1
|
Angelicola S, Giunchi F, Ruzzi F, Frascino M, Pitzalis M, Scalambra L, Semprini MS, Pittino OM, Cappello C, Siracusa I, Chillico IC, Di Noia M, Turato C, De Siervi S, Lescai F, Ciavattini T, Lopatriello G, Bertoli L, De Jonge H, Iamele L, Altimari A, Gruppioni E, Ardizzoni A, Rossato M, Gelsomino F, Lollini PL, Palladini A. PD-L1 and IFN-γ modulate Non-Small Cell Lung Cancer (NSCLC) cell plasticity associated to immune checkpoint inhibitor (ICI)-mediated hyperprogressive disease (HPD). J Transl Med 2025; 23:2. [PMID: 39748404 PMCID: PMC11697469 DOI: 10.1186/s12967-024-06023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND Non-Small Cell Lung Cancer (NSCLC) is the leading cause of cancer death worldwide. Although immune checkpoint inhibitors (ICIs) have shown remarkable clinical efficacy, they can also induce a paradoxical cancer acceleration, known as hyperprogressive disease (HPD), whose causative mechanisms are still unclear. METHODS This study investigated the mechanisms of ICI resistance in an HPD-NSCLC model. Two primary cell cultures were established from samples of a NSCLC patient, before ICI initiation ("baseline", NSCLC-B) and during HPD ("hyperprogression", NSCLC-H). The cell lines were phenotypically and molecularly characterized through immunofluorescence, Western Blotting and RNA-Seq analysis. To assess cell plasticity and aggressiveness, cellular growth patterns were evaluated both in vitro and in vivo through 2D and 3D cell growth assays and patient-derived xenografts establishment. In vitro investigations, including the evaluation of cell sensitivity to interferon-gamma (IFN-γ) and cell response to PD-L1 modulation, were conducted to explore the influence of these factors on cell plasticity regulation. RESULTS NSCLC-H exhibited increased expression of specific CD44 isoforms and a more aggressive phenotype, including organoid formation ability, compared to NSCLC-B. Plastic changes in NSCLC-H were well described by a deep transcriptome shift, that also affected IFN-γ-related genes, including PD-L1. IFN-γ-mediated cell growth inhibition was compromised in both 2D-cultured NSCLC-B and NSCLC-H cells. Further, the cytokine induced a partial activation of both type I and type II IFN-pathway mediators, together with a striking increase in NSCLC-B growth in 3D cell culture systems. Finally, low IFN-γ doses and PD-L1 modulation both promoted plastic changes in NSCLC-B, increasing CD44 expression and its ability to produce spheres. CONCLUSIONS Our findings identified plasticity as a relevant hallmark of ICI-mediated HPD by demonstrating that ICIs can modulate the IFN-γ and PD-L1 pathways, driving tumor cell plasticity and fueling HPD development.
Collapse
Affiliation(s)
- Stefania Angelicola
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Francesca Giunchi
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | | - Mary Pitzalis
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Laura Scalambra
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Maria Sofia Semprini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Olga Maria Pittino
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Chiara Cappello
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Irene Siracusa
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Martina Di Noia
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Cristian Turato
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Silvia De Siervi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Francesco Lescai
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | | | | | - Luca Bertoli
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Hugo De Jonge
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Luisa Iamele
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Annalisa Altimari
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Elisa Gruppioni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrea Ardizzoni
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Marzia Rossato
- Department of Biotechnology, University of Verona, Verona, Italy
- Genartis S.R.L., Verona, Italy
| | - Francesco Gelsomino
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy
| | - Arianna Palladini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Unità Operativa di Oncologia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| |
Collapse
|
2
|
Tecalco-Cruz AC, Medina-Abreu KH, Oropeza-Martínez E, Zepeda-Cervantes J, Vázquez-Macías A, Macías-Silva M. Deregulation of interferon-gamma receptor 1 expression and its implications for lung adenocarcinoma progression. World J Clin Oncol 2024; 15:195-207. [PMID: 38455133 PMCID: PMC10915940 DOI: 10.5306/wjco.v15.i2.195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/05/2024] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Interferon-gamma (IFN-γ) plays a dual role in cancer; it is both a pro- and an antitumorigenic cytokine, depending on the type of cancer. The deregulation of the IFN-γ canonic pathway is associated with several disorders, including vulnerability to viral infections, inflammation, and cancer progression. In particular, the interplay between lung adenocarcinoma (LUAD) and viral infections appears to exist in association with the deregulation of IFN-γ signaling. In this mini-review, we investigated the status of the IFN-γ signaling pathway and the expression level of its components in LUAD. Interestingly, a reduction in IFNGR1 expression seems to be associated with LUAD progression, affecting defenses against viruses such as severe acute respiratory syndrome coronavirus 2. In addition, alterations in the expression of IFNGR1 may inhibit the antiproliferative action of IFN-γ signaling in LUAD.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX 03100, Mexico
| | - Karen H Medina-Abreu
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, CDMX 03100, Mexico
| | | | - Jesus Zepeda-Cervantes
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, CDMX 04510, Mexico
| | - Aleida Vázquez-Macías
- Colegio de Ciencias y Humanidades, Universidad Autónoma de la Ciudad de México, CDMX 03100, Mexico
| | - Marina Macías-Silva
- Instituo de Fisiología Celular, Universidad Nacional Autónoma de México, CDMX 04510, Mexico
| |
Collapse
|
3
|
Ji L, Li T, Chen H, Yang Y, Lu E, Liu J, Qiao W, Chen H. The crucial regulatory role of type I interferon in inflammatory diseases. Cell Biosci 2023; 13:230. [PMID: 38124132 PMCID: PMC10734085 DOI: 10.1186/s13578-023-01188-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/16/2023] [Indexed: 12/23/2023] Open
Abstract
Type I interferon (IFN-I) plays crucial roles in the regulation of inflammation and it is associated with various inflammatory diseases including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and periodontitis, impacting people's health and quality of life. It is well-established that IFN-Is affect immune responses and inflammatory factors by regulating some signaling. However, currently, there is no comprehensive overview of the crucial regulatory role of IFN-I in distinctive pathways as well as associated inflammatory diseases. This review aims to provide a narrative of the involvement of IFN-I in different signaling pathways, mainly mediating the related key factors with specific targets in the pathways and signaling cascades to influence the progression of inflammatory diseases. As such, we suggested that IFN-Is induce inflammatory regulation through the stimulation of certain factors in signaling pathways, which displays possible efficient treatment methods and provides a reference for the precise control of inflammatory diseases.
Collapse
Affiliation(s)
- Ling Ji
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
| | - Tianle Li
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
| | - Huimin Chen
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
| | - Yanqi Yang
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
- Division of Pediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, China
| | - Jieying Liu
- Department of Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Qiao
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China.
- Applied Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Level 3, 34 Hospital Road, Sai Ying Pun, Hong Kong, SAR, People's Republic of China.
| | - Hui Chen
- Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong, SAR, People's Republic of China.
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Level 3, 34 Hospital Road, Sai Ying Pun, Hong Kong, SAR, People's Republic of China.
| |
Collapse
|
4
|
Lagumdzic E, Pernold CPS, Ertl R, Palmieri N, Stadler M, Sawyer S, Stas MR, Kreutzmann H, Rümenapf T, Ladinig A, Saalmüller A. Gene expression of peripheral blood mononuclear cells and CD8 + T cells from gilts after PRRSV infection. Front Immunol 2023; 14:1159970. [PMID: 37409113 PMCID: PMC10318438 DOI: 10.3389/fimmu.2023.1159970] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a positive-stranded RNA virus, which emerged in Europe and U.S.A. in the late 1980s and has since caused huge economic losses. Infection with PRRSV causes mild to severe respiratory and reproductive clinical symptoms in pigs. Alteration of the host immune response by PRRSV is associated with the increased susceptibility to secondary viral and bacterial infections resulting in more serious and chronic disease. However, the expression profiles underlying innate and adaptive immune responses to PRRSV infection are yet to be further elucidated. In this study, we investigated gene expression profiles of PBMCs and CD8+ T cells after PRRSV AUT15-33 infection. We identified the highest number of differentially expressed genes in PBMCs and CD8+ T cells at 7 dpi and 21 dpi, respectively. The gene expression profile of PBMCs from infected animals was dominated by a strong innate immune response at 7 dpi which persisted through 14 dpi and 21 dpi and was accompanied by involvement of adaptive immunity. The gene expression pattern of CD8+ T cells showed a strong adaptive immune response to PRRSV, leading to the formation of highly differentiated CD8+ T cells starting from 14 dpi. The hallmark of the CD8+ T-cell response was the increased expression of effector and cytolytic genes (PRF1, GZMA, GZMB, GZMK, KLRK1, KLRD1, FASL, NKG7), with the highest levels observed at 21 dpi. Temporal clustering analysis of DEGs of PBMCs and CD8+ T cells from PRRSV-infected animals revealed three and four clusters, respectively, suggesting tight transcriptional regulation of both the innate and the adaptive immune response to PRRSV. The main cluster of PBMCs was related to the innate immune response to PRRSV, while the main clusters of CD8+ T cells represented the initial transformation and differentiation of these cells in response to the PRRSV infection. Together, we provided extensive transcriptomics data explaining gene signatures of the immune response of PBMCs and CD8+ T cells after PRRSV infection. Additionally, our study provides potential biomarker targets useful for vaccine and therapeutics development.
