151
|
Amezcua Vesely MC, Rodríguez C, Gruppi A, Acosta Rodríguez EV. Interleukin-17 mediated immunity during infections with Trypanosoma cruzi and other protozoans. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165706. [PMID: 31987839 PMCID: PMC7071987 DOI: 10.1016/j.bbadis.2020.165706] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/05/2019] [Accepted: 01/22/2020] [Indexed: 12/31/2022]
Abstract
Host resistance during infection with Trypanosoma cruzi, and other protozoans, is dependent on a balanced immune response. Robust immunity against these pathogens requires of the concerted action of many innate and adaptive cell populations including macrophages, neutrophils, dendritic cells, CD4+, and CD8+ T cells and B cells among others. Indeed, during most protozoan infections only a balanced production of inflammatory (TH1) and anti-inflammatory (TH2/regulatory) cytokines will allow the control of parasite spreading without compromising host tissue integrity. The description of TH17 cells, a novel effector helper T cell lineage that produced IL-17 as signature cytokine, prompted the revision of our knowledge about the mechanisms that mediate protection and immunopathology during protozoan infections. In this manuscript we discuss the general features of IL-17 mediated immune responses as well as the cellular sources, effector mechanisms and overall role of IL-17 in the immune response to T. cruzi and other protozoan infections.
Collapse
Affiliation(s)
- María Carolina Amezcua Vesely
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Constanza Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | - Eva Virginia Acosta Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI)-CONICET, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina.
| |
Collapse
|
152
|
Kadekar D, Agerholm R, Rizk J, Neubauer HA, Suske T, Maurer B, Viñals MT, Comelli EM, Taibi A, Moriggl R, Bekiaris V. The neonatal microenvironment programs innate γδ T cells through the transcription factor STAT5. J Clin Invest 2020; 130:2496-2508. [PMID: 32281944 PMCID: PMC7190909 DOI: 10.1172/jci131241] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 01/29/2020] [Indexed: 01/18/2023] Open
Abstract
IL-17-producing RORγt+ γδ T cells (γδT17 cells) are innate lymphocytes that participate in type 3 immune responses during infection and inflammation. Herein, we show that γδT17 cells rapidly proliferate within neonatal lymph nodes and gut, where, upon entry, they upregulate T-bet and coexpress IL-17, IL-22, and IFN-γ in a STAT3- and retinoic acid-dependent manner. Neonatal expansion was halted in mice conditionally deficient in STAT5, and its loss resulted in γδT17 cell depletion from all adult organs. Hyperactive STAT5 mutant mice showed that the STAT5A homolog had a dominant role over STAT5B in promoting γδT17 cell expansion and downregulating gut-associated T-bet. In contrast, STAT5B preferentially expanded IFN-γ-producing γδ populations, implying a previously unknown differential role of STAT5 gene products in lymphocyte lineage regulation. Importantly, mice lacking γδT17 cells as a result of STAT5 deficiency displayed a profound resistance to experimental autoimmune encephalomyelitis. Our data identify that the neonatal microenvironment in combination with STAT5 is critical for post-thymic γδT17 development and tissue-specific imprinting, which is essential for infection and autoimmunity.
Collapse
Affiliation(s)
- Darshana Kadekar
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Rasmus Agerholm
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - John Rizk
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Heidi A. Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tobias Suske
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara Maurer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Elena M. Comelli
- Department of Nutritional Sciences and
- Department of Nutritional Sciences and Joannah and Brian Lawson Centre for Child Nutrition, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Vasileios Bekiaris
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
153
|
Abstract
Innate lymphocyte populations are emerging as key effectors in tissue homeostasis, microbial defense, and inflammatory skin disease. The cells are evolutionarily ancient and carry conserved principles of function, which can be achieved through shared or unique specific mechanisms. Recent technological and treatment advances have provided insight into heterogeneity within and between individuals and species. Similar pathways can extend through to adaptive lymphocytes, which softens the margins with innate lymphocyte populations and allows investigation of nonredundant pathways of immunity and inflammation that might be amenable to therapeutic intervention. Here, we review advances in understanding of innate lymphocyte biology with a focus on skin disease and the roles of commensal and pathogen responses and tissue homeostasis.
Collapse
Affiliation(s)
- Yi-Ling Chen
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Headington, Oxford, OX3 9DS, United Kingdom
| | - Clare S Hardman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Headington, Oxford, OX3 9DS, United Kingdom
| | - Koshika Yadava
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Headington, Oxford, OX3 9DS, United Kingdom
| | - Graham Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, Headington, Oxford, OX3 9DS, United Kingdom
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals, Headington, Oxford OX3 7LE, United Kingdom;
| |
Collapse
|
154
|
Wei XS, Pei XB, Liu YL, Wu XZ, Shi HZ, Zhou Q. IL-17A-Producing γδT Cells Inhibit the Formation of Malignant Pleural Effusions. Am J Respir Cell Mol Biol 2020; 61:174-184. [PMID: 30608868 DOI: 10.1165/rcmb.2018-0201oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
γδT cells are an important source of IL-17A and play an anti- or protumor role depending on the surrounding microenvironment. The precise role of γδT cells in the development of malignant pleural effusions (MPE) remains unknown. Using flow cytometry, we analyzed the distribution and differentiation of γδT cells in wild-type (WT) and IL-10-∕- mice. We carefully elucidated the influence of γδT cells on the formation of MPE by depleting γδT cells from WT, IL-10-∕-, and IL-17a-∕- mice. The distribution of γδT17 cells in human MPE and peripheral blood was also determined. Our data showed that both γδT cells and IL-17A-producing γδT (γδT17) cells accumulated in murine MPE, and IL-10 deficiency enhanced their accumulation. γδT cells were the main source of IL-17A in MPE for both WT and IL-10-∕- mice. IL-10 inhibited the chemotactic response of γδT cells to MPE and the proliferation of these cells. IL-10 suppressed γδT cell secretion of IL-17A via RORγt. The ablation of γδT cells accelerated MPE accumulation in both WT and IL-10-∕- mice, but it did not influence MPE accumulation in IL-17a-∕- mice. Patients with higher frequencies of γδT17 cells had significantly longer survival times than patients with lower frequencies of γδT17 cells. Taken together, our data demonstrate that γδT17 cells play an inhibitory role in the progression of MPE, and the accumulation of γδT17 cells in MPE is suppressed by IL-10.
Collapse
Affiliation(s)
- Xiao-Shan Wei
- 1Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | | | - Ya-Lan Liu
- 2Center of Medical Research and.,3Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiu-Zhi Wu
- 2Center of Medical Research and.,3Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Huan-Zhong Shi
- 2Center of Medical Research and.,3Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qiong Zhou
- 1Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| |
Collapse
|
155
|
Dienz O, DeVault VL, Musial SC, Mistri SK, Mei L, Baraev A, Dragon JA, Krementsov D, Veillette A, Boyson JE. Critical Role for SLAM/SAP Signaling in the Thymic Developmental Programming of IL-17- and IFN-γ-Producing γδ T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1521-1534. [PMID: 32024701 PMCID: PMC7065973 DOI: 10.4049/jimmunol.1901082] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022]
Abstract
During thymic development, mouse γδ T cells commit to either an IFN-γ- or an IL-17-producing phenotype through mechanisms that remain unclear. In this study, we investigated the extent to which the SLAM/SAP signaling pathway regulates the functional programming of γδ T cells. Characterization of SLAM family receptor expression revealed that thymic γδ T cell subsets were each marked by distinct coexpression profiles of SLAMF1, SLAMF4, and SLAMF6. In the thymus, Vγ1 and Vγ4 T cells that exhibited an SLAMF1+SLAMF6+ double positive phenotype were largely contained within immature CD24+CD73- and CD24+CD73+ subsets, whereas SLAMF1 single positive, SLAMF6 single positive, or SLAMF1SLAMF6 double negative cells were found within mature CD24-CD73+ and CD24-CD73- subsets. In the periphery, SLAMF1 and SLAMF6 expression distinguished IL-17- and IFN-γ-producing γδ T cells, respectively. Disruption of SLAM family receptor signaling through deletion of SAP resulted in impaired thymic Vγ1 and Vγ4 T cell maturation at the CD24+CD73-SLAMF1+SLAMF6+ double positive stage that was associated with a decreased frequency of CD44+RORγt+ γδ T cells. Impaired development was in turn associated with decreased γδ T cell IL-17 and IFN-γ production in the thymus as well as in peripheral tissues. The role for SAP was subset-specific, as Vγ1Vδ6.3, Vγ4, Vγ5, but not Vγ6 subsets were SAP-dependent. Together, these data suggest that the SLAM/SAP signaling pathway plays a larger role in γδ T cell development than previously appreciated and represents a critical checkpoint in the functional programming of both IL-17- and IFN-γ-producing γδ T cell subsets.
Collapse
Affiliation(s)
- Oliver Dienz
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Victoria L DeVault
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Shawn C Musial
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Somen K Mistri
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Linda Mei
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Aleksandr Baraev
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT 05405
| | - Julie A Dragon
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT 05405
| | - Dimitry Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT 05405; and
| | - Andre Veillette
- Montreal Clinical Research Institute, Montreal, Quebec H2W 1R7, Canada
| | - Jonathan E Boyson
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, VT 05405;
| |
Collapse
|
156
|
Deng Z, Wang H, Chen Z, Wang T. Bibliometric Analysis of Dendritic Epidermal T Cell (DETC) Research From 1983 to 2019. Front Immunol 2020; 11:259. [PMID: 32226424 PMCID: PMC7080701 DOI: 10.3389/fimmu.2020.00259] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 01/31/2020] [Indexed: 12/12/2022] Open
Abstract
Dendritic epidermal T cells (DETC) are a group of immune cells expressing canonical γδ TCR in the murine epidermis. Similar to γδ T cells in the human epidermis, DETC serve an important barrier cell in the skin and participate in skin immune surveillance, immune regulation, skin homeostasis, tissue protection, and other activities. Since its discovery in 1983, research on DETC has grown rapidly and unevenly. To evaluate DETC research trends and map the DETC knowledge structure, we have applied bibliometric methods and techniques. A total of 384 DETC-related articles obtained from the Scopus database published between 1983 and 2019 were analyzed using indicators of publication and citation metrics, country and international cooperation, author and co-authorship, and keyword co-occurrence cluster. The present research status, the emerging global trends and the future development direction are also visualized and discussed. In summary, this study provides novel and useful data for the DETC research scientific community, and will help researchers explore DETC more intuitively and effectively.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongwei Wang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiye Chen
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
157
|
Polese B, Zhang H, Thurairajah B, King IL. Innate Lymphocytes in Psoriasis. Front Immunol 2020; 11:242. [PMID: 32153574 PMCID: PMC7047158 DOI: 10.3389/fimmu.2020.00242] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/29/2020] [Indexed: 12/12/2022] Open
Abstract
Skin is a fundamental component of our host defense system that provides a dynamic physical and chemical barrier against pathogen invasion and environmental insults. Cutaneous barrier function is mediated by complex interactions between structural cells such as keratinocytes and diverse lineages of immune cells. In contrast to the protective role of these intercellular interactions, uncontrolled immune activation can lead to keratinocyte dysfunction and psoriasis, a chronic inflammatory disease affecting 2% of the global population. Despite some differences between human and murine skin, animal models of psoriasiform inflammation have greatly informed clinical approaches to disease. These studies have helped to identify the interleukin (IL)-23-IL-17 axis as a central cytokine network that drives disease. In addition, they have led to the recent description of long-lived, skin-resident innate lymphocyte and lymphoid cells that accumulate in psoriatic lesions. Although not completely defined, these populations have both overlapping and unique functions compared to antigen-restricted αβ T lymphocytes, the latter of which are well-known to contribute to disease pathogenesis. In this review, we describe the diversity of innate lymphocytes and lymphoid cells found in mammalian skin with a special focus on αβ T cells, Natural Killer T cells and Innate Lymphoid cells. In addition, we discuss the effector functions of these unique leukocyte subsets and how each may contribute to different stages of psoriasis. A more complete understanding of these cell types that bridge the innate and adaptive immune system will hopefully lead to more targeted therapies that mitigate or prevent disease progression.
