employed the computational cosolvent mapping analysis to identify a shared mode of interaction between niacin, salicylic acid, and ST2. The mode of interaction was further confirmed by four analogous compounds that exhibited similar or improved activities. Our study provided the evidence of inhibition of ST2 and IL33 binding by salicylic acid and analogs. The results suggest that biological activity of salicylic acid may be partly mediated through modulating extracellular cytokine receptors and cytokine interaction.
Collapse
Affiliation(s)
- Xinrui Yuan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | | | - Jeanne A Stuckey
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Chao-Yie Yang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
122
|
Steinhoff M, Ahmad F, Pandey A, Datsi A, AlHammadi A, Al-Khawaga S, Al-Malki A, Meng J, Alam M, Buddenkotte J. Neuro-immune communication regulating pruritus in atopic dermatitis. J Allergy Clin Immunol 2022; 149:1875-1898. [PMID: 35337846 DOI: 10.1016/j.jaci.2022.03.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 02/13/2022] [Accepted: 03/10/2022] [Indexed: 11/26/2022]
Abstract
Atopic dermatitis (AD) is a common, chronic-relapsing inflammatory skin disease with significant disease burden. Genetic and environmental trigger factors contribute to AD, activating two of our largest organs, the nervous and immune system. Dysregulation of neuro-immune circuits plays a key role in the pathophysiology of AD causing inflammation, pruritus, pain, and barrier dysfunction. Sensory nerves can be activated by environmental or endogenous trigger factors transmitting itch stimuli to the brain. Upon stimulation, sensory nerve endings also release neuromediators into the skin contributing again to inflammation, barrier dysfunction and itch. Additionally, dysfunctional peripheral and central neuronal structures contribute to neuroinflammation, sensitization, nerve elongation, neuropathic itch, thus chronification and therapy-resistance. Consequently, neuro-immune circuits in skin and central nervous system may be targets to treat pruritus in AD. Cytokines, chemokines, proteases, lipids, opioids, ions excite/sensitize sensory nerve endings not only induce itch but further aggravate/perpetuate inflammation, skin barrier disruption, and pruritus. Thus, targeted therapies for neuro-immune circuits as well as pathway inhibitors (e.g., kinase inhibitors) may be beneficial to control pruritus in AD either in systemic and/or topical form. Understanding neuro-immune circuits and neuronal signaling will optimize our approach to control all pathological mechanisms in AD, inflammation, barrier dysfunction and pruritus.
Collapse
Affiliation(s)
- Martin Steinhoff
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Department of Dermatology, Weill Cornell Medicine-Qatar, Doha, Qatar; Qatar University, College of Medicine, Doha, Qatar; Department of Dermatology, Weill Cornell Medicine, New York, USA.
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Atul Pandey
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Angeliki Datsi
- Institute for Transplantational Diagnostics and Cell Therapeutics, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Ayda AlHammadi
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Sara Al-Khawaga
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Aysha Al-Malki
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Jianghui Meng
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Majid Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Joerg Buddenkotte
- Department of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar; Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
123
|
Meier HCS, Sandler DP, Wilkerson J, Miller FW, Dinse GE, Parks CG. Hygiene Hypothesis Indicators and Prevalence of Antinuclear Antibodies in US Adolescents. Front Immunol 2022; 13:789379. [PMID: 35154106 PMCID: PMC8832391 DOI: 10.3389/fimmu.2022.789379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Autoimmunity prevalence, as measured by antinuclear antibodies (ANA), is increasing in U.S. adolescents. Improved hygiene and cleaner environments in childhood may reduce exposure to infections and other immune challenges, resulting in improper immune responses to later-life exposures. We examined associations of hygiene hypothesis indicators, including asthma, allergies, and antibodies to infectious agents, with ANA prevalence, measured by HEp-2 immunofluorescence, in adolescents (aged 12-19 years) over a 25-year time span in the National Health and Nutrition Examination Survey (NHANES) (N=2,709), adjusting for age, sex, race/ethnicity, body mass index, education and survey cycle, overall and within individual time periods, using logistic regression. Prevalence of ANA in adolescents increased from 5.0% in 1988-1991 to 12.8% in 2011-2012. ANA were positively associated with diagnosis of asthma in early childhood (OR: 2.07, CI: 1.09-3.99) and the effect estimate for current hay fever was elevated but not statistically significant (OR: 1.55, CI: 0.85-2.84). Fewer than 2% of those with ANA in 1988-1991 had been diagnosed with asthma, compared with 18% in 1999-2000, and 27% in 2003-2004 and 2011-2012. ANA trended negatively with Helicobacter pylori antibodies (OR: 0.49, CI: 0.24-0.99). ANA may be useful as an additional indicator of inadequate immune education in adolescence, a critical period of growth and development.
Collapse
Affiliation(s)
- Helen C S Meier
- Population, Neurodevelopment and Genetics Program, Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, United States
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), Durham, NC, United States
| | - Jesse Wilkerson
- Public Health & Scientific Research, Social and Scientific Systems, Durham, NC, United States
| | - Frederick W Miller
- Environmental Autoimmunity Group, National Institute of Environmental Health Sciences (NIEHS), Durham, NC, United States
| | - Gregg E Dinse
- Public Health & Scientific Research, Social and Scientific Systems, Durham, NC, United States
| | - Christine G Parks
- Epidemiology Branch, National Institute of Environmental Health Sciences (NIEHS), Durham, NC, United States
| |
Collapse
|
124
|
The Hodgkin Lymphoma Immune Microenvironment: Turning Bad News into Good. Cancers (Basel) 2022; 14:cancers14051360. [PMID: 35267668 PMCID: PMC8909875 DOI: 10.3390/cancers14051360] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/09/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
The classic Hodgkin lymphoma (cHL) tumor microenvironment (TME) is by far the most abundant component of tumors and is responsible for most of their biological and clinical characteristics. Recent advances in our knowledge of these networks in cellular interactions allow us to understand that the neoplastic Hodgkin and Reed Sternberg (HRS) cells, although they are in the minority, are the main architects of this dysregulated immune milieu. Here, we review the major changes that have happened in recent years: from TME as a helpless bystander, reflecting an ineffective immune response, to a dynamic tumor-promoting and immunosuppressive element. The HRS cells promote survival through interconnected intrinsic and extrinsic alterations, boosting pro-tumoral signaling pathways through genetic aberrations and autocrine growth signals, in parallel with abnormal cytokine secretion for the recruitment and selection of the best cell partners for this immunosuppressive TME. In turn, cHL is already proving to be the perfect model with which to address an immune checkpoint blockade. Preliminary data demonstrate the utility of druggable key signaling pathways in this ensemble, such as JAK-STAT, NF-κB, and others. In addition, myriad biomarkers predicting a response await validation by new in situ multiplex analytical methods, single-cell gene expression, and other techniques. Together, these components will define the functional phenotypes with which we will elucidate the molecular pathogenesis of the disease and improve the survival of patients who are refractory to conventional therapies.