Collapse
Affiliation(s)
- Emil Lagumdzic
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Clara P. S. Pernold
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Reinhard Ertl
- VetCore Facility for Research, University of Veterinary Medicine, Vienna, Austria
| | - Nicola Palmieri
- University Clinic for Poultry and Fish Medicine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Spencer Sawyer
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Melissa R. Stas
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Heinrich Kreutzmann
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Till Rümenapf
- Institute of Virology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| | - Andrea Ladinig
- University Clinic for Swine, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
5
|
Thorlacius GE, Björk A, Wahren-Herlenius M. Genetics and epigenetics of primary Sjögren syndrome: implications for future therapies. Nat Rev Rheumatol 2023; 19:288-306. [PMID: 36914790 PMCID: PMC10010657 DOI: 10.1038/s41584-023-00932-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2023] [Indexed: 03/14/2023]
Abstract
In primary Sjögren syndrome (pSS), chronic inflammation of exocrine glands results in tissue destruction and sicca symptoms, primarily of the mouth and eyes. Fatigue, arthralgia and myalgia are also common symptoms, whereas extraglandular manifestations that involve the respiratory, nervous and vascular systems occur in a subset of patients. The disease predominantly affects women, with an estimated female to male ratio of 14 to 1. The aetiology of pSS, however, remains incompletely understood, and effective treatment is lacking. Large-scale genetic and epigenetic investigations have revealed associations between pSS and genes in both innate and adaptive immune pathways. The genetic variants mediate context-dependent effects, and both sex and environmental factors can influence the outcome. As such, genetic and epigenetic studies can provide insight into the dysregulated molecular mechanisms, which in turn might reveal new therapeutic possibilities. This Review discusses the genetic and epigenetic features that have been robustly connected with pSS, putting them into the context of cellular function, carrier sex and environmental challenges. In all, the observations point to several novel opportunities for early detection, treatment development and the pathway towards personalized medicine.
Collapse
Affiliation(s)
- Gudny Ella Thorlacius
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Albin Björk
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Rheumatology, Academic Specialist Center, Stockholm, Sweden
| | - Marie Wahren-Herlenius
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
6
|
Yu Y, Li M, Zhao Y, Fan F, Wu W, Gao Y, Bai C. Immune cell-derived extracellular vesicular microRNAs induce pancreatic beta cell apoptosis. Heliyon 2022; 8:e11995. [PMID: 36561684 PMCID: PMC9763775 DOI: 10.1016/j.heliyon.2022.e11995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/01/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
Background Type 1 diabetes mellitus (T1DM) is an autoimmune disease caused by an autoimmune response against pancreatic islet β cells. Increasing evidence indicates that specific microRNAs (miRNAs) from immune cells extracellular vesicles are involved in islet β cells apoptosis. Methods In this study, the microarray datasets GSE27997 and GSE137637 were downloaded from the Gene Expression Omnibus (GEO) database. miRNAs that promote islet β cells apoptosis in T1DM were searched in PubMed. We used the FunRich tool to determine the miRNA expression in extracellular vesicles derived from immune cells associated with islet β cell apoptosis, of which we selected candidate miRNAs based on fold change expression. Potential upstream transcription factors and downstream target genes of candidate miRNAs were predicted using TransmiR V2.0 and starBase database, respectively. Results Candidate miRNAs expressed in extracellular vesicles derived from T cells, pro-inflammatory macrophages, B cells, and dendritic cells were analyzed to identify the miRNAs involved in β cells apoptosis. Based on these candidate miRNAs, 25 downstream candidate genes, which positively regulate β cell functions, were predicted and screened; 17 transcription factors that positively regulate the candidate miRNAs were also identified. Conclusions Our study demonstrated that immune cell-derived extracellular vesicular miRNAs could promote islet β cell dysfunction and apoptosis. Based on these findings, we have constructed a transcription factor-miRNA-gene regulatory network, which provides a theoretical basis for clinical management of T1DM. This study provides novel insights into the mechanism underlying immune cell-derived extracellular vesicle-mediated islet β cell apoptosis.
Collapse
Affiliation(s)
- Yueyang Yu
- Institute of Precision Medicine, Jining Medical University, Jining, Shandong 272067, PR China
| | - Mengyin Li
- Department of Otorhinolaryngology Head and Neck Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, 272067, PR China
| | - Yuxuan Zhao
- Institute of Precision Medicine, Jining Medical University, Jining, Shandong 272067, PR China
| | - Fangzhou Fan
- Institute of Precision Medicine, Jining Medical University, Jining, Shandong 272067, PR China
| | - Wenxiang Wu
- Institute of Precision Medicine, Jining Medical University, Jining, Shandong 272067, PR China
| | - Yuhua Gao
- Institute of Precision Medicine, Jining Medical University, Jining, Shandong 272067, PR China
- Corresponding author.
| | - Chunyu Bai
- Institute of Precision Medicine, Jining Medical University, Jining, Shandong 272067, PR China
- Corresponding author.
| |
Collapse
|
7
|
Pathogenic Biohacking: Induction, Modulation and Subversion of Host Transcriptional Responses by Listeria monocytogenes. Toxins (Basel) 2020; 12:toxins12050294. [PMID: 32380645 PMCID: PMC7290974 DOI: 10.3390/toxins12050294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 12/23/2022] Open
Abstract
During infection, the foodborne bacterial pathogen Listeria monocytogenes dynamically influences the gene expression profile of host cells. Infection-induced transcriptional changes are a typical feature of the host-response to bacteria and contribute to the activation of protective genes such as inflammatory cytokines. However, by using specialized virulence factors, bacterial pathogens can target signaling pathways, transcription factors, and epigenetic mechanisms to alter host gene expression, thereby reprogramming the response to infection. Therefore, the transcriptional profile that is established in the host is delicately balanced between antibacterial responses and pathogenesis, where any change in host gene expression might significantly influence the outcome of infection. In this review, we discuss the known transcriptional and epigenetic processes that are engaged during Listeria monocytogenes infection, the virulence factors that can remodel them, and the impact these processes have on the outcome of infection.
Collapse
|
8
|
Eshleman EM, Bortell N, McDermott DS, Crisler WJ, Lenz LL. Myeloid cell responsiveness to interferon-gamma is sufficient for initial resistance to Listeria monocytogenes. CURRENT RESEARCH IN IMMUNOLOGY 2020; 1:1-9. [PMID: 34337387 PMCID: PMC8323841 DOI: 10.1016/j.crimmu.2020.01.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The type II interferon (IFNγ) promotes resistance to intracellular pathogens. Most immune and somatic cells also express the IFNγ receptor (IFNGR) and respond to IFNγ. While myeloid cell have been implicated as important targets of IFNγ, it remains unknown if IFNγ signaling to myeloid cell types suffices for resistance to infection. Here, we addressed this question by generating mice in which IFNGR1 is selectively expressed by myeloid cells. These “MSGR1” (myeloid selective IFNGR1) mice express an epitope-tagged Ifngr1 transgene (fGR1) from the myeloid-specific c-fms promoter in a background lacking endogenous Ifngr1. IFNGR staining was selectively observed on myeloid cells in the MSGR1 mice and correlated with responsiveness of these cells to IFNγ. During systemic infection by the bacterium Listeria monocytogenes, activation marker staining was comparable on monocytes from MSGR1 and control B6 mice. Bacterial burdens and survival were also equivalent in MSGR1 and wildtype B6 animals at a timepoint when B6.Ifngr1−/− mice began to succumb. These data confirm that activation of inflammatory monocytes and neutrophils is a key mechanism by which IFNγ promotes innate anti-bacterial immunity and suggest that IFNγ targeting of myeloid cells is largely sufficient to mediate protection against systemic L. monocytogenes. Expression of IFNGR1 is restricted to monocytes and neutrophils in “MSGR1” (myeloid selective IFNGR1) mice. Myeloid cells from MSGR1 mice are responsive to IFNγ and show elevated activation compared to cells from B6.Ifngr1−/− mice. MSGR1 myeloid cells respond to Listeria monocytogenes infection and promote early resistance. IFNγ stimulation of myeloid cells can thus protect against infection independent of effects on other hematopoietic and non-hematopoietic cell populations. Particularly in female mice, IFNγ stimulation of non-myeloid cells may also contribute to improved survival.
Collapse
Affiliation(s)
- Emily M Eshleman
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Nikki Bortell
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Daniel S McDermott
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - William J Crisler
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Laurel L Lenz
- Immunology and Microbiology Department, University of Colorado School of Medicine, Aurora, CO 80045 USA
| |
Collapse
|
9
|
Yang C, McDermot DS, Pasricha S, Brown AS, Bedoui S, Lenz LL, van Driel IR, Hartland EL. IFNγ receptor down-regulation facilitates Legionella survival in alveolar macrophages. J Leukoc Biol 2020; 107:273-284. [PMID: 31793076 PMCID: PMC8015206 DOI: 10.1002/jlb.4ma1019-152r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/10/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022] Open
Abstract
Legionella pneumophila is an opportunistic human pathogen and causative agent of the acute pneumonia known as Legionnaire's disease. Upon inhalation, the bacteria replicate in alveolar macrophages (AM), within an intracellular vacuole termed the Legionella-containing vacuole. We recently found that, in vivo, IFNγ was required for optimal clearance of intracellular L. pneumophila by monocyte-derived cells (MC), but the cytokine did not appear to influence clearance by AM. Here, we report that during L. pneumophila lung infection, expression of the IFNγ receptor subunit 1 (IFNGR1) is down-regulated in AM and neutrophils, but not MC, offering a possible explanation for why AM are unable to effectively restrict L. pneumophila replication in vivo. To test this, we used mice that constitutively express IFNGR1 in AM and found that prevention of IFNGR1 down-regulation enhanced the ability of AM to restrict L. pneumophila intracellular replication. IFNGR1 down-regulation was independent of the type IV Dot/Icm secretion system of L. pneumophila indicating that bacterial effector proteins were not involved. In contrast to previous work, we found that signaling via type I IFN receptors was not required for IFNGR1 down-regulation in macrophages but rather that MyD88- or Trif- mediated NF-κB activation was required. This work has uncovered an alternative signaling pathway responsible for IFNGR1 down-regulation in macrophages during bacterial infection.