Collapse
Affiliation(s)
- Barbara Polese
- Meakins-Christie Laboratories, Department of Microbiology and Immunology, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Hualin Zhang
- Meakins-Christie Laboratories, Department of Microbiology and Immunology, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Bavanitha Thurairajah
- Meakins-Christie Laboratories, Department of Microbiology and Immunology, McGill University Health Centre Research Institute, Montreal, QC, Canada
| | - Irah L King
- Meakins-Christie Laboratories, Department of Microbiology and Immunology, McGill University Health Centre Research Institute, Montreal, QC, Canada.,Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre Research Institute, Montreal, QC, Canada
| |
Collapse
|
158
|
Spidale NA, Malhotra N, Frascoli M, Sylvia K, Miu B, Freeman C, Stadinski BD, Huseby E, Kang J. Neonatal-derived IL-17 producing dermal γδ T cells are required to prevent spontaneous atopic dermatitis. eLife 2020; 9:e51188. [PMID: 32065580 PMCID: PMC7025821 DOI: 10.7554/elife.51188] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 02/11/2020] [Indexed: 01/01/2023] Open
Abstract
Atopic Dermatitis (AD) is a T cell-mediated chronic skin disease and is associated with altered skin barrier integrity. Infants with mutations in genes involved in tissue barrier fitness are predisposed towards inflammatory diseases, but most do not develop or sustain the diseases, suggesting that there exist regulatory immune mechanisms to prevent aberrant inflammation. The absence of one single murine dermal cell type, the innate neonatal-derived IL-17 producing γδ T (Tγδ17) cells, from birth resulted in spontaneous, highly penetrant AD with many of the major hallmarks of human AD. In Tγδ17 cell-deficient mice, basal keratinocyte transcriptome was altered months in advance of AD induction. Tγδ17 cells respond to skin commensal bacteria and the fulminant disease in their absence was driven by skin commensal bacteria dysbiosis. AD in this model was characterized by highly expanded dermal αβ T clonotypes that produce the type three cytokines, IL-17 and IL-22. These results demonstrate that neonatal Tγδ17 cells are innate skin regulatory T cells that are critical for skin homeostasis, and that IL-17 has dual homeostatic and inflammatory function in the skin.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Autoantigens/genetics
- Cell Differentiation
- Dermatitis, Atopic/genetics
- Dermatitis, Atopic/immunology
- Dermatitis, Atopic/prevention & control
- Disease Models, Animal
- Gene Expression
- Interleukin-17/biosynthesis
- Interleukins/biosynthesis
- Keratinocytes/cytology
- Keratinocytes/metabolism
- Lymphocyte Activation
- Mice
- Mice, Knockout
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Skin/metabolism
- Skin/microbiology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Interleukin-22
Collapse
Affiliation(s)
- Nicholas A Spidale
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Nidhi Malhotra
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Michela Frascoli
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Katelyn Sylvia
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Bing Miu
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Coral Freeman
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Brian D Stadinski
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Eric Huseby
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| | - Joonsoo Kang
- Department of PathologyUniversity of Massachusetts Medical SchoolWorcesterUnited States
| |
Collapse
|
159
|
Cao Y, Guimaraes AO, Peck MC, Mayba O, Ruffin F, Hong K, Carrasco-Triguero M, Fowler VG, Maskarinec SA, Rosenberger CM. Risk stratification biomarkers for Staphylococcus aureus bacteraemia. Clin Transl Immunology 2020; 9:e1110. [PMID: 32082571 PMCID: PMC7018520 DOI: 10.1002/cti2.1110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/18/2019] [Accepted: 01/23/2020] [Indexed: 11/28/2022] Open
Abstract
Objectives To identify risk stratification biomarkers to enrich for the subset of Staphylococcus aureus bacteraemia patients who develop deep‐seated tissue infections with high morbidity and mortality to guide clinical trial enrolment and clinical management. Methods We evaluated the prognostic value of eight biomarkers for persistent bacteraemia, mortality and endovascular infection foci in a validation cohort of 160 patients with S. aureus bacteraemia enrolled consecutively over 3 years. Results High levels of IL‐17A, IL‐10 or soluble E‐selectin at bacteraemia diagnosis correlated with the duration of positive blood cultures. When thresholds defined in an independent cohort were applied, these biomarkers were robust predictors of persistent bacteraemia or endovascular infection. High serum levels of IL‐17A and IL‐10 often preceded the radiographic diagnosis of infective endocarditis, suggesting potential utility for prioritising diagnostic radiographic imaging. High IL‐8 was prognostic for all‐cause mortality, while IL‐17A and IL‐10 were superior to clinical metrics in discriminating between attributable mortality and non‐attributable mortality. High IL‐17A and IL‐10 identified more patients who developed microbiological failure or mortality than were identified by infective endocarditis diagnosis. Conclusion These biomarkers offer potential utility to identify patients at risk of persistent bacteraemia to guide diagnostic imaging and clinical management. Low biomarker levels could be used to rule out the need for more invasive TEE imaging in patients at lower risk of infective endocarditis. These biomarkers could enable clinical trials by enriching for patients with the greatest need for novel therapies.
Collapse
Affiliation(s)
- Yi Cao
- Bioinformatics and Computational Biology Genentech, Inc. South San Francisco CA USA
| | | | - Melicent C Peck
- Clinical Sciences Genentech, Inc. South San Francisco CA USA
| | - Oleg Mayba
- Bioinformatics and Computational Biology Genentech, Inc. South San Francisco CA USA
| | - Felicia Ruffin
- Division of Infectious Diseases Duke University Durham NC USA
| | - Kyu Hong
- BioAnalytical Sciences Genentech, Inc. South San Francisco CA USA.,BioAnalysis, Immune-Onc Therapeutics Palo Alto CA USA
| | | | - Vance G Fowler
- Division of Infectious Diseases Duke University Durham NC USA
| | | | | |
Collapse
|
160
|
Liu X, Mao Y, Kang Y, He L, Zhu B, Zhang W, Lu Y, Wu Q, Xu D, Shi L. MicroRNA-127 Promotes Anti-microbial Host Defense through Restricting A20-Mediated De-ubiquitination of STAT3. iScience 2020; 23:100763. [PMID: 31958753 PMCID: PMC6992901 DOI: 10.1016/j.isci.2019.100763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/29/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
The increasing rising of multiple drug-resistant Staphylococcus aureus has become a major public health concern, underscoring a pressing need for developing therapies essentially based on the understanding of host defensive mechanism. In the present study, we showed that microRNA (miR)-127 played a key role in controlling bacterial infection and conferred a profound protection against staphylococcal pneumonia. The protective effect of miR-127 was largely dependent on its regulation of macrophage bactericidal activity and the generation of IL-22, IL-17, and anti-microbial peptides (AMPs), the pathway primarily driven by STAT3. Importantly, we revealed that the ubiquitin-editing enzyme A20, a genuine target of miR-127, specifically interacted with and repressed K63-ubiquitination of STAT3, thereby compromising its phosphorylation upon bacterial infection. Thus, our data not only identify miR-127 as a non-coding molecule with anti-bacterial activity but also delineate an unappreciated mechanism whereby A20 regulates STAT3-driven anti-microbial signaling via modulating its ubiquitination.
Collapse
Affiliation(s)
- Xiaoyi Liu
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Mao
- Key Laboratory of Inflammation and Immunoregulation, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Yanhua Kang
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Inflammation and Immunoregulation, Hangzhou Normal University School of Medicine, Hangzhou, China
| | - Long He
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Zhu
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Zhang
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qinan Wu
- Collaborative Innovation Centers of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Hudson Institute of Medical Research, Department of Molecular and Translational Science, Monash University, Clayton, VIC 3800, Australia
| | - Liyun Shi
- Department of Immunology and Medical Microbiology, Nanjing University of Chinese Medicine, Nanjing, China; Key Laboratory of Inflammation and Immunoregulation, Hangzhou Normal University School of Medicine, Hangzhou, China.
| |
Collapse
|
161
|
Zhou J, Wang Y, Fan Q, Liu Y, Liu H, Yan J, Li M, Dong W, Li W. High levels of fucosylation and sialylation of milk N-glycans from mothers with gestational diabetes mellitus alter the offspring gut microbiome and immune balance in mice. FASEB J 2020; 34:3715-3731. [PMID: 31944389 DOI: 10.1096/fj.201901674r] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 12/09/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022]
Abstract
Gestational diabetes mellitus (GDM) is significantly associated with allergen sensitization in early childhood, and this may influence the gut microbiome and immune system of the children. In addition to mother-to-child transmission of microbes, milk glycans play a pivotal role in shaping the gut microbiome of infants. A previous study has demonstrated alterations in the major milk N-glycans of mothers with GDM. However, the impact of these changes on the gut microbiome and immune response of the neonates has yet to be studied. Here, we aimed to compare the glycosylation levels of various milk glycans between normal and GDM mice, and to characterize the intestinal microbiome and immune responses of the offspring after weaning. We found that GDM mouse milk contained significantly higher concentrations of fucosylated and sialylated N-glycans than control mice, but there was no difference in the concentration of milk oligosaccharides between the groups. The differences in milk N-glycans had direct effects on the intestinal microbiome of the offspring, which in turn affected their immune response upon challenge with ovalbumin (OVA), with disruptions in the Th1/Th2 and Th17/Treg cell balances. This study lays the foundation for further research and development of specific nutritional care for the offspring of GDM mothers.
Collapse
Affiliation(s)
- Jiaorui Zhou
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yue Wang
- Department of Biochemistry, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qingjie Fan
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - He Liu
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Jingyu Yan
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Ming Li
- Department of Microecology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Weijie Dong
- Department of Biochemistry, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Wenzhe Li
- Department of Biochemistry, College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|
162
|
David BA, Kubes P. Exploring the complex role of chemokines and chemoattractants in vivo on leukocyte dynamics. Immunol Rev 2020; 289:9-30. [PMID: 30977202 DOI: 10.1111/imr.12757] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/05/2019] [Accepted: 02/07/2019] [Indexed: 12/14/2022]
Abstract
Chemotaxis is fundamental for leukocyte migration in immunity and inflammation and contributes to the pathogenesis of many human diseases. Although chemokines and various other chemoattractants were initially appreciated as important mediators of acute inflammation, in the past years they have emerged as critical mediators of cell migration during immune surveillance, organ development, and cancer progression. Such advances in our knowledge in chemokine biology have paved the way for the development of specific pharmacological targets with great therapeutic potential. Chemoattractants may belong to different classes, including a complex chemokine system of approximately 50 endogenous molecules that bind to G protein-coupled receptors, which are expressed by a wide variety of cell types. Also, an unknown number of other chemoattractants may be generated by pathogens and damaged/dead cells. Therefore, blocking chemotaxis without causing side effects is an extremely challenging task. In this review, we focus on recent advances in understanding how the chemokine system orchestrates immune cell migration and positioning at the whole organ level in homeostasis, inflammation, and infection.
