1
|
Melchior K, Gerner RR, Hossain S, Nuccio SP, Moreira CG, Raffatellu M. IL-22-dependent responses and their role during Citrobacter rodentium infection. Infect Immun 2024; 92:e0009924. [PMID: 38557196 PMCID: PMC11075456 DOI: 10.1128/iai.00099-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
The mouse pathogen Citrobacter rodentium is utilized as a model organism for studying infections caused by the human pathogens enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) and to elucidate mechanisms of mucosal immunity. In response to C. rodentium infection, innate lymphoid cells and T cells secrete interleukin (IL)-22, a cytokine that promotes mucosal barrier function. IL-22 plays a pivotal role in enabling mice to survive and recover from C. rodentium infection, although the exact mechanisms involved remain incompletely understood. Here, we investigated whether particular components of the host response downstream of IL-22 contribute to the cytokine's protective effects during C. rodentium infection. In line with previous research, mice lacking the IL-22 gene (Il22-/- mice) were highly susceptible to C. rodentium infection. To elucidate the role of specific antimicrobial proteins modulated by IL-22, we infected the following knockout mice: S100A9-/- (calprotectin), Lcn2-/- (lipocalin-2), Reg3b-/- (Reg3β), Reg3g-/- (Reg3γ), and C3-/- (C3). All knockout mice tested displayed a considerable level of resistance to C. rodentium infection, and none phenocopied the lethality observed in Il22-/- mice. By investigating another arm of the IL-22 response, we observed that C. rodentium-infected Il22-/- mice exhibited an overall decrease in gene expression related to intestinal barrier integrity as well as significantly elevated colonic inflammation, gut permeability, and pathogen levels in the spleen. Taken together, these results indicate that host resistance to lethal C. rodentium infection may depend on multiple antimicrobial responses acting in concert, or that other IL-22-regulated processes, such as tissue repair and maintenance of epithelial integrity, play crucial roles in host defense to attaching and effacing pathogens.
Collapse
Affiliation(s)
- Karine Melchior
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Romana R. Gerner
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- School of Life Sciences, ZIEL – Institute for Food and Health, Freising-Weihenstephan, Technical University of Munich, Munich, Germany
- Department of Internal Medicine III, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Suzana Hossain
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Sean-Paul Nuccio
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Cristiano Gallina Moreira
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines (CU-UCSD cMAV), La Jolla, California, USA
| |
Collapse
|
2
|
Yamamura Y, Nakashima C, Otsuka A. Interplay of cytokines in the pathophysiology of atopic dermatitis: insights from Murin models and human. Front Med (Lausanne) 2024; 11:1342176. [PMID: 38590314 PMCID: PMC10999685 DOI: 10.3389/fmed.2024.1342176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 04/10/2024] Open
Abstract
The pathogenesis of atopic dermatitis (AD) is understood to be crucially influenced by three main factors: dysregulation of the immune response, barrier dysfunction, and pruritus. In the lesional skin of AD, various innate immune cells, including Th2 cells, type 2 innate lymphoid cells (ILC2s), and basophils, produce Th2 cytokines [interleukin (IL)-4, IL-5, IL-13, IL-31]. Alarmins such as TSLP, IL-25, and IL-33 are also produced by epidermal keratinocytes, amplifying type 2 inflammation. In the chronic phase, not only Th2 cells but also Th22 and Th17 cells increase in number, leading to suppression of filaggrin expression by IL-4, IL-13, and IL-22, which further deteriorates the epidermal barrier function. Dupilumab, which targets IL-4 and IL-13, has shown efficacy in treating moderate to severe AD. Nemolizumab, targeting IL-31RA, effectively reduces pruritus in AD patients. In addition, clinical trials with fezakinumab, targeting IL-22, have demonstrated promising results, particularly in severe AD cases. Conversely, in murine models of AD, several cytokines, initially regarded as promising therapeutic targets, have not demonstrated sufficient efficacy in clinical trials. IL-33 has been identified as a potent activator of immune cells, exacerbating AD in murine models and correlating with disease severity in human patients. However, treatments targeting IL-33 have not shown sufficient efficacy in clinical trials. Similarly, thymic stromal lymphopoietin (TSLP), integral to type 2 immune responses, induces dermatitis in animal models and is elevated in human AD, yet clinical treatments like tezepelumab exhibit limited efficacy. Therapies targeting IL-1α, IL-5, and IL-17 also failed to achieve sufficient efficacy in clinical trials. It has become clear that for treating AD, IL-4, IL-13, and IL-31 are relevant therapeutic targets during the acute phase, while IL-22 emerges as a target in more severe cases. This delineation underscores the necessity of considering distinct pathophysiological aspects and therapeutic targets in AD between mouse models and humans. Consequently, this review delineates the distinct roles of cytokines in the pathogenesis of AD, juxtaposing their significance in human AD from clinical trials against insights gleaned from AD mouse models. This approach will improve our understanding of interspecies variation and facilitate a deeper insight into the pathogenesis of AD in humans.
Collapse
Affiliation(s)
| | - Chisa Nakashima
- Department of Dermatology, Faculty of Medicine, Kindai University Hospital, Osaka, Japan
| | | |
Collapse
|
3
|
Devvanshi H, Kachhwaha R, Manhswita A, Bhatnagar S, Kshetrapal P. Immunological Changes in Pregnancy and Prospects of Therapeutic Pla-Xosomes in Adverse Pregnancy Outcomes. Front Pharmacol 2022; 13:895254. [PMID: 35517798 PMCID: PMC9065684 DOI: 10.3389/fphar.2022.895254] [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: 03/13/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Stringent balance of the immune system is a key regulatory factor in defining successful implantation, fetal development, and timely parturition. Interference in these primary regulatory mechanisms, either at adolescence or prenatal state led to adverse pregnancy outcomes. Fertility restoration with the help of injectable gonadotrophins/progesterone, ovulation-inducing drugs, immunomodulatory drugs (corticosteroids), and reproductive surgeries provides inadequate responses, which manifest its own side effects. The development of a potential diagnostic biomarker and an effectual treatment for adverse pregnancy outcomes is a prerequisite to maternal and child health. Parent cell originated bi-layered-intraluminal nano-vesicles (30-150 nm) also known as exosomes are detected in all types of bodily fluids like blood, saliva, breast milk, urine, etc. Exosomes being the most biological residual structures with the least cytotoxicity are loaded with cargo in the form of RNAs (miRNAs), proteins (cytokines), hormones (estrogen, progesterone, etc.), cDNAs, and metabolites making them chief molecules of cell-cell communication. Their keen involvement in the regulation of biological processes has portrayed them as the power shots of cues to understand the disease's pathophysiology and progression. Recent studies have demonstrated the role of immunexosomes (immunomodulating exosomes) in maintaining unwavering immune homeostasis between the mother and developing fetus for a healthy pregnancy. Moreover, the concentration and size of the exosomes are extensively studied in adverse pregnancies like preeclampsia, gestational diabetes mellitus (GDM), and preterm premature rupture of membrane (pPROMs) as an early diagnostic marker, thus giving in-depth information about their pathophysiology. Exosomes have also been engineered physically as well as genetically to enhance their encapsulation efficiency and specificity in therapy for cancer and adverse pregnancies. Successful bench to bedside discoveries and interventions in cancer has motivated developmental biologists to investigate the role of immunexosomes and their active components. Our review summarizes the pre-clinical studies for the use of these power-shots as therapeutic agents. We envisage that these studies will pave the path for the use of immunexosomes in clinical settings for reproductive problems that arise due to immune perturbance in homeostasis either at adolescence or prenatal state.
Collapse
Affiliation(s)
- Himadri Devvanshi
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Rohit Kachhwaha
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Anima Manhswita
- School of Agriculture and Food Science, The University of Queensland, Brisbane, QLD, Australia
| | - Shinjini Bhatnagar
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| | - Pallavi Kshetrapal
- Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
4
|
Maehara T, Fujimori K. Inhibition of Prostaglandin F 2α Receptors Exaggerates HCl-Induced Lung Inflammation in Mice. Int J Mol Sci 2021; 22:ijms222312843. [PMID: 34884648 PMCID: PMC8657597 DOI: 10.3390/ijms222312843] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 01/11/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are severe respiratory disorders that are caused by aspiration, sepsis, trauma, and pneumonia. A clinical feature of ALI/ARDS is the acute onset of severe hypoxemia, and the mortality rate, which is estimated at 38-50%, remains high. Although prostaglandins (PGs) are detected in the bronchoalveolar lavage fluid of patients with ALI/ARDS, the role of PGF2α in ALI remains unclear. We aimed to clarify the role of PGF2α/PGF2α receptor (FP) signaling in acid-induced ALI using an FP receptor antagonist, AL8810. Intratracheal injection of hydrochloric acid (HCl) increased neutrophil migration into the lungs, leading to respiratory dysfunction. Pre-administration of AL8810 further increased these features. Moreover, pre-treatment with AL8810 enhanced the HCl-induced expression of pro-inflammatory cytokines and neutrophil migratory factors in the lungs. Administration of HCl decreased the gene expression of lung surfactant proteins, which was further reduced by co-administration of AL8810. Administration of AL8810 also increased lung edema and reduced mRNA expression of epithelial sodium channel in the lungs, indicating that AL8810 reduced fluid clearance. Furthermore, AL8810 also increased lipopolysaccharide-induced expression of adhesion molecules such as intracellular adhesion molecule-1 and E-selectin in human umbilical vein endothelial cells. These results indicate that inhibition of FP receptors by AL8810 exacerbated HCl-induced ALI.