Collapse
Affiliation(s)
- Chao Yang
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Daniel S McDermot
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Shivani Pasricha
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
| | - Andrew S Brown
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Sammy Bedoui
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Laurel L Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Denver, Colorado, USA
| | - Ian R van Driel
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Elizabeth L Hartland
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Crisler WJ, Eshleman EM, Lenz LL. Ligand-induced IFNGR1 down-regulation calibrates myeloid cell IFNγ responsiveness. Life Sci Alliance 2019; 2:e201900447. [PMID: 31585982 PMCID: PMC6778285 DOI: 10.26508/lsa.201900447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/04/2023] Open
Abstract
The type II IFN (IFNγ) enhances antimicrobial activity yet also drives expression of genes that amplify inflammatory responses. Hence, excessive IFNγ stimulation can be pathogenic. Here, we describe a previously unappreciated mechanism whereby IFNγ itself dampens myeloid cell activation. Staining of monocytes from Listeria monocytogenes-infected mice provided evidence of type I IFN-independent reductions in IFNGR1. IFNγ was subsequently found to reduce surface IFNGR1 on cultured murine myeloid cells and human CD14+ peripheral blood mononuclear cells. IFNγ-driven reductions in IFNGR1 were not explained by ligand-induced receptor internalization. Rather, IFNγ reduced macrophage Ifngr1 transcription by altering chromatin structure at putative Ifngr1 enhancer sites. This is a distinct mechanism from that used by type I IFNs. Ligand-induced reductions in IFNGR1 altered myeloid cell sensitivity to IFNγ, blunting activation of STAT1 and 3. Our data, thus, reveal a mechanism by which IFNGR1 abundance and myeloid cell sensitivity to IFNγ can be modulated in the absence of type I IFNs. Multiple mechanisms, thus, exist to calibrate macrophage IFNGR1 abundance, likely permitting the fine tuning of macrophage activation and inflammation.
Collapse
Affiliation(s)
- William J Crisler
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Emily M Eshleman
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Laurel L Lenz
- Department of Immunology & Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
11
|
Ali S, Mann-Nüttel R, Schulze A, Richter L, Alferink J, Scheu S. Sources of Type I Interferons in Infectious Immunity: Plasmacytoid Dendritic Cells Not Always in the Driver's Seat. Front Immunol 2019; 10:778. [PMID: 31031767 PMCID: PMC6473462 DOI: 10.3389/fimmu.2019.00778] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/25/2019] [Indexed: 12/28/2022] Open
Abstract
Type I Interferons (IFNs) are hallmark cytokines produced in immune responses to all classes of pathogens. Type I IFNs can influence dendritic cell (DC) activation, maturation, migration, and survival, but also directly enhance natural killer (NK) and T/B cell activity, thus orchestrating various innate and adaptive immune effector functions. Therefore, type I IFNs have long been considered essential in the host defense against virus infections. More recently, it has become clear that depending on the type of virus and the course of infection, production of type I IFN can also lead to immunopathology or immunosuppression. Similarly, in bacterial infections type I IFN production is often associated with detrimental effects for the host. Although most cells in the body are thought to be able to produce type I IFN, plasmacytoid DCs (pDCs) have been termed the natural "IFN producing cells" due to their unique molecular adaptations to nucleic acid sensing and ability to produce high amounts of type I IFN. Findings from mouse reporter strains and depletion experiments in in vivo infection models have brought new insights and established that the role of pDCs in type I IFN production in vivo is less important than assumed. Production of type I IFN, especially the early synthesized IFNβ, is rather realized by a variety of cell types and cannot be mainly attributed to pDCs. Indeed, the cell populations responsible for type I IFN production vary with the type of pathogen, its tissue tropism, and the route of infection. In this review, we summarize recent findings from in vivo models on the cellular source of type I IFN in different infectious settings, ranging from virus, bacteria, and fungi to eukaryotic parasites. The implications from these findings for the development of new vaccination and therapeutic designs targeting the respectively defined cell types are discussed.
Collapse
Affiliation(s)
- Shafaqat Ali
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
- Cluster of Excellence EXC 1003, Cells in Motion, Münster, Germany
| | - Ritu Mann-Nüttel
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| | - Anja Schulze
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| | - Lisa Richter
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| | - Judith Alferink
- Cluster of Excellence EXC 1003, Cells in Motion, Münster, Germany
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
12
|
Sequential conditioning-stimulation reveals distinct gene- and stimulus-specific effects of Type I and II IFN on human macrophage functions. Sci Rep 2019; 9:5288. [PMID: 30918279 PMCID: PMC6437173 DOI: 10.1038/s41598-019-40503-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/11/2019] [Indexed: 12/21/2022] Open
Abstract
Macrophages orchestrate immune responses by sensing and responding to pathogen-associated molecules. These responses are modulated by prior conditioning with cytokines such as interferons (IFNs). Type I and II IFN have opposing functions in many biological scenarios, yet macrophages directly stimulated with Type I or II IFN activate highly overlapping gene expression programs. We hypothesized that a sequential conditioning-stimulation approach would reveal with greater specificity the differential effects of Type I and II IFN on human macrophages. By first conditioning with IFN then stimulating with toll-like receptor ligands and cytokines, followed by genome-wide RNA-seq analysis, we identified 713 genes whose expression was unaffected by IFN alone but showed potentiated or diminished responses to a stimulus after conditioning. For example, responses to the cytokine TNF were restricted by Type II IFN conditioning but potentiated by Type I IFN conditioning. We observed that the effects of IFN were not uniformly pro- or anti-inflammatory, but highly gene-specific and stimulus-specific. By assessing expression levels of key signal transducers and characterizing chromatin accessibility by ATAC-seq, we identify the likely molecular mechanisms underlying Type I and Type II-specific effects, distinguishing between modulation of cytoplasmic signaling networks and the nuclear epigenome that synergistically regulate macrophage immune responses.
Collapse
|
13
|
Acosta-Herrera M, Kerick M, González-Serna D, Wijmenga C, Franke A, Gregersen PK, Padyukov L, Worthington J, Vyse TJ, Alarcón-Riquelme ME, Mayes MD, Martin J. Genome-wide meta-analysis reveals shared new loci in systemic seropositive rheumatic diseases. Ann Rheum Dis 2019; 78:311-319. [PMID: 30573655 PMCID: PMC6800208 DOI: 10.1136/annrheumdis-2018-214127] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/18/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Immune-mediated inflammatory diseases (IMIDs) are heterogeneous and complex conditions with overlapping clinical symptoms and elevated familial aggregation, which suggests the existence of a shared genetic component. In order to identify this genetic background in a systematic fashion, we performed the first cross-disease genome-wide meta-analysis in systemic seropositive rheumatic diseases, namely, systemic sclerosis, systemic lupus erythematosus, rheumatoid arthritis and idiopathic inflammatory myopathies. METHODS We meta-analysed ~6.5 million single nucleotide polymorphisms in 11 678 cases and 19 704 non-affected controls of European descent populations. The functional roles of the associated variants were interrogated using publicly available databases. RESULTS Our analysis revealed five shared genome-wide significant independent loci that had not been previously associated with these diseases: NAB1, KPNA4-ARL14, DGQK, LIMK1 and PRR12. All of these loci are related with immune processes such as interferon and epidermal growth factor signalling, response to methotrexate, cytoskeleton dynamics and coagulation cascade. Remarkably, several of the associated loci are known key players in autoimmunity, which supports the validity of our results. All the associated variants showed significant functional enrichment in DNase hypersensitivity sites, chromatin states and histone marks in relevant immune cells, including shared expression quantitative trait loci. Additionally, our results were significantly enriched in drugs that are being tested for the treatment of the diseases under study. CONCLUSIONS We have identified shared new risk loci with functional value across diseases and pinpoint new potential candidate loci that could be further investigated. Our results highlight the potential of drug repositioning among related systemic seropositive rheumatic IMIDs.
Collapse
Affiliation(s)
| | - Martin Kerick
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS Granada, Granada, Spain
| | - David González-Serna
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS Granada, Granada, Spain
| | - Cisca Wijmenga
- Department of Genetics, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Peter K Gregersen
- Robert S Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Leonid Padyukov
- Rheumatology Unit, Department of Medicine, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Jane Worthington
- Manchester NIHR Biomedical Research Centre, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Timothy James Vyse
- Division of Genetics and Molecular Medicine, King's College London, London, UK
- Division of Immunology, Infection and Inflammatory Disease, King's College London, London, UK
| | - Marta Eugenia Alarcón-Riquelme
- Centre for Genomics and Oncological Research (GENYO), Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
| | - Maureen D Mayes
- Department of Internal Medicine, Division of Rheumatology, The University of Texas Health Science Center-Houston, Houston, Texas, USA
| | - Javier Martin
- Institute of Parasitology and Biomedicine López-Neyra, IPBLN-CSIC, PTS Granada, Granada, Spain
| |
Collapse
|
14
|
Li J, Wang J, Liu Y, Yang J, Guo L, Ren S, Chen Z, Liu Z, Zhang Y, Qiu W, Li Y, Zhang S, Yu J, Wu J. Porcine reproductive and respiratory syndrome virus NADC30-like strain accelerates Streptococcus suis serotype 2 infection in vivo and in vitro. Transbound Emerg Dis 2018; 66:729-742. [PMID: 30427126 DOI: 10.1111/tbed.13072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/24/2018] [Accepted: 11/03/2018] [Indexed: 12/27/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS), an economically significant pandemic disease, commonly results in increased impact of bacterial infections, including those by Streptococcus suis (S. suis). In recent years, PRRS virus (PRRSV) NADC30-like strain has emerged in different regions of China, and coinfected with S. suis and PRRSV has also gradually increased in clinical performance. However, the mechanisms involved in host innate responses towards S. suis and their implications of coinfection with NADC30-like strain remain unknown. Therefore, the pathogenicity of NADC30-like strain and S. suis serotype 2 (SS2) coinfection in vivo and in vitro was investigated in this study. The results showed that NADC30-like increased the invasion and proliferation of SS2 in blood and tissues, resulting in more severe pneumonia, myocarditis, and peritonitisas well as higher mortality rate in pigs. In vitro, NADC30-like strain increased the invasion and survival of SS2 in porcine alveolar macrophages (PAM) cells, causing more drastic expression of inflammatory cytokines and activation of NF-ĸB signalling. These results pave the way for understanding the interaction of S. suis with the swine immune system and their modulation in a viral coinfection.