Collapse
Affiliation(s)
- Bruna A David
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Paul Kubes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.,Department of Microbiology and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
163
|
Su Q, Bouteau A, Cardenas J, Uthra B, Wang Y, Smitherman C, Gu J, Igyártó BZ. Brief communication: Long-term absence of Langerhans cells alters the gene expression profile of keratinocytes and dendritic epidermal T cells. PLoS One 2020; 15:e0223397. [PMID: 31923202 PMCID: PMC6953782 DOI: 10.1371/journal.pone.0223397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/17/2019] [Indexed: 11/18/2022] Open
Abstract
Tissue-resident and infiltrating immune cells are continuously exposed to molecules derived from the local cells that often come in form of secreted factors, such as cytokines. These factors are known to impact the immune cells’ biology. However, very little is known about whether the tissue resident immune cells in return also affect the local environment. In this study, with the help of RNA-sequencing, we show for the first time that long-term absence of epidermal resident Langerhans cells led to significant gene expression changes in the local keratinocytes and resident dendritic epidermal T cells. Thus, immune cells might play an active role in maintaining tissue homeostasis, which should be taken in consideration at data interpretation.
Collapse
Affiliation(s)
- Qingtai Su
- Baylor Scott & White Research Institute, Baylor Institute for Immunology Research, Dallas, Texas, United States of America
| | - Aurélie Bouteau
- Baylor Scott & White Research Institute, Baylor Institute for Immunology Research, Dallas, Texas, United States of America
- Baylor University, Institute of Biomedical Studies, Waco, Texas, United States of America
| | - Jacob Cardenas
- Baylor Scott & White Research Institute, Dallas, Texas, United States of America
| | - Balaji Uthra
- Baylor Scott & White Research Institute, Dallas, Texas, United States of America
| | - Yuanyaun Wang
- Baylor Scott & White Research Institute, Dallas, Texas, United States of America
| | - Cynthia Smitherman
- Baylor Scott & White Research Institute, Dallas, Texas, United States of America
| | - Jinghua Gu
- Baylor Scott & White Research Institute, Dallas, Texas, United States of America
| | - Botond Z. Igyártó
- Baylor Scott & White Research Institute, Baylor Institute for Immunology Research, Dallas, Texas, United States of America
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
164
|
Archer NK, Wang Y, Ortines RV, Liu H, Nolan SJ, Liu Q, Alphonse MP, Dikeman DA, Mazhar M, Miller RJ, Anderson LS, Francis KP, Simon SI, Miller LS. Preclinical Models and Methodologies for Monitoring Staphylococcus aureus Infections Using Noninvasive Optical Imaging. Methods Mol Biol 2020; 2069:197-228. [PMID: 31523776 PMCID: PMC7745539 DOI: 10.1007/978-1-4939-9849-4_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In vivo whole-animal optical (bioluminescence and fluorescence) imaging of Staphylococcus aureus infections has provided the opportunity to noninvasively and longitudinally monitor the dynamics of the bacterial burden and ensuing host immune responses in live anesthetized animals. Herein, we describe several different mouse models of S. aureus skin infection, skin inflammation, incisional/excisional wound infections, as well as mouse and rabbit models of orthopedic implant infection, which utilized this imaging technology. These animal models and imaging methodologies provide insights into the pathogenesis of these infections and innate and adaptive immune responses, as well as the preclinical evaluation of diagnostic and treatment modalities. Noninvasive approaches to investigate host-pathogen interactions are extremely important as virulent community-acquired methicillin-resistant S. aureus strains (CA-MRSA) are spreading through the normal human population, becoming more antibiotic resistant and creating a serious threat to public health.
Collapse
Affiliation(s)
- Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sabrina J Nolan
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dustin A Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Momina Mazhar
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leif S Anderson
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | | | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
165
|
Nur S, Sparber F, Lemberg C, Guiducci E, Schweizer TA, Zwicky P, Becher B, LeibundGut-Landmann S. IL-23 supports host defense against systemic Candida albicans infection by ensuring myeloid cell survival. PLoS Pathog 2019; 15:e1008115. [PMID: 31887131 PMCID: PMC6957211 DOI: 10.1371/journal.ppat.1008115] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 01/13/2020] [Accepted: 10/01/2019] [Indexed: 01/21/2023] Open
Abstract
The opportunistic fungal pathogen Candida albicans can cause invasive infections in susceptible hosts and the innate immune system, in particular myeloid cell-mediated immunity, is critical for rapid immune protection and host survival during systemic candidiasis. Using a mouse model of the human disease, we identified a novel role of IL-23 in antifungal defense. IL-23-deficient mice are highly susceptible to systemic infection with C. albicans. We found that this results from a drastic reduction in all subsets of myeloid cells in the infected kidney, which in turn leads to rapid fungal overgrowth and renal tissue injury. The loss in myeloid cells is not due to a defect in emergency myelopoiesis or the recruitment of newly generated cells to the site of infection but, rather, is a consequence of impaired survival of myeloid cells at the site of infection. In fact, the absence of a functional IL-23 pathway causes massive myeloid cell apoptosis upon C. albicans infection. Importantly, IL-23 protects myeloid cells from apoptosis independently of the IL-23-IL-17 immune axis and independently of lymphocytes and innate lymphoid cells. Instead, our results suggest that IL-23 acts in a partially autocrine but not cell-intrinsic manner within the myeloid compartment to promote host protection from systemic candidiasis. Collectively, our data highlight an unprecedented and non-canonical role of IL-23 in securing survival of myeloid cells, which is key for maintaining sufficient numbers of cells at the site of infection to ensure efficient host protection. Linked to advances in medical technology and the resulting increase in the number of intensive care patients, nosocomial infections with Candida albicans are on the rise. In patients suffering from invasive candidiasis the innate immune response is typically severely impaired. Strengthening the innate immune system has become a promising approach complementing the use of antifungal drugs. Our findings identify an unexpected and IL-17-independent role of IL-23 that prevents rapid death of myeloid cells during systemic candidiasis and thereby promotes optimal protection from disease. As such, IL-23 represents an important new piece in the puzzle of the finely tuned network of cytokines that regulates the innate immune response to fungal infection. Our results contribute to a better understanding of myeloid cell regulation during infection and thereby open new perspectives for future immunotherapeutic applications that may improve patient outcome.
Collapse
Affiliation(s)
- Selim Nur
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Florian Sparber
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Christina Lemberg
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Eva Guiducci
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Tiziano A. Schweizer
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | | |
Collapse
|
166
|
Zabotti A, Goletti D, Lubrano E, Cantini F. The impact of the interleukin 12/23 inhibitor ustekinumab on the risk of infections in patients with psoriatic arthritis. Expert Opin Drug Saf 2019; 19:69-82. [PMID: 31847608 DOI: 10.1080/14740338.2020.1703946] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Psoriatic arthritis (PsA) is characterized by chronic inflammation mediated by pro-inflammatory cytokines, with clinical features resulting from dysfunctional integrated signaling pathways affecting different constituents of the immune system. Increased understanding of the processes responsible for enthesitis, synovial inflammation, joint erosion, and new bone formation during PsA has led to development of biologic therapies targeting these cytokines. There is an increased risk of opportunistic infections in patients with PsA, and this risk is increased further with targeted biologic therapy.Areas covered: This paper reviews the role of the interleukin (IL)-12, IL-23 and IL-17 axis in the pathogenesis of PsA. The data suggest that ustekinumab is associated with a low risk of infections in patients with PsA, including tuberculosis or hepatitis reactivation. No live vaccines can be safely administered; ustekinumab is contraindicated/cannot be given with live vaccines. However, long-term treatment with ustekinumab does not impair the immune response to these vaccines when administered after an appropriate interval.Expert opinion: Ustekinumab is associated with a low risk of serious and opportunistic infections. More research is needed to confirm these findings specifically in patients with PsA, and comparative studies are needed to investigate the relative risk of infection with different biologics.
Collapse
Affiliation(s)
- Alen Zabotti
- Department of Medical and Biological Sciences, Rheumatology Clinic, University of Udine, Udine, Italy
| | - Delia Goletti
- Translational Research Unit, Epidemiology and Preclinical Research Department, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Ennio Lubrano
- Dipartimento di Medicina e Scienze della Salute "Vincenzo Tiberio", Università degli Studi del Molise, Campobasso, Italy
| | | |
Collapse
|
167
|
Miller RJ, Crosby HA, Schilcher K, Wang Y, Ortines RV, Mazhar M, Dikeman DA, Pinsker BL, Brown ID, Joyce DP, Zhang J, Archer NK, Liu H, Alphonse MP, Czupryna J, Anderson WR, Bernthal NM, Fortuno-Miranda L, Bulte JWM, Francis KP, Horswill AR, Miller LS. Development of a Staphylococcus aureus reporter strain with click beetle red luciferase for enhanced in vivo imaging of experimental bacteremia and mixed infections. Sci Rep 2019; 9:16663. [PMID: 31723175 PMCID: PMC6853927 DOI: 10.1038/s41598-019-52982-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/26/2019] [Indexed: 02/03/2023] Open
Abstract
In vivo bioluminescence imaging has been used to monitor Staphylococcus aureus infections in preclinical models by employing bacterial reporter strains possessing a modified lux operon from Photorhabdus luminescens. However, the relatively short emission wavelength of lux (peak 490 nm) has limited tissue penetration. To overcome this limitation, the gene for the click beetle (Pyrophorus plagiophtalamus) red luciferase (luc) (with a longer >600 emission wavelength), was introduced singly and in combination with the lux operon into a methicillin-resistant S. aureus strain. After administration of the substrate D-luciferin, the luc bioluminescent signal was substantially greater than the lux signal in vitro. The luc signal had enhanced tissue penetration and improved anatomical co-registration with infected internal organs compared with the lux signal in a mouse model of S. aureus bacteremia with a sensitivity of approximately 3 × 104 CFU from the kidneys. Finally, in an in vivo mixed bacterial wound infection mouse model, S. aureus luc signals could be spectrally unmixed from Pseudomonas aeruginosa lux signals to noninvasively monitor the bacterial burden of both strains. Therefore, the S. aureus luc reporter may provide a technological advance for monitoring invasive organ dissemination during S. aureus bacteremia and for studying bacterial dynamics during mixed infections.
Collapse
Affiliation(s)
- Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Heidi A Crosby
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Katrin Schilcher
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Momina Mazhar
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dustin A Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Bret L Pinsker
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Isabelle D Brown
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel P Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey Zhang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | - Nicholas M Bernthal
- Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Lea Fortuno-Miranda
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Jeff W M Bulte
- Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Department of Chemical & Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, Maryland, 21205, USA.,Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Kevin P Francis
- PerkinElmer, Hopkinton, Massachusetts, USA.,Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, Santa Monica, California, USA
| | - Alexander R Horswill
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, 80045, USA.,Denver VA Healthcare System, Denver, Colorado, USA
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA. .,Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA. .,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA. .,Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, 21218, USA.
| |
Collapse
|
168
|
Livingston ET, Mursalin MH, Callegan MC. A Pyrrhic Victory: The PMN Response to Ocular Bacterial Infections. Microorganisms 2019; 7:E537. [PMID: 31703354 PMCID: PMC6920826 DOI: 10.3390/microorganisms7110537] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/03/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Some tissues of the eye are susceptible to damage due to their exposure to the outside environment and inability to regenerate. Immune privilege, although beneficial to the eye in terms of homeostasis and protection, can be harmful when breached or when an aberrant response occurs in the face of challenge. In this review, we highlight the role of the PMN (polymorphonuclear leukocyte) in different bacterial ocular infections that invade the immune privileged eye at the anterior and posterior segments: keratitis, conjunctivitis, uveitis, and endophthalmitis. Interestingly, the PMN response from the host seems to be necessary for pathogen clearance in ocular disease, but the inflammatory response can also be detrimental to vision retention. This "Pyrrhic Victory" scenario is explored in each type of ocular infection, with details on PMN recruitment and response at the site of ocular infection. In addition, we emphasize the differences in PMN responses between each ocular disease and its most common corresponding bacterial pathogen. The in vitro and animal models used to identify PMN responses, such as recruitment, phagocytosis, degranulation, and NETosis, are also outlined in each ocular infection. This detailed study of the ocular acute immune response to infection could provide novel therapeutic strategies for blinding diseases, provide more general information on ocular PMN responses, and reveal areas of bacterial ocular infection research that lack PMN response studies.