Collapse
|
5
|
Lücke J, Sabihi M, Zhang T, Bauditz LF, Shiri AM, Giannou AD, Huber S. The good and the bad about separation anxiety: roles of IL-22 and IL-22BP in liver pathologies. Semin Immunopathol 2021; 43:591-607. [PMID: 33851257 PMCID: PMC8443499 DOI: 10.1007/s00281-021-00854-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/23/2021] [Indexed: 12/17/2022]
Abstract
The human liver fulfills several vital tasks daily and possesses an impressive ability to self-regenerate. However, the capacity of this self-healing process can be exhausted by a variety of different liver diseases, such as alcoholic liver damage, viral hepatitis, or hepatocellular carcinoma. Over time, all these diseases generally lead to progressive liver failure that can become fatal if left untreated. Thus, a great effort has been directed towards the development of innovative therapies. The most recently discovered therapies often involve modifying the patient's immune system to enhance a beneficial immune response. Current data suggest that, among others, the cytokine IL-22 might be a promising therapeutical candidate. IL-22 and its endogenous antagonist, IL-22BP, have been under thorough scientific investigation for nearly 20 years. While IL-22 is mainly produced by TH22 cells, ILC3s, NKT cells, or γδ T cells, sources of IL-22BP include dendritic cells, eosinophils, and CD4+ cells. In many settings, IL-22 was shown to promote regenerative potential and, thus, could protect tissues from pathogens and damage. However, the effects of IL-22 during carcinogenesis are more ambiguous and depend on the tumor entity and microenvironment. In line with its capabilities of neutralizing IL-22 in vivo, IL-22BP possesses often, but not always, an inverse expression pattern compared to its ligand. In this comprehensive review, we will summarize past and current findings regarding the roles of IL-22 and IL-22BP in liver diseases with a particular focus on the leading causes of advanced liver failure, namely, liver infections, liver damage, and liver malignancies.
Collapse
Affiliation(s)
- Jöran Lücke
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Morsal Sabihi
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tao Zhang
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Lennart Fynn Bauditz
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ahmad Mustafa Shiri
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Anastasios D Giannou
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| | - Samuel Huber
- Section of Molecular Immunology und Gastroenterology, I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
6
|
Arshad T, Mansur F, Palek R, Manzoor S, Liska V. A Double Edged Sword Role of Interleukin-22 in Wound Healing and Tissue Regeneration. Front Immunol 2020; 11:2148. [PMID: 33042126 PMCID: PMC7527413 DOI: 10.3389/fimmu.2020.02148] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
Wound healing and tissue regeneration is an intricate biological process that involves repair of cellular damage and maintenance of tissue integrity. Cascades involved in wound healing and tissue regeneration highly overlap with cancer causing pathways. Usually, subsequent tissue damage events include release of a number of cytokines to accomplish post-trauma restoration. IL-22 is one of the cytokines that are immediately produced to initiate immune response against several tissue impairments. IL-22 is a fundamental mediator in inflammation, mucous production, protective role against pathogens, wound healing, and tissue regeneration. However, accumulating evidence suggests pivotal role of IL-22 in instigation of various cancers due to its pro-inflammatory and tissue repairing activity. In this review, we summarize how healing effects of IL-22, when executed in an uncontrollable fashion can lead to carcinogenesis.
Collapse
Affiliation(s)
- Tanzeela Arshad
- Molecular Virology and Immunology Research Group, Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Fizzah Mansur
- Molecular Virology and Immunology Research Group, Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Richard Palek
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
- Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Sobia Manzoor
- Molecular Virology and Immunology Research Group, Atta-ur-Rahman School of Applied Bio-Sciences, National University of Sciences and Technology, Islamabad, Pakistan
- Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| | - Vaclav Liska
- Department of Surgery, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
- Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Prague, Czechia
| |
Collapse
|
7
|
Wang W, Sung N, Gilman-Sachs A, Kwak-Kim J. T Helper (Th) Cell Profiles in Pregnancy and Recurrent Pregnancy Losses: Th1/Th2/Th9/Th17/Th22/Tfh Cells. Front Immunol 2020; 11:2025. [PMID: 32973809 PMCID: PMC7461801 DOI: 10.3389/fimmu.2020.02025] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
During pregnancy, various immune effectors and molecules participating in the immune-microenvironment establish specific maternal tolerance toward the semi-allogeneic fetus. Activated maternal immune effectors by the trophoblast antigens, such as T helper (Th), T cytotoxic (Tc), T regulatory (Treg), and B cells, are involved in the regulation of adaptive immunity. Recognition of active signal through the T cell receptors stimulate the differentiation of naive CD3+CD4+ T cells into specific T cell subsets, such as Th1, Th2, Th9, Th17, Th22, and follicular Th cells (Tfh). Each of these subsets has a significant and distinct role in human pregnancy. Th1 immunity, characterized by immune-inflammatory responses, becomes dominant during the peri-implantation period, and the “controlled” Th1 immunity benefits the invading trophoblasts rather than harm. Quickly after the placental implantation, the early inflammatory Th1 immunity is shifted to the Th2 anti-inflammatory immune responses. The predominant Th2 immunity, which overrules the Th1 immunity at the placental implantation site, protects a fetus by balancing Th1 immunity and accommodate fetal and placental development. Moreover, Treg and Th9 cells regulate local inflammatory immune responses, potentially detrimental to the fetus. Th17 cells induce protective immunity against extracellular microbes during pregnancy. However, excessive Th17 immunity may induce uncontrolled neutrophil infiltration at the maternal-fetal interface. Other Th cell subsets such as Tfh cells, also contribute to pregnancy by setting up favorable humoral immunity during pregnancy. However, dysregulation of Th cell immunity during pregnancy may result in obstetrical complications, such as recurrent pregnancy losses (RPL) and preeclampsia (PE). With this review, we intend to deliver a comprehensive overview of CD4+ Th cell subsets, including Th1, Th2, Th9, Th17, Th22, and Tfh cells, in human pregnancy by reviewing their roles in normal and pathological pregnancies.
Collapse
Affiliation(s)
- Wenjuan Wang
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Department of Clinical Sciences, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Reproductive Medicine Center, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Nayoung Sung
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Department of Clinical Sciences, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Alice Gilman-Sachs
- Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Clinical Immunology Laboratory, Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Department of Clinical Sciences, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States.,Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
8
|
Pian Y, Chai Q, Ren B, Wang Y, Lv M, Qiu J, Zhu M. Type 3 Innate Lymphoid Cells Direct Goblet Cell Differentiation via the LT-LTβR Pathway during Listeria Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:853-863. [PMID: 32591396 DOI: 10.4049/jimmunol.2000197] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022]
Abstract
As a specialized subset of intestinal epithelial cells (IECs), goblet cells (GCs) play an important role during the antibacterial response via mucin production. However, the regulatory mechanisms involved in GC differentiation and function during infection, particularly the role of immune cell-IEC cross-talk, remain largely unknown. In this study, using Villin∆Ltbr conditional knockout mice, we demonstrate that LTβR, expressed on IECs, is required for GC hyperplasia and mucin 2 (MUC2) expression during Listeria infection for host defense but not homeostatic maintenance in the naive state. Analysis of single gene-deficient mice revealed that the ligand lymphotoxin (LT), but not LIGHT, and type 3 innate lymphoid cells (ILC3s), but not conventional T cells, are required for MUC2-dependent Listeria control. Conditional deficiency of LT in ILC3s further confirmed the importance of LT signals derived from ILC3s. Lack of ILC3-derived LT or IEC-derived LTβR resulted in the defective expression of genes related to GC differentiation but was not correlated with IEC proliferation and cell death, which were found to be normal by Ki-67 and Annexin V staining. In addition, the alternative NF-κB signaling pathway (involving RelB) in IECs was found to be required for the expression of GC differentiation-related genes and Muc2 and required for the anti-Listeria response. Therefore, our data together suggest a previously unrecognized ILC3-IEC interaction and LT-LTβR-RelB signaling axis governing GC differentiation and function during Listeria infection for host defense.
Collapse
Affiliation(s)
- Yaya Pian
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qian Chai
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China;
| | - Boyang Ren
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Yue Wang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Mengjie Lv
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mingzhao Zhu
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; .,College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; and
| |
Collapse
|
9
|
Logiodice F, Lombardelli L, Kullolli O, Haller H, Maggi E, Rukavina D, Piccinni MP. Decidual Interleukin-22-Producing CD4+ T Cells (Th17/Th0/IL-22+ and Th17/Th2/IL-22+, Th2/IL-22+, Th0/IL-22+), Which Also Produce IL-4, Are Involved in the Success of Pregnancy. Int J Mol Sci 2019; 20:E428. [PMID: 30669479 PMCID: PMC6359245 DOI: 10.3390/ijms20020428] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/17/2022] Open
Abstract
Trophoblast expressing paternal HLA-C resembles a semiallograft, and could be rejected by maternal T cells. IL-22 seems to be involved in allograft rejection and thus could be responsible for miscarriages. We examined the role of decidual IL-22-producing CD4+ T on human pregnancy. In those experiencing successful pregnancy and those experiencing unexplained recurrent abortion (URA), the levels of IL-22 produced by decidual CD4+ T cells are higher than those of peripheral blood T cells. We found a correlation of IL-22 and IL-4 produced by decidual CD4+ T cells in those experiencing successful pregnancy, not in those experiencing URA. The correlation of IL-22 and IL-4 was also found in the serum of successful pregnancy. A prevalence of CD4+ T cells producing IL-22 and IL-4 (Th17/Th2/IL-22+, Th17/Th0/IL-22+, Th17/Th2/IL-22+, and Th0/IL-22+ cells) was observed in decidua of those experiencing successful pregnancy, whereas Th17/Th1/IL-22+ cells, which do not produce IL-4, are prevalent in those experiencing URA. Th17/Th2/IL-22+ and Th17/Th0/IL-22+ cells are exclusively present at the embryo implantation site where IL-4, GATA-3, IL-17A, ROR-C, IL-22, and AHR mRNA are expressed. T-bet and IFN-γ mRNA are found away from the implantation site. There is no pathogenic role of IL-22 when IL-4 is also produced by decidual CD4+ cells. Th17/Th2/IL-22+ and Th17/Th0/IL-22+ cells seem to be crucial for embryo implantation.