Collapse
Affiliation(s)
- Jianda Li
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jinbao Wang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,School of Life Sciences, Shandong Normal University, Jinan, China.,School of Life Sciences, Shandong University, Jinan, China
| | - Yueyue Liu
- School of Life Sciences, Shandong University, Jinan, China
| | - Jie Yang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Lihui Guo
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Sufang Ren
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Zhi Chen
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Zhaoshan Liu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,School of Life Sciences, Shandong University, Jinan, China
| | - Yuyu Zhang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Wenbin Qiu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,School of Life Sciences, Shandong Normal University, Jinan, China
| | - Yubao Li
- School of Life Sciences, Liaocheng University, Liaocheng, China
| | - Shujin Zhang
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,School of Life Sciences, Liaocheng University, Liaocheng, China
| | - Jiang Yu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Jiaqiang Wu
- Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China.,School of Life Sciences, Shandong Normal University, Jinan, China.,School of Life Sciences, Shandong University, Jinan, China.,School of Life Sciences, Liaocheng University, Liaocheng, China
| |
Collapse
|
15
|
Márquez A, Kerick M, Zhernakova A, Gutierrez-Achury J, Chen WM, Onengut-Gumuscu S, González-Álvaro I, Rodriguez-Rodriguez L, Rios-Fernández R, González-Gay MA, Mayes MD, Raychaudhuri S, Rich SS, Wijmenga C, Martín J. Meta-analysis of Immunochip data of four autoimmune diseases reveals novel single-disease and cross-phenotype associations. Genome Med 2018; 10:97. [PMID: 30572963 PMCID: PMC6302306 DOI: 10.1186/s13073-018-0604-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/22/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In recent years, research has consistently proven the occurrence of genetic overlap across autoimmune diseases, which supports the existence of common pathogenic mechanisms in autoimmunity. The objective of this study was to further investigate this shared genetic component. METHODS For this purpose, we performed a cross-disease meta-analysis of Immunochip data from 37,159 patients diagnosed with a seropositive autoimmune disease (11,489 celiac disease (CeD), 15,523 rheumatoid arthritis (RA), 3477 systemic sclerosis (SSc), and 6670 type 1 diabetes (T1D)) and 22,308 healthy controls of European origin using the R package ASSET. RESULTS We identified 38 risk variants shared by at least two of the conditions analyzed, five of which represent new pleiotropic loci in autoimmunity. We also identified six novel genome-wide associations for the diseases studied. Cell-specific functional annotations and biological pathway enrichment analyses suggested that pleiotropic variants may act by deregulating gene expression in different subsets of T cells, especially Th17 and regulatory T cells. Finally, drug repositioning analysis evidenced several drugs that could represent promising candidates for CeD, RA, SSc, and T1D treatment. CONCLUSIONS In this study, we have been able to advance in the knowledge of the genetic overlap existing in autoimmunity, thus shedding light on common molecular mechanisms of disease and suggesting novel drug targets that could be explored for the treatment of the autoimmune diseases studied.
Collapse
Affiliation(s)
- Ana Márquez
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, PTS Granada, Granada, Spain
- Systemic Autoimmune Disease Unit, Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| | - Martin Kerick
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, PTS Granada, Granada, Spain
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Wei-Min Chen
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA USA
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA USA
| | | | | | - Raquel Rios-Fernández
- Systemic Autoimmune Diseases Unit, Complejo Hospitalario de Granada, Hospital Campus de la Salud, Granada, Spain
| | - Miguel A. González-Gay
- Epidemiology, Genetics and Atherosclerosis Research Group on Systemic Inflammatory Diseases, IDIVAL, Santander, Spain
| | - Maureen D. Mayes
- Division of Rheumatology and Clinical Immunogenetics, The University of Texas Health Science Center-Houston, Houston, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
- Division of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA USA
| | - Cisca Wijmenga
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Javier Martín
- Instituto de Parasitología y Biomedicina “López-Neyra”, CSIC, PTS Granada, Granada, Spain
| |
Collapse
|
16
|
Crisler WJ, Lenz LL. Crosstalk between type I and II interferons in regulation of myeloid cell responses during bacterial infection. Curr Opin Immunol 2018; 54:35-41. [PMID: 29886270 DOI: 10.1016/j.coi.2018.05.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 05/22/2018] [Accepted: 05/22/2018] [Indexed: 12/29/2022]
Abstract
Type I and type II interferons (IFNα/β and IFNγ) are cytokines that play indispensable roles in directing myeloid cell activity during inflammatory and immune responses. Each IFN type binds a distinct receptor (IFNAR or IFNGR) to transduce signals that reshape gene expression and function of myeloid and other cell types. In the context of murine models and human bacterial infections, production of IFNγ generally promotes resistance while production of IFNα/β is associated with increased host susceptibility. Here, we review mechanisms of crosstalk between type I and II IFNs in myeloid cells and their impact on myeloid cell activation and anti-microbial function.
Collapse
Affiliation(s)
- William J Crisler
- Department of Microbiology and Immunology, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Laurel L Lenz
- Department of Microbiology and Immunology, University of Colorado School of Medicine, Aurora, CO 80045, United States.
| |
Collapse
|
17
|
Londino JD, Gulick DL, Lear TB, Suber TL, Weathington NM, Masa LS, Chen BB, Mallampalli RK. Post-translational modification of the interferon-gamma receptor alters its stability and signaling. Biochem J 2017; 474:3543-3557. [PMID: 28883123 PMCID: PMC5967388 DOI: 10.1042/bcj20170548] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/28/2017] [Accepted: 09/05/2017] [Indexed: 12/28/2022]
Abstract
The IFN gamma receptor 1 (IFNGR1) binds IFN-γ and activates gene transcription pathways crucial for controlling bacterial and viral infections. Although decreases in IFNGR1 surface levels have been demonstrated to inhibit IFN-γ signaling, little is known regarding the molecular mechanisms controlling receptor stability. Here, we show in epithelial and monocytic cell lines that IFNGR1 displays K48 polyubiquitination, is proteasomally degraded, and harbors three ubiquitin acceptor sites at K277, K279, and K285. Inhibition of glycogen synthase kinase 3 beta (GSK3β) destabilized IFNGR1 while overexpression of GSK3β increased receptor stability. We identified critical serine and threonine residues juxtaposed to ubiquitin acceptor sites that impacted IFNGR1 stability. In CRISPR-Cas9 IFNGR1 generated knockout cell lines, cellular expression of IFNGR1 plasmids encoding ubiquitin acceptor site mutations demonstrated significantly impaired STAT1 phosphorylation and decreased STAT1-dependent gene induction. Thus, IFNGR1 undergoes rapid site-specific polyubiquitination, a process modulated by GSK3β. Ubiquitination appears to be necessary for efficient IFNGR1-dependent gamma gene induction and represents a relatively uncharacterized regulatory mechanism for this receptor.
Collapse
Affiliation(s)
- James D Londino
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Dexter L Gulick
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Travis B Lear
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Tomeka L Suber
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Nathaniel M Weathington
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Luke S Masa
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Bill B Chen
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A
| | - Rama K Mallampalli
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, U.S.A.
- Medical Specialty Service Line, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, U.S.A
- Department of Cell Biology and Physiology and Bioengineering, University of Pittsburgh, Pittsburgh, PA, U.S.A
| |
Collapse
|
18
|
Ye Z, Ren L, Tang Z, Deng Y, Xie X, Fu Z, Luo Z, Xu F, Zang N, Liu E. Pulmonary C-fiber degeneration downregulates IFN-γ receptor 1 via IFN-α induction to attenuate RSV-induced airway hyperresponsiveness. Virology 2017; 510:262-272. [PMID: 28772166 DOI: 10.1016/j.virol.2017.06.034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/25/2017] [Accepted: 06/29/2017] [Indexed: 12/19/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of respiratory infection in infants. Unfortunately, no effective vaccine or treatment against RSV is currently available. Pulmonary C-fibers (PCFs) are critical for regulating pulmonary inflammation and airway hyperresponsiveness (AHR). We previously reported that IFN-γ partially mediated RSV-induced airway disorders. In this study, we found that PCF degeneration alleviated RSV-induced airway inflammation, especially AHR by downregulating IFN-γ receptor 1 (IFNGR1), but had no effect on IFN-γ induction. In contrast, PCF degeneration actually increased IFN-α/β levels, as were the levels of STAT1 and phosphorylated STAT1 (pSTAT1). Exogenous IFN-α treatment induced STAT1 activation and downregulated IFNGR1 expression. These results suggest that PCFs affect IFNGR1 expression by inducing IFN-α to regulate IFN-γ-mediated airway inflammation and AHR. Thus, targeting PCFs activation may help control RSV-induced airway disorders, especially AHR, even with the presence of inflammation.