Collapse
Affiliation(s)
- Erin T. Livingston
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (E.T.L.); (M.H.M.)
| | - Md Huzzatul Mursalin
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (E.T.L.); (M.H.M.)
| | - Michelle C. Callegan
- Department of Microbiology and Immunology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (E.T.L.); (M.H.M.)
- Department of Ophthalmology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Oklahoma Center for Neuroscience, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, Oklahoma City, OK 73104, USA
| |
Collapse
|
169
|
Cunha P, Vern YL, Gitton C, Germon P, Foucras G, Rainard P. Expansion, isolation and first characterization of bovine Th17 lymphocytes. Sci Rep 2019; 9:16115. [PMID: 31695097 PMCID: PMC6834651 DOI: 10.1038/s41598-019-52562-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022] Open
Abstract
Interleukin 17A-producing T helper cells (Th17) are CD4+ T cells that are crucial to immunity to extracellular bacteria. The roles of these cells in the bovine species are poorly defined, because the characterization of bovine Th17 cells lags behind for want of straightforward cultivation and isolation procedures. We have developed procedures to differentiate, expand, and isolate bovine Th17 cells from circulating CD4+ T cells of adult cows. Using polyclonal stimulation with antibodies to CD3 and CD28, we expanded IL-17A-positive CD4+ T cells in a serum-free cell culture medium supplemented with TGF-β1, IL-6 and IL-2. Populations of CD4+ T cells producing IL-17A or IFN-γ or both cytokines were obtained. Isolation of IL-17A-secreting CD4+ T cells was performed by labelling surface IL-17A, followed by flow cytometry cell sorting. The sorted Th17 cells were restimulated and could be expanded for several weeks. These cells were further characterized by cytokine profiling at transcriptomic and protein levels. They produced high amounts of IL-17A and IL-17F, and moderate amounts of IL-22 and IFN-γ. The techniques developed will be useful to characterize the phenotypic and functional properties of bovine Th17 cells.
Collapse
Affiliation(s)
- Patricia Cunha
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France
| | - Yves Le Vern
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France
| | | | - Pierre Germon
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France
| | - Gilles Foucras
- IHAP, INRA, ENVT, Université de Toulouse, Toulouse, France
| | - Pascal Rainard
- ISP, INRA, Université de Tours, UMR1282, Nouzilly, France.
| |
Collapse
|
170
|
Agerholm R, Rizk J, Viñals MT, Bekiaris V. STAT3 but not STAT4 is critical for γδT17 cell responses and skin inflammation. EMBO Rep 2019; 20:e48647. [PMID: 31549795 PMCID: PMC6832010 DOI: 10.15252/embr.201948647] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/23/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022] Open
Abstract
The transcription factors STAT3 and STAT4 are essential for lymphocyte differentiation and function. Interleukin (IL)-17 producing γδ T (γδT17) cells are innate lymphocytes important for anti-bacterial and inflammatory responses at barrier surfaces. Herein, we examine the role of STAT3 and STAT4 in regulating the homeostasis, activation, and pathogenicity of γδT17 cells. We show that STAT3 sustains γδT17 numbers in the skin but not in the lymph nodes, while STAT4 deficiency does not affect their homeostasis. Similarly, STAT3 but not STAT4 is essential for IL-23-induced IL-22 production by γδT17 cells. Concomitantly, mice lacking STAT3 expression in γδT17 cells develop significantly reduced psoriasis-like inflammation. STAT3-deficient γδT17 cells fail to expand and to upregulate IL-17A, IL-17F, and IL-22 in response to psoriatic stimuli. Although STAT4-deficient animals develop psoriasis-like disease, γδT17 cells in these mice are defective in IL-17F production. Collectively, our data demonstrate for the first time a critical role for STAT3 in orchestrating the homeostasis and pathogenicity of γδT17 cells and provide evidence for the requirement of STAT4 for optimal cytokine responses during inflammation.
Collapse
Affiliation(s)
- Rasmus Agerholm
- Department of Health TechnologyTechnical University of DenmarkKgs LyngbyDenmark
| | - John Rizk
- Department of Health TechnologyTechnical University of DenmarkKgs LyngbyDenmark
| | | | - Vasileios Bekiaris
- Department of Health TechnologyTechnical University of DenmarkKgs LyngbyDenmark
| |
Collapse
|
171
|
Ferraro A, Buonocore SM, Auquier P, Nicolas I, Wallemacq H, Boutriau D, van der Most RG. Role and plasticity of Th1 and Th17 responses in immunity to Staphylococcus aureus. Hum Vaccin Immunother 2019; 15:2980-2992. [PMID: 31149870 PMCID: PMC6930085 DOI: 10.1080/21645515.2019.1613126] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The human commensal Staphylococcus aureus (SA) is a leading cause of skin/soft tissue and surgical-site infections, and bacteremia. Functional antibodies and T-cell-mediated immunity, particularly Th1/Th17 responses, are thought to mediate protection. Vaccine development may be hindered by modulation of vaccine-induced T cells by pathogen-activated immunoregulatory responses, e.g., via IL-10.We screened SA proteins for CD4+ T-cell-activating and IL-10/IL-17-inducing capacities using healthy donor-derived PBMCs. Responses were characterized (Th1/Th17/Th22/immunosuppressive IL-10-producing cells) using intracellular cytokine staining and flow cytometry. Phenotypic plasticity of Th1/Th17 cells was evaluated under pro- or anti-inflammatory conditions using modulatory cytokines. The impact of vaccination on SA-specific memory responses was assessed using samples from a clinical trial evaluating AS03-adjuvanted and non-adjuvanted multicomponent (CPS5/CPS8/α-toxin/ClfA) vaccines (NCT01160172).The donors exhibited SA-specific memory T-cell responses, indicative of pre-existing immunity to SA. We identified effective activators of Th1 responses (EbhA/IsaA/SdrE/MntC/Aaa/α-toxin), and Th17 and Th1/Th17 responses (EbhA/IsaA/SdrE and, to a lesser extent, α-toxin), but not of Th22 responses or IL-10 production. MRPII, IsdA, and ClfA were inefficient CD4+ T-cell activators in our assays. IL-10, likely produced by innate immune cells, influenced mainly Th1 cells by suppressing IFN-γ production. The memory CD4+ T-cells observed after long-term stimulation with α-toxin and ClfA indicated that vaccination with these proteins had induced expansion of pre-existing Th1 but not Th17 responses, without apparent adjuvant effect, confirming the trial data. The Th1/Th17-driving proteins (EbhA/IsaA/SdrE) shared low IL-10-promoting abilities and restricted phenotypic plasticity under pro- and anti-inflammatory conditions.Given the complex immunopathology and multiple virulence factors, identification of Th1/Th17-driving antigens, adjuvants and administration routes, and delineation of the role of memory responses, may advance vaccine development.
Collapse
|
172
|
Interleukin 17 (IL-17) manipulates mouse bone marrow- derived neutrophils in response to acute lung inflammation. Comp Immunol Microbiol Infect Dis 2019; 67:101356. [PMID: 31634721 DOI: 10.1016/j.cimid.2019.101356] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 09/07/2019] [Accepted: 09/23/2019] [Indexed: 12/30/2022]
Abstract
Interleukin 17 (IL-17) mediates neutrophil migration to the lungs during acute inflammation, potentially leading to lung tissue damage. In the present study, we evaluated whether IL-17 could facilitate certain neutrophil functions in a mouse model. Mice were divided into four groups and intranasally challenged with PBS (1 = Control), Influenza A (H1N1) and Klebsiella pneumoniae (2 = Mix), Influenza A alone (3 = Flu), or K. pneumoniae (4 = KP) alone. Bone marrow, BAL cells, and lung specimens were collected seven days post-challenge for analysis. Mice in the Flu group showed the highest mortality rate. Neutrophils were the prominent cell type in BAL from Mix and KP, whereas lymphocytes were most numerous in Flu. Lesions in the lungs revealed considerably damage in the Mix, Flu, and KP groups. Isolated bone marrow-derived neutrophils were in vitro primed with mouse recombinant IL-17A protein (rIL-17A) followed by various functional assays. The reactive oxygen species (ROS) levels in rIL-17A primed cells showed significant elevations in all groups. Phagocytosis and bacterial destruction showed no significant difference between (+) or (-) rIL-17A groups. The formation of neutrophil extracellular traps (NETs) in rIL-17A-primed neutrophils showed elevated NET production. We next monitored expressions of genes in neutrophils. IL-17A mRNA expression was significantly increased in Mix and Flu; IL-1β mRNA only significantly increased in Flu, and IL-17RA showed constitutive expressions in all groups. In summary, neutrophils may cause tissue damage during lung inflammation through specific functions influenced by IL-17.
Collapse
|
173
|
Ahmadi K, Pouladfar G, Kalani M, Faezi S, Pourmand MR, Hasanzadeh S, Mafakher L, Aslani MM, Mahdavi M. Epitope-based immunoinformatics study of a novel Hla-MntC-SACOL0723 fusion protein from Staphylococcus aureus: Induction of multi-pattern immune responses. Mol Immunol 2019; 114:88-99. [DOI: 10.1016/j.molimm.2019.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/13/2019] [Accepted: 05/28/2019] [Indexed: 01/10/2023]
|
174
|
Zhang H, Carnevale G, Polese B, Simard M, Thurairajah B, Khan N, Gentile ME, Fontes G, Vinh DC, Pouliot R, King IL. CD109 Restrains Activation of Cutaneous IL-17-Producing γδ T Cells by Commensal Microbiota. Cell Rep 2019; 29:391-405.e5. [DOI: 10.1016/j.celrep.2019.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/26/2019] [Accepted: 08/30/2019] [Indexed: 01/01/2023] Open
|
175
|
Interleukin-10 (IL-10) Produced by Mutant Toxic Shock Syndrome Toxin 1 Vaccine-Induced Memory T Cells Downregulates IL-17 Production and Abrogates the Protective Effect against Staphylococcus aureus Infection. Infect Immun 2019; 87:IAI.00494-19. [PMID: 31358568 DOI: 10.1128/iai.00494-19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 07/23/2019] [Indexed: 11/20/2022] Open
Abstract
Development of long-term memory is crucial for vaccine-induced adaptive immunity against infectious diseases such as Staphylococcus aureus infection. Toxic shock syndrome toxin 1 (TSST-1), one of the superantigens produced by S. aureus, is a possible vaccine candidate against infectious diseases caused by this pathogen. We previously reported that vaccination with less toxic mutant TSST-1 (mTSST-1) induced T helper 17 (Th17) cells and elicited interleukin-17A (IL-17A)-mediated protection against S. aureus infection 1 week after vaccination. In the present study, we investigated the host immune response induced by mTSST-1 vaccination in the memory phase, 12 weeks after the final vaccination. The protective effect and IL-17A production after vaccination with mTSST-1 were eliminated because of IL-10 production. In the presence of IL-10-neutralizing monoclonal antibody (mAb), IL-17A production was restored in culture supernatants of CD4+ T cells and macrophages sorted from the spleens of vaccinated mice. Vaccinated mice treated with anti-IL-10 mAb were protected against systemic S. aureus infection in the memory phase. From these results, it was suggested that IL-10 produced in the memory phase suppresses the IL-17A-dependent vaccine effect through downregulation of IL-17A production.