Collapse
|
125
|
Dai M, Zhu X, Yu J, Yuan J, Zhu Y, Bao Y, Yong X. CCR3 gene knockout in bone marrow cells ameliorates combined allergic rhinitis and asthma syndrome (CARAS) by reducing airway inflammatory cell infiltration and Th2 cytokines expression in mice model. Int Immunopharmacol 2022; 104:108509. [PMID: 34998035 DOI: 10.1016/j.intimp.2021.108509] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/17/2021] [Accepted: 12/28/2021] [Indexed: 12/15/2022]
Abstract
The present study aims to investigate the effects of CCR3 gene knockout in bone marrow cells (CCR3-KO) on the mouse model of combined allergic rhinitis and asthma syndrome (CARAS). It was found that CCR3-KO significantly reduced eosinophil (EOS) migration into the nasal (NALF) and bronchoalveolar (BALF) cavities of mice, and decreased Th2 cytokines (such as, IL-4, IL-5 and IL-13) levels in nasal mucosa and lung tissues. In addition, histological analysis showed that the damage degree of nasal mucosa structure in ovalbumin (OVA) modulated CCR3-KO mice was significantly less than that in OVA modulated Wild type (WT) mice, with reduced inflammatory cell infiltration and nasal mucus secretion. The infiltration of inflammatory cells in lung tissue was significantly reduced, and the proliferation of lung smooth muscle layer and extracellular matrix (ECM) production were decreased. Symptom analysis showed that CCR3-KO can reduced allergic rhinitis (AR) signals as nose scratching and sneezing. It was also found CCR3-KO reduce OVA-induced weight loss. The results showed that CCR3-KO could reduce the symptoms of allergic inflammation in CARAS mice by reducing airway inflammatory cell infiltration and down-regulating the expression of Th2 cytokines, and CCR3 gene could be used as a target gene for the treatment of CARAS.
Collapse
Affiliation(s)
- MeiNa Dai
- The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330000, China.
| | - XinHua Zhu
- The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330000, China.
| | - Juan Yu
- The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330000, China.
| | - JiaSheng Yuan
- The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330000, China.
| | - Yv Zhu
- The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330000, China.
| | - YouWei Bao
- The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang City, Jiangxi Province, 330000, China.
| | - XiaoZhuang Yong
- Institute of Translational Medicine, Nanchang University, 1299 Xuefu Avenue, Honggutan New District, Nanchang City, Jiangxi Province, 330000, China.
| |
Collapse
|
126
|
Kim HS, Yang HJ, Song DJ, Lee YJ, Suh DI, Shim JY, Yoo Y, Kim CK, Ahn YM, Kim JT. Eosinophil-derived neurotoxin: An asthma exacerbation biomarker in children. Allergy Asthma Proc 2022; 43:133-139. [PMID: 35317890 DOI: 10.2500/aap.2022.43.210001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background: Asthma is a heterogeneous disease, characterized by chronic airway inflammation. Asthma exacerbations (AE) are episodes characterized by a progressive increase in symptoms of shortness of breath, cough, wheezing, or chest tightness with a decrease in lung function. There have been previous studies that examined the role of eosinophil-derived neurotoxin (EDN) in asthma, but there have been no studies of the role of EDN in children experiencing AE. Objective: In this study, we aimed to examine the association of EDN with lung function and prognosis in children admitted for severe AE. Methods: We enrolled 82 children who were admitted for severe AE at two different university hospitals in South Korea between January 2018 and December 2019. Blood tests, including white blood cell count, myeloperoxidase (MPO), total eosinophil count, EDN, C-reactive protein (CRP) level, and interleukin (IL) 4, IL-5, IL-10 values, and lung function were measured on admission and at discharge in each patient. Results: We observed significant decreases in the levels of MPO, EDN, CRP, and IL-4, with significant improvement in lung function after treatment. We then classified the subjects into two groups of different clinical phenotypes: eosinophilic asthma exacerbation (EAE) group and non-EAE group. EDN levels were higher and lung functions were lower in the EAE group. Also, we found that the EDN level was a significant biomarker useful for predicting the number of days for hospital stay. Conclusion: We found that EDN can act as a biomarker that reflects lung function, and that EDN could act as a prognostic biomarker, which demonstrated the complex role of EDN in children experiencing AE.
Collapse
Affiliation(s)
- Hwan Soo Kim
- From the Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyeon-Jong Yang
- Department of Pediatrics, Pediatric Allergy and Respiratory Center, Soonchunhyang University College of Medicine, Seoul, South Korea
| | - Dae Jin Song
- Department of Pediatrics, Korea University College of Medicine, Seoul, South Korea
| | - Yong Ju Lee
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong In Suh
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea
| | - Jung Yeon Shim
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Young Yoo
- Department of Pediatrics, Korea University College of Medicine, Seoul, South Korea
| | - Chang Keun Kim
- Department of Pediatrics, Asthma and Allergy Center, Inje University Sanggye Paik Hospital, Seoul, South Korea; and
| | - Young Min Ahn
- Department of Pediatrics, Jang's Hospital, Seoul, South Korea
| | - Jin Tack Kim
- From the Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
127
|
González‐Rodríguez MI, Nurminen N, Kummola L, Laitinen OH, Oikarinen S, Parajuli A, Salomaa T, Mäkelä I, Roslund MI, Sinkkonen A, Hyöty H, Junttila IS. Effect of inactivated nature‐derived microbial composition on mouse immune system. Immun Inflamm Dis 2022; 10:e579. [PMID: 34873877 PMCID: PMC8926502 DOI: 10.1002/iid3.579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/04/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction The hygiene hypothesis suggests that decrease in early life infections due to increased societal‐level hygiene standards subjects one to allergic and autoimmune diseases. In this report, we have studied the effect of sterilized forest soil and plant‐based material on mouse immune system and gut microbiome. Methods Inbred C57Bl/6 mice maintained in normal sterile environment were subjected to autoclaved forest soil‐derived powder in their bedding for 1 h a day for 3 weeks. Immune response was measured by immune cell flow cytometry, serum cytokine enzyme‐linked immunoassay (ELISA) and quantitative polymerase chain reaction (qPCR) analysis. Furthermore, the mouse gut microbiome was analyzed by sequencing. Results When compared to control mice, mice treated with soil‐derived powder had decreased level of pro‐inflammatory cytokines namely interleukin (IL)−17F and IL‐21 in the serum. Furthermore, splenocytes from mice treated with soil‐derived powder expressed less IL‐1b, IL‐5, IL‐6, IL‐13, and tumor necrosis factor (TNF) upon cell activation. Gut microbiome appeared to be stabilized by the treatment. Conclusions These results provide insights on the effect of biodiversity on murine immune system in sterile environment. Subjecting mice to soil‐based plant and microbe structures appears to elicit immune response that could be beneficial, for example, in type 2 inflammation‐related diseases, that is, allergic diseases.
Collapse
Affiliation(s)
| | - Noora Nurminen
- Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Laura Kummola
- Faculty of Medicine and Health Technology Tampere University Tampere Finland
- Department of Clinical Microbiology Fimlab Laboratories Finland
| | - Olli H. Laitinen
- Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Sami Oikarinen
- Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Anirudra Parajuli
- Department of Medicine, Karolinska Institutet Center for infectious medicine (CIM) Huddinge Sweden
| | - Tanja Salomaa
- Faculty of Medicine and Health Technology Tampere University Tampere Finland
| | - Iida Mäkelä
- Department of Garden Technologies, Horticulture Technologies Natural Resources Institute Finland Finland
| | - Marja I. Roslund
- Ecosystems and Environment Research Programme University of Helsinki Helsinki Finland
| | - Aki Sinkkonen
- Department of Garden Technologies, Horticulture Technologies Natural Resources Institute Finland Finland
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology Tampere University Tampere Finland
- Department of Clinical Microbiology Fimlab Laboratories Finland
| | - Ilkka S. Junttila
- Faculty of Medicine and Health Technology Tampere University Tampere Finland
- Department of Clinical Microbiology Fimlab Laboratories Finland
| | | |
Collapse
|
128
|
Identification of Key Determinants of Cerebral Malaria Development and Inhibition Pathways. mBio 2022; 13:e0370821. [PMID: 35073748 PMCID: PMC8787489 DOI: 10.1128/mbio.03708-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cerebral malaria (CM), coma caused by Plasmodium falciparum-infected red blood cells (iRBCs), is the deadliest complication of malaria. The mechanisms that lead to CM development are incompletely understood. Here we report on the identification of activation and inhibition pathways leading to mouse CM with supporting evidence from the analysis of human specimens. We find that CM suppression can be induced by vascular injury when sporozoites exit the circulation to infect the liver and that CM suppression is mediated by the release of soluble factors into the circulation. Among these factors is insulin like growth factor 1 (IGF1), administration of which inhibits CM development in mice. IMPORTANCE Liver infection by Plasmodium sporozoites is a required step for infection of the organism. We found that alternate pathways of sporozoite liver infection differentially influence cerebral malaria (CM) development. CM is one of the primary causes of death following malaria infection. To date, CM research has focused on how CM phenotypes develop but no successful therapeutic treatment or prognostic biomarkers are available. Here we show for the first time that sporozoite liver invasion can trigger CM-inhibitory immune responses. Importantly, we identified a number of early-stage prognostic CM inhibitory biomarkers, many of which had never been associated with CM development. Serological markers identified using a mouse model are directly relevant to human CM.