Collapse
Affiliation(s)
- Federica Logiodice
- Department of Experimental and Clinical Medicine and DENOTHE Excellence Center, University of Florence, 50134 Florence, Italy.
| | - Letizia Lombardelli
- Department of Experimental and Clinical Medicine and DENOTHE Excellence Center, University of Florence, 50134 Florence, Italy.
| | - Ornela Kullolli
- Department of Experimental and Clinical Medicine and DENOTHE Excellence Center, University of Florence, 50134 Florence, Italy.
| | - Herman Haller
- Department of Gynecology and Obstetrics, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia.
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine and DENOTHE Excellence Center, University of Florence, 50134 Florence, Italy.
- Immunology Area, IRCCS Bambino Gesù Children's Hospital, 00165 Rome, Italy.
| | - Daniel Rukavina
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia.
| | - Marie-Pierre Piccinni
- Department of Experimental and Clinical Medicine and DENOTHE Excellence Center, University of Florence, 50134 Florence, Italy.
| |
Collapse
|
10
|
Zheng Y, Li T. Interleukin-22, a potent target for treatment of non-autoimmune diseases. Hum Vaccin Immunother 2018; 14:2811-2819. [PMID: 30335564 DOI: 10.1080/21645515.2018.1509649] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Interleukin -22 (IL-22) is a member of interleukin-10 (IL-10) family cytokines that is produced by different types of lymphocytes included in both innate and adaptive immune systems. These lymphocytes include activated T cells, most notably Th17 and Th22 cells, as well as NK cells, γδ T cells, etc. IL-22 mediate its effects via the IL-22-IL-22R complex and subsequent Janus Kinase-signal transduces and activators transcription (JAK-STAT) signaling pathway. According to recent evidence, IL-22 played a critical role in the pathogenesis of many non-autoimmune diseases. In this review, we mainly discussed the recent findings and advancements of the role of IL-22 in several non-autoimmune diseases, such as acute lung injury, atherosclerosis and some bacterial infections, suggesting that IL-22 may have therapeutic potential for treating non-autoimmune diseases.
Collapse
Affiliation(s)
- Yue Zheng
- a Cardiology , The Third Central Clinical College of Tianjin Medical University , Tianjin , China.,b Cardiology , Tianjin Key Laboratory of Artificial Cell.,c Artificial Cell Engineering Technology Research Center of Public Health Ministry , Tianjin , China.,d Tianjin Institute of Hepatobiliary Disease , Tianjin , China
| | - Tong Li
- b Cardiology , Tianjin Key Laboratory of Artificial Cell.,c Artificial Cell Engineering Technology Research Center of Public Health Ministry , Tianjin , China.,d Tianjin Institute of Hepatobiliary Disease , Tianjin , China.,e The Third Central Hospital of Tianjin , Tianjin , China
| |
Collapse
|
11
|
Disson O, Blériot C, Jacob JM, Serafini N, Dulauroy S, Jouvion G, Fevre C, Gessain G, Thouvenot P, Eberl G, Di Santo JP, Peduto L, Lecuit M. Peyer's patch myeloid cells infection by Listeria signals through gp38 + stromal cells and locks intestinal villus invasion. J Exp Med 2018; 215:2936-2954. [PMID: 30355616 PMCID: PMC6219733 DOI: 10.1084/jem.20181210] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/05/2018] [Accepted: 09/28/2018] [Indexed: 12/12/2022] Open
Abstract
The foodborne pathogen Listeria monocytogenes (Lm) crosses the intestinal villus epithelium via goblet cells (GCs) upon the interaction of Lm surface protein InlA with its receptor E-cadherin. Here, we show that Lm infection accelerates intestinal villus epithelium renewal while decreasing the number of GCs expressing luminally accessible E-cadherin, thereby locking Lm portal of entry. This novel innate immune response to an enteropathogen is triggered by the infection of Peyer's patch CX3CR1+ cells and the ensuing production of IL-23. It requires STAT3 phosphorylation in epithelial cells in response to IL-22 and IL-11 expressed by lamina propria gp38+ stromal cells. Lm-induced IFN-γ signaling and STAT1 phosphorylation in epithelial cells is also critical for Lm-associated intestinal epithelium response. GC depletion also leads to a decrease in colon mucus barrier thickness, thereby increasing host susceptibility to colitis. This study unveils a novel innate immune response to an enteropathogen, which implicates gp38+ stromal cells and locks intestinal villus invasion, but favors colitis.
Collapse
Affiliation(s)
- Olivier Disson
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Camille Blériot
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Jean-Marie Jacob
- Institut Pasteur, Stroma, Inflammation and Tissue Repair Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1224, Paris, France
| | - Nicolas Serafini
- Institut Pasteur, Innate Immunity Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1223, Paris, France
| | - Sophie Dulauroy
- Institut National de la Santé et de la Recherche Médicale U1224, Paris, France.,Institut Pasteur, Microenvironnement and Immunity Unit, Paris, France
| | - Grégory Jouvion
- Institut Pasteur, Human Histopathology and Animal Models Unit, Paris, France
| | - Cindy Fevre
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Grégoire Gessain
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Pierre Thouvenot
- Institut Pasteur, Biology of Infection Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France
| | - Gérard Eberl
- Institut National de la Santé et de la Recherche Médicale U1224, Paris, France.,Institut Pasteur, Microenvironnement and Immunity Unit, Paris, France
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1223, Paris, France
| | - Lucie Peduto
- Institut Pasteur, Stroma, Inflammation and Tissue Repair Unit, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1224, Paris, France
| | - Marc Lecuit
- Institut Pasteur, Biology of Infection Unit, Paris, France .,Institut National de la Santé et de la Recherche Médicale U1117, Paris, France.,Paris Descartes University, Department of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, APHP, Institut Imagine, Paris, France
| |
Collapse
|
12
|
Cabinian A, Sinsimer D, Tang M, Jang Y, Choi B, Laouar Y, Laouar A. Gut symbiotic microbes imprint intestinal immune cells with the innate receptor SLAMF4 which contributes to gut immune protection against enteric pathogens. Gut 2018; 67:847-859. [PMID: 28341747 PMCID: PMC5890651 DOI: 10.1136/gutjnl-2016-313214] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 02/27/2017] [Accepted: 03/04/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND Interactions between host immune cells and gut microbiota are crucial for the integrity and function of the intestine. How these interactions regulate immune cell responses in the intestine remains a major gap in the field. AIM We have identified the signalling lymphocyte activation molecule family member 4 (SLAMF4) as an immunomodulator of the intestinal immunity. The aim is to determine how SLAMF4 is acquired in the gut and what its contribution to intestinal immunity is. METHODS Expression of SLAMF4 was assessed in mice and humans. The mechanism of induction was studied using GFPtg bone marrow chimaera mice, lymphotoxin α and TNLG8A-deficient mice, as well as gnotobiotic mice. Role in immune protection was revealed using oral infection with Listeria monocytogenes and Cytobacter rodentium. RESULTS SLAMF4 is a selective marker of intestinal immune cells of mice and humans. SLAMF4 induction occurs directly in the intestinal mucosa without the involvement of the gut-associated lymphoid tissue. Gut bacterial products, particularly those of gut anaerobes, and gut-resident antigen-presenting cell (APC) TNLG8A are key contributors of SLAMF4 induction in the intestine. Importantly, lack of SLAMF4 expression leads the increased susceptibility of mice to infection by oral pathogens culminating in their premature death. CONCLUSIONS SLAMF4 is a marker of intestinal immune cells which contributes to the protection against enteric pathogens and whose expression is dependent on the presence of the gut microbiota. This discovery provides a possible mechanism for answering the long-standing question of how the intertwining of the host and gut microbial biology regulates immune cell responses in the gut.
Collapse
Affiliation(s)
- Allison Cabinian
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Daniel Sinsimer
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - May Tang
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| | - Youngsoon Jang
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Bongkum Choi
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Yasmina Laouar
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Amale Laouar
- The Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey, USA
| |
Collapse
|
13
|
Yoon J, Leyva-Castillo JM, Wang G, Galand C, Oyoshi MK, Kumar L, Hoff S, He R, Chervonsky A, Oppenheim JJ, Kuchroo VK, van den Brink MRM, Malefyt RDW, Tessier PA, Fuhlbrigge R, Rosenstiel P, Terhorst C, Murphy G, Geha RS. IL-23 induced in keratinocytes by endogenous TLR4 ligands polarizes dendritic cells to drive IL-22 responses to skin immunization. J Exp Med 2016; 213:2147-66. [PMID: 27551155 PMCID: PMC5032726 DOI: 10.1084/jem.20150376] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/25/2016] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is a Th2-dominated inflammatory skin disease characterized by epidermal thickening. Serum levels of IL-22, a cytokine known to induce keratinocyte proliferation, are elevated in AD, and Th22 cells infiltrate AD skin lesions. We show that application of antigen to mouse skin subjected to tape stripping, a surrogate for scratching, induces an IL-22 response that drives epidermal hyperplasia and keratinocyte proliferation in a mouse model of skin inflammation that shares many features of AD. DC-derived IL-23 is known to act on CD4(+) T cells to induce IL-22 production. However, the mechanisms that drive IL-23 production by skin DCs in response to cutaneous sensitization are not well understood. We demonstrate that IL-23 released by keratinocytes in response to endogenous TLR4 ligands causes skin DCs, which selectively express IL-23R, to up-regulate their endogenous IL-23 production and drive an IL-22 response in naive CD4(+) T cells that mediates epidermal thickening. We also show that IL-23 is released in human skin after scratching and polarizes human skin DCs to drive an IL-22 response, supporting the utility of IL-23 and IL-22 blockade in AD.