Collapse
|
176
|
Liu C, Ouyang W, Xia J, Sun X, Zhao L, Xu F. Tumor Necrosis Factor-α Is Required for Mast Cell-Mediated Host Immunity Against Cutaneous Staphylococcus aureus Infection. J Infect Dis 2019; 218:64-74. [PMID: 29741644 DOI: 10.1093/infdis/jiy149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 04/17/2018] [Indexed: 01/09/2023] Open
Abstract
Background Mast cells (MCs) play a key role in immune process response to invading pathogens. Methods This study assessed the involvement of MCs in controlling Staphylococcus aureus infection in a cutaneous infection model of MC-deficient (KitW-sh/W-sh) mice. Results KitW-sh/W-sh mice developed significantly larger skin lesions after the cutaneous S. aureus challenge, when compared to wild-type (WT) mice, while MC dysfunction reduced the inflammation response to S. aureus. The levels of tumor necrosis factor (TNF)-α in skin tissues were significantly decreased in KitW-sh/W-sh mice upon infection. Moreover, the exogenous administration of MCs or recombinant TNF-α effectively restored the immune response against S. aureus in KitW-sh/W-sh mice via the recruitment of neutrophils to the infected site. These results indicate that the effects of MC deficiency are largely attributed to the decrease in production of TNF-α in cutaneous S. aureus infection. In addition, S. aureus-induced MC activation was dependent on the c-kit receptor-activated phosphoinositide 3-kinase (PI3K)/AKT/P65-nuclear factor (NF-κB) pathway, which was confirmed by treatment with Masitinib (a c-kit receptor inhibitor), Wortmannin (a PI3K inhibitor), and pyrrolidine dithiocarbamate (a NF-κB inhibitor), respectively. Conclusions The present study identifies the critical role of MCs in the host defense against S. aureus infection.
Collapse
Affiliation(s)
- Chao Liu
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Ouyang
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingyan Xia
- Department of Radiation Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoru Sun
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Liying Zhao
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Xu
- Department of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
177
|
Broad and Effective Protection against Staphylococcus aureus Is Elicited by a Multivalent Vaccine Formulated with Novel Antigens. mSphere 2019; 4:4/5/e00362-19. [PMID: 31484738 PMCID: PMC6731528 DOI: 10.1128/msphere.00362-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The demand for a prophylactic vaccine against methicillin-resistant Staphylococcus aureus (MRSA) has motivated numerous dedicated research groups to design and develop such a vaccine. In this study, we have developed a multivalent vaccine, Sta-V5, composed of five conserved antigens involved in three important virulence mechanisms. This prototype vaccine conferred up to 100% protection against multiple epidemiologically relevant S. aureus isolates in five different murine disease models. The vaccine not only elicits functional antibodies that mediate opsonophagocytic killing of S. aureus but also mounts robust antigen-specific T-cell responses. In addition, our data implied that γδ T cells contribute to the protection induced by Sta-V5 in a murine skin infection model.IMPORTANCE Staphylococcus aureus infections, especially MRSA infections, are becoming a major global health issue and are resulting in mortality rates that are increasing every year. However, an effective vaccine is lacking due to the complexity of the infection process of S. aureus In this study, we found that the addition of two novel protein components to three well-studied vaccine candidates significantly improved the efficacy of the combined vaccine. Furthermore, the five-component vaccine not only elicits a robust antibody response but also induces cytokine secretion by T cells, making it a promising vaccine candidate to fill the void.
Collapse
|
178
|
Kobayashi T, Naik S, Nagao K. Choreographing Immunity in the Skin Epithelial Barrier. Immunity 2019; 50:552-565. [PMID: 30893586 DOI: 10.1016/j.immuni.2019.02.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/21/2022]
Abstract
The skin interfaces with the external environment and is home to a myriad of immune cells that patrol the barrier to ward off harmful agents and aid in tissue repair. The formation of the cutaneous immune arsenal begins before birth and evolves throughout our lifetime, incorporating exogenous cues from microbes and inflammatory encounters, to achieve optimal fitness and function. Here, we discuss the context-specific signals that drive productive immune responses in the skin epithelium, highlighting key modulators of these reactions, including hair follicles, neurons, and commensal microbes. We thus also discuss the causal and mechanistic underpinning of inflammatory skin diseases that have been revealed in recent years. Finally, we discuss the non-canonical functions of cutaneous immune cells including their burgeoning role in epithelial regeneration and repair. The rapidly growing field of cutaneous immunity is revealing immune mechanisms and functions that can be harnessed to boost skin health and treat disease.
Collapse
Affiliation(s)
- Tetsuro Kobayashi
- Cutaneous Leukocyte Biology Section, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shruti Naik
- Department of Pathology, Ronald O. Perelman Department of Dermatology, and Department of Medicine, New York University School of Medicine, New York, NY, USA.
| | - Keisuke Nagao
- Cutaneous Leukocyte Biology Section, National Institutes of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
179
|
Wu KC, Zhong Y, Maher J. Predicting Human Infection Risk: Do Rodent Host Resistance Models Add Value? Toxicol Sci 2019; 170:260-272. [DOI: 10.1093/toxsci/kfz116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AbstractUse of genetically engineered rodents is often considered a valuable exercise to assess potential safety concerns associated with the inhibition of a target pathway. When there are potential immunomodulatory risks associated with the target, these genetically modified animals are often challenged with various pathogens in an acute setting to determine the risk to humans. However, the applicability of the results from infection models is seldom assessed when significant retrospective human data become available. Thus, the purpose of the current review is to compare the outcomes of infectious pathogen challenge in mice with genetic deficiencies in TNF-α, IL17, IL23, or Janus kinase pathways with infectious outcomes caused by inhibitors of these pathways in humans. In general, mouse infection challenge models had modest utility for hazard identification and were generally only able to predict overall trends in infection risk. These models did not demonstrate significant value in evaluating specific types of pathogens that are either prevalent (ie rhinoviruses) or of significant concern (ie herpes zoster). Similarly, outcomes in mouse models tended to overestimate the severity of infection risk in human patients. Thus, there is an emerging need for more human-relevant models that have better predictive value. Large meta-analyses of multiple clinical trials or post-marketing evaluations remains the gold-standard for characterizing the true infection risk to patients.
Collapse
Affiliation(s)
- Kai Connie Wu
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| | - Yu Zhong
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| | - Jonathan Maher
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
180
|
McGonagle DG, McInnes IB, Kirkham BW, Sherlock J, Moots R. The role of IL-17A in axial spondyloarthritis and psoriatic arthritis: recent advances and controversies. Ann Rheum Dis 2019; 78:1167-1178. [PMID: 31278139 PMCID: PMC6788885 DOI: 10.1136/annrheumdis-2019-215356] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/17/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022]
Abstract
Although the pathogenic mechanisms underlying axial spondyloarthritis (axSpA) and psoriatic arthritis (PsA) are not fully elucidated, several lines of evidence suggest that immune responses mediated by interleukin 17A (IL-17A) play a pivotal role in both diseases. This is best highlighted by the significant clinical efficacy shown with inhibitors of IL-17A in treating axSpA and PsA. Nevertheless, a number of knowledge gaps exist regarding the role of IL-17A in the pathophysiology of spondyloarthritis in man, including its cellular origin, its precise role in discrete disease processes such enthesitis, bone erosion, and bone formation, and the reasons for the discrepant responses to IL-17A inhibition observed in certain other spondyloarthritis manifestations. In this review, we focus on the latest data from studies investigating the role of IL-17A in ankylosing spondylitis (AS) and PsA that build on existing and emerging scientific knowledge in the field. Key remaining research questions are also highlighted to guide future research.
Collapse
Affiliation(s)
- Dennis G McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.,NIHR Leeds Musculoskeletal Biomedical Research Unit, Chapel Allerton, Leeds Teaching Hospital Trust, Leeds, UK
| | - Iain B McInnes
- Institute of Infection Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Bruce W Kirkham
- Rheumatology Department, Guy's and Saint Thomas' NHS Foundation Trust, London, UK
| | - Jonathan Sherlock
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Robert Moots
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK .,Department of Academic Rheumatology, Aintree University Hospital, Liverpool, UK
| |
Collapse
|
181
|
Weiss DI, Ma F, Merleev AA, Maverakis E, Gilliet M, Balin SJ, Bryson BD, Ochoa MT, Pellegrini M, Bloom BR, Modlin RL. IL-1β Induces the Rapid Secretion of the Antimicrobial Protein IL-26 from Th17 Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:911-921. [PMID: 31235553 DOI: 10.4049/jimmunol.1900318] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022]
Abstract
Th17 cells play a critical role in the adaptive immune response against extracellular bacteria, and the possible mechanisms by which they can protect against infection are of particular interest. In this study, we describe, to our knowledge, a novel IL-1β dependent pathway for secretion of the antimicrobial peptide IL-26 from human Th17 cells that is independent of and more rapid than classical TCR activation. We find that IL-26 is secreted 3 hours after treating PBMCs with Mycobacterium leprae as compared with 48 hours for IFN-γ and IL-17A. IL-1β was required for microbial ligand induction of IL-26 and was sufficient to stimulate IL-26 release from Th17 cells. Only IL-1RI+ Th17 cells responded to IL-1β, inducing an NF-κB-regulated transcriptome. Finally, supernatants from IL-1β-treated memory T cells killed Escherichia coli in an IL-26-dependent manner. These results identify a mechanism by which human IL-1RI+ "antimicrobial Th17 cells" can be rapidly activated by IL-1β as part of the innate immune response to produce IL-26 to kill extracellular bacteria.
Collapse
Affiliation(s)
- David I Weiss
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095.,Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095
| | - Feiyang Ma
- Molecular Biology Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, CA 90095.,Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095
| | - Alexander A Merleev
- Department of Dermatology, University of California, Davis, Sacramento, CA 95817
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, CA 95817
| | - Michel Gilliet
- Department of Dermatology, University Hospital Lausanne, 1005 Lausanne, Switzerland
| | - Samuel J Balin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Maria Teresa Ochoa
- Department of Dermatology, University of Southern California School of Medicine, Los Angeles, CA 90033
| | - Matteo Pellegrini
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095.,Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA 90095
| | - Barry R Bloom
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115; and
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, CA 90095; .,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
182
|
Nye TM, Jacob KM, Holley EK, Nevarez JM, Dawid S, Simmons LA, Watson ME. DNA methylation from a Type I restriction modification system influences gene expression and virulence in Streptococcus pyogenes. PLoS Pathog 2019; 15:e1007841. [PMID: 31206562 PMCID: PMC6597129 DOI: 10.1371/journal.ppat.1007841] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/27/2019] [Accepted: 05/14/2019] [Indexed: 11/26/2022] Open
Abstract
DNA methylation is pervasive across all domains of life. In bacteria, the presence of N6-methyladenosine (m6A) has been detected among diverse species, yet the contribution of m6A to the regulation of gene expression is unclear in many organisms. Here we investigated the impact of DNA methylation on gene expression and virulence within the human pathogen Streptococcus pyogenes, or Group A Streptococcus. Single Molecule Real-Time sequencing and subsequent methylation analysis identified 412 putative m6A sites throughout the 1.8 Mb genome. Deletion of the Restriction, Specificity, and Methylation gene subunits (ΔRSM strain) of a putative Type I restriction modification system lost all detectable m6A at the recognition sites and failed to prevent transformation with foreign-methylated DNA. RNA-sequencing identified 20 genes out of 1,895 predicted coding regions with significantly different gene expression. All of the differentially expressed genes were down regulated in the ΔRSM strain relative to the parent strain. Importantly, we found that the presence of m6A DNA modifications affected expression of Mga, a master transcriptional regulator for multiple virulence genes, surface adhesins, and immune-evasion factors in S. pyogenes. Using a murine subcutaneous infection model, mice infected with the ΔRSM strain exhibited an enhanced host immune response with larger skin lesions and increased levels of pro-inflammatory cytokines compared to mice infected with the parent or complemented mutant strains, suggesting alterations in m6A methylation influence virulence. Further, we found that the ΔRSM strain showed poor survival within human neutrophils and reduced adherence to human epithelial cells. These results demonstrate that, in addition to restriction of foreign DNA, gram-positive bacteria also use restriction modification systems to regulate the expression of gene networks important for virulence. DNA methylation is common among many bacterial species, yet the contribution of DNA methylation to the regulation of gene expression is unclear outside of a limited number of gram-negative species. We characterized sites of DNA methylation throughout the genome of the gram-positive pathogen Streptococcus pyogenes or Group A Streptococcus. We determined that the gene products of a functional restriction modification system are responsible for genome-wide m6A. The mutant strain lacking DNA methylation showed altered gene expression compared to the parent strain, with several genes important for causing human disease down regulated. Furthermore, we showed that the mutant strain lacking DNA methylation exhibited altered virulence properties compared to the parent strain using various models of pathogenesis. The mutant strain was attenuated for both survival within human neutrophils and adherence to human epithelial cells, and was unable to suppress the host immune response in a murine subcutaneous infection model. Together, these results show that bacterial m6A contributes to differential gene expression and influences the ability of Group A Streptococcus to cause disease. DNA methylation is a conserved feature among bacteria and may represent a potential target for intervention in effort to interfere with the ability of bacteria to cause human disease.