Collapse
|
129
|
Moretti L, Stalfort J, Barker TH, Abebayehu D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J Biol Chem 2022; 298:101530. [PMID: 34953859 PMCID: PMC8784641 DOI: 10.1016/j.jbc.2021.101530] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
Various forms of fibrosis, comprising tissue thickening and scarring, are involved in 40% of deaths across the world. Since the discovery of scarless functional healing in fetuses prior to a certain stage of development, scientists have attempted to replicate scarless wound healing in adults with little success. While the extracellular matrix (ECM), fibroblasts, and inflammatory mediators have been historically investigated as separate branches of biology, it has become increasingly necessary to consider them as parts of a complex and tightly regulated system that becomes dysregulated in fibrosis. With this new paradigm, revisiting fetal scarless wound healing provides a unique opportunity to better understand how this highly regulated system operates mechanistically. In the following review, we navigate the four stages of wound healing (hemostasis, inflammation, repair, and remodeling) against the backdrop of adult versus fetal wound healing, while also exploring the relationships between the ECM, effector cells, and signaling molecules. We conclude by singling out recent findings that offer promising leads to alter the dynamics between the ECM, fibroblasts, and inflammation to promote scarless healing. One factor that promises to be significant is fibroblast heterogeneity and how certain fibroblast subpopulations might be predisposed to scarless healing. Altogether, reconsidering fetal wound healing by examining the interplay of the various factors contributing to fibrosis provides new research directions that will hopefully help us better understand and address fibroproliferative diseases, such as idiopathic pulmonary fibrosis, liver cirrhosis, systemic sclerosis, progressive kidney disease, and cardiovascular fibrosis.
Collapse
Affiliation(s)
- Leandro Moretti
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Jack Stalfort
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Thomas Harrison Barker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel Abebayehu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
130
|
Inflammatory Arthritis and Bone Metabolism Regulated by Type 2 Innate and Adaptive Immunity. Int J Mol Sci 2022; 23:ijms23031104. [PMID: 35163028 PMCID: PMC8834748 DOI: 10.3390/ijms23031104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
While type 2 immunity has traditionally been associated with the control of parasitic infections and allergic reactions, increasing evidence suggests that type 2 immunity exerts regulatory functions on inflammatory diseases such as arthritis, and also on bone homeostasis. This review summarizes the current evidence of the regulatory role of type 2 immunity in arthritis and bone. Key type 2 cytokines, like interleukin (IL)-4 and IL-13, but also others such as IL-5, IL-9, IL-25, and IL-33, exert regulatory properties on arthritis, dampening inflammation and inducing resolution of joint swelling. Furthermore, these cytokines share anti-osteoclastogenic properties and thereby reduce bone resorption and protect bone. Cellular effectors of this action are both T cells (i.e., Th2 and Th9 cells), but also non-T cells, like type 2 innate lymphoid cells (ILC2). Key regulatory actions mediated by type 2 cytokines and immune cells on both inflammation as well as bone homeostasis are discussed.
Collapse
|
131
|
Dolitzky A, Shapira G, Grisaru-Tal S, Hazut I, Avlas S, Gordon Y, Itan M, Shomron N, Munitz A. Transcriptional Profiling of Mouse Eosinophils Identifies Distinct Gene Signatures Following Cellular Activation. Front Immunol 2022; 12:802839. [PMID: 34970274 PMCID: PMC8712732 DOI: 10.3389/fimmu.2021.802839] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/22/2021] [Indexed: 01/21/2023] Open
Abstract
Eosinophils are multifunctional, evolutionary conserved leukocytes that are involved in a plethora of responses ranging from regulation of tissue homeostasis to host defense and cancer. Eosinophils have been studied mostly in the context of Type 2 inflammatory responses such as those found in allergy. Nonetheless, it is now evident that they participate in Type 1 inflammatory responses and can respond to Type 1 cytokines such as IFN-γ. Recent data suggest that the pleotropic roles of eosinophils are due to heterogeneous responses to environmental cues. Despite this, the activation profile of eosinophils, in response to various stimuli is yet to be defined. To better understand the transcriptional spectrum of eosinophil activation, we exposed eosinophils to Type 1 (e.g. IFN-γ, E. coli) vs. Type 2 (e.g. IL-4) conditions and subjected them to global RNA sequencing. Our analyses show that IL-4, IFN-γ, E. coli and IFN-γ in the presence of E. coli (IFN-γ/E. coli)-stimulated eosinophils acquire distinct transcriptional profiles, which polarize them towards what we termed Type 1 and Type 2 eosinophils. Bioinformatics analyses using Gene Ontology based on biological processes revealed that different stimuli induced distinct pathways in eosinophils. These pathways were confirmed using functional assays by assessing cytokine/chemokine release (i.e. CXCL9, CCL24, TNF-α and IL-6) from eosinophils following activation. In addition, analysis of cell surface markers highlighted CD101 and CD274 as potential cell surface markers that distinguish between Type 1 and Type 2 eosinophils, respectively. Finally, the transcriptome signature of Type 1 eosinophils resembled that of eosinophils that were obtained from mice with experimental colitis whereas the transcriptome signature of Type 2 eosinophils resembled that of eosinophils from experimental asthma. Our data demonstrate that eosinophils are polarized to distinct “Type 1” and “Type 2” phenotypes following distinct stimulations. These findings provide fundamental knowledge regarding the heterogeneity of eosinophils and support the presence of transcriptional differences between Type 1 and Type 2 cells that are likely reflected by their pleotropic activities in diverse disease settings.