Collapse
Affiliation(s)
- Zhixu Ye
- Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, CSTC2009CA5002, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing 400014, China; Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Luo Ren
- Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, CSTC2009CA5002, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing 400014, China
| | - Zhengzhen Tang
- Ministry of Education Key Laboratory of Child Development and Disorders, Key Laboratory of Pediatrics in Chongqing, CSTC2009CA5002, Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing 400014, China; Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yu Deng
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Xiaohong Xie
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Zhou Fu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - ZhengXiu Luo
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Fadi Xu
- Pathophysiology Program, Lovelace Respiratory Research Institute, Albuquerque, NM, 87108, USA
| | - Na Zang
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| | - Enmei Liu
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.
| |
Collapse
|
19
|
Awuh JA, Flo TH. Molecular basis of mycobacterial survival in macrophages. Cell Mol Life Sci 2017; 74:1625-1648. [PMID: 27866220 PMCID: PMC11107535 DOI: 10.1007/s00018-016-2422-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/06/2016] [Accepted: 11/14/2016] [Indexed: 12/31/2022]
Abstract
Macrophages play an essential role in the immune system by ingesting and degrading invading pathogens, initiating an inflammatory response and instructing adaptive immune cells, and resolving inflammation to restore homeostasis. More interesting is the fact that some bacteria have evolved to use macrophages as a natural habitat and tools of spread in the host, e.g., Mycobacterium tuberculosis (Mtb) and some non-tuberculous mycobacteria (NTM). Mtb is considered one of humanity's most successful pathogens and is the causal agent of tuberculosis, while NTMs cause opportunistic infections all of which are of significant public health concern. Here, we describe mechanisms by which intracellular pathogens, with an emphasis on mycobacteria, manipulate macrophage functions to circumvent killing and live inside these cells even under considerable immunological pressure. Such macrophage functions include the selective evasion or engagement of pattern recognition receptors, production of cytokines, reactive oxygen and nitrogen species, phagosome maturation, as well as other killing mechanisms like autophagy and cell death. A clear understanding of host responses elicited by a specific pathogen and strategies employed by the microbe to evade or exploit these is of significant importance for the development of effective vaccines and targeted immunotherapy against persistent intracellular infections like tuberculosis.
Collapse
Affiliation(s)
- Jane Atesoh Awuh
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, PB 8905, 7491, Trondheim, Norway
| | - Trude Helen Flo
- Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, PB 8905, 7491, Trondheim, Norway.
| |
Collapse
|
20
|
Eshleman EM, Delgado C, Kearney SJ, Friedman RS, Lenz LL. Down regulation of macrophage IFNGR1 exacerbates systemic L. monocytogenes infection. PLoS Pathog 2017; 13:e1006388. [PMID: 28542482 PMCID: PMC5457163 DOI: 10.1371/journal.ppat.1006388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 06/02/2017] [Accepted: 04/27/2017] [Indexed: 12/21/2022] Open
Abstract
Interferons (IFNs) target macrophages to regulate inflammation and resistance to microbial infections. The type II IFN (IFNγ) acts on a cell surface receptor (IFNGR) to promote gene expression that enhance macrophage inflammatory and anti-microbial activity. Type I IFNs can dampen macrophage responsiveness to IFNγ and are associated with increased susceptibility to numerous bacterial infections. The precise mechanisms responsible for these effects remain unclear. Type I IFNs silence macrophage ifngr1 transcription and thus reduce cell surface expression of IFNGR1. To test how these events might impact macrophage activation and host resistance during bacterial infection, we developed transgenic mice that express a functional FLAG-tagged IFNGR1 (fGR1) driven by a macrophage-specific promoter. Macrophages from fGR1 mice expressed physiologic levels of cell surface IFNGR1 at steady state and responded equivalently to WT C57Bl/6 macrophages when treated with IFNγ alone. However, fGR1 macrophages retained cell surface IFNGR1 and showed enhanced responsiveness to IFNγ in the presence of type I IFNs. When fGR1 mice were infected with the bacterium Listeria monocytogenes their resistance was significantly increased, despite normal type I and II IFN production. Enhanced resistance was dependent on IFNγ and associated with increased macrophage activation and antimicrobial function. These results argue that down regulation of myeloid cell IFNGR1 is an important mechanism by which type I IFNs suppress inflammatory and anti-bacterial functions of macrophages.
Collapse
Affiliation(s)
- Emily M. Eshleman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO United States of America
| | - Christine Delgado
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO United States of America
| | - Staci J. Kearney
- Department of Biomedical Sciences, National Jewish Health, Denver, CO United States of America
| | - Rachel S. Friedman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO United States of America
- Department of Biomedical Sciences, National Jewish Health, Denver, CO United States of America
| | - Laurel L. Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO United States of America
- Department of Biomedical Sciences, National Jewish Health, Denver, CO United States of America
| |
Collapse
|
21
|
Snyder DT, Hedges JF, Jutila MA. Getting "Inside" Type I IFNs: Type I IFNs in Intracellular Bacterial Infections. J Immunol Res 2017; 2017:9361802. [PMID: 28529959 PMCID: PMC5424489 DOI: 10.1155/2017/9361802] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/23/2022] Open
Abstract
Type I interferons represent a unique and complex group of cytokines, serving many purposes during innate and adaptive immunity. Discovered in the context of viral infections, type I IFNs are now known to have myriad effects in infectious and autoimmune disease settings. Type I IFN signaling during bacterial infections is dependent on many factors including whether the infecting bacterium is intracellular or extracellular, as different signaling pathways are activated. As such, the repercussions of type I IFN induction can positively or negatively impact the disease outcome. This review focuses on type I IFN induction and downstream consequences during infection with the following intracellular bacteria: Chlamydia trachomatis, Listeria monocytogenes, Mycobacterium tuberculosis, Salmonella enterica serovar Typhimurium, Francisella tularensis, Brucella abortus, Legionella pneumophila, and Coxiella burnetii. Intracellular bacterial infections are unique because the bacteria must avoid, circumvent, and even co-opt microbial "sensing" mechanisms in order to reside and replicate within a host cell. Furthermore, life inside a host cell makes intracellular bacteria more difficult to target with antibiotics. Because type I IFNs are important immune effectors, modulating this pathway may improve disease outcomes. But first, it is critical to understand the context-dependent effects of the type I IFN pathway in intracellular bacterial infections.
Collapse
Affiliation(s)
- Deann T. Snyder
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Jodi F. Hedges
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - Mark A. Jutila
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| |
Collapse
|
22
|
Silva-Barrios S, Stäger S. Protozoan Parasites and Type I IFNs. Front Immunol 2017; 8:14. [PMID: 28154565 PMCID: PMC5243830 DOI: 10.3389/fimmu.2017.00014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/05/2017] [Indexed: 12/16/2022] Open
Abstract
For many years, the role of interferon (IFN)-I has been characterized primarily in the context of viral infections. However, regulatory functions mediated by IFN-I have also been described against bacterial infections and in tumor immunology. Only recently, the interest in understanding the immune functions mediated by IFN-I has dramatically increased in the field of protozoan infections. In this review, we discuss the discrete role of IFN-I in the immune response against major protozoan infections: Plasmodium, Leishmania, Trypanosoma, and Toxoplasma.
Collapse
Affiliation(s)
- Sasha Silva-Barrios
- INRS-Institut Armand Frappier, Center for Host-Parasite Interactions , Laval, QC , Canada
| | - Simona Stäger
- INRS-Institut Armand Frappier, Center for Host-Parasite Interactions , Laval, QC , Canada
| |
Collapse
|
23
|
Morita K, Okamura T, Sumitomo S, Iwasaki Y, Fujio K, Yamamoto K. Emerging roles of Egr2 and Egr3 in the control of systemic autoimmunity. Rheumatology (Oxford) 2016; 55:ii76-ii81. [DOI: 10.1093/rheumatology/kew342] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 08/23/2016] [Indexed: 01/04/2023] Open
|
24
|
Zibara K, Zeidan A, Bjeije H, Kassem N, Badran B, El-Zein N. ROS mediates interferon gamma induced phosphorylation of Src, through the Raf/ERK pathway, in MCF-7 human breast cancer cell line. J Cell Commun Signal 2016; 11:57-67. [PMID: 27838900 DOI: 10.1007/s12079-016-0362-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/03/2016] [Indexed: 01/05/2023] Open
Abstract
Interferon gamma (IFN-ɣ) is a pleiotropic cytokine which plays dual contrasting roles in cancer. Although IFN-ɣ has been clinically used to treat various malignancies, it was recently shown to have protumorigenic activities. Reactive oxygen species (ROS) are overproduced in cancer cells, mainly due to NADPH oxidase activity, which results into several changes in signaling pathways. In this study, we examined IFN-ɣ effect on the phosphorylation levels of key signaling proteins, through ROS production, in the human breast cancer cell line MCF-7. After treatment by IFN-ɣ, results showed a significant increase in the phosphorylation of STAT1, Src, raf, AKT, ERK1/2 and p38 signaling molecules, in a time specific manner. Src and Raf were found to be involved in early stages of IFN-ɣ signaling since their phosphorylation increased very rapidly. Selective inhibition of Src-family kinases resulted in an immediate significant decrease in the phosphorylation status of Raf and ERK1/2, but not p38 and AKT. On the other hand, IFN-ɣ resulted in ROS generation, through H2O2 production, whereas pre-treatment with the ROS inhibitor NAC caused ROS inhibition and a significant decrease in the phosphorylation levels of AKT, ERK1/2, p38 and STAT1. Moreover, pretreatment with a selective NOX1 inhibitor resulted in a significant decrease of AKT phosphorylation. Finally, no direct relationship was found between ROS production and calcium mobilization. In summary, IFN-ɣ signaling in MCF-7 cell line is ROS-dependent and follows the Src/Raf/ERK pathway whereas its signaling through the AKT pathway is highly dependent on NOX1.
Collapse
Affiliation(s)
- Kazem Zibara
- ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon.,Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Asad Zeidan
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Bjeije
- ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon
| | - Nouhad Kassem
- ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon
| | - Bassam Badran
- Biochemistry Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Nabil El-Zein
- ER045, PRASE, DSST, Lebanese University, Beirut, Lebanon. .,Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon.
| |
Collapse
|
25
|
Abstract
Type I interferons (IFNs) are pleiotropic cytokines well recognized for their role in the induction of a potent antiviral gene program essential for host defense against viruses. They also modulate innate and adaptive immune responses. However, the role of type I IFNs in host defense against bacterial infections is enigmatic. Depending on the bacterium, they exert seemingly opposite and capricious functions. In this review, we summarize the effect of type I IFNs on specific bacterial infections and highlight the effector mechanisms regulated by type I IFNs in an attempt to elucidate new avenues to understanding their role.