Collapse
Affiliation(s)
- Taylor M. Nye
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States of America
| | - Kristin M. Jacob
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
| | - Elena K. Holley
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
| | - Juan M. Nevarez
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
| | - Suzanne Dawid
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
| | - Lyle A. Simmons
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States of America
| | - Michael E. Watson
- Division of Pediatric Infectious Diseases, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, United States of America
- * E-mail:
| |
Collapse
|
183
|
Marchitto MC, Dillen CA, Liu H, Miller RJ, Archer NK, Ortines RV, Alphonse MP, Marusina AI, Merleev AA, Wang Y, Pinsker BL, Byrd AS, Brown ID, Ravipati A, Zhang E, Cai SS, Limjunyawong N, Dong X, Yeaman MR, Simon SI, Shen W, Durum SK, O'Brien RL, Maverakis E, Miller LS. Clonal Vγ6 +Vδ4 + T cells promote IL-17-mediated immunity against Staphylococcus aureus skin infection. Proc Natl Acad Sci U S A 2019; 116:10917-10926. [PMID: 31088972 PMCID: PMC6561199 DOI: 10.1073/pnas.1818256116] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
T cell cytokines contribute to immunity against Staphylococcus aureus, but the predominant T cell subsets involved are unclear. In an S. aureus skin infection mouse model, we found that the IL-17 response was mediated by γδ T cells, which trafficked from lymph nodes to the infected skin to induce neutrophil recruitment, proinflammatory cytokines IL-1α, IL-1β, and TNF, and host defense peptides. RNA-seq for TRG and TRD sequences in lymph nodes and skin revealed a single clonotypic expansion of the encoded complementarity-determining region 3 amino acid sequence, which could be generated by canonical nucleotide sequences of TRGV5 or TRGV6 and TRDV4 However, only TRGV6 and TRDV4 but not TRGV5 sequences expanded. Finally, Vγ6+ T cells were a predominant γδ T cell subset that produced IL-17A as well as IL-22, TNF, and IFNγ, indicating a broad and substantial role for clonal Vγ6+Vδ4+ T cells in immunity against S. aureus skin infections.
Collapse
Affiliation(s)
- Mark C Marchitto
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Carly A Dillen
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Robert J Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Roger V Ortines
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Alina I Marusina
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817
| | - Alexander A Merleev
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817
| | - Yu Wang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Bret L Pinsker
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Angel S Byrd
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Isabelle D Brown
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Advaitaa Ravipati
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Emily Zhang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Shuting S Cai
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231
| | - Nathachit Limjunyawong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- The Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Xinzhong Dong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- The Center for Sensory Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Michael R Yeaman
- Division of Molecular Medicine, Harbor-UCLA Medical Center, Torrance, CA 90502
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA 90502
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095
- Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA 90502
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Wei Shen
- Cytokines and Immunity Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Scott K Durum
- Cytokines and Immunity Section, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Rebecca L O'Brien
- Department of Biomedical Research, National Jewish Health, Denver, CO 80206
- Department of Immunology and Microbiology, University of Colorado Health Sciences Center, Aurora, CO 80206
| | - Emanual Maverakis
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA 95817
| | - Lloyd S Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21231;
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218
| |
Collapse
|
184
|
Borbón TY, Scorza BM, Clay GM, Lima Nobre de Queiroz F, Sariol AJ, Bowen JL, Chen Y, Zhanbolat B, Parlet CP, Valadares DG, Cassel SL, Nauseef WM, Horswill AR, Sutterwala FS, Wilson ME. Coinfection with Leishmania major and Staphylococcus aureus enhances the pathologic responses to both microbes through a pathway involving IL-17A. PLoS Negl Trop Dis 2019; 13:e0007247. [PMID: 31107882 PMCID: PMC6527190 DOI: 10.1371/journal.pntd.0007247] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 02/15/2019] [Indexed: 12/12/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is a parasitic disease causing chronic, ulcerating skin lesions. Most humans infected with the causative Leishmania protozoa are asymptomatic. Leishmania spp. are usually introduced by sand flies into the dermis of mammalian hosts in the presence of bacteria from either the host skin, sand fly gut or both. We hypothesized that bacteria at the dermal inoculation site of Leishmania major will influence the severity of infection that ensues. A C57BL/6 mouse ear model of single or coinfection with Leishmania major, Staphylococcus aureus, or both showed that single pathogen infections caused localized lesions that peaked after 2–3 days for S. aureus and 3 weeks for L. major infection, but that coinfection produced lesions that were two-fold larger than single infection throughout 4 weeks after coinfection. Coinfection increased S. aureus burdens over 7 days, whereas L. major burdens (3, 7, 28 days) were the same in singly and coinfected ears. Inflammatory lesions throughout the first 4 weeks of coinfection had more neutrophils than did singly infected lesions, and the recruited neutrophils from early (day 1) lesions had similar phagocytic and NADPH oxidase capacities. However, most neutrophils were apoptotic, and transcription of immunomodulatory genes that promote efferocytosis was not upregulated, suggesting that the increased numbers of neutrophils may, in part, reflect defective clearance and resolution of the inflammatory response. In addition, the presence of more IL-17A-producing γδ and non-γδ T cells in early lesions (1–7 days), and L. major antigen-responsive Th17 cells after 28 days of coinfection, with a corresponding increase in IL-1β, may recruit more naïve neutrophils into the inflammatory site. Neutralization studies suggest that IL-17A contributed to an enhanced inflammatory response, whereas IL-1β has an important role in controlling bacterial replication. Taken together, these data suggest that coinfection of L. major infection with S. aureus exacerbates disease, both by promoting more inflammation and neutrophil recruitment and by increasing neutrophil apoptosis and delaying resolution of the inflammatory response. These data illustrate the profound impact that coinfecting microorganisms can exert on inflammatory lesion pathology and host adaptive immune responses. Cutaneous leishmaniasis (CL) is a vector-borne ulcerating skin disease affecting several million people worldwide. The causative Leishmania spp. protozoa are transmitted by infected phlebotomine sand flies. During a sand fly bite, bacteria can be coincidentally inoculated into the dermis with the parasite. Staphylococcus aureus is the most common bacterium in CL skin lesions. Symptomatic CL is characterized by papulonodular skin lesions that ulcerate and resolve with scarring, although most cutaneous Leishmania infections are asymptomatic. We sought to explore factors that determine whether infection with a cutaneous Leishmania species would result in symptomatic CL rather than asymptomatic infection. We hypothesized that local bacteria promote the development of symptomatic CL lesions during infection with Leishmania major. We discovered that cutaneous lesions were significantly larger in mice inoculated simultaneously with S. aureus and L. major than in mice infected with either organism alone. Coinfection led to increased S. aureus growth in skin lesions, whereas L. major parasite numbers were unchanged by coinfection. The size of the exacerbated lesion correlated with early increased numbers of neutrophils and elevated levels of proinflammatory cytokines IL-1β and IL-17A during the first 7 days, and with sustained increases in IL-17A through 28 days of coinfection. Neutralizing antibody experiments suggested IL-17A was partially responsible for lesion exacerbation during coinfection, whereas IL-1β was important for both control of early lesion exacerbation and promotion of IL-17A production. These data suggest that treatment of symptomatic CL targeting the parasite, local commensal bacteria, and host proinflammatory IL-17A immune responses might improve the outcome of CL.
Collapse
Affiliation(s)
- Tiffany Y. Borbón
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Breanna M. Scorza
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Gwendolyn M. Clay
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Molecular Medicine, University of Iowa, Iowa City, IA, United States of America
| | | | - Alan J. Sariol
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Jayden L. Bowen
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Yani Chen
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Bayan Zhanbolat
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Corey P. Parlet
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Diogo G. Valadares
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
- Conselho Nacional de Desenvolvimento Cientifico e Tecnológico (CNPq), Brasilia, Brazil
| | - Suzanne L. Cassel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - William M. Nauseef
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado Denver—Anschutz Medical Campus, Aurora, CO, United States of America
| | - Fayyaz S. Sutterwala
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Mary E. Wilson
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Molecular Medicine, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
- * E-mail:
| |
Collapse
|
185
|
Richardson JR, Armbruster NS, Günter M, Biljecki M, Klenk J, Heumos S, Autenrieth SE. PSM Peptides From Community-Associated Methicillin-Resistant Staphylococcus aureus Impair the Adaptive Immune Response via Modulation of Dendritic Cell Subsets in vivo. Front Immunol 2019; 10:995. [PMID: 31134074 PMCID: PMC6524657 DOI: 10.3389/fimmu.2019.00995] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/17/2019] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) are key players of the immune system and thus a target for immune evasion by pathogens. We recently showed that the virulence factors phenol-soluble-modulins (PSMs) produced by community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) strains induce tolerogenic DCs upon Toll-like receptor activation via the p38-CREB-IL-10 pathway in vitro. Here, we addressed the hypothesis that S. aureus PSMs disturb the adaptive immune response via modulation of DC subsets in vivo. Using a systemic mouse infection model we found that S. aureus reduced the numbers of splenic DC subsets, mainly CD4+ and CD8+ DCs independently of PSM secretion. S. aureus infection induced upregulation of the C-C motif chemokine receptor 7 (CCR7) on the surface of all DC subsets, on CD4+ DCs in a PSM-dependent manner, together with increased expression of MHCII, CD86, CD80, CD40, and the co-inhibitory molecule PD-L2, with only minor effects of PSMs. Moreover, PSMs increased IL-10 production in the spleen and impaired TNF production by CD4+ DCs. Besides, S. aureus PSMs reduced the number of CD4+ T cells in the spleen, whereas CD4+CD25+Foxp3+ regulatory T cells (Tregs) were increased. In contrast, Th1 and Th17 priming and IFN-γ production by CD8+ T cells were impaired by S. aureus PSMs. Thus, PSMs from highly virulent S. aureus strains modulate the adaptive immune response in the direction of tolerance by affecting DC functions.