Collapse
Affiliation(s)
- Avishay Dolitzky
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Shapira
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Grisaru-Tal
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Hazut
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shmulik Avlas
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yaara Gordon
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Micahl Itan
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Noam Shomron
- Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Munitz
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
132
|
Bermick J, Schaller M. Epigenetic regulation of pediatric and neonatal immune responses. Pediatr Res 2022; 91:297-327. [PMID: 34239066 DOI: 10.1038/s41390-021-01630-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
Epigenetic regulation of transcription is a collective term that refers to mechanisms known to regulate gene transcription without changing the underlying DNA sequence. These mechanisms include DNA methylation and histone tail modifications which influence chromatin accessibility, and microRNAs that act through post-transcriptional gene silencing. Epigenetics is known to regulate a variety of biological processes, and the role of epigtenetics in immunity and immune-mediated diseases is becoming increasingly recognized. While DNA methylation is the most widely studied, each of these systems play an important role in the development and maintenance of appropriate immune responses. There is clear evidence that epigenetic mechanisms contribute to developmental stage-specific immune responses in a cell-specific manner. There is also mounting evidence that prenatal exposures alter epigenetic profiles and subsequent immune function in exposed offspring. Early life exposures that are associated with poor long-term health outcomes also appear to impact immune specific epigenetic patterning. Finally, each of these epigenetic mechanisms contribute to the pathogenesis of a wide variety of diseases that manifest during childhood. This review will discuss each of these areas in detail. IMPACT: Epigenetics, including DNA methylation, histone tail modifications, and microRNA expression, dictate immune cell phenotypes. Epigenetics influence immune development and subsequent immune health. Prenatal, perinatal, and postnatal exposures alter immune cell epigenetic profiles and subsequent immune function. Numerous pediatric-onset diseases have an epigenetic component. Several successful strategies for childhood diseases target epigenetic mechanisms.
Collapse
Affiliation(s)
- Jennifer Bermick
- Department of Pediatrics, Division of Neonatology, University of Iowa, Iowa City, IA, USA. .,Iowa Inflammation Program, University of Iowa, Iowa City, IA, USA.
| | - Matthew Schaller
- Department of Pulmonary, Critical Care & Sleep Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
133
|
Ferrante G, Tenero L, Piazza M, Piacentini G. Severe pediatric asthma therapy: Dupilumab. Front Pediatr 2022; 10:963610. [PMID: 36483465 PMCID: PMC9723147 DOI: 10.3389/fped.2022.963610] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022] Open
Abstract
Severe asthma is a rare disease affecting <5% of children with asthma. This group of patients account for about 50% of the costs of healthcare for children with asthma. Nowadays, several biological agents are available for pediatric severe asthma. One of these is dupilumab, a monoclonal antibody against the Interleukin (IL)-4 receptor α-subunit that acts as an antagonist against both IL-4 and IL-13. Dupilumab binds the subunit of the IL-4 receptor, at the level of the subunit shared by the IL-13 receptor, blocking the inflammatory cascade of these two cytokines and the progression of the Th2-inflammatory pathway. The efficacy and safety of dupilumab have been investigated in recently published randomized controlled trials including pediatric patients with asthma. Currently, its use in asthma is approved in adults, adolescents, and children with severe asthma with type 2 inflammation, that are not controlled in spite of high-dose inhaled corticosteroids plus another maintenance drug. Studies are warranted for the evaluation of long-term treatment with dupilumab, including steroid sparing effect and discontinuation of treatment. Further research should also be planned in order to investigate dupilumab potential ability to interfere with the natural history of atopy since early childhood.
Collapse
Affiliation(s)
- Giuliana Ferrante
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Division, University of Verona, Verona, Italy
| | - Laura Tenero
- Pediatric Division, University Hospital of Verona, Verona, Italy
| | - Michele Piazza
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Division, University of Verona, Verona, Italy
| | - Giorgio Piacentini
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Division, University of Verona, Verona, Italy
| |
Collapse
|
134
|
Choi BY, Han M, Kwak JW, Kim TH. Genetics and Epigenetics in Allergic Rhinitis. Genes (Basel) 2021; 12:2004. [PMID: 34946955 PMCID: PMC8700872 DOI: 10.3390/genes12122004] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of allergic rhinitis is associated with genetic, environmental, and epigenetic factors. Genotyping of single nucleotide polymorphisms (SNPs) is an advanced technique in the field of molecular genetics that is closely correlated with genome-wide association studies (GWASs) in large population groups with allergic diseases. Many recent studies have paid attention to the role of epigenetics, including alteration of DNA methylation, histone acetylation, and miRNA levels in the pathogenesis of allergic rhinitis. In this review article, genetics and epigenetics of allergic rhinitis, including information regarding functions and significance of previously known and newly-discovered genes, are summarized. Directions for future genetic and epigenetic studies of allergic rhinitis are also proposed.
Collapse
Affiliation(s)
| | | | | | - Tae Hoon Kim
- Department of Otorhinolaryngology-Head & Neck Surgery, College of Medicine, Korea University, Seoul 02841, Korea; (B.Y.C.); (M.H.); (J.W.K.)
| |
Collapse
|
135
|
Zhao Y, Qi C, Li X, Lu M, Zhang H, Zhou J, Dang H, Chen J, Li S, Sun J, Yu R, Li D. Prevention of Atopic Dermatitis in Mice by Lactobacillus Reuteri Fn041 Through Induction of Regulatory T Cells and Modulation of the Gut Microbiota. Mol Nutr Food Res 2021; 66:e2100699. [PMID: 34825773 DOI: 10.1002/mnfr.202100699] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 11/11/2021] [Indexed: 11/11/2022]
Abstract
SCOPE The development of atopic dermatitis (AD) in infants is closely related to the lagging development of intestinal microbiota, including that inoculated by breast milk bacteria, and immune development. Lactobacillus reuteri Fn041 is a secretory immunoglobulin A (sIgA) -coated bacterium derived from human milk. METHODS AND RESULTS We intervened with L. reuteri Fn041 in maternal and offspring BALB/C mice during late gestation and lactation and after weaning of the pups, respectively. AD was then induced with MC903. L. reuteri Fn041 significantly suppressed AD symptoms such as skin swelling, mast cell and eosinophil infiltration. This effect was attributed to the regulation of the systemic Th1 and Th2 cytokine ratios and the promotion of CD4+ CD25+ Foxp3+ regulatory T cell proliferation in mesenteric lymph nodes. It is also associated with the regulation of intestinal microbiota, particularly promoting Lactobacillus and Akkermansia. CONCLUSIONS Our study strengthens the understanding that breast milk-derived sIgA coated potential probiotics are involved in the development of infant intestinal microbiota, thus promoting immune development and preventing allergic diseases, and expanding the knowledge of breast milk sIgA and bacterial interactions on infant immune development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yuning Zhao
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Xinyue Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Mengyao Lu
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Haowen Zhang
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Jingbo Zhou
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Hongyang Dang
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Jie Chen
- Department of Pediatric Cardiology Nephrology and Rheumatism, The Affiliated, Hospital of Qingdao University Medical College, Qingdao, 266003, China
| | - Shuangqi Li
- Guangzhou Fine Nutrition Research Center, Guangzhou, 510700, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, China
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, Qingdao, 266071, China
| |
Collapse
|
136
|
Liu Z, Huo JH, Dong WT, Sun GD, Li FJ, Zhang YN, Qin ZW, Pengna J, Wang WM. A Study Based on Metabolomics, Network Pharmacology, and Experimental Verification to Explore the Mechanism of Qinbaiqingfei Concentrated Pills in the treatment of Mycoplasma Pneumonia. Front Pharmacol 2021; 12:761883. [PMID: 34803705 PMCID: PMC8599429 DOI: 10.3389/fphar.2021.761883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
Qinbaiqingfei concentrated pills (QB) are a commonly used medicine for the treatment of mycoplasma pneumonia in China, and the mechanism of action of QB needs to be studied further. Therefore, we use a combination of metabolomics and network pharmacology to clarify the mechanism of QB. Nontarget metabolomics studies were performed on rat serum, urine, and lung tissues, and 56 therapeutic biomarkers were found. Subsequently, the components of QB absorbed into the blood and lung tissues were clarified, and based on this finding, the core target of network pharmacology was predicted. The enrichment analysis of biomarkers–genes finally confirmed their close relationship with the NF-κB signaling pathway. By western blotting expression of the proteins in the lung tissue–related signaling pathways, it is finally confirmed that QB inhibits the NF-κB signaling pathway through SIRT1, IL-10 and MMP9, CTNNB1, EGFR, and other targets. It plays a role in regulating immunity, regulating metabolism, and treating diseases.