Collapse
Affiliation(s)
- Juhan Yoon
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Juan Manuel Leyva-Castillo
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Guoxing Wang
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Claire Galand
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Michiko K Oyoshi
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Lalit Kumar
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Sabine Hoff
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Rui He
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | | | - Joost J Oppenheim
- Laboratory of Molecular Immunoregulation, National Cancer Institute, Frederick, MD 21702
| | - Vijay K Kuchroo
- Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Marcel R M van den Brink
- Department of Immunology and Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | | | - Philippe A Tessier
- Centre de Recherche du Centre Hospitalier de l'Université Laval, Sainte-Foy, Quebec QC G1V 4G2, Canada
| | - Robert Fuhlbrigge
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - George Murphy
- Department of Dermatology, Harvard Medical School, Boston, MA 02115 Division of Dermatopathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Raif S Geha
- Division of Immunology, Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
14
|
Dixon BREA, Radin JN, Piazuelo MB, Contreras DC, Algood HMS. IL-17a and IL-22 Induce Expression of Antimicrobials in Gastrointestinal Epithelial Cells and May Contribute to Epithelial Cell Defense against Helicobacter pylori. PLoS One 2016; 11:e0148514. [PMID: 26867135 PMCID: PMC4750979 DOI: 10.1371/journal.pone.0148514] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 01/19/2016] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori colonization of the human stomach can lead to adverse clinical outcomes including gastritis, peptic ulcers, or gastric cancer. Current data suggest that in addition to bacterial virulence factors, the magnitude and types of immune responses influence the outcome of colonization. Specifically, CD4+ T cell responses impact the pathology elicited in response to H. pylori. Because gastritis is believed to be the initiating host response to more detrimental pathological outcomes, there has been a significant interest in pro-inflammatory T cell cytokines, including the cytokines produced by T helper 17 cells. Th17 cells produce IL-17A, IL-17F, IL-21 and IL-22. While these cytokines have been linked to inflammation, IL-17A and IL-22 are also associated with anti-microbial responses and control of bacterial colonization. The goal of this research was to determine the role of IL-22 in activation of antimicrobial responses in models of H. pylori infection using human gastric epithelial cell lines and the mouse model of H. pylori infection. Our data indicate that IL-17A and IL-22 work synergistically to induce antimicrobials and chemokines such as IL-8, components of calprotectin (CP), lipocalin (LCN) and some β-defensins in both human and primary mouse gastric epithelial cells (GEC) and gastroids. Moreover, IL-22 and IL-17A-activated GECs were capable of inhibiting growth of H. pylori in vitro. While antimicrobials were activated by IL-17A and IL-22 in vitro, using a mouse model of H. pylori infection, the data herein indicate that IL-22 deficiency alone does not render mice more susceptible to infection, change their antimicrobial gene transcription, or significantly change their inflammatory response.
Collapse
Affiliation(s)
- Beverly R. E. A. Dixon
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jana N. Radin
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - M. Blanca Piazuelo
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Diana C. Contreras
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Holly M. Scott Algood
- Veterans Affairs Tennessee Valley Healthcare Services, Nashville, Tennessee, United States of America
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
15
|
Okita Y, Shiono T, Yahagi A, Hamada S, Umemura M, Matsuzaki G. Interleukin-22-Induced Antimicrobial Phospholipase A2 Group IIA Mediates Protective Innate Immunity of Nonhematopoietic Cells against Listeria monocytogenes. Infect Immun 2016; 84:573-9. [PMID: 26644377 PMCID: PMC4730562 DOI: 10.1128/iai.01000-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/30/2015] [Indexed: 01/07/2023] Open
Abstract
Listeria monocytogenes is a bacterial pathogen which establishes intracellular parasitism in various cells, including macrophages and nonhematopoietic cells, such as hepatocytes. It has been reported that several proinflammatory cytokines have pivotal roles in innate protection against L. monocytogenes infection. We found that a proinflammatory cytokine, interleukin 22 (IL-22), was expressed by CD3(+) CD4(+) T cells at an early stage of L. monocytogenes infection in mice. To assess the influence of IL-22 on L. monocytogenes infection in hepatocytes, cells of a human hepatocellular carcinoma line, HepG2, were treated with IL-22 before L. monocytogenes infection in vitro. Gene expression analysis of the IL-22-treated HepG2 cells identified phospholipase A2 group IIA (PLA2G2A) as an upregulated antimicrobial molecule. Addition of recombinant PLA2G2A to the HepG2 culture significantly suppressed L. monocytogenes infection. Culture supernatant of the IL-22-treated HepG2 cells contained bactericidal activity against L. monocytogenes, and the activity was abrogated by a specific PLA2G2A inhibitor, demonstrating that HepG2 cells secreted PLA2G2A, which killed extracellular L. monocytogenes. Furthermore, colocalization of PLA2G2A and L. monocytogenes was detected in the IL-22-treated infected HepG2 cells, which suggests involvement of PLA2G2A in the mechanism of intracellular killing of L. monocytogenes by HepG2 cells. These results suggest that IL-22 induced at an early stage of L. monocytogenes infection enhances innate immunity against L. monocytogenes in the liver by stimulating hepatocytes to produce an antimicrobial molecule, PLA2G2A.
Collapse
Affiliation(s)
- Yamato Okita
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Takeru Shiono
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Ayano Yahagi
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Satoru Hamada
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Masayuki Umemura
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Goro Matsuzaki
- Molecular Microbiology Group, Department of Infectious Diseases, Tropical Biosphere Research Center, and Department of Host Defense, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| |
Collapse
|
16
|
Nasal vaccination stimulates CD8(+) T cells for potent protection against mucosal Brucella melitensis challenge. Immunol Cell Biol 2016; 94:496-508. [PMID: 26752510 PMCID: PMC4879022 DOI: 10.1038/icb.2016.5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 12/10/2015] [Accepted: 01/05/2016] [Indexed: 12/31/2022]
Abstract
Brucellosis remains a significant zoonotic threat worldwide. Humans and animals acquire infection via their oropharynx and upper respiratory tract following oral or aerosol exposure. After mucosal infection, brucellosis develops into a systemic disease. Mucosal vaccination could offer a viable alternative to conventional injection practices to deter disease. Using a nasal vaccination approach, the ΔznuA B. melitensis was found to confer potent protection against pulmonary Brucella challenge, and reduce colonization of spleens and lungs by more than 2500-fold, with more than 50% of vaccinated mice showing no detectable brucellae. Furthermore, tenfold more brucellae-specific, IFN-γ-producing CD8+ T cells than CD4+ T cells were induced in the spleen and respiratory lymph nodes. Evaluation of pulmonary and splenic CD8+ T cells from mice vaccinated with ΔznuA B. melitensis revealed that these expressed an activated effector memory (CD44hiCD62LloCCR7lo) T cells producing elevated levels of IFN-γ, TNF-α, perforin, and granzyme B. To assess the relative importance of these increased numbers of CD8+ T cells, CD8−/− mice were challenged with virulent B. melitensis, and they showed markedly increased bacterial loads in organs in contrast to similarly challenged CD4−/− mice. Only ΔznuA B. melitensis- and Rev-1-vaccinated CD4−/− and wild-type mice, not CD8−/− mice, were completely protected against Brucella challenge. Determination of cytokines responsible for conferring protection showed the relative importance of IFN-γ, but not IL-17. Unlike wild-type mice, IL-17 was greatly induced in IFN-γ−/− mice, but IL-17 could not substitute for IFN-γ’s protection, although an increase in brucellae dissemination was observed upon in vivo IL-17 neutralization. These results show that nasal ΔznuA B. melitensis vaccination represents an attractive means to stimulate systemic and mucosal immune protection via CD8+ T cell engagement.
Collapse
|
17
|
Pichavant M, Sharan R, Le Rouzic O, Olivier C, Hennegrave F, Rémy G, Pérez-Cruz M, Koné B, Gosset P, Just N, Gosset P. IL-22 Defect During Streptococcus pneumoniae Infection Triggers Exacerbation of Chronic Obstructive Pulmonary Disease. EBioMedicine 2015; 2:1686-96. [PMID: 26870795 PMCID: PMC4740310 DOI: 10.1016/j.ebiom.2015.09.040] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 12/22/2022] Open
Abstract
Progression of chronic obstructive pulmonary disease (COPD) is linked to episodes of exacerbations caused by bacterial infections due to Streptococcus pneumoniae. Our objective was to identify during COPD, factors of susceptibility to bacterial infections among cytokine network and their role in COPD exacerbations. S. pneumoniae was used to sub-lethally challenge mice chronically exposed to air or cigarette smoke (CS) and to stimulate peripheral blood mononuclear cells (PBMC) from non-smokers, smokers and COPD patients. The immune response and the cytokine production were evaluated. Delayed clearance of the bacteria and stronger lung inflammation observed in infected CS-exposed mice were associated with an altered production of IL-17 and IL-22 by innate immune cells. This defect was related to a reduced production of IL-1β and IL-23 by antigen presenting cells. Importantly, supplementation with recombinant IL-22 restored bacterial clearance in CS-exposed mice and limited lung alteration. In contrast with non-smokers, blood NK and NKT cells from COPD patients failed to increase IL-17 and IL-22 levels in response to S. pneumoniae, in association with a defect in IL-1β and IL-23 secretion. This study identified IL-17 and IL-22 as susceptibility factors in COPD exacerbation. Therefore targeting such cytokines could represent a potent strategy to control COPD exacerbation.