Collapse
Affiliation(s)
| | - Nicole S Armbruster
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Manina Günter
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Michelle Biljecki
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Juliane Klenk
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| | - Simon Heumos
- Quantitative Biology Center, University of Tübingen, Tübingen, Germany
| | - Stella E Autenrieth
- Department of Internal Medicine II, University of Tübingen, Tübingen, Germany
| |
Collapse
|
186
|
Clumping factor B is an important virulence factor during Staphylococcus aureus skin infection and a promising vaccine target. PLoS Pathog 2019; 15:e1007713. [PMID: 31009507 PMCID: PMC6497315 DOI: 10.1371/journal.ppat.1007713] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 05/02/2019] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Staphylococcus aureus expresses a number of cell wall-anchored proteins that mediate adhesion and invasion of host cells and tissues and promote immune evasion, consequently contributing to the virulence of this organism. The cell wall-anchored protein clumping factor B (ClfB) has previously been shown to facilitate S. aureus nasal colonization through high affinity interactions with the cornified envelope in the anterior nares. However, the role of ClfB during skin and soft tissue infection (SSTI) has never been investigated. This study reveals a novel role for ClfB during SSTIs. ClfB is crucial in determining the abscess structure and bacterial burden early in infection and this is dependent upon a specific interaction with the ligand loricrin which is expressed within the abscess tissue. Targeting ClfB using a model vaccine that induced both protective humoral and cellular responses, leads to protection during S. aureus skin infection. This study therefore identifies ClfB as an important antigen for future SSTI vaccines. Staphylococcus aureus is the leading cause of skin and soft tissue infections (SSTIs), the treatment of which is becoming increasingly difficult due to antibiotic resistance. An anti-S. aureus vaccine offers a potential solution, but a better understanding of how S. aureus causes pathology during SSTI is required to identify effective vaccine targets. Here, we identify an important virulence determinant during S. aureus SSTI. Clumping factor B (ClfB), a surface protein expressed by S. aureus is shown to promote skin abscess formation by binding to the host protein loricrin. Targeting ClfB using a model vaccine conferred significant protection during S. aureus SSTI. In this study, we uncover an entirely novel mechanism by which S. aureus forms abscesses during skin infection, identifying an important therapeutic target for treating S. aureus SSTI.
Collapse
|
187
|
Nowicka D, Grywalska E. Staphylococcus aureus and Host Immunity in Recurrent Furunculosis. Dermatology 2019; 235:295-305. [DOI: 10.1159/000499184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/28/2019] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus is one of the severest and most persistent bacterial pathogens. The most frequent S. aureus infections include impetigo, folliculitis, furuncles, furunculosis, abscesses, hidradenitis suppurativa, and mastitis. S. aureus produces a great variety of cellular and extracellular factors responsible for its invasiveness and ability to cause pathological lesions. Their expression depends on the growth phase, environmental factors, and location of the infection. Susceptibility to staphylococcal infections is rooted in multiple mechanisms of host immune responses and reactions to bacterial colonization. Immunological and inflammatory processes of chronic furunculosis are based on the pathogenicity of S. aureus as well as innate and acquired immunity. In-depth knowledge about them may help to discover the whole pathomechanism of the disease and to develop effective therapeutic options. In this review, we focus on the S. aureus-host immune interactions in the pathogenesis of recurrent furunculosis according to the most recent experimental and clinical findings.
Collapse
|
188
|
Tait Wojno ED, Hunter CA, Stumhofer JS. The Immunobiology of the Interleukin-12 Family: Room for Discovery. Immunity 2019; 50:851-870. [PMID: 30995503 PMCID: PMC6472917 DOI: 10.1016/j.immuni.2019.03.011] [Citation(s) in RCA: 291] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/12/2022]
Abstract
The discovery of interleukin (IL)-6 and its receptor subunits provided a foundation to understand the biology of a group of related cytokines: IL-12, IL-23, and IL-27. These family members utilize shared receptors and cytokine subunits and influence the outcome of cancer, infection, and inflammatory diseases. Consequently, many facets of their biology are being therapeutically targeted. Here, we review the landmark discoveries in this field, the combinatorial biology inherent to this family, and how patient datasets have underscored the critical role of these pathways in human disease. We present significant knowledge gaps, including how similar signals from these cytokines can mediate distinct outcomes, and discuss how a better understanding of the biology of the IL-12 family provides new therapeutic opportunities.
Collapse
Affiliation(s)
- Elia D Tait Wojno
- Baker Institute for Animal Health and Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, 235 Hungerford Hill Rd., Ithaca, NY 14853, USA
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 380 South University Ave., Philadelphia, PA 19104-4539, USA.
| | - Jason S Stumhofer
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, 4301 West Markham St., Little Rock, AR 72205, USA.
| |
Collapse
|
189
|
The Dynamics of the Skin's Immune System. Int J Mol Sci 2019; 20:ijms20081811. [PMID: 31013709 PMCID: PMC6515324 DOI: 10.3390/ijms20081811] [Citation(s) in RCA: 317] [Impact Index Per Article: 63.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/09/2019] [Accepted: 04/09/2019] [Indexed: 12/12/2022] Open
Abstract
The skin is a complex organ that has devised numerous strategies, such as physical, chemical, and microbiological barriers, to protect the host from external insults. In addition, the skin contains an intricate network of immune cells resident to the tissue, crucial for host defense as well as tissue homeostasis. In the event of an insult, the skin-resident immune cells are crucial not only for prevention of infection but also for tissue reconstruction. Deregulation of immune responses often leads to impaired healing and poor tissue restoration and function. In this review, we will discuss the defensive components of the skin and focus on the function of skin-resident immune cells in homeostasis and their role in wound healing.
Collapse
|
190
|
Ebrahim AA, Mustafa AI, El‐Abd AM. Serum interleukin‐17 as a novel biomarker in patients with acne vulgaris. J Cosmet Dermatol 2019; 18:1975-1979. [DOI: 10.1111/jocd.12934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 02/20/2019] [Indexed: 01/09/2023]
Affiliation(s)
- Adel A. Ebrahim
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine Benha University Benha Egypt
| | - Amany Ibrahim Mustafa
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine Benha University Benha Egypt
| | - Awad Mohamed El‐Abd
- Department of Medical Biochemistry, Faculty of Medicine Benha University Benha Egypt
| |
Collapse
|
191
|
Li H, Hong W, Jin X, Li G, Zhou G, Fan L. The aryl hydrocarbon receptor is a novel negative regulator of interleukin-17-mediated signaling and inflammation in vitro. FEBS Lett 2019; 593:952-961. [PMID: 30953345 DOI: 10.1002/1873-3468.13380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/24/2019] [Accepted: 03/06/2019] [Indexed: 12/02/2022]
Abstract
Interleukin (IL)-17 plays a critical role in the pathogenesis of inflammation and autoimmune diseases. The aryl hydrocarbon receptor (AHR) is a transcription factor responsible for the elimination of xenobiotic chemicals. However, it remains unknown whether AHR is involved in IL-17 signaling. Here, we demonstrate that knockdown of AHR significantly enhances, while overexpression or activation of AHR inhibits IL-17-induced inflammation in Hela cells. AHR specifically suppresses IL-17-induced p38 activation, and inhibition of p38 activity markedly reverses the effect of AHR knockdown on IL-17-induced inflammation. Mechanistically, AHR physically interacts with TAK1 and mitogen-activated protein kinase kinase 3/6 (MKK3/6) and disrupts TAK1-MKK3/6 interaction, leading to impaired IL-17 signaling. Thus, our study indicates that AHR negatively regulates IL-17-mediated signaling and inflammation at least partially through interfering with the interaction between TAK1 and MKK3/6.
Collapse
Affiliation(s)
- Hui Li
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wei Hong
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Xiangyu Jin
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Guangliang Li
- Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Guoming Zhou
- Departments of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Liping Fan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
192
|
Petrović J, Silva JR, Bannerman CA, Segal JP, Marshall AS, Haird CM, Gilron I, Ghasemlou N. γδ T Cells Modulate Myeloid Cell Recruitment but Not Pain During Peripheral Inflammation. Front Immunol 2019; 10:473. [PMID: 30936874 PMCID: PMC6431614 DOI: 10.3389/fimmu.2019.00473] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
Circulating immune cells, which are recruited to the site of injury/disease, secrete various inflammatory mediators that are critical to nociception and pain. The role of tissue-resident immune cells, however, remains poorly characterized. One of the first cells to be activated in peripheral tissues following injury are γδT cells, which serve important roles in infection, disease, and wound healing. Using a mouse line lacking these cells, we sought to identify their contribution to inflammatory pain. Three distinct models of peripheral inflammatory pain were used: intraplantar injection of formalin (spontaneous inflammatory pain), incisional wound (acute inflammatory pain), and intraplantar injection of complete Freund's adjuvant (chronic inflammatory pain). Our results show that absence of γδT cells does not alter baseline sensitivity, nor does it result in changes to mechanical or thermal hypersensitivity after tissue injury. Myeloid cell recruitment did show differential changes between models of acute and chronic inflammatory pain. These results were consistent in both male and female mice, suggesting that there are no sex differences in these outcomes. This comprehensive characterization suggests that γδT cells do not contribute to basal sensitivity or the development and maintenance of inflammatory pain.
Collapse
Affiliation(s)
- Jelena Petrović
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Jaqueline Raymondi Silva
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Anesthesiology & Perioperative Medicine, Queen's University, Kingston, ON, Canada
| | - Courtney A. Bannerman
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Julia P. Segal
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Abigail S. Marshall
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Cortney M. Haird
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Ian Gilron
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Anesthesiology & Perioperative Medicine, Queen's University, Kingston, ON, Canada
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| | - Nader Ghasemlou
- Department of Biomedical & Molecular Sciences, Queen's University, Kingston, ON, Canada
- Department of Anesthesiology & Perioperative Medicine, Queen's University, Kingston, ON, Canada
- Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| |
Collapse
|
193
|
CCR2 mediates increased susceptibility to post-H1N1 bacterial pneumonia by limiting dendritic cell induction of IL-17. Mucosal Immunol 2019; 12:518-530. [PMID: 30498200 PMCID: PMC6375750 DOI: 10.1038/s41385-018-0106-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 10/06/2018] [Accepted: 10/23/2018] [Indexed: 02/06/2023]
Abstract
Post influenza bacterial pneumonia is associated with significant mortality and morbidity. Dendritic cells (DCs) play a crucial role in host defense against bacterial pneumonia, but their contribution to post influenza-susceptibility to secondary bacterial pneumonia is incompletely understood. WT and CCR2-/- mice were infected with 100 plaque forming units (pfu) H1N1 intranasally alone or were challenged on day 5 with 7 × 107 colony forming units (cfu) methicillin-resistant Staphylococcus aureus intratracheally. WT mice express abundant CCL2 mRNA and protein post-H1N1 alone or dual infection. CCR2-/- mice had significantly higher survival as compared to WT mice, associated with significantly improved bacterial clearance at 24 and 48 h (10-fold and 14-fold, respectively) post bacterial challenge. There was robust upregulation of IL-23 and IL-17 as well as downregulation of IL-27 expression in CCR2-/- mice following sequential infection as compared to WT mice, which was also associated with significantly greater accumulation of CD103+ DC. Finally, WT mice treated with a CCR2 inhibitor showed improved bacterial clearance in association with similar cytokine profiles as CCR2-/- mice. Thus, CCR2 significantly contributes to increased susceptibility to bacterial infection after influenza pneumonia likely via altered dendritic cell responses and thus, CCR2 antagonism represents a potential therapeutic strategy.