Collapse
Affiliation(s)
- Gayle M Boxx
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
26
|
Auray G, Lachance C, Wang Y, Gagnon CA, Segura M, Gottschalk M. Transcriptional Analysis of PRRSV-Infected Porcine Dendritic Cell Response to Streptococcus suis Infection Reveals Up-Regulation of Inflammatory-Related Genes Expression. PLoS One 2016; 11:e0156019. [PMID: 27213692 PMCID: PMC4877111 DOI: 10.1371/journal.pone.0156019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
The porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important swine pathogens and often serves as an entry door for other viral or bacterial pathogens, of which Streptococcus suis is one of the most common. Pre-infection with PRRSV leads to exacerbated disease caused by S. suis infection. Very few studies have assessed the immunological mechanisms underlying this higher susceptibility. Since antigen presenting cells play a major role in the initiation of the immune response, the in vitro transcriptional response of bone marrow-derived dendritic cells (BMDCs) and monocytes in the context of PRRSV and S. suis co-infection was investigated. BMDCs were found to be more permissive than monocytes to PRRSV infection; S. suis phagocytosis by PRRSV-infected BMDCs was found to be impaired, whereas no effect was found on bacterial intracellular survival. Transcription profile analysis, with a major focus on inflammatory genes, following S. suis infection, with and without pre-infection with PRRSV, was then performed. While PRRSV pre-infection had little effect on monocytes response to S. suis infection, a significant expression of several pro-inflammatory molecules was observed in BMDCs pre-infected with PRRSV after a subsequent infection with S. suis. While an additive effect could be observed for CCL4, CCL14, CCL20, and IL-15, a distinct synergistic up-regulatory effect was observed for IL-6, CCL5 and TNF-α after co-infection. This increased pro-inflammatory response by DCs could participate in the exacerbation of the disease observed during PRRSV and S. suis co-infection.
Collapse
Affiliation(s)
- Gaël Auray
- Groupe de recherche sur les maladies infectieuses en production animale (GREMIP), Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2
| | - Claude Lachance
- Groupe de recherche sur les maladies infectieuses en production animale (GREMIP), Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2
| | - Yingchao Wang
- Groupe de recherche sur les maladies infectieuses en production animale (GREMIP), Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2
| | - Carl A. Gagnon
- Groupe de recherche sur les maladies infectieuses en production animale (GREMIP), Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2
| | - Mariela Segura
- Groupe de recherche sur les maladies infectieuses en production animale (GREMIP), Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2
| | - Marcelo Gottschalk
- Groupe de recherche sur les maladies infectieuses en production animale (GREMIP), Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2
- * E-mail:
| |
Collapse
|
27
|
Lienard J, Movert E, Valfridsson C, Sturegård E, Carlsson F. ESX-1 exploits type I IFN-signalling to promote a regulatory macrophage phenotype refractory to IFNγ-mediated autophagy and growth restriction of intracellular mycobacteria. Cell Microbiol 2016; 18:1471-85. [PMID: 27062290 DOI: 10.1111/cmi.12594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 03/14/2016] [Accepted: 03/16/2016] [Indexed: 12/20/2022]
Abstract
The ability of macrophages to eradicate intracellular pathogens is normally greatly enhanced by IFNγ, a cytokine produced mainly after onset of adaptive immunity. However, adaptive immunity is unable to provide sterilizing immunity against mycobacteria, suggesting that mycobacteria have evolved virulence strategies to inhibit the bactericidal effect of IFNγ-signalling in macrophages. Still, the host-pathogen interactions and cellular mechanisms responsible for this feature have remained elusive. We demonstrate that the ESX-1 type VII secretion systems of Mycobacterium tuberculosis and Mycobacterium marinum exploit type I IFN-signalling to promote an IL-12(low) /IL-10(high) regulatory macrophage phenotype characterized by secretion of IL-10, IL-27 and IL-6. This mechanism had no impact on intracellular growth in the absence of IFNγ but suppressed IFNγ-mediated autophagy and growth restriction, indicating that the regulatory phenotype extends to function. The IFNγ-refractory phenotype was partly mediated by IL-27-signalling, establishing functional relevance for this downstream cytokine. These findings identify a novel macrophage-modulating function for the ESX-1 secretion system that may contribute to suppress the efficacy of adaptive immunity and provide mechanistic insight into the antagonistic cross talk between type I IFNs and IFNγ in mycobacterial infection.
Collapse
Affiliation(s)
- Julia Lienard
- Section for Immunology, Department of Experimental Medical Science, Lund University, BMC D14, 221 84, Lund, Sweden
| | - Elin Movert
- Section for Immunology, Department of Experimental Medical Science, Lund University, BMC D14, 221 84, Lund, Sweden
| | - Christine Valfridsson
- Section for Immunology, Department of Experimental Medical Science, Lund University, BMC D14, 221 84, Lund, Sweden
| | - Erik Sturegård
- Section for Medical Microbiology, Department of Laboratory Medicine, Lund University, Jan Waldenströms gata 59, 205 02, Malmö, Sweden
| | - Fredric Carlsson
- Section for Immunology, Department of Experimental Medical Science, Lund University, BMC D14, 221 84, Lund, Sweden.
| |
Collapse
|
28
|
Pitts MG, Myers-Morales T, D'Orazio SEF. Type I IFN Does Not Promote Susceptibility to Foodborne Listeria monocytogenes. THE JOURNAL OF IMMUNOLOGY 2016; 196:3109-16. [PMID: 26895837 DOI: 10.4049/jimmunol.1502192] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 01/20/2016] [Indexed: 01/01/2023]
Abstract
Type I IFN (IFN-α/β) is thought to enhance growth of the foodborne intracellular pathogen Listeria monocytogenes by promoting mechanisms that dampen innate immunity to infection. However, the type I IFN response has been studied primarily using methods that bypass the stomach and, therefore, fail to replicate the natural course of L. monocytogenes infection. In this study, we compared i.v. and foodborne transmission of L. monocytogenes in mice lacking the common type I IFN receptor (IFNAR1(-/-)). Contrary to what was observed using i.v. infection, IFNAR1(-/-) and wild-type mice had similar bacterial burdens in the liver and spleen following foodborne infection. Splenocytes from wild-type mice infected i.v. produced significantly more IFN-β than did those infected by the foodborne route. Consequently, the immunosuppressive effects of type I IFN signaling, which included T cell death, increased IL-10 secretion, and repression of neutrophil recruitment to the spleen, were all observed following i.v. but not foodborne transmission of L. monocytogenes. Type I IFN was also previously shown to cause a loss of responsiveness to IFN-γ through downregulation of the IFN-γ receptor α-chain on macrophages and dendritic cells. However, we detected a decrease in surface expression of IFN-γ receptor α-chain even in the absence of IFN-α/β signaling, suggesting that in vivo, this infection-induced phenotype is not type I IFN-dependent. These results highlight the importance of using the natural route of infection for studies of host-pathogen interactions and suggest that the detrimental effects of IFN-α/β signaling on the innate immune response to L. monocytogenes may be an artifact of the i.v. infection model.
Collapse
Affiliation(s)
- Michelle G Pitts
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536
| | - Tanya Myers-Morales
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536
| | - Sarah E F D'Orazio
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
29
|
Stifter SA, Bhattacharyya N, Pillay R, Flórido M, Triccas JA, Britton WJ, Feng CG. Functional Interplay between Type I and II Interferons Is Essential to Limit Influenza A Virus-Induced Tissue Inflammation. PLoS Pathog 2016; 12:e1005378. [PMID: 26731100 PMCID: PMC4701664 DOI: 10.1371/journal.ppat.1005378] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 12/09/2015] [Indexed: 01/28/2023] Open
Abstract
Host control of influenza A virus (IAV) is associated with exuberant pulmonary inflammation characterized by the influx of myeloid cells and production of proinflammatory cytokines including interferons (IFNs). It is unclear, however, how the immune system clears the virus without causing lethal immunopathology. Here, we demonstrate that in addition to its known anti-viral activity, STAT1 signaling coordinates host inflammation during IAV infection in mice. This regulatory mechanism is dependent on both type I IFN and IFN-γ receptor signaling and, importantly, requires the functional interplay between the two pathways. The protective function of type I IFNs is associated with not only the recruitment of classical inflammatory Ly6Chi monocytes into IAV-infected lungs, but also the prevention of excessive monocyte activation by IFN-γ. Unexpectedly, type I IFNs preferentially regulate IFN-γ signaling in Ly6Clo rather than inflammatory Ly6Chi mononuclear cell populations. In the absence of type I IFN signaling, Ly6Clo monocytes/macrophages, become phenotypically and functionally more proinflammatory than Ly6Chi cells, revealing an unanticipated function of the Ly6Clo mononuclear cell subset in tissue inflammation. In addition, we show that type I IFNs employ distinct mechanisms to regulate monocyte and neutrophil trafficking. Type I IFN signaling is necessary, but not sufficient, for preventing neutrophil recruitment into the lungs of IAV-infected mice. Instead, the cooperation of type I IFNs and lymphocyte-produced IFN-γ is required to regulate the tissue neutrophilic response to IAV. Our study demonstrates that IFN interplay links innate and adaptive anti-viral immunity to orchestrate tissue inflammation and reveals an additional level of complexity for IFN-dependent regulatory mechanisms that function to prevent excessive immunopathology while preserving anti-microbial functions. Influenza A virus (IAV) is a leading cause of respiratory infection and induces a strong acute inflammation manifested by the recruitment of monocytes and neutrophils as well as the production of proinflammatory cytokines in infected lungs. The interferons (IFNs) are strongly induced by IAV and are known to mediate host resistance to the infection. However, in contrast to their well-studied inhibitory effect on viral replication, the effects of IFNs on host inflammatory responses are less well understood. In this manuscript, we demonstrate that anti-viral IFN signaling is also required for the orchestration of a tissue response associated with the protection against IAV infection in mice. Importantly, we identify that type I IFNs cross-regulate and cooperate with IFN-γ to inhibit monocyte activation and neutrophil infiltration, respectively. This study also demonstrates that Ly6Clo monocytes/macrophages can potentially mediate influenza virus-induced inflammation, suggesting that IFNs dictate the homeostasis versus inflammatory function of mononuclear phagocytes in viral infection. Our study reveals a novel IFN-dependent regulatory mechanism designed to prevent the excessive immunopathology while preserving its anti-microbial functions. Moreover, these observations have particular relevance for understanding the mechanisms underlying the strong inflammatory response associated with lethal IAV strains and have implications for the development of new immunotherapies to treat influenza.
Collapse
Affiliation(s)
- Sebastian A. Stifter
- Immunology and Host Defense Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- Mycobacterial Research Program, The Centenary Institute, Camperdown, New South Wales, Australia
| | - Nayan Bhattacharyya
- Immunology and Host Defense Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Roman Pillay
- Immunology and Host Defense Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Manuela Flórido
- Mycobacterial Research Program, The Centenary Institute, Camperdown, New South Wales, Australia
| | - James A. Triccas
- Mycobacterial Research Program, The Centenary Institute, Camperdown, New South Wales, Australia
- Microbial Pathogenesis and Immunity Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Warwick J. Britton
- Mycobacterial Research Program, The Centenary Institute, Camperdown, New South Wales, Australia
- Discipline of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- Department of Medicine, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Carl G. Feng
- Immunology and Host Defense Group, Discipline of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- Mycobacterial Research Program, The Centenary Institute, Camperdown, New South Wales, Australia
- * E-mail:
| |
Collapse
|
30
|
Robinson MW, Aranday‐Cortes E, Gatherer D, Swann R, Liefhebber JMP, Filipe ADS, Sigruener A, Barclay ST, Mills PR, Patel AH, McLauchlan J. Viral genotype correlates with distinct liver gene transcription signatures in chronic hepatitis C virus infection. Liver Int 2015; 35:2256-64. [PMID: 25800823 PMCID: PMC4949513 DOI: 10.1111/liv.12830] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/16/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Chronic hepatitis C virus (HCV) infection of the liver with either genotype 1 or genotype 3 gives rise to distinct pathologies, and the two viral genotypes respond differently to antiviral therapy. METHODS To understand these clinical differences, we compared gene transcription profiles in liver biopsies from patients infected with either gt1 or gt3, and uninfected controls. RESULTS Gt1-infected biopsies displayed elevated levels of transcripts regulated by type I and type III interferons (IFN), including genes that predict response to IFN-α therapy. In contrast, genes controlled by IFN-γ were induced in gt3-infected biopsies. Moreover, IFN-γ levels were higher in gt3-infected biopsies. Analysis of hepatocyte-derived cell lines confirmed that the genes upregulated in gt3 infection were preferentially induced by IFN-γ. The transcriptional profile of gt3 infection was unaffected by IFNL4 polymorphisms, providing a rationale for the reduced predictive power of IFNL genotyping in gt3-infected patients. CONCLUSIONS The interactions between HCV genotypes 1 and 3 and hepatocytes are distinct. These unique interactions provide avenues to explore the biological mechanisms that drive viral genotype-specific differences in disease progression and treatment response. A greater understanding of the distinct host-pathogen interactions of the different HCV genotypes is required to facilitate optimal management of HCV infection.
Collapse
Affiliation(s)
- Mark W. Robinson
- MRC – University of Glasgow Centre for Virus ResearchGlasgowUK
- School of Biochemistry and ImmunologyTrinity College DublinDublinIreland
| | | | - Derek Gatherer
- MRC – University of Glasgow Centre for Virus ResearchGlasgowUK
- Division of Biomedical and Life SciencesLancaster UniversityLancasterUK
| | - Rachael Swann
- MRC – University of Glasgow Centre for Virus ResearchGlasgowUK
- Gartnavel General HospitalNHS Greater Glasgow and ClydeGlasgowUK
| | | | | | - Alex Sigruener
- Institute of Clinical Chemistry and Laboratory MedicineRegensburg University Medical CenterRegensburgGermany
| | | | - Peter R. Mills
- Gartnavel General HospitalNHS Greater Glasgow and ClydeGlasgowUK
| | - Arvind H. Patel
- MRC – University of Glasgow Centre for Virus ResearchGlasgowUK
| | - John McLauchlan
- MRC – University of Glasgow Centre for Virus ResearchGlasgowUK
| |
Collapse
|
31
|
Stifter SA, Feng CG. Interfering with immunity: detrimental role of type I IFNs during infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:2455-65. [PMID: 25747907 DOI: 10.4049/jimmunol.1402794] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type I IFNs are known to inhibit viral replication and mediate protection against viral infection. However, recent studies revealed that these cytokines play a broader and more fundamental role in host responses to infections beyond their well-established antiviral function. Type I IFN induction, often associated with microbial evasion mechanisms unique to virulent microorganisms, is now shown to increase host susceptibility to a diverse range of pathogens, including some viruses. This article presents an overview of the role of type I IFNs in infections with bacterial, fungal, parasitic, and viral pathogens and discusses the key mechanisms mediating the regulatory function of type I IFNs in pathogen clearance and tissue inflammation.
Collapse
Affiliation(s)
- Sebastian A Stifter
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney 2006, New South Wales, Australia; and Mycobacterial Research Program, Centenary Institute, Sydney 2050, New South Wales, Australia
| | - Carl G Feng
- Immunology and Host Defense Group, Department of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney 2006, New South Wales, Australia; and Mycobacterial Research Program, Centenary Institute, Sydney 2050, New South Wales, Australia
| |
Collapse
|
32
|
Abstract
The pathogenesis of Alzheimer's disease (AD) is a critical unsolved question; and although recent studies have demonstrated a strong association between altered brain immune responses and disease progression, the mechanistic cause of neuronal dysfunction and death is unknown. We have previously described the unique CVN-AD mouse model of AD, in which immune-mediated nitric oxide is lowered to mimic human levels, resulting in a mouse model that demonstrates the cardinal features of AD, including amyloid deposition, hyperphosphorylated and aggregated tau, behavioral changes, and age-dependent hippocampal neuronal loss. Using this mouse model, we studied longitudinal changes in brain immunity in relation to neuronal loss and, contrary to the predominant view that AD pathology is driven by proinflammatory factors, we find that the pathology in CVN-AD mice is driven by local immune suppression. Areas of hippocampal neuronal death are associated with the presence of immunosuppressive CD11c(+) microglia and extracellular arginase, resulting in arginine catabolism and reduced levels of total brain arginine. Pharmacologic disruption of the arginine utilization pathway by an inhibitor of arginase and ornithine decarboxylase protected the mice from AD-like pathology and significantly decreased CD11c expression. Our findings strongly implicate local immune-mediated amino acid catabolism as a novel and potentially critical mechanism mediating the age-dependent and regional loss of neurons in humans with AD.
Collapse
|
33
|
McNab F, Mayer-Barber K, Sher A, Wack A, O'Garra A. Type I interferons in infectious disease. Nat Rev Immunol 2015; 15:87-103. [PMID: 25614319 DOI: 10.1038/nri3787] [Citation(s) in RCA: 1829] [Impact Index Per Article: 182.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Type I interferons (IFNs) have diverse effects on innate and adaptive immune cells during infection with viruses, bacteria, parasites and fungi, directly and/or indirectly through the induction of other mediators. Type I IFNs are important for host defence against viruses. However, recently, they have been shown to cause immunopathology in some acute viral infections, such as influenza virus infection. Conversely, they can lead to immunosuppression during chronic viral infections, such as lymphocytic choriomeningitis virus infection. During bacterial infections, low levels of type I IFNs may be required at an early stage, to initiate cell-mediated immune responses. High concentrations of type I IFNs may block B cell responses or lead to the production of immunosuppressive molecules, and such concentrations also reduce the responsiveness of macrophages to activation by IFNγ, as has been shown for infections with Listeria monocytogenes and Mycobacterium tuberculosis. Recent studies in experimental models of tuberculosis have demonstrated that prostaglandin E2 and interleukin-1 inhibit type I IFN expression and its downstream effects, demonstrating that a cross-regulatory network of cytokines operates during infectious diseases to provide protection with minimum damage to the host.
Collapse
Affiliation(s)
- Finlay McNab
- 1] Allergic Inflammation Discovery Performance Unit, Respiratory Disease Respiratory Research and Development, GlaxoSmithKline, Stevenage, Hertfordshire SG1 2NY, UK. [2] Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Katrin Mayer-Barber
- Immunobiology Section, Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases (LPD), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland 20892, USA
| | - Andreas Wack
- Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Anne O'Garra
- 1] Division of Immunoregulation, Medical Research Council (MRC) National Institute for Medical Research, Mill Hill, London NW7 1AA, UK. [2] National Heart and Lung Institute (NHLI), Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
34
|
Davidson S, Maini MK, Wack A. Disease-promoting effects of type I interferons in viral, bacterial, and coinfections. J Interferon Cytokine Res 2015; 35:252-64. [PMID: 25714109 PMCID: PMC4389918 DOI: 10.1089/jir.2014.0227] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
While type I interferons (IFNs) are universally acknowledged for their antiviral and immunostimulatory functions, there is increasing appreciation of the detrimental effects of inappropriate, excessive, or mistimed type I IFN responses in viral and bacterial infections. The underlying mechanisms by which type I IFNs promote susceptibility or severity include direct tissue damage by apoptosis induction or suppression of proliferation in tissue cells, immunopathology due to excessive inflammation, and cell death induced by TRAIL- and Fas-expressing immune cells, as well as immunosuppression through IL-10, IL-27, PD-L1, IL-1Ra, and other regulatory molecules that antagonize the induction or action of IL-1, IL-12, IL-17, IFN-γ, KC, and other effectors of the immune response. Bacterial superinfections following influenza infection are a prominent example of a situation where type I IFNs can misdirect the immune response. This review discusses current understanding of the parameters of signal strength, duration, timing, location, and cellular recipients that determine whether type I IFNs have beneficial or detrimental effects in infection.
Collapse
Affiliation(s)
- Sophia Davidson
- 1 Division of Immunoregulation, MRC National Institute for Medical Research , Mill Hill, London, United Kingdom
| | | | | |
Collapse
|
35
|
Zdrenghea MT, Makrinioti H, Muresan A, Johnston SL, Stanciu LA. The role of macrophage IL-10/innate IFN interplay during virus-induced asthma. Rev Med Virol 2014; 25:33-49. [PMID: 25430775 PMCID: PMC4316183 DOI: 10.1002/rmv.1817] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/25/2014] [Accepted: 10/14/2014] [Indexed: 12/18/2022]
Abstract
Activation through different signaling pathways results in two functionally different types of macrophages, the pro-inflammatory (M1) and the anti-inflammatory (M2). The polarization of macrophages toward the pro-inflammatory M1 phenotype is considered to be critical for efficient antiviral immune responses in the lung. Among the various cell types that are present in the asthmatic airways, macrophages have emerged as significant participants in disease pathogenesis, because of their activation during both the inflammatory and resolution phases, with an impact on disease progression. Polarized M1 and M2 macrophages are able to reversibly undergo functional redifferentiation into anti-inflammatory or pro-inflammatory macrophages, respectively, and therefore, macrophages mediate both processes. Recent studies have indicated a predominance of M2 macrophages in asthmatic airways. During a virus infection, it is likely that M2 macrophages would secrete higher amounts of the suppressor cytokine IL-10, and less innate IFNs. However, the interactions between IL-10 and innate IFNs during virus-induced exacerbations of asthma have not been well studied. The possible role of IL-10 as a therapy in allergic asthma has already been suggested, but the divergent roles of this suppressor molecule in the antiviral immune response raise concerns. This review attempts to shed light on macrophage IL-10-IFNs interactions and discusses the role of IL-10 in virus-induced asthma exacerbations. Whereas IL-10 is important in terminating pro-inflammatory and antiviral immune responses, the presence of this immune regulatory cytokine at the beginning of virus infection could impair the response to viruses and play a role in virus-induced asthma exacerbations.
Collapse
Affiliation(s)
- Mihnea T Zdrenghea
- Ion Chiricuta Oncology InstituteCluj-Napoca, Romania
- Iuliu Hatieganu, University of Medicine and PharmacyCluj-Napoca, Romania
| | - Heidi Makrinioti
- Airways Disease Infection Section, National Heart and Lung Institute, Imperial College LondonLondon, UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of AsthmaLondon, UK
- Centre for Respiratory InfectionsLondon, UK
| | - Adriana Muresan
- Iuliu Hatieganu, University of Medicine and PharmacyCluj-Napoca, Romania
| | - Sebastian L Johnston
- Airways Disease Infection Section, National Heart and Lung Institute, Imperial College LondonLondon, UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of AsthmaLondon, UK
- Centre for Respiratory InfectionsLondon, UK
| | - Luminita A Stanciu
- Iuliu Hatieganu, University of Medicine and PharmacyCluj-Napoca, Romania
- Airways Disease Infection Section, National Heart and Lung Institute, Imperial College LondonLondon, UK
- Medical Research Council and Asthma UK Centre in Allergic Mechanisms of AsthmaLondon, UK
- Centre for Respiratory InfectionsLondon, UK
- *
Correspondence to: Dr. L. A. Stanciu, MD, PhD, Airway Disease Infection Section, Imperial College London, London, UK., E-mail:
| |
Collapse
|
36
|
Eshleman EM, Lenz LL. Type I interferons in bacterial infections: taming of myeloid cells and possible implications for autoimmunity. Front Immunol 2014; 5:431. [PMID: 25309533 PMCID: PMC4161047 DOI: 10.3389/fimmu.2014.00431] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 08/23/2014] [Indexed: 01/13/2023] Open
Abstract
Type I interferons (IFNs) were first described for their ability to protect the host from viral infections and may also have beneficial effects under specific conditions within some bacterial infections. Yet, these pleiotropic cytokines are now known to exacerbate infections by numerous life-threatening bacteria, including the intracellular pathogens Listeria monocytogenes and Mycobacterium tuberculosis. The evidence that such detrimental effects occur during bacterial infections in both animals and humans argues for selective pressure. In this review, we summarize the evidence demonstrating a pro-bacterial role for type I IFNs and discuss possible mechanisms that have been proposed to explain such effects. The theme emerges that type I IFNs act to suppress myeloid cell immune responses. The evolutionary conservation of such anti-inflammatory effects, particularly in the context of infections, suggests they may be important for limiting chronic inflammation. Given the effectiveness of type I IFNs in treatment of certain autoimmune diseases, their production may also act to raise the threshold for activation of immune responses to self-antigens.
Collapse
Affiliation(s)
- Emily M Eshleman
- Department of Immunology and Microbiology, University of Colorado School of Medicine , Aurora, CO , USA
| | - Laurel L Lenz
- Department of Immunology and Microbiology, University of Colorado School of Medicine , Aurora, CO , USA ; Department of Biomedical Research, National Jewish Health , Denver, CO , USA
| |
Collapse
|
37
|
McNab FW, Ewbank J, Howes A, Moreira-Teixeira L, Martirosyan A, Ghilardi N, Saraiva M, O'Garra A. Type I IFN induces IL-10 production in an IL-27-independent manner and blocks responsiveness to IFN-γ for production of IL-12 and bacterial killing in Mycobacterium tuberculosis-infected macrophages. THE JOURNAL OF IMMUNOLOGY 2014; 193:3600-12. [PMID: 25187652 PMCID: PMC4170673 DOI: 10.4049/jimmunol.1401088] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tuberculosis, caused by the intracellular bacterium Mycobacterium tuberculosis, currently causes ∼1.4 million deaths per year, and it therefore remains a leading global health problem. The immune response during tuberculosis remains incompletely understood, particularly regarding immune factors that are harmful rather than protective to the host. Overproduction of the type I IFN family of cytokines is associated with exacerbated tuberculosis in both mouse models and in humans, although the mechanisms by which type I IFN promotes disease are not well understood. We have investigated the effect of type I IFN on M. tuberculosis–infected macrophages and found that production of host-protective cytokines such as TNF-α, IL-12, and IL-1β is inhibited by exogenous type I IFN, whereas production of immunosuppressive IL-10 is promoted in an IL-27–independent manner. Furthermore, much of the ability of type I IFN to inhibit cytokine production was mediated by IL-10. Additionally, type I IFN compromised macrophage activation by the lymphoid immune response through severely disrupting responsiveness to IFN-γ, including M. tuberculosis killing. These findings describe important mechanisms by which type I IFN inhibits the immune response during tuberculosis.
Collapse
Affiliation(s)
- Finlay W McNab
- Division of Immunoregulation, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom;
| | - John Ewbank
- Division of Immunoregulation, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Ashleigh Howes
- Division of Immunoregulation, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Lucia Moreira-Teixeira
- Division of Immunoregulation, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom; Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal; Life and Health Sciences Research Institute and Biomaterials, Biodegradables and Biomimetics Research Group, Portugal Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal; and
| | - Anna Martirosyan
- Division of Immunoregulation, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom
| | - Nico Ghilardi
- Department of Immunology, Genentech, Inc., South San Francisco, CA 94080
| | - Margarida Saraiva
- Life and Health Sciences Research Institute, School of Health Sciences, University of Minho, 4710-057 Braga, Portugal; Life and Health Sciences Research Institute and Biomaterials, Biodegradables and Biomimetics Research Group, Portugal Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal; and
| | - Anne O'Garra
- Division of Immunoregulation, Medical Research Council National Institute for Medical Research, London NW7 1AA, United Kingdom
| |
Collapse
|
38
|
Dussurget O, Bierne H, Cossart P. The bacterial pathogen Listeria monocytogenes and the interferon family: type I, type II and type III interferons. Front Cell Infect Microbiol 2014; 4:50. [PMID: 24809023 PMCID: PMC4009421 DOI: 10.3389/fcimb.2014.00050] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/04/2014] [Indexed: 12/14/2022] Open
Abstract
Interferons (IFNs) are secreted proteins of the cytokine family that regulate innate and adaptive immune responses to infection. Although the importance of IFNs in the antiviral response has long been appreciated, their role in bacterial infections is more complex and is currently a major focus of investigation. This review summarizes our current knowledge of the role of these cytokines in host defense against the bacterial pathogen Listeria monocytogenes and highlights recent discoveries on the molecular mechanisms evolved by this intracellular bacterium to subvert IFN responses.
Collapse
Affiliation(s)
- Olivier Dussurget
- Unité des Interactions Bactéries-Cellules, Institut PasteurParis, France
- Inserm, U604Paris, France
- INRA, USC2020Paris, France
- University of Paris Diderot, Sorbonne Paris CitéParis, France
| | - Hélène Bierne
- Unité des Interactions Bactéries-Cellules, Institut PasteurParis, France
- Inserm, U604Paris, France
- INRA, USC2020Paris, France
| | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Institut PasteurParis, France
- Inserm, U604Paris, France
- INRA, USC2020Paris, France
| |
Collapse
|