Collapse
Affiliation(s)
- Zheng Liu
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Jin-Hai Huo
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Wen-Ting Dong
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Guo-Dong Sun
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Feng-Jin Li
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Ya-Nan Zhang
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Zhi-Wei Qin
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| | - Jiang Pengna
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei-Ming Wang
- Heilongjiang Academy of Chinese Medicine, Institute of Chinese Materia Medica, Harbin, China
| |
Collapse
|
137
|
Fang X, Li M, Zhang W, Li J, Zhu T. Thrombin induces pro-inflammatory and anti-inflammatory cytokines secretion from human mast cell line (HMC-1) via protease-activated receptors. Mol Immunol 2021; 141:60-69. [PMID: 34808483 DOI: 10.1016/j.molimm.2021.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/24/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023]
Abstract
Thrombin-induced mast cell activation represents cross-talk between coagulation and inflammation. However, there is still controversy concerning the pro- or anti-inflammatory effects mast cells have in response to thrombin signaling. Human mast cell HMC-1 was incubated with 0.2 U/mL thrombin. Cells and supernatants were collected. Production of pro- and anti-inflammatory mediators was determined by quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA). Expression of proteinase-activated receptor-1 (PAR1) and -4 (PAR4) mRNA in HMC-1 cells was analyzed by qPCR. Activation of mitogen-activated protein kinases (MAPKs) was measured by immunoblotting. Furthermore, the impact of PAR1 inhibitor (SCH79797) and agonist (TFLLR-NH2), PAR4 inhibitor (BMS986120) and agonist (AYPGKF-NH2), and MAPK inhibitors (SB203580, PD98059, and SP600125) on the production of mediators was evaluated using qPCR and ELISA. Thrombin exposure increased pro- and anti-inflammatory mediators, expression of PAR1 and PAR4 mRNA, and phosphorylation of JNK, p38, and ERK1/2 MAPKs in HMC-1 cells. SCH79797, BMS986120, and MAPK inhibitors (SB203580, PD98059, and SP600125) were inhibited, while TFLLR-NH2 and AYPGKF-NH2 promoted pro- and anti-inflammatory cytokines in this process. HMC-1 produces pro- and anti-inflammatory cytokines after thrombin incubation, namely PAR1 and PAR4. Alongside HMC-1, MAPK signaling pathways are involved in the production of these mediators. The mast cells showed dual activation after thrombin stimulation.
Collapse
Affiliation(s)
- Xiaobin Fang
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, Sichuan 610041, China.
| | - Mengmeng Li
- Department of Dermatovenereology, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Weiyi Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, Sichuan 610041, China.
| | - Jingyi Li
- Department of Dermatovenereology, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Unit of West China (2018RU012), Chinese Academy of Medical Science, Chengdu, Sichuan 610041, China.
| |
Collapse
|
138
|
Yang W, Lin J, Zhou J, Zheng Y, Jiang S, He S, Li D. Innate Lymphoid Cells and Myocardial Infarction. Front Immunol 2021; 12:758272. [PMID: 34867998 PMCID: PMC8636005 DOI: 10.3389/fimmu.2021.758272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/27/2021] [Indexed: 11/13/2022] Open
Abstract
Myocardial infarction results from obstruction of a coronary artery that causes insufficient blood supply to the myocardium and leads to ischemic necrosis. It is one of the most common diseases threatening human health and is characterized by high morbidity and mortality. Atherosclerosis is the pathological basis of myocardial infarction, and its pathogenesis has not been fully elucidated. Innate lymphoid cells (ILCs) are an important part of the human immune system and participate in many processes, including inflammation, metabolism and tissue remodeling, and play an important role in atherosclerosis. However, their specific roles in myocardial infarction are unclear. This review describes the current understanding of the relationship between innate lymphoid cells and myocardial infarction during the acute phase of myocardial infarction, myocardial ischemia-reperfusion injury, and heart repair and regeneration following myocardial infarction. We suggest that this review may provide new potential intervention targets and ideas for treatment and prevention of myocardial infarction.
Collapse
Affiliation(s)
| | | | | | | | | | - Shaolin He
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dazhu Li
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
139
|
IL-4 and IL-13 Promote Proliferation of Mammary Epithelial Cells through STAT6 and IRS-1. Int J Mol Sci 2021; 22:ijms222112008. [PMID: 34769439 PMCID: PMC8584551 DOI: 10.3390/ijms222112008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/08/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022] Open
Abstract
T helper (Th)2 cytokines such as interleukin (IL)-4 and IL-13 control immune function by acting on leukocytes. They also regulate multiple responses in non-hematopoietic cells. During pregnancy, IL-4 and IL-13 facilitate alveologenesis of mammary glands. This particular morphogenesis generates alveoli from existing ducts and requires substantial cell proliferation. Using 3D cultures of primary mouse mammary epithelial cells, we demonstrate that IL-4 and IL-13 promote cell proliferation, leading to enlargement of mammary acini with partially filled lumens. The mitogenic effects of IL-4 and IL-13 are mediated by STAT6 as inhibition of STAT6 suppresses cell proliferation and improves lumen formation. In addition, IL-4 and IL-13 stimulate tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1). Prolonged treatment with these cytokines leads to increased IRS-1 abundance, which, in turn, amplifies IL-4- and IL-13-stimulated IRS-1 tyrosine phosphorylation. Through signaling crosstalk between IL-4/IL-13 and insulin, a hormone routinely included in mammary cultures, IRS-1 tyrosine phosphorylation is further enhanced. Lowering IRS-1 expression reduces cell proliferation, suggesting that IRS-1 is involved in IL-4- and IL-13-stimulated cell proliferation. Thus, a Th2-dominant cytokine milieu during pregnancy confers mammary gland development by promoting cell proliferation.
Collapse
|
140
|
Iwaszko M, Biały S, Bogunia-Kubik K. Significance of Interleukin (IL)-4 and IL-13 in Inflammatory Arthritis. Cells 2021; 10:cells10113000. [PMID: 34831223 PMCID: PMC8616130 DOI: 10.3390/cells10113000] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/26/2021] [Accepted: 10/30/2021] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-4 and IL-13 belong to the T helper 2 (Th2) cytokine family, along with IL-3, IL-5, and IL-9. These cytokines are key mediators of allergic inflammation. They have important immunomodulatory activities and exert influence on a wide variety of immune cells, such as B cells, eosinophils, basophils, monocytes, fibroblasts, endothelial cells, airway epithelial cells, smooth muscle cells, and keratinocytes. Recent studies have implicated IL-4 and IL-13 in the development of various autoimmune diseases. Additionally, these cytokines have emerged as potential players in pathogenesis of inflammatory arthritis. Recent findings suggest that the IL-4 and IL-13 might play a significant role in the downregulation of inflammatory processes underlying RA pathology, and beneficially modulate the course of the disease. This review summarizes the biological features of the IL-4 and IL-13 and provides current knowledge regarding the role of these cytokines in inflammatory arthritis.
Collapse
|
141
|
Katz BZ, Benisty D, Kay S, Herzlich J, Raskind C, Marom R. Comprehensive Morphological Assessment of Cord Blood: Normal Values and the Prevalence of Morphologically Aberrant Leukocytes. Acta Haematol 2021; 145:184-192. [PMID: 34727546 DOI: 10.1159/000520638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 11/01/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Cord blood (CB) is becoming a valuable source for stem cells utilized in a variety of cell therapy applications, as well as for newborn diagnostics. Some parameters of the CB cellular components can be provided by automated analyzers, while others, such as immature or aberrant cells, require blood film morphological assessment. The objectives of the study were to establish normal CB morphology and to determine the prevalence of morphologically aberrant leukocytes in CB. METHODS We performed a comprehensive morphological analysis of 100 CB samples taken from healthy term and appropriate-for-gestational-age neonates born to healthy mothers, preterm neonates, neonates of diabetic mothers, and small-for-gestational-age neonates. Blood counts were assessed, and manual morphological analyses were performed by laboratory specialists. RESULTS The manual differential count of normal CB samples established the following values: 47.8 ± 10.7% neutrophils, 31.2 ± 9.8% lymphocytes, 10.0 ± 4.0% monocytes, and 3.0 ± 2.5% eosinophils, with no significant sex-related differences. Blasts were observed in 44/100 samples with an average of 0.5 ± 0.7% per sample, and only a minor left shift was observed. There were significant populations of large granular lymphocytes (19.1 ± 10.6% of the total lymphocytes) and morphologically aberrant lymphocytes (12.4 ± 5.4% of the total lymphocytes) in the samples, irrespective of neonatal status. The differentials of preterm CB samples differ significantly from normal term CB samples, including the reverse of neutrophils/lymphocytes ratio, and the lack of basophils. CONCLUSIONS Normal values and unique morphological features in the CB of neonates are described. The abundant morphologically aberrant lymphocytes in CB may represent an immature state of the immune system at birth.
Collapse
Affiliation(s)
- Ben-Zion Katz
- Division of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dan Benisty
- Division of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Sigi Kay
- Division of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Jacky Herzlich
- Department of Neonatology, Dana Dwek Children Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Craig Raskind
- Department of Neonatology, Dana Dwek Children Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Ronella Marom
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Neonatology, Dana Dwek Children Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
142
|
Hong T, Li S, Guo X, Wei Y, Zhang J, Su X, Zhou M, Jin H, Miao Q, Shen L, Zhu M, He B. IL-13 Derived Type 2 Innate Lymphocytes Ameliorates Cardiomyocyte Apoptosis Through STAT3 Signaling Pathway. Front Cell Dev Biol 2021; 9:742662. [PMID: 34616745 PMCID: PMC8488199 DOI: 10.3389/fcell.2021.742662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/30/2021] [Indexed: 12/23/2022] Open
Abstract
The involvement of cardiomyopathy during sepsis means higher mortality and prolonged length of hospital stay. Many efforts have been made to alleviate the apoptosis of cardiomyocytes in sepsis. The huge potential of IL-13 in tissue repair has attracted increasing attention. In the present study, we used LPS-treated mice or primary cardiomyocytes as a sepsis model to explore the anti-apoptotic ability of IL-13. It was found that an increased level of exogenous IL-13 was beneficial to the recovery of heart function in sepsis, and this anti-apoptotic effect of IL-13 was probably through enhancing the phosphorylation of STAT3 Ser727. In addition, we identified that the heart protective effect of IL-13 was associated with type 2 innate lymphocytes (ILC2). All these findings may provide a potential promising treatment for sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
- Ting Hong
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Saiqi Li
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyu Guo
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yazhong Wei
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jingjing Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohui Su
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Miao Zhou
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haizhen Jin
- Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Miao
- Departments of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Shen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minfang Zhu
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bin He
- Department of Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
143
|
Wang L, Yang F, Qiu Y, Ye L, Song D, Huang D. The Potential Roles of T Cells in Periapical Lesions. J Endod 2021; 48:70-79. [PMID: 34627784 DOI: 10.1016/j.joen.2021.09.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Periapical lesions are inflammatory diseases mainly caused by microbial infection from the root canal system, affecting the integrity of alveolar bone, periapical cementum, and periodontal ligament. The invasion of pathogenic microorganisms activates local inflammation and host immune response, especially the recruitment and differentiation of T cells. Many studies have discussed the fundamental roles of T cell-related immunological regulation and the possible clinical significance of cytokine disorders in periapical lesions. However, oral pathogen-mediated T cell immune response is far more clarified. Therefore, the aim of this study was to discuss the research status of T cell-related immunology involved in the progression of periapical lesions and potential future directions. METHODS We conducted a literature review focusing on T cell-related immunology in periapical lesions by searching PubMed, Web of Science, Scopus and ScienceDirect online databases. RESULTS In total 108 articles were involved in this narrative review. During the development of periapical lesions, the infiltrated number of different types of T cells and the secretion of T cell-related cytokines in root apex region reflected the inflammatory status of periapical lesions. In addition, it was also highly correlated with the periapical bone destruction. Future study could attempt to provide a wider and deeper study on the expression profile and regulatory function of T cells in the development of periapical lesions. CONCLUSION This review would help us understand the essence of the T cell-related pathology of periapical lesions and raise the potential therapeutic targets for the treatment of apical periodontitis.
Collapse
Affiliation(s)
- Liu Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fan Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Qiu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ling Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dongzhe Song
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Dingming Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
144
|
da Silva Antunes R, Sutherland A, Frazier A, Schulten V, Pomés A, Glesner J, Calatroni A, Altman MC, Wood RA, O'Connor GT, Pongracic JA, Khurana Hershey GK, Kercsmar CM, Gruchalla RS, Gill M, Liu AH, Zoratti E, Kattan M, Busse PJ, Bacharier LB, Teach SJ, Wheatley LM, Togias A, Busse WW, Jackson DJ, Sette A. Heterogeneity of magnitude, allergen immunodominance, and cytokine polarization of cockroach allergen-specific T cell responses in allergic sensitized children. Clin Transl Allergy 2021; 11:e12073. [PMID: 34691392 PMCID: PMC8514843 DOI: 10.1002/clt2.12073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/13/2021] [Accepted: 10/03/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Characterization of allergic responses to cockroach (CR), a common aeroallergen associated with asthma, has focused mainly on IgE reactivity, but little is known about T cell responses, particularly in children. We conducted a functional evaluation of CR allergen-specific T cell reactivity in a cohort of CR allergic children with asthma. METHODS Peripheral blood mononuclear cells (PBMCs) were obtained from 71 children, with mild-to-moderate asthma who were enrolled in a CR immunotherapy (IT) clinical trial, prior to treatment initiation. PBMC were stimulated with peptide pools derived from 11 CR allergens, and CD4+ T cell responses assessed by intracellular cytokine staining. RESULTS Highly heterogeneous responses in T cell reactivity were observed among participants, both in terms of the magnitude of cytokine response and allergen immunodominance. Reactivity against Bla g 9 and Bla g 5 was most frequent. The phenotype of the T cell response was dominated by IL-4 production and a Th2 polarized profile in 54.9% of participants, but IFNγ production and Th1 polarization was observed in 25.3% of the participants. The numbers of regulatory CD4+ T cells were also highly variable and the magnitude of effector responses and Th2 polarization were positively correlated with serum IgE levels specific to a clinical CR extract. CONCLUSIONS Our results demonstrate that in children with mild-to-moderate asthma, CR-specific T cell responses display a wide range of magnitude, allergen dominance, and polarization. These results will enable examination of whether any of the variables measured are affected by IT and/or are predictive of clinical outcomes.
Collapse
Affiliation(s)
| | - Aaron Sutherland
- Division of Vaccine DiscoveryLa Jolla Institute for ImmunologyLa JollaCaliforniaUSA
| | - April Frazier
- Division of Vaccine DiscoveryLa Jolla Institute for ImmunologyLa JollaCaliforniaUSA
| | - Veronique Schulten
- Division of Vaccine DiscoveryLa Jolla Institute for ImmunologyLa JollaCaliforniaUSA
| | - Anna Pomés
- Basic ResearchIndoor Biotechnologies, Inc.CharlottesvilleVirginiaUSA
| | - Jill Glesner
- Basic ResearchIndoor Biotechnologies, Inc.CharlottesvilleVirginiaUSA
| | | | - Matthew C. Altman
- Benaroya Research Institute Systems Immunology DivisionDepartment of MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Robert A. Wood
- Division of Pediatric Allergy, Immunology and RheumatologyDepartment of PediatricsJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - George T. O'Connor
- Boston University School of MedicinePulmonary CenterBostonMassachusettsUSA
| | - Jacqueline A. Pongracic
- Advanced General Pediatrics and Primary CareAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | | | - Carolyn M. Kercsmar
- Division of Pulmonary MedicineCincinnati Children's HospitalCincinnatiOhioUSA
| | - Rebecca S. Gruchalla
- Divisions of Infectious Diseases and Pulmonary Vascular BiologyDepartment of PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Michelle Gill
- Divisions of Infectious Diseases and Pulmonary Vascular BiologyDepartment of PediatricsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Andrew H. Liu
- Department of PediatricsChildren's Hospital ColoradoUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Edward Zoratti
- Henry Ford Health System and Wayne State University School of MedicineDetroitMichiganUSA
| | - Meyer Kattan
- College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Paula J. Busse
- Division of Clinical Immunology and AllergyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Leonard B. Bacharier
- Department of PediatricsMonroe Carell Jr Children's Hospital at Vanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Stephen J. Teach
- Center for Translational ResearchChildren's National HospitalWashingtonDCUSA
| | - Lisa M. Wheatley
- Division of Allergy, Immunology, and TransplantationNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthRockvilleMarylandUSA
| | - Alkis Togias
- Division of Allergy, Immunology, and TransplantationNational Institute of Allergy and Infectious DiseasesNational Institutes of HealthRockvilleMarylandUSA
| | - William W. Busse
- Departments of Pediatrics and MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Daniel J. Jackson
- Departments of Pediatrics and MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Alessandro Sette
- Division of Vaccine DiscoveryLa Jolla Institute for ImmunologyLa JollaCaliforniaUSA
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
145
|
Okamoto K, Morio T. Inborn errors of immunity with eosinophilia. Allergol Int 2021; 70:415-420. [PMID: 34456137 DOI: 10.1016/j.alit.2021.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022] Open
Abstract
Monogenic diseases of the immune system, also known as inborn errors of immunity (IEIs), are caused by single-gene mutations and result in immune deficiency and dysregulation. More than 400 monogenic diseases have been described to date, and this number is rapidly expanding. The increasing availability of next-generation sequencing is now facilitating the diagnosis of IEIs. It is known that IEIs can predispose a person to not only infectious diseases but also cancer and immune disorders, such as inflammatory, autoimmune, and atopic diseases. IEIs with eosinophilia and atopic diseases can occur in several disorders. IEIs with eosinophilia have provided insights into human immunity and the pathogenesis of allergic diseases. Eosinophilia is not a rare finding in clinical practice, and it often poses problems in terms of etiologic research and differential diagnoses. Secondary eosinophilia is the most common form. The main underlying conditions are infectious diseases such as parasitic infections, allergic disorders, drug reactions, and of course IEIs. In clinical settings, the recognition of IEIs in the context of an allergic phenotype with eosinophilia is critical for prompt diagnosis and appropriate treatment aimed at modulating pathophysiological mechanisms and improving clinical symptoms.
Collapse
Affiliation(s)
- Keisuke Okamoto
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
146
|
Tian X, Huang Q, Liang J, Wang J, Zhang J, Yang Y, Ye Q, He S, Li J, Wu Z, Liu Y. A review of the mechanisms of keratinocytes damage caused by Staphylococcus aureus infection in patients with atopic dermatitis. J Leukoc Biol 2021; 110:1163-1169. [PMID: 34585438 DOI: 10.1002/jlb.3mr0921-030rrr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 01/15/2023] Open
Abstract
The dysregulation of skin microflora in patients with atopic dermatitis (AD) has become a research hotspot in recent years. Metagenomic studies have shown that microbial diversity is decreased, whereas the Staphylococcus aureus infection is increased in AD. Keratinocytes are the primary barrier against the invasion of external pathogenic microorganisms. Staphylococcus aureus infection can abnormally activate innate and adaptive immune responses in keratinocytes, resulting in a vicious cycle between Staphylococcus aureus infection and AD. This article reviews the mechanisms of inflammatory damage of keratinocytes induced by Staphylococcus aureus infection in patients with AD, providing a theoretical basis for the study of new targeted drugs. This review also suggests for the management of Staphylococcus aureus infection in patients with AD.
Collapse
Affiliation(s)
- Xin Tian
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Qiongxiao Huang
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Jingyao Liang
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Jianqin Wang
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Jing Zhang
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Yan Yang
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Qianru Ye
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Suling He
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Junlong Li
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| | - Zhouwei Wu
- Department of Dermatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yumei Liu
- Institute of Dermatology, Guangzhou Medical University, Guangzhou, China.,Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, China
| |
Collapse
|
147
|
Ding M, Dong X, Sun YL, Sokolowska M, Akdis M, van de Veen W, Azkur AK, Azkur D, Akdis CA, Gao YD. Recent advances and developments in COVID-19 in the context of allergic diseases. Clin Transl Allergy 2021; 11:e12065. [PMID: 34582102 PMCID: PMC8646609 DOI: 10.1002/clt2.12065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Since the first reports of coronavirus disease 2019 (COVID-19) in Wuhan, China, in December 2019, there have been 198 million confirmed cases worldwide as of August 2021. The scientific community has joined efforts to gain knowledge of the newly emerged virus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the immunopathological mechanisms leading to COVID-19, and its significance for patients with allergies and asthma. METHODS Based on the current literature, recent advances and developments in COVID-19 in the context of allergic diseases were reviewed. RESULTS AND CONCLUSIONS In this review, we discuss the prevalence of COVID-19 in subjects with asthma, attacks of hereditary angioedema, and other allergic diseases during COVID-19. Underlying mechanisms suggest a protective role of allergy in COVID-19, involving eosinophilia, SARS-CoV-2 receptors expression, interferon responses, and other immunological events, but further studies are needed to fully understand those associations. There has been significant progress in disease evaluation and management of COVID-19, and allergy care should continue during the COVID-19 pandemic. The European Academy of Allergy & Clinical Immunology (EAACI) launched a series of statements and position papers providing recommendations on the organization of the allergy clinic, handling of allergen immunotherapy, asthma, drug hypersensitivity, allergic rhinitis, and other allergic diseases. Treatment of allergies using biologics during the COVID-19 pandemic has also been discussed. Allergic reactions to the COVID-19 vaccines, including severe anaphylaxis, have been reported. Vaccination is a prophylactic strategy that can lead to a significant reduction in the mortality and morbidity associated with SARS-CoV-2 infection, and in this review, we discuss the proposed culprit components causing rare adverse reactions and recommendations to mitigate the risk of anaphylactic events during the administration of the vaccines.
Collapse
Affiliation(s)
- Mei Ding
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| | - Xiang Dong
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| | - Yuan-Li Sun
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ahmet Kursat Azkur
- Department of Virology, Faculty of Veterinary Medicine, University of Kirikkale, Kirikkale, Turkey
| | - Dilek Azkur
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, University of Kirikkale, Kirikkale, Turkey
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.,Christine Kühne - Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Ya-Dong Gao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, China
| |
Collapse
|
148
|
Wohlrab J, Stintzing D, Schultz L, Jügelt K, Schroeder OHU. Influence of Janus Kinase Inhibitors on the Neuronal Activity as a Proof-of-Concept Model for Itch. Skin Pharmacol Physiol 2021; 35:94-101. [PMID: 34530431 DOI: 10.1159/000519669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 09/02/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Itching is considered to be a subjective symptom of the activation of neurosensory structures by different signal molecules and trigger factors. The signaling cascades responsible for it are closely linked to inflammatory processes. This explains why itching also occurs in many inflammatory diseases. One of these signaling cascades is mediated by Janus kinases (JAKs). Recently, it could be shown on a molecular level that Janus kinase 1 (JAK1) directly activates frontal cortex neurons and thus can cause chronic itching. OBJECTIVES This study deals with the influence of different JAK inhibitors (JAKi) on the activity of chip-based neural networks of cultured frontal cortex neurons by investigating neurophysiological activity parameters. This in vitro model provides information on dose-dependent effects of model substances with different specificity regarding the inhibition of different JAKs. METHODS Tofacitinib (pan-JAKi), baricitinib (JAK1/2i), and upadacitinib (JAK1i) in a concentration range from 10 nmol/L to 50 μmol/L were tested in a microelectrode array neurochip culture system. RESULTS The results show that the inhibition of the neuronal activity of frontal cortex neurons increases with JAK1 selectivity and is dependent on concentration. CONCLUSION These observations are supported by data from clinical studies in atopic dermatitis and psoriasis. The clinical relevance of these results must be proven by further clinical studies with subjective and objective parameters for itching.
Collapse
Affiliation(s)
- Johannes Wohlrab
- Department of Dermatology and Venereology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.,Institute of Applied Dermatopharmacy, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - David Stintzing
- Institute of Applied Dermatopharmacy, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | | | | | | |
Collapse
|
149
|
Gao S, Duan Y, Chen J, Wang J. Evaluation of Blood Markers at Admission for Predicting Community Acquired Pneumonia in Chronic Obstructive Pulmonary Disease. COPD 2021; 18:557-566. [PMID: 34511022 DOI: 10.1080/15412555.2021.1976739] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Acute exacerbations of Chronic Obstructive Pulmonary Disease (AECOPD) and community acquired pneumonia (CAP) are two common acute attacks in COPD patients and it is not always easy to determine whether a COPD patient at admission has parenchymal infection or bronchial infection. Comprehensive comparison between AECOPD patients and CAP patients with COPD (COPD + CAP) can help us understand them better. We retrospectively collected the medical records of AECOPD and COPD + CAP patients. Systemic inflammation, eosinophilic inflammation, damage to other organs, common chronic comorbidities, structural changes, phenotype and endotype distributions and coagulation functions between two groups were compared and correlations of these characteristics in total subjects, AECOPD patients and COPD + CAP patients were analyzed. Logistic regression analysis was performed to select helpful biomarkers for distinguishing between them. Receiver operator characteristic (ROC) curve was plotted to assess the diagnostic value of selected biomarkers and their combination. A nomogram was established for the differential diagnosis of AECOPD and COPD + CAP. A total of 206 patients were included into our analysis. In these subjects, 104 patients were classified as AECOPD group and 102 patients were considered to have COPD + CAP mainly based on their chest CT scan results. The counts of eosinophils (EOS), basophils (BAS) and lymphocytes (LYM) and percentage of total white blood cell count, hemoglobin and hematocrit were increased in AECOPD patients compared with COPD + CAP patients. The counts of neutrophils (NEU) and percentage of total white blood cell count, C-reactive protein (CRP), Erythrocyte sedimentation rate (ESR), fibrinogen, D-dimer and N-Terminal pro-brain natriuretic peptide (NT-proBNP) levels were increased in COPD + CAP patients. After logistic regression analysis, EOS < 0.5 × 109/L, ESR ≥ 8 mm/H and NT-proBNP ≥ 100 pg/mL were selected as helpful biomarkers for diagnosis of COPD + CAP instead of AECOPD. Area under the ROC curve (AUC) of the combination of selected biomarkers was 0.764(0.698-0.829). A nomogram was established and the calibration curve suggested that fitting efficiency of the nomogram was good. AECOPD and COPD + CAP are markedly different, mainly reflected in eosinophilic inflammation, systemic inflammation and coagulation function. Correlations between some common inflammatory biomarkers are also different in the two groups. A nomogram was established to offer help to clinicians for differential diagnosis of these two diseases.
Collapse
Affiliation(s)
- Shupei Gao
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifei Duan
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinqing Chen
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianmiao Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
150
|
Barrett DE, Estensoro I, Sitjà-Bobadilla A, Bartholomew JL. Intestinal Transcriptomic and Histologic Profiling Reveals Tissue Repair Mechanisms Underlying Resistance to the Parasite Ceratonova shasta. Pathogens 2021; 10:1179. [PMID: 34578212 PMCID: PMC8467531 DOI: 10.3390/pathogens10091179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Myxozoan parasites infect fish worldwide causing significant disease or death in many economically important fish species, including rainbow trout and steelhead trout (Oncorhynchus mykiss). The myxozoan Ceratonova shasta is a parasite of salmon and trout that causes ceratomyxosis, a disease characterized by severe inflammation in the intestine resulting in hemorrhaging and necrosis. Populations of O. mykiss that are genetically fixed for resistance or susceptibility to ceratomyxosis exist naturally, offering a tractable system for studying the immune response to myxozoans. The aim of this study was to understand how steelhead trout that are resistant to the disease respond to C. shasta once it has become established in the intestine and identify potential mechanisms of resistance. RESULTS Sequencing of intestinal mRNA from resistant steelhead trout with severe C. shasta infections identified 417 genes differentially expressed during the initial stage of the infection compared to uninfected control fish. A strong induction of interferon-gamma and interferon-stimulated genes was evident, along with genes involved in cell adhesion and migration. A total of 11,984 genes were differentially expressed during the late stage of the infection, most notably interferon-gamma, interleukin-6, and immunoglobulin transcripts. A distinct hardening of the intestinal tissue and a strong inflammatory reaction in the intestinal submucosa including severe hyperplasia and inflammatory cell infiltrates were observed in response to the infection. The massive upregulation of caspase-14 early in the infection, a protein involved in keratinocyte differentiation might reflect the rapid onset of epithelial repair mechanisms, and the collagenous stratum compactum seemed to limit the spread of C. shasta within the intestinal layers. These observations could explain the ability of resistant fish to eventually recover from the infection. CONCLUSIONS Our results suggest that resistance to ceratomyxosis involves both a rapid induction of key immune factors and a tissue response that limits the spread of the parasite and the subsequent tissue damage. These results improve our understanding of the myxozoan-host dialogue and provide a framework for future studies investigating the infection dynamics of C. shasta and other myxozoans.
Collapse
Affiliation(s)
- Damien E. Barrett
- Department of Microbiology, Oregon State University, Corvallis, OR 97331-3804, USA;
| | - Itziar Estensoro
- Fish Pathology Group, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, 12595 Castellón, Spain; (I.E.); (A.S.-B.)
| | - Ariadna Sitjà-Bobadilla
- Fish Pathology Group, Instituto de Acuicultura Torre de la Sal, Consejo Superior de Investigaciones Científicas, 12595 Castellón, Spain; (I.E.); (A.S.-B.)
| | - Jerri L. Bartholomew
- Department of Microbiology, Oregon State University, Corvallis, OR 97331-3804, USA;
| |
Collapse
|