Collapse
|
194
|
O’Brien EC, McLoughlin RM. Considering the ‘Alternatives’ for Next-Generation Anti-Staphylococcus aureus Vaccine Development. Trends Mol Med 2019; 25:171-184. [DOI: 10.1016/j.molmed.2018.12.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/20/2018] [Accepted: 12/28/2018] [Indexed: 12/14/2022]
|
195
|
Kim HK, Garcia AB, Siu E, Tilstam P, Das R, Roberts S, Leng L, Bucala R. Macrophage migration inhibitory factor regulates innate γδ T-cell responses via IL-17 expression. FASEB J 2019; 33:6919-6932. [PMID: 30817226 DOI: 10.1096/fj.201802433r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
T cells expressing invariant γδ antigen receptors (γδ T cells) bridge innate and adaptive immunity and facilitate barrier responses to pathogens. Macrophage migration inhibitory factor (MIF) is an upstream mediator of host defense that up-regulates the expression of pattern recognition receptors and sustains inflammatory responses by inhibiting activation-induced apoptosis in monocytes and macrophages. Surprisingly, Mif-/- γδ T cells, when compared with wild type, were observed to produce >10-fold higher levels of the proinflammatory cytokine IL-17 after stimulation with gram-positive exotoxins. High-IL-17 expression was associated with the characteristic features of IL-17-producing γδ T (γδ17) cells, including expression of IL-23R, IL-1R1, and the transcription factors RORγt and Sox13. In the gram-positive model of shock mediated by toxic shock syndrome toxin (TSST-1), Mif-/- mice succumbed to death more quickly with increased pulmonary neutrophil accumulation and higher production of cytokines, including IL-1β and IL-23. Mif-/- γδ T cells also produced high levels of IL-17 in response to Mycobacterium lipomannan, and depletion of γδ T cells improved survival from acutely lethal Mycobacterium infection or TSST-1 administration. These data indicate that MIF deficiency is associated with a compensatory amplification of γδ17 cell responses, with implications for innate immunity and IL-17-mediated pathology in situations such as gram-positive toxic shock or Mycobacterium infection.-Kim, H. K., Garcia, A. B., Siu, E., Tilstam, P., Das, R., Roberts, S., Leng, L., Bucala, R. Macrophage migration inhibitory factor regulates innate γδ T-cell responses via IL-17 expression.
Collapse
Affiliation(s)
- Hee Kyung Kim
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - Alvaro Baeza Garcia
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - Edwin Siu
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - Pathricia Tilstam
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - Rita Das
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA; and.,Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Scott Roberts
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - Lin Leng
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut, USA; and
| | - Richard Bucala
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
196
|
Strbo N, Pastar I, Romero L, Chen V, Vujanac M, Sawaya AP, Jozic I, Ferreira ADF, Wong LL, Head C, Stojadinovic O, Garcia D, O'Neill K, Drakulich S, Taller S, Kirsner RS, Tomic-Canic M. Single cell analyses reveal specific distribution of anti-bacterial molecule Perforin-2 in human skin and its modulation by wounding and Staphylococcus aureus infection. Exp Dermatol 2019; 28:225-232. [PMID: 30609079 DOI: 10.1111/exd.13870] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 12/03/2018] [Accepted: 01/02/2019] [Indexed: 01/13/2023]
Abstract
Perforin-2 (P-2) is a recently described antimicrobial protein with unique properties to kill intracellular bacteria. We investigated P-2 expression pattern and cellular distribution in human skin and its importance in restoration of barrier function during wound healing process and infection with the common wound pathogen Staphylococcus aureus. We describe a novel approach for the measurement of P-2 mRNA within individual skin cells using an amplified fluorescence in situ hybridization (FISH) technique. The unique aspect of this approach is simultaneous detection of P-2 mRNA in combination with immune-phenotyping for cell surface proteins using fluorochrome-conjugated antibodies. We detected P-2 transcript in both hematopoietic (CD45+ ) and non-hematopoietic (CD45- ) cutaneous cell populations, confirming the P-2 expression in both professional and non-professional phagocytes. Furthermore, we found an induction of P-2 during wound healing. P-2 overexpression resulted in a reduction of intracellular S. aureus, while infection of human wounds by this pathogen resulted in P-2 suppression, revealing a novel mechanism by which S. aureus may escape cutaneous immunity to cause persistent wound infections.
Collapse
Affiliation(s)
- Natasa Strbo
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
| | - Irena Pastar
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Laura Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
| | - Vivien Chen
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Milos Vujanac
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
| | - Andrew P Sawaya
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Ivan Jozic
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Andrea D F Ferreira
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Lulu L Wong
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Cheyanne Head
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Olivera Stojadinovic
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Denisse Garcia
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
| | - Katelyn O'Neill
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida
| | - Stefan Drakulich
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Seth Taller
- Plastic Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Robert S Kirsner
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| | - Marjana Tomic-Canic
- Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
197
|
Shen F, Verma AH, Volk A, Jones B, Coleman BM, Loza MJ, Malaviya R, Moore B, Weinstock D, Elloso MM, Gaffen SL, Ort T. Combined Blockade of TNF-α and IL-17A Alleviates Progression of Collagen-Induced Arthritis without Causing Serious Infections in Mice. THE JOURNAL OF IMMUNOLOGY 2019; 202:2017-2026. [PMID: 30745461 DOI: 10.4049/jimmunol.1801436] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/15/2019] [Indexed: 02/06/2023]
Abstract
The cytokines TNF-α and IL-17A are elevated in a variety of autoimmune diseases, including rheumatoid arthritis. Both cytokines are targets of several biologic drugs used in the clinic, but unfortunately many patients are refractory to these therapies. IL-17A and TNF-α are known to mediate signaling synergistically to drive expression of inflammatory genes. Hence, combined blockade of TNF-α and IL-17A represents an attractive treatment strategy in autoimmune settings where monotherapy is not fully effective. However, a major concern with this approach is the potential predisposition to opportunistic infections that might outweigh any clinical benefits. Accordingly, we examined the impact of individual versus combined neutralization of TNF-α and IL-17A in a mouse model of rheumatoid arthritis (collagen-induced arthritis) and the concomitant susceptibility to infections that are likely to manifest as side effects of blocking these cytokines (oral candidiasis or tuberculosis). Our findings indicate that combined neutralization of TNF-α and IL-17A was considerably more effective than monotherapy in improving collagen-induced arthritis disease even when administered at a minimally efficacious dose. Encouragingly, however, dual cytokine blockade did not cooperatively impair antimicrobial host defenses, as mice given combined IL-17A and TNF-α neutralization displayed infectious profiles and humoral responses comparable to mice given high doses of individual anti-TNF-α or anti-IL-17A mAbs. These data support the idea that combined neutralization of TNF-α and IL-17A for refractory autoimmunity is likely to be associated with acceptable and manageable risks of opportunistic infections associated with these cytokines.
Collapse
Affiliation(s)
- Fang Shen
- Immunology Discovery, Janssen Research and Development, LLC, Spring House, PA 19477
| | - Akash H Verma
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Amy Volk
- Immuno-Toxicology, Janssen Research and Development, LLC, Spring House, PA 19477; and
| | - Brian Jones
- Immunology Discovery, Janssen Research and Development, LLC, Spring House, PA 19477
| | - Bianca M Coleman
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Matthew J Loza
- Immunology Biomarker, Janssen Research and Development, LLC, Spring House, PA 19477
| | - Ravi Malaviya
- Immunology Discovery, Janssen Research and Development, LLC, Spring House, PA 19477
| | - Beverley Moore
- Immunology Discovery, Janssen Research and Development, LLC, Spring House, PA 19477
| | - Daniel Weinstock
- Immuno-Toxicology, Janssen Research and Development, LLC, Spring House, PA 19477; and
| | - M Merle Elloso
- Immunology Discovery, Janssen Research and Development, LLC, Spring House, PA 19477
| | - Sarah L Gaffen
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA 15261;
| | - Tatiana Ort
- Immunology Discovery, Janssen Research and Development, LLC, Spring House, PA 19477;
| |
Collapse
|
198
|
Efficacy of photoactivated Myrciaria cauliflora extract against Staphylococcus aureus infection – A pilot study. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 191:107-115. [DOI: 10.1016/j.jphotobiol.2018.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 12/12/2018] [Accepted: 12/16/2018] [Indexed: 01/11/2023]
|
199
|
Copin R, Sause WE, Fulmer Y, Balasubramanian D, Dyzenhaus S, Ahmed JM, Kumar K, Lees J, Stachel A, Fisher JC, Drlica K, Phillips M, Weiser JN, Planet PJ, Uhlemann AC, Altman DR, Sebra R, van Bakel H, Lighter J, Torres VJ, Shopsin B. Sequential evolution of virulence and resistance during clonal spread of community-acquired methicillin-resistant Staphylococcus aureus. Proc Natl Acad Sci U S A 2019; 116:1745-1754. [PMID: 30635416 PMCID: PMC6358666 DOI: 10.1073/pnas.1814265116] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The past two decades have witnessed an alarming expansion of staphylococcal disease caused by community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA). The factors underlying the epidemic expansion of CA-MRSA lineages such as USA300, the predominant CA-MRSA clone in the United States, are largely unknown. Previously described virulence and antimicrobial resistance genes that promote the dissemination of CA-MRSA are carried by mobile genetic elements, including phages and plasmids. Here, we used high-resolution genomics and experimental infections to characterize the evolution of a USA300 variant plaguing a patient population at increased risk of infection to understand the mechanisms underlying the emergence of genetic elements that facilitate clonal spread of the pathogen. Genetic analyses provided conclusive evidence that fitness (manifest as emergence of a dominant clone) changed coincidently with the stepwise emergence of (i) a unique prophage and mutation of the regulator of the pyrimidine nucleotide biosynthetic operon that promoted abscess formation and colonization, respectively, thereby priming the clone for success; and (ii) a unique plasmid that conferred resistance to two topical microbiocides, mupirocin and chlorhexidine, frequently used for decolonization and infection prevention. The resistance plasmid evolved through successive incorporation of DNA elements from non-S. aureus spp. into an indigenous cryptic plasmid, suggesting a mechanism for interspecies genetic exchange that promotes antimicrobial resistance. Collectively, the data suggest that clonal spread in a vulnerable population resulted from extensive clinical intervention and intense selection pressure toward a pathogen lifestyle that involved the evolution of consequential mutations and mobile genetic elements.
Collapse
Affiliation(s)
- Richard Copin
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - William E Sause
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| | - Yi Fulmer
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Divya Balasubramanian
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| | - Sophie Dyzenhaus
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| | - Jamil M Ahmed
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Krishan Kumar
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - John Lees
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| | - Anna Stachel
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Jason C Fisher
- Division of Pediatric Surgery, Department of Surgery, New York University School of Medicine, New York, NY 10016
| | - Karl Drlica
- Public Health Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers University, Newark, NJ 07103
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Rutgers University, Newark, NJ 07103
| | - Michael Phillips
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, NY 10016
| | - Jeffrey N Weiser
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| | - Paul J Planet
- Department of Pediatric Infectious Disease, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Anne-Catrin Uhlemann
- Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY 10032
| | - Deena R Altman
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY 10029
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jennifer Lighter
- Division of Pediatric Infectious Diseases, Department of Pediatrics, New York University School of Medicine, New York, NY 10016
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016;
| | - Bo Shopsin
- Division of Infectious Diseases and Immunology, Department of Medicine, New York University School of Medicine, New York, NY 10016;
- Department of Microbiology, New York University School of Medicine, New York, NY 10016
| |
Collapse
|
200
|
Abstract
Psoriasis (PsO) is one of the most common chronic inflammatory skin diseases with a world prevalence of 2%-4%. The increasing knowledge of the mechanisms driving PsO has raised focus on existing links to metabolic syndrome and type 2 diabetes (T2D). We reviewed the existing literature of the prevalence and risk of T2D in patients with PsO. The studies reviewed were mainly large retrospective cohort and case-control studies, showing an increased prevalence of T2D in PsO patients compared to controls, particularly in late onset (type 2) PsO. T2D prevalence did not correlate to patient age or severity of PsO in the reviewed studies. Conclusively, T2D was found to be more prevalent in patients with PsO compared to the background population. Several mechanisms involved in lipid transportation seem to be upregulated in PsO patients. Physicians play a key role concerning information about known comorbidity and promotion of early prophylaxis in patients with PsO.
Collapse
Affiliation(s)
- Jesper Grønlund Holm
- Department of Dermato-Venereology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark,
| | - Simon Francis Thomsen
- Department of Dermato-Venereology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark, .,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|