1
|
Piccaro G, Aquino G, Gigantino V, Tirelli V, Sanchez M, Iorio E, Matarese G, Cassone A, Palma C. Mycobacterium tuberculosis antigen 85B modifies BCG-induced antituberculosis immunity and favors pathogen survival. J Leukoc Biol 2024; 115:1053-1069. [PMID: 38242866 DOI: 10.1093/jleuko/qiae014] [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: 04/20/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/21/2024] Open
Abstract
Tuberculosis is one of the deadliest infectious diseases worldwide. Mycobacterium tuberculosis has developed strategies not only to evade host immunity but also to manipulate it for its survival. We investigated whether Mycobacterium tuberculosis exploited the immunogenicity of Ag85B, one of its major secretory proteins, to redirect host antituberculosis immunity to its advantage. We found that administration of Ag85B protein to mice vaccinated with Bacillus Calmette-Guérin impaired the protection elicited by vaccination, causing a more severe infection when mice were challenged with Mycobacterium tuberculosis. Ag85B administration reduced Bacillus Calmette-Guérin-induced CD4 T-cell activation and IFN-γ, CCL-4, and IL-22 production in response to Mycobacterium tuberculosis-infected cells. On the other hand, it promoted robust Ag85B-responsive IFN-γ-producing CD4 T cells, expansion of a subset of IFN-γ/IL-10-producing CD4+FOXP3+Treg cells, differential activation of IL-17/IL-22 responses, and activation of regulatory and exhaustion pathways, including programmed death ligand 1 expression on macrophages. All this resulted in impaired intracellular Mycobacterium tuberculosis growth control by systemic immunity, both before and after the Mycobacterium tuberculosis challenge. Interestingly, Mycobacterium tuberculosis infection itself generated Ag85B-reactive inflammatory immune cells incapable of clearing Mycobacterium tuberculosis in both unvaccinated and Bacillus Calmette-Guérin-vaccinated mice. Our data suggest that Mycobacterium tuberculosis can exploit the strong immunogenicity of Ag85B to promote its own survival and spread. Since Ag85B is normally secreted by replicating bacteria and is commonly found in the lungs of the Mycobacterium tuberculosis-infected host, our findings may advance the understanding on the mechanisms of Mycobacterium tuberculosis pathogenesis and immune evasion.
Collapse
Affiliation(s)
- Giovanni Piccaro
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Gabriella Aquino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, Via Mariano Semmola 53, 80131 Naples, Italy
| | - Valentina Tirelli
- Core Facilities-Flow Cytometry Area, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Massimo Sanchez
- Core Facilities-Flow Cytometry Area, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Egidio Iorio
- Core Facilities-High Resolution NMR Unit, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie mediche, Università di Napoli "Federico II," Via Sergio Pansini 5, 80131 Naples, Italy
| | - Antonio Cassone
- Polo d'innovazione della Genomica, Genetica e Biologia, Via Fiorentina 1, 53100 Siena, Italy
| | - Carla Palma
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
2
|
Yu F, Zhu X, Li Q, Xu W, Gao Y, Wen Y, Zhang Q, Dou J. Elevated IL-35 level and iTr35 subset increase the bacterial burden and lung lesions in Mycobacterium tuberculosis-infected mice. Open Life Sci 2022; 17:312-320. [PMID: 35434368 PMCID: PMC8974395 DOI: 10.1515/biol-2022-0025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/02/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
This study aimed to investigate the relationship between interleukin (IL)-35 level and IL-35-producing regulatory T cells (iTr35 subset) in Mycobacterium tuberculosis (Mtb)-infected mice. After the mice were injected with Mtb strain H37R via tail vein, the bacterial burden, lung lesions, and the impact of immune suppression on the infected mice were respectively assessed. The results, when compared with the control mice, showed that the mRNA expression levels of the p35 and Epstein-Barr virus-induced gene 3 of IL-35 were significantly increased in the Mtb-infected mouse spleen at 4 or 8 weeks post-infection and their protein expression levels were concurrently increased in the lungs of the mice, especially in 8 week infected mice. In addition, the levels of serum IL-35 and the iTr35 subset in the spleen of mice were also increased in 4 or 8 weeks post-infection compared with the control mice. Importantly, the high bacterial burden and lung lesions and the low mouse weight were found at 8 week post-infection. Therefore, the mice infected with Mtb resulted in elevating IL-35 level and iTr35 subset and increasing bacterial burden and lung lesions. The findings from the study suggest IL-35 and iTr35 cells may exert an immune suppression role in chronic Mtb-infected mice.
Collapse
Affiliation(s)
- Fangliu Yu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College , Wuhu 241001 , PR China
| | - Xinying Zhu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College , Wuhu 241001 , PR China
| | - Qingdeng Li
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College , Wuhu 241001 , PR China
| | - Wenqin Xu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College , Wuhu 241001 , PR China
| | - Yunxing Gao
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College , Wuhu 241001 , PR China
| | - Yufeng Wen
- School of Public Health, Wannan Medical College , Wuhu 241001 , PR China
| | - Qiong Zhang
- Center of Disease Control and Prevention , Wuhu , Anhui, 241001 , PR China
| | - Jun Dou
- Departments of Pathogenic Biology and Immunology, Medical School, Southeast University , 87 Ding Jiaqiao Rd. , Nanjing 210009 , PR China
| |
Collapse
|
3
|
Wiche Salinas TR, Gosselin A, Raymond Marchand L, Moreira Gabriel E, Tastet O, Goulet JP, Zhang Y, Vlad D, Touil H, Routy JP, Bego MG, El-Far M, Chomont N, Landay AL, Cohen ÉA, Tremblay C, Ancuta P. IL-17A reprograms intestinal epithelial cells to facilitate HIV-1 replication and outgrowth in CD4+ T cells. iScience 2021; 24:103225. [PMID: 34712922 PMCID: PMC8531570 DOI: 10.1016/j.isci.2021.103225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 08/09/2021] [Accepted: 10/01/2021] [Indexed: 12/25/2022] Open
Abstract
The crosstalk between intestinal epithelial cells (IECs) and Th17-polarized CD4+ T cells is critical for mucosal homeostasis, with HIV-1 causing significant alterations in people living with HIV (PLWH) despite antiretroviral therapy (ART). In a model of IEC and T cell co-cultures, we investigated the effects of IL-17A, the Th17 hallmark cytokine, on IEC ability to promote de novo HIV infection and viral reservoir reactivation. Our results demonstrate that IL-17A acts in synergy with TNF to boost IEC production of CCL20, a Th17-attractant chemokine, and promote HIV trans-infection of CD4+ T cells and viral outgrowth from reservoir cells of ART-treated PLWH. Importantly, the Illumina RNA-sequencing revealed an IL-17A-mediated pro-inflammatory and pro-viral molecular signature, including a decreased expression of type I interferon (IFN-I)-induced HIV restriction factors. These findings point to the deleterious features of IL-17A and raise awareness for caution when designing therapies aimed at restoring the paucity of mucosal Th17 cells in ART-treated PLWH. IL-17A acts in synergy with TNF to enhance CCL20 production in IEC exposed to HIV IL-17A/TNF-activated IEC efficiently promote HIV trans-infection of CD4+ T cells IL-17A reprograms IEC to boost HIV outgrowth from CD4+ T cells of ART-treated PLWH IL-17A decreases the expression of IFN-I-induced HIV restriction factors in IEC
Collapse
Affiliation(s)
- Tomas Raul Wiche Salinas
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Annie Gosselin
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | | | - Etiene Moreira Gabriel
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Olivier Tastet
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | | | - Yuwei Zhang
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | - Dragos Vlad
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | - Hanane Touil
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Jean-Pierre Routy
- Chronic Viral Illness Service and Division of Hematology, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Mariana G. Bego
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada
| | - Mohamed El-Far
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
| | - Nicolas Chomont
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Alan L. Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Éric A. Cohen
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
- Institut de Recherches Cliniques de Montréal, Montréal, QC, Canada
| | - Cécile Tremblay
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Petronela Ancuta
- CHUM-Research Centre, 900 rue Saint-Denis, Tour Viger R, room R09.416, Montreal, QC H2X 0A9, Canada
- Département de microbiologie, infectiologie et immunologie, Faculté de médecine, Université de Montréal, Montréal, QC, Canada
- Corresponding author
| |
Collapse
|
4
|
Ensoli B, Moretti S, Borsetti A, Maggiorella MT, Buttò S, Picconi O, Tripiciano A, Sgadari C, Monini P, Cafaro A. New insights into pathogenesis point to HIV-1 Tat as a key vaccine target. Arch Virol 2021; 166:2955-2974. [PMID: 34390393 PMCID: PMC8363864 DOI: 10.1007/s00705-021-05158-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023]
Abstract
Despite over 30 years of enormous effort and progress in the field, no preventative and/or therapeutic vaccines against human immunodeficiency virus (HIV) are available. Here, we briefly summarize the vaccine strategies and vaccine candidates that in recent years advanced to efficacy trials with mostly unsatisfactory results. Next, we discuss a novel and somewhat contrarian approach based on biological and epidemiological evidence, which led us to choose the HIV protein Tat for the development of preventive and therapeutic HIV vaccines. Toward this goal, we review here the role of Tat in the virus life cycle as well as experimental and epidemiological evidence supporting its key role in the natural history of HIV infection and comorbidities. We then discuss the preclinical and clinical development of a Tat therapeutic vaccine, which, by improving the functionality and homeostasis of the immune system and by reducing the viral reservoir in virologically suppressed vaccinees, helps to establish key determinants for intensification of combination antiretroviral therapy (cART) and a functional cure. Future developments and potential applications of the Tat therapeutic vaccine are also discussed, as well as the rationale for its use in preventative strategies. We hope this contribution will lead to a reconsideration of the current paradigms for the development of HIV/AIDS vaccines, with a focus on targeting of viral proteins with key roles in HIV pathogenesis.
Collapse
Affiliation(s)
- Barbara Ensoli
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.
| | - Sonia Moretti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Alessandra Borsetti
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