Collapse
Key Words
- AM, alveolar macrophages
- APC, antigen presenting cells
- BAL, broncho-alveolar lavage
- Bacterial infection
- CFU, colony forming unit
- COPD, chronic obstructive pulmonary disease
- CS, cigarette smoke
- Chronic obstructive pulmonary disease
- DC, dendritic cells
- IL-22
- Innate immunity
- NK, natural killer cells
- NKT, natural killer T cells
- PBMC, peripheral blood mononuclear cells
- Sp, Streptococcus pneumoniae
Collapse
Affiliation(s)
- Muriel Pichavant
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Riti Sharan
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Olivier Le Rouzic
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
- Service de Pneumologie, Hôpital Calmette, CHRU, Lille, France
| | - Cécile Olivier
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
- Service de Pneumologie, Hôpital Calmette, CHRU, Lille, France
| | - Florence Hennegrave
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
- Service de Pneumologie, Hôpital Calmette, CHRU, Lille, France
| | - Gaëlle Rémy
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Magdiel Pérez-Cruz
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Bachirou Koné
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| | - Pierre Gosset
- Service d'Anatomo-Pathologie, Hôpital Saint Vincent de Paul, Lille, France
| | - Nicolas Just
- Service de Pneumologie, Hôpital Victor Provo, Roubaix, France
| | - Philippe Gosset
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, F-59019 Lille, France
- Université Lille Nord de France, F-59000 Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, F-59021 Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, F-59019 Lille, France
- Institut Fédératif de Recherche 142, F-59019 Lille, France
| |
Collapse
|
18
|
Zhao D, Long XD, Lu TF, Wang T, Zhang WW, Liu YX, Cui XL, Dai HJ, Xue F, Xia Q. Metformin decreases IL-22 secretion to suppress tumor growth in an orthotopic mouse model of hepatocellular carcinoma. Int J Cancer 2015; 136:2556-65. [PMID: 25370454 DOI: 10.1002/ijc.29305] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/21/2014] [Indexed: 12/17/2022]
Abstract
Epidemiological, preclinical and cellular studies in the last 5 years have shown that metformin exerts anti-tumoral properties, but its mode of action in cancer remains unclear. Here, we investigated the effects of metformin on a mouse hepatocellular carcinoma (HCC) model and tumor-associated T cell immune responses. Oral metformin administration led to a significant reduction of tumor growth, which was accompanied by decreased interleukin-22 (IL-22). Meanwhile, IL-22-induced STAT3 phosphorylation and upregulation of downstream genes Bcl-2 and cyclin D1 were inhibited by metformin. At the cellular level, metformin attenuated Th1- and Th17-derived IL-22 production. Furthermore, metformin inhibited de novo generation of Th1 and Th17 cells from naive CD4(+) cells. These observations were further supported by the fact that metformin treatment inhibited CD3/CD28-induced IFN-γ and IL-17A expression along with the transcription factors that drive their expression (T-bet [Th1] and ROR-γt [Th17], respectively). The effects of metformin on T cell differentiation were mediated by downregulated STAT3 and STAT4 phosphorylation via the AMP-activated kinase-mammalian target of rapamycin complex 1 pathway. Notably, metformin led to a reduction in glucose transporter Glut1 expression, resulting in less glucose uptake, which is critical to regulate CD4(+) T cell fate. Taken together, these findings provide evidence for the growth-inhibitory and immune-modulatory effects of metformin in HCC and thus, broaden our understanding about the action of metformin in liver cancer treatment.
Collapse
Affiliation(s)
- Dong Zhao
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wittkopf N, Pickert G, Billmeier U, Mahapatro M, Wirtz S, Martini E, Leppkes M, Neurath MF, Becker C. Activation of intestinal epithelial Stat3 orchestrates tissue defense during gastrointestinal infection. PLoS One 2015; 10:e0118401. [PMID: 25799189 PMCID: PMC4370566 DOI: 10.1371/journal.pone.0118401] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 12/14/2014] [Indexed: 01/21/2023] Open
Abstract
Gastrointestinal infections with EHEC and EPEC are responsible for outbreaks of diarrheal diseases and represent a global health problem. Innate first-line-defense mechanisms such as production of mucus and antimicrobial peptides by intestinal epithelial cells are of utmost importance for host control of gastrointestinal infections. For the first time, we directly demonstrate a critical role for Stat3 activation in intestinal epithelial cells upon infection of mice with Citrobacter rodentium – a murine pathogen that mimics human infections with attaching and effacing Escherichia coli. C. rodentium induced transcription of IL-6 and IL-22 in gut samples of mice and was associated with activation of the transcription factor Stat3 in intestinal epithelial cells. C. rodentium infection induced expression of several antimicrobial peptides such as RegIIIγ and Pla2g2a in the intestine which was critically dependent on Stat3 activation. Consequently, mice with specific deletion of Stat3 in intestinal epithelial cells showed increased susceptibility to C. rodentium infection as indicated by high bacterial load, severe gut inflammation, pronounced intestinal epithelial cell death and dissemination of bacteria to distant organs. Together, our data implicate an essential role for Stat3 activation in intestinal epithelial cells during C. rodentium infection. Stat3 concerts the host response to bacterial infection by controlling bacterial growth and suppression of apoptosis to maintain intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Nadine Wittkopf
- Department of Medicine 1, Friedrich-Alexander-University, 91052 Erlangen, Germany
| | - Geethanjali Pickert
- Institute of Translational Immunology, Johannes Gutenberg-University, 55131 Mainz, Germany
| | - Ulrike Billmeier
- Department of Medicine 1, Friedrich-Alexander-University, 91052 Erlangen, Germany
| | - Mousumi Mahapatro
- Department of Medicine 1, Friedrich-Alexander-University, 91052 Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-University, 91052 Erlangen, Germany
| | - Eva Martini
- Department of Medicine 1, Friedrich-Alexander-University, 91052 Erlangen, Germany
| | - Moritz Leppkes
- Department of Medicine 1, Friedrich-Alexander-University, 91052 Erlangen, Germany
| | | | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-University, 91052 Erlangen, Germany
- * E-mail:
| |
Collapse
|
20
|
Abstract
Interleukin-22 (IL-22) is a recently described IL-10 family cytokine that is produced by T helper (Th) 17 cells, γδ T cells, NKT cells, and newly described innate lymphoid cells (ILCs). Knowledge of IL-22 biology has evolved rapidly since its discovery in 2000, and a role for IL-22 has been identified in numerous tissues, including the intestines, lung, liver, kidney, thymus, pancreas, and skin. IL-22 primarily targets nonhematopoietic epithelial and stromal cells, where it can promote proliferation and play a role in tissue regeneration. In addition, IL-22 regulates host defense at barrier surfaces. However, IL-22 has also been linked to several conditions involving inflammatory tissue pathology. In this review, we assess the current understanding of this cytokine, including its physiologic and pathologic effects on epithelial cell function.
Collapse
|
21
|
Pham TAN, Clare S, Goulding D, Arasteh JM, Stares MD, Browne HP, Keane JA, Page AJ, Kumasaka N, Kane L, Mottram L, Harcourt K, Hale C, Arends MJ, Gaffney DJ, Dougan G, Lawley TD. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization resistance to an opportunistic pathogen. Cell Host Microbe 2014; 16:504-16. [PMID: 25263220 PMCID: PMC4190086 DOI: 10.1016/j.chom.2014.08.017] [Citation(s) in RCA: 228] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/12/2014] [Accepted: 08/18/2014] [Indexed: 12/17/2022]
Abstract
Our intestinal microbiota harbors a diverse microbial community, often containing opportunistic bacteria with virulence potential. However, mutualistic host-microbial interactions prevent disease by opportunistic pathogens through poorly understood mechanisms. We show that the epithelial interleukin-22 receptor IL-22RA1 protects against lethal Citrobacter rodentium infection and chemical-induced colitis by promoting colonization resistance against an intestinal opportunistic bacterium, Enterococcus faecalis. Susceptibility of Il22ra1(-/-) mice to C. rodentium was associated with preferential expansion and epithelial translocation of pathogenic E. faecalis during severe microbial dysbiosis and was ameloriated with antibiotics active against E. faecalis. RNA sequencing analyses of primary colonic organoids showed that IL-22RA1 signaling promotes intestinal fucosylation via induction of the fucosyltransferase Fut2. Additionally, administration of fucosylated oligosaccharides to C. rodentium-challenged Il22ra1(-/-) mice attenuated infection and promoted E. faecalis colonization resistance by restoring the diversity of anaerobic commensal symbionts. These results support a model whereby IL-22RA1 enhances host-microbiota mutualism to limit detrimental overcolonization by opportunistic pathogens.
Collapse
Affiliation(s)
- Tu Anh N Pham
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Simon Clare
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - David Goulding
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Julia M Arasteh
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Mark D Stares
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Hilary P Browne
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Jacqueline A Keane
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Andrew J Page
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Natsuhiko Kumasaka
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Leanne Kane
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Lynda Mottram
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Katherine Harcourt
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Christine Hale
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Mark J Arends
- Department of Pathology, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Daniel J Gaffney
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Gordon Dougan
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK
| | - Trevor D Lawley
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, CB10 1SA, UK.
| |
Collapse
|
22
|
Hradetzky S, Roesner LM, Balaji H, Heratizadeh A, Mittermann I, Valenta R, Werfel T. Cytokine effects induced by the human autoallergen α-NAC. J Invest Dermatol 2014; 134:1570-1578. [PMID: 24441101 DOI: 10.1038/jid.2014.25] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 12/03/2013] [Accepted: 12/16/2013] [Indexed: 12/20/2022]
Abstract
Autoallergy is a phenomenon found in a subgroup of patients with atopic dermatitis (AD). These patients exhibit serum IgE reactivity toward autoantigens like the alpha-chain of the nascent polypeptide-associated complex (α-NAC; Hom s 2). α-NAC has been shown before to induce T-cell proliferation and secretion of IFN-γ. To elucidate the immune modulating functions α-NAC may exert, we analyzed its effects on cytokine transcription and secretion in peripheral blood mononuclear cells (PBMCs), monocytes, and CD4+ T cells. Transcription and secretion of IFN-γ, IL-17, and IL-22 were increased in α-NAC-stimulated PBMCs. As IL-17 was significantly upregulated by α-NAC, we assessed signal transduction in PBMCs and found signal transducer and activator of transcription 3 phosphorylation in α-NAC-stimulated cells. Furthermore, we could show the importance of monocyte activation by α-NAC, as isolated T cells reacted only weakly toward the stimulation. Inhibition of IL-23 p19 led to lower amounts of IL-17 in the PBMC supernatants after α-NAC stimulation. α-NAC stimulation of PBMCs from non-allergic donors resulted in secretion of IL-10, which was greatly reduced in PBMCs from α-NAC-sensitized AD patients. Our findings provide insights into the mechanisms of autoallergy, investigating the interplay of immune cells, signaling events, and cytokines, which are known to be relevant in atopic skin inflammation.
Collapse
Affiliation(s)
- Susanne Hradetzky
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany.
| | - Lennart M Roesner
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Hari Balaji
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Annice Heratizadeh
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Irene Mittermann
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Rudolf Valenta
- Division of Immunopathology, Department of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Thomas Werfel
- Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
23
|
Peng Y, Gao X, Yang J, Shekhar S, Wang S, Fan Y, Zhao W, Yang X. Interleukin-22 promotes T helper 1 (Th1)/Th17 immunity in chlamydial lung infection. Mol Med 2014; 20:109-19. [PMID: 24531835 DOI: 10.2119/molmed.2013.00115] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 01/30/2014] [Indexed: 01/10/2023] Open
Abstract
The role of interleukin-22 (IL-22) in intracellular bacterial infections is a controversial issue, although the contribution of this cytokine to host defense against extracellular bacterial pathogens has been well established. In this study, we focused on an intra-cellular bacterium, Chlamydia, and evaluated the production and function of IL-22 in host defense against chlamydial lung infection using a mouse model. We found that Chlamydia muridarum infection elicited quick IL-22 responses in the lung, which increased during infection and were reduced when bacterial loads decreased. More importantly, blockade of endogenous IL-22 using neutralizing anti-IL-22 monoclonal antibodies (mAb) resulted in more severe disease in the mice, leading to significantly higher weight loss and bacterial growth and much more severe pathological changes than treatment with isotype control antibody. Immunological analyses identified significantly lower T helper 1 (Th1) and Th17 responses in the IL-22-neutralized mice. In contrast, intranasal administration of exogenous IL-22 significantly enhanced protection following chlamydial lung infection, which was associated with a significant increase of Th17 response. The data demonstrate that IL-22 is a critical cytokine, mediating host defense against chlamydial lung infection and coordinating the function of distinct Th-cell subsets, particularly Th1 and Th17, in the process.
Collapse
Affiliation(s)
- Ying Peng
- Departments of Medical Microbiology and Immunology, University of Manitoba, Winnipeg, Manitoba, Canada Department of Medical Microbiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Xiaoling Gao
- Departments of Medical Microbiology and Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jie Yang
- Departments of Medical Microbiology and Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sudhanshu Shekhar
- Departments of Medical Microbiology and Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Shuhe Wang
- Departments of Medical Microbiology and Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Yijun Fan
- Departments of Medical Microbiology and Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Weiming Zhao
- Department of Medical Microbiology, Shandong University School of Medicine, Jinan, Shandong, PR China
| | - Xi Yang
- Departments of Medical Microbiology and Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
24
|
Behnsen J, Jellbauer S, Wong CP, Edwards RA, George MD, Ouyang W, Raffatellu M. The cytokine IL-22 promotes pathogen colonization by suppressing related commensal bacteria. Immunity 2014; 40:262-73. [PMID: 24508234 PMCID: PMC3964146 DOI: 10.1016/j.immuni.2014.01.003] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 11/27/2013] [Indexed: 12/16/2022]
Abstract
Interleukin-22 (IL-22) is highly induced in response to infections with a variety of pathogens, and its main functions are considered to be tissue repair and host defense at mucosal surfaces. Here we showed that IL-22 has a unique role during infection in that its expression suppressed the intestinal microbiota and enhanced the colonization of a pathogen. IL-22 induced the expression of antimicrobial proteins, including lipocalin-2 and calprotectin, which sequester essential metal ions from microbes. Because Salmonella enterica ser. Typhimurium can overcome metal ion starvation mediated by lipocalin-2 and calprotectin via alternative pathways, IL-22 boosted its colonization of the inflamed intestine by suppressing commensal Enterobacteriaceae, which are susceptible to the antimicrobial proteins. Thus, IL-22 tipped the balance between pathogenic and commensal bacteria in favor of a pathogen. Taken together, IL-22 induction can be exploited by pathogens to suppress the growth of their closest competitors, thereby enhancing pathogen colonization of mucosal surfaces.
Collapse
Affiliation(s)
- Judith Behnsen
- Department of Microbiology, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| | - Stefan Jellbauer
- Department of Microbiology, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| | - Christina P Wong
- Department of Microbiology, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| | - Robert A Edwards
- Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA; Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael D George
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, CA 95616, USA
| | - Wenjun Ouyang
- Department of Immunology, Genentech, South San Francisco, CA 94080, USA
| | - Manuela Raffatellu
- Department of Microbiology, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
25
|
Abstract
Interleukin-22 (IL-22) is a key effector molecule that is produced by activated T cells, including T helper 22 (TH22) cells, TH17 cells and TH1 cells, as well as subsets of innate lymphoid cells. Although IL-22 can act synergistically with IL-17 or tumour necrosis factor, some important functions of IL-22 are unique to this cytokine. Data obtained over the past few years indicate that the IL-22-IL-22 receptor subunit 1 (IL-22R1) system has a high potential clinical relevance in psoriasis, ulcerative colitis, graft-versus-host disease, certain infections and tumours, as well as in liver and pancreas damage. This Review highlights current knowledge of the biology of the IL-22-IL-22R1 system, its role in inflammation, tissue protection, regeneration and antimicrobial defence, as well as the positive and potentially negative consequences of its therapeutic modulation.
Collapse
Affiliation(s)
- Robert Sabat
- 1] Interdisciplinary Group of Molecular Immunopathology, Institute of Medical Immunology, Department of Dermatology and Allergy, University Medicine Charité, Charitéplatz 1, D-10117 Berlin, Germany. [2] Research Center Immunosciences, University Hospital Charité, Hessische Strasse 3-4, D-10115 Berlin, Germany
| | - Wenjun Ouyang
- Department of Immunology, Genentech, 1 DNA Way, South San Francisco, California 94080, USA
| | - Kerstin Wolk
- 1] Interdisciplinary Group of Molecular Immunopathology, Institute of Medical Immunology, Department of Dermatology and Allergy, University Medicine Charité, Charitéplatz 1, D-10117 Berlin, Germany. [2] Research Center Immunosciences, University Hospital Charité, Hessische Strasse 3-4, D-10115 Berlin, Germany
| |
Collapse
|
26
|
Jia L, Wu C. The biology and functions of Th22 cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 841:209-30. [PMID: 25261209 DOI: 10.1007/978-94-017-9487-9_8] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
T helper (Th) cells develop from naïve CD4(+) T cells under lineage-specific culture conditions and are nominated by their lineage-specific cytokines. Th22 cells, new players in adoptive immune responses, are identified by the production of interleukin (IL)-22. Plenty of observations are obtained over the past few years indicating that IL-22 is produced by activated T cells including Th22 cells, Th17 cells, Th1 cells, innate lymphoid cells and some nonlymphocytes. IL-22 functions synergistically with IL-17 or tumor necrosis factor (TNF), however, it plays different roles by IL-22/IL-22 receptor signal transductions in pathologic processes, including inflammations, autoimmunity, tumor, and digestive organs damages. In this chapter, we focus on the biology of IL-22, the generation and regulation of Th22 cells, the possible signal pathways that involved in the functions of Th22 cells, as well as the relationship between Th22 cells and various diseases.
Collapse
Affiliation(s)
- Lei Jia
- Key Laboratory of Tropical Disease Control Research of Ministry of Education, Zhongshan School of Medicine, Institute of Immunology, Sun Yat-Sen University, 74th, Zhongshan 2nd Road, Guangzhou, 510080, China
| | | |
Collapse
|
27
|
Kimura A, Abe H, Tsuruta S, Chiba S, Fujii-Kuriyama Y, Sekiya T, Morita R, Yoshimura A. Aryl hydrocarbon receptor protects against bacterial infection by promoting macrophage survival and reactive oxygen species production. Int Immunol 2013; 26:209-20. [PMID: 24343818 DOI: 10.1093/intimm/dxt067] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) is crucial for various immune responses. The relationship between AhR and infection with the intracellular bacteria Listeria monocytogenes (LM) is poorly understood. Here, we show that in response to LM infection, AhR is required for bacterial clearance by promoting macrophage survival and reactive oxygen species (ROS) production. AhR-deficient mice were more susceptible to listeriosis, and AhR deficiency enhances bacterial growth in vivo and in vitro. On the other hand, pro-inflammatory cytokines were increased in AhR-deficient macrophages infected with LM despite enhanced susceptibility to LM infection in AhR-deficient mice. Subsequent studies demonstrate that AhR protects against macrophage cell death induced by LM infection through the induction of the antiapoptotic factor, the apoptosis inhibitor of macrophages, which promotes macrophage survival in the setting of LM infection. Furthermore, AhR promotes ROS production for bacterial clearance. Our results demonstrate that AhR is essential to the resistance against LM infection as it promotes macrophage survival and ROS production. This suggests that the activation of AhR by its ligands may be an effective strategy against listeriosis.
Collapse
Affiliation(s)
- Akihiro Kimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Frazer LC, Scurlock AM, Zurenski MA, Riley MM, Mintus M, Pociask DA, Sullivan JE, Andrews CW, Darville T. IL-23 induces IL-22 and IL-17 production in response to Chlamydia muridarum genital tract infection, but the absence of these cytokines does not influence disease pathogenesis. Am J Reprod Immunol 2013; 70:472-84. [PMID: 24238108 PMCID: PMC3852156 DOI: 10.1111/aji.12171] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 10/03/2013] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Chlamydia trachomatis infections are a significant cause of reproductive tract pathology. Protective and pathological immune mediators must be differentiated to design a safe and effective vaccine. METHODS Wild-type mice and mice deficient in IL-22 and IL-23 were infected intravaginally with Chlamydia muridarum, and their course of infection and oviduct pathology were compared. Local genital tract and draining lymph node immune responses were also examined in IL-23-deficient mice. RESULTS IL-22- and IL-23-deficient mice exhibited normal susceptibility to infection and oviduct pathology. IL-23 was required for the development of a Chlamydia-specific Th17 response in the lymph nodes and for production of IL-22 and IL-17 in the genital tract. However, influx of Th1 and innate immune cells was not compromised in the absence of IL-23. CONCLUSION IL-22 and IL-23 play either redundant or minimal roles in the pathogenesis of Chlamydia infection in the mouse model. Induction of Th17-associated cytokines by a Chlamydia vaccine should be avoided as these responses are not central to resolution of infection and have pathologic potential.
Collapse
Affiliation(s)
- Lauren C. Frazer
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15201
- Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Amy M. Scurlock
- Department of Pediatrics, University of Arkansas for Medical Sciences and Arkansas Children’s Hospital Research Institute, Little Rock, Arkansas 72202
| | - Matthew A. Zurenski
- Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Melissa M. Riley
- Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Margaret Mintus
- Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Derek A. Pociask
- Richard King Mellon Foundation Institute for Pediatric Research, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Jeanne E. Sullivan
- Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | | | - Toni Darville
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15201
- Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| |
Collapse
|
29
|
Stange J, Veldhoen M. The aryl hydrocarbon receptor in innate T cell immunity. Semin Immunopathol 2013; 35:645-55. [PMID: 24030775 DOI: 10.1007/s00281-013-0389-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 07/01/2013] [Indexed: 01/07/2023]
Abstract
Recent studies highlight an important role of the aryl hydrocarbon receptor (AhR) at mucosal barriers. Surprisingly, activation of the AhR, required for the maintenance of lymphocytes as well as lymphoid architecture, can be achieved via cues derived from the external environment. This environment contains both beneficial and harmful microorganisms as well as a diverse array of compounds, and the epithelia must offer very sophisticated levels of defence. This is achieved via multifaceted immune recognition diversity and cellular complexity. Mucosal associated tissues, particularly in the gastrointestinal tract, constitute a complex immune organ for local lymphocytes and contain highly organised lymphoid structures. We will discuss the recent observations concerning the AhR in relation to the function and maintenance of innate T cells, with focus on γδ T cells found enriched at epithelial barriers.
Collapse
Affiliation(s)
- Jörg Stange
- Laboratory for Lymphocyte Signalling and Development, The Babraham Institute, Cambridge, CB22 3AT, UK
| | | |
Collapse
|
30
|
Chung Y, Yamazaki T, Kim BS, Zhang Y, Reynolds JM, Martinez GJ, Chang SH, Lim H, Birkenbach M, Dong C. Epstein Barr virus-induced 3 (EBI3) together with IL-12 negatively regulates T helper 17-mediated immunity to Listeria monocytogenes infection. PLoS Pathog 2013; 9:e1003628. [PMID: 24068935 PMCID: PMC3777861 DOI: 10.1371/journal.ppat.1003628] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2013] [Accepted: 08/02/2013] [Indexed: 12/23/2022] Open
Abstract
Although the protective functions by T helper 17 (Th17) cytokines against extracellular bacterial and fungal infection have been well documented, their importance against intracellular bacterial infection remains unclear. Here, we investigated the contribution of Th17 responses to host defense against intracellular bacteria Listeria monocytogenes and found that Th17 cell generation was suppressed in this model. Unexpectedly, mice lacking both p35 and EBI3 cleared L. monocytogenes as efficiently as wild-type mice, whereas p35-deficient mice failed to do so. Furthermore, both innate cells and pathogen-specific T cells from double-deficient mice produced significantly higher IL-17 and IL-22 compared to wild-type mice. The bacterial burden in the liver of double-deficient mice treated with anti-IL-17 was significantly increased compared to those receiving a control Ab. Transfer of Th17 cells specific for listeriolysin O as well as administration of IL-17 and IL-22 significantly suppressed bacterial growth in p35-deficient mice, indicating the critical contribution of Th17 responses to host defense against the intracellular pathogen in the absence of IL-12 and proper Th1 responses. Our findings unveil a novel immune evasion mechanism whereby the intracellular bacteria exploit IL-27EBI3 to suppress Th17-mediated protective immunity.
Collapse
Affiliation(s)
- Yeonseok Chung
- Department of Immunology and Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, The University of Texas Medical School at Houston, Houston, Texas, United States of America
- The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Tomohide Yamazaki
- Department of Immunology and Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Byung-Seok Kim
- Department of Immunology and Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Yongliang Zhang
- Department of Immunology and Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Joseph M. Reynolds
- Department of Immunology and Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Gustavo J. Martinez
- Department of Immunology and Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Seon Hee Chang
- Department of Immunology and Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Hoyong Lim
- Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, The University of Texas Medical School at Houston, Houston, Texas, United States of America
| | - Mark Birkenbach
- Department of Medicine, Section of Rheumatology, Temple University, Philadelphia, Pennsylvania, United States of America
| | - Chen Dong
- Department of Immunology and Center for Inflammation and Cancer, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States of America
- The Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
31
|
Ivanov S, Renneson J, Fontaine J, Barthelemy A, Paget C, Fernandez EM, Blanc F, De Trez C, Van Maele L, Dumoutier L, Huerre MR, Eberl G, Si-Tahar M, Gosset P, Renauld JC, Sirard JC, Faveeuw C, Trottein F. Interleukin-22 reduces lung inflammation during influenza A virus infection and protects against secondary bacterial infection. J Virol 2013; 87:6911-24. [PMID: 23596287 PMCID: PMC3676141 DOI: 10.1128/jvi.02943-12] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 04/04/2013] [Indexed: 12/30/2022] Open
Abstract
Interleukin-22 (IL-22) has redundant, protective, or pathogenic functions during autoimmune, inflammatory, and infectious diseases. Here, we addressed the potential role of IL-22 in host defense and pathogenesis during lethal and sublethal respiratory H3N2 influenza A virus (IAV) infection. We show that IL-22, as well as factors associated with its production, are expressed in the lung tissue during the early phases of IAV infection. Our data indicate that retinoic acid receptor-related orphan receptor-γt (RORγt)-positive αβ and γδ T cells, as well as innate lymphoid cells, expressed enhanced Il22 transcripts as early as 2 days postinfection. During lethal or sublethal IAV infections, endogenous IL-22 played no role in the control of IAV replication and in the development of the IAV-specific CD8(+) T cell response. During lethal infection, where wild-type (WT) mice succumbed to severe pneumonia, the lack of IL-22 did not accelerate or delay IAV-associated pathogenesis and animal death. In stark contrast, during sublethal IAV infection, IL-22-deficient animals had enhanced lung injuries and showed a lower airway epithelial integrity relative to WT littermates. Of importance, the protective effect of endogenous IL-22 in pulmonary damages was associated with a more controlled secondary bacterial infection. Indeed, after challenge with Streptococcus pneumoniae, IAV-experienced Il22(-/-) animals were more susceptible than WT controls in terms of survival rate and bacterial burden in the lungs. Together, IL-22 plays no major role during lethal influenza but is beneficial during sublethal H3N2 IAV infection, where it limits lung inflammation and subsequent bacterial superinfections.
Collapse
Affiliation(s)
- Stoyan Ivanov
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Joelle Renneson
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Josette Fontaine
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Adeline Barthelemy
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Christophe Paget
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Elodie Macho Fernandez
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Fany Blanc
- Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U874, Paris, France
| | - Carl De Trez
- Vlaams Interuniversitair Instituut voor Biotechnologie, Vrije Universiteit Brussel, Brussels, Belgium
| | - Laurye Van Maele
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Laure Dumoutier
- Ludwig Institute for Cancer Research, Université Catholique de Louvain, Brussels
| | - Michel-René Huerre
- Institut Pasteur, Paris, France
- Unite de Recherche et d'Expertise Histotechnologie et Pathologie, Paris, France
| | - Gérard Eberl
- Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique, URA 1961, Paris, France
| | - Mustapha Si-Tahar
- Institut Pasteur, Paris, France
- Institut National de la Santé et de la Recherche Médicale, U874, Paris, France
| | - Pierre Gosset
- Hopital Saint Vincent, Groupe Hospitalier de l'Institut Catholique de Lille, Université Catholique de Lille, Lille, France
| | | | - Jean Claude Sirard
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - Christelle Faveeuw
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| | - François Trottein
- Institut Pasteur de Lille, Centre d'Infection et d'Immunité de Lille, Lille, France
- Université Lille Nord de France, Lille, France
- Centre National de la Recherche Scientifique, UMR 8204, Lille, France
- Institut National de la Santé et de la Recherche Médicale, U1019, Lille, France
- Institut Fédératif de Recherche 142, Lille, France
| |
Collapse
|
32
|
Poulsen KP, Faith NG, Steinberg H, Czuprynski CJ. Bacterial load and inflammation in fetal tissues is not dependent on IL-17a or IL-22 in 10-14 day pregnant mice infected with Listeria monocytogenes. Microb Pathog 2013; 56:47-52. [PMID: 23178254 PMCID: PMC3578105 DOI: 10.1016/j.micpath.2012.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 11/06/2012] [Accepted: 11/08/2012] [Indexed: 01/23/2023]
Abstract
In this study, we first assessed the effect of intragastric infection of pregnant mice with Listeria monocytogenes on relative expression of select genes associated with T cell subsets. Relative gene expression was moderately increased in placental tissues for IFNγ, IL-4, IL-17a, IL-22, CD3, and FoxP3. To assess the roles of IL-17a and IL-22 in resistance to listeriosis during pregnancy, we compared the severity of maternal and fetal infection in IL-17a((-/-)), IL-22((-/-)), and IL-17a((-/-))/IL-22((-/-)) mice with that of wild type C57BL/6 mice. Intragastric infection with modest numbers of bacterial cells (10(5) CFU) caused reproducible maternal and fetal infection in all four mouse strains. We recovered greater numbers of CFU from the bloodstream of pregnant IL-22((-/-)) mice than pregnant wild type mice. Otherwise we found no significant difference in bacterial load in maternal or fetal tissues (spleen, liver, fetoplacental units) from pregnant IL-17a((-/-)), IL-22((-/-)), or IL-17a((-/-))/IL-22((-/-)) or wild type mice. Nor did we observe histopathologic differences in severity of inflammation in maternal or fetal tissues from the various groups of mice. Although IL-17a and IL-22 are up-regulated in placental tissue, our study suggests that antibacterial resistance and the host inflammatory response are not dependent on IL-17a or IL-22 during infection of mice with L. monocytogenes at 10-14 days of gestation.
Collapse
Affiliation(s)
- Keith P. Poulsen
- Department of Clinical Sciences, College of Veterinary Medicine, Oregon State University, 700 SW 30 Street, Corvallis, Oregon, 97331, USA
| | - Nancy G. Faith
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
- Food Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Howard Steinberg
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Charles J. Czuprynski
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
- Food Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| |
Collapse
|
33
|
Pennino D, Bhavsar PK, Effner R, Avitabile S, Venn P, Quaranta M, Marzaioli V, Cifuentes L, Durham SR, Cavani A, Eyerich K, Chung KF, Schmidt-Weber CB, Eyerich S. IL-22 suppresses IFN-γ-mediated lung inflammation in asthmatic patients. J Allergy Clin Immunol 2013; 131:562-70. [PMID: 23174657 DOI: 10.1016/j.jaci.2012.09.036] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 09/16/2012] [Accepted: 09/27/2012] [Indexed: 11/25/2022]
Abstract
BACKGROUND IL-22 controls tissue homeostasis by both proinflammatory and anti-inflammatory effects. However, the anti-inflammatory mechanisms of IL-22 remain poorly investigated. OBJECTIVE We sought to investigate the anti-inflammatory role for IL-22 in human asthma. METHODS T-cell lines derived from lung biopsy specimens of asthmatic patients were characterized by means of flow cytometry. Human bronchial epithelial cells from healthy and asthmatic subjects were stimulated with IL-22, IFN-γ, or the combination of both cytokines. Effects of cytokine stimulation were investigated by using whole-genome analysis, ELISA, and flow cytometry. The functional consequence of cytokine stimulation was evaluated in an in vitro wound repair model and T cell-mediated cytotoxicity experiments. In vivo cytokine expression was measured by using immunohistochemistry and Luminex assays in bronchoalveolar lavage fluid of healthy and asthmatic patients. RESULTS The current study identifies a tissue-restricted antagonistic interplay of IL-22 and the proinflammatory cytokine IFN-γ. On the one hand, IFN-γ antagonized IL-22-mediated induction of the antimicrobial peptide S100A7 and epithelial cell migration in bronchial epithelial cells. On the other hand, IL-22 decreased epithelial susceptibility to T cell-mediated cytotoxicity by inhibiting the IFN-γ-induced expression of MHC-I, MHC-II, and CD54/intercellular adhesion molecule 1 molecules. Likewise, IL-22 inhibited IFN-γ-induced secretion of the proinflammatory chemokines CCL5/RANTES and CXCL10/interferon-inducible protein 10 in vitro. Consistently, the IL-22 expression in bronchoalveolar lavage fluid of asthmatic patients inversely correlated with the expression of CCL5/RANTES and CXCL10/interferon-inducible protein 10 in vivo. CONCLUSIONS IL-22 might control the extent of IFN-γ-mediated lung inflammation and therefore play a tissue-restricted regulatory role.
Collapse
Affiliation(s)
- Davide Pennino
- ZAUM-Center of Allergy and Environment, Technische Universität and Helmholtz Center Munich, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Listeria monocytogenes causes several clinical manifestations in humans and domestic animals. This bacterium is a saprophyte in soil and ensiled feeds, which are sources of infection for food producing animals (i.e. ruminants). The most common route of infection for people is via ingestion of contaminated ready-to-eat food products such as produce, soft cheeses and deli meats. In the United States, L. monocytogenes causes relatively few cases of clinical disease compared to other food-borne pathogens. However, clinical listeriosis is associated with high mortality, especially in immunocompromised patients, pregnant women, neonates, and the elderly. Listeria is an intracellular pathogen, which has been widely used in basic research to elucidate mechanisms of molecular pathogenesis and protective cell-mediated immunity. Despite the sizeable knowledge on L. monocytogenes pathogenesis, key points regarding listeriosis during pregnancy and the perinatal period remain unknown. This review summarizes listeriosis in humans and domestic animals during pregnancy, and animal models used to study the pathogenesis and immune response to L. monocytogenes infection during these periods.
Collapse
|
35
|
Sotolongo J, Ruiz J, Fukata M. The role of innate immunity in the host defense against intestinal bacterial pathogens. Curr Infect Dis Rep 2012; 14:15-23. [PMID: 22139594 DOI: 10.1007/s11908-011-0234-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Eradication of infectious disease is our global health challenge. After encountering intestinal infection with a bacterial pathogen, the host defense program is initiated by local antigen-presenting cells (APCs) that eliminate invading pathogens by phagocytosis and establish localized inflammation by secreting cytokines and chemokines. These pathogen-experienced APCs migrate to the mesenteric lymph nodes, where host immune responses are precisely orchestrated. Initiation and regulation of this defense program appear to be largely dependent on innate immunity which is antigen non-specific and provides a rapid defense against broader targets. On the other hand, many bacterial enteropathogens have evoked abilities to modify the host defense program to their advantage. Therefore, better understanding of the host-pathogen interactions is essential to establish effective eradication strategies for enteric infectious diseases. In this review, we will discuss the current understanding of innate immune regulation of the host defense mechanisms against intestinal infection by bacterial pathogens.
Collapse
Affiliation(s)
- John Sotolongo
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Post Office Box 016960 (D-149), Miami, FL, 33101, USA
| | | | | |
Collapse
|
36
|
Inflammatory monocyte recruitment is regulated by interleukin-23 during systemic bacterial infection. Infect Immun 2012; 80:4099-105. [PMID: 22966045 DOI: 10.1128/iai.00589-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Listeria monocytogenes is a gram-positive intracellular pathogen that causes meningitis and septicemia in immunocompromised individuals and spontaneous abortion in pregnant women. The innate immune response against L. monocytogenes is primarily mediated by neutrophils and monocytes. Interleukin-23 (IL-23) is an important proinflammatory cytokine well known for its role in neutrophil recruitment in various infectious and autoimmune diseases. We have previously shown that IL-23 is required for host resistance against L. monocytogenes and for neutrophil recruitment to the liver, but not the spleen, during infection. Despite efficient neutrophil recruitment to the spleen, IL-23p19 knockout (KO) mice have an increased bacterial burden in this organ, suggesting that IL-23 may regulate the recruitment/function of another cell type to the spleen. In this study, we show that specific depletion of neutrophils abrogated the differences in bacterial burdens in the livers but not the spleens of C57BL/6 (B6) and IL-23p19 KO mice. Interestingly, L. monocytogenes-infected IL-23p19 KO mice had fewer monocytes in the spleen than B6 mice, as well as a reduction in the monocyte-recruiting chemokines CCL2 and CCL7. Additionally, the overall concentrations of tumor necrosis factor alpha (TNF-α) and nitric oxide (NO(•)), as well as the percentages and total numbers of monocytes producing TNF-α and NO(•), were reduced in IL-23p19 KO mice compared to levels in B6 mice, leading to increased bacterial burdens in the spleens of L. monocytogenes-infected IL-23p19 KO mice. Collectively, our data establish that IL-23 is required for the optimal recruitment of TNF-α- and NO(•)-producing inflammatory monocytes, thus revealing a novel mechanism by which this proinflammatory cytokine provides protection against bacterial infection.
Collapse
|
37
|
Gessner MA, Werner JL, Lilly LM, Nelson MP, Metz AE, Dunaway CW, Chan YR, Ouyang W, Brown GD, Weaver CT, Steele C. Dectin-1-dependent interleukin-22 contributes to early innate lung defense against Aspergillus fumigatus. Infect Immun 2012; 80:410-7. [PMID: 22038916 PMCID: PMC3255669 DOI: 10.1128/iai.05939-11] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 10/15/2011] [Indexed: 12/30/2022] Open
Abstract
We have previously reported that mice deficient in the beta-glucan receptor Dectin-1 displayed increased susceptibility to Aspergillus fumigatus lung infection in the presence of lower interleukin 23 (IL-23) and IL-17A production in the lungs and have reported a role for IL-17A in lung defense. As IL-23 is also thought to control the production of IL-22, we examined the role of Dectin-1 in IL-22 production, as well as the role of IL-22 in innate host defense against A. fumigatus. Here, we show that Dectin-1-deficient mice demonstrated significantly reduced levels of IL-22 in the lungs early after A. fumigatus challenge. Culturing cells from enzymatic lung digests ex vivo further demonstrated Dectin-1-dependent IL-22 production. IL-22 production was additionally found to be independent of IL-1β, IL-6, or IL-18 but required IL-23. The addition of recombinant IL-23 augmented IL-22 production in wild-type (WT) lung cells and rescued IL-22 production by lung cells from Dectin-1-deficient mice. In vivo neutralization of IL-22 in the lungs of WT mice resulted in impaired A. fumigatus lung clearance. Moreover, mice deficient in IL-22 also demonstrated a higher lung fungal burden after A. fumigatus challenge in the presence of impaired IL-1α, tumor necrosis factor alpha (TNF-α), CCL3/MIP-1α, and CCL4/MIP-1β production and lower neutrophil recruitment, yet intact IL-17A production. We further show that lung lavage fluid collected from both A. fumigatus-challenged Dectin-1-deficient and IL-22-deficient mice had compromised anti-fungal activity against A. fumigatus in vitro. Although lipocalin 2 production was observed to be Dectin-1 and IL-22 dependent, lipocalin 2-deficient mice did not demonstrate impaired A. fumigatus clearance. Moreover, lung S100a8, S100a9, and Reg3g mRNA expression was not lower in either Dectin-1-deficient or IL-22-deficient mice. Collectively, our results indicate that early innate lung defense against A. fumigatus is mediated by Dectin-1-dependent IL-22 production.
Collapse
Affiliation(s)
- Melissa A. Gessner
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jessica L. Werner
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lauren M. Lilly
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael P. Nelson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Allison E. Metz
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chad W. Dunaway
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yvonne R. Chan
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wenjun Ouyang
- Department of Immunology, Genentech, Inc., South San Francisco, California, USA
| | - Gordon D. Brown
- Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Casey T. Weaver
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chad Steele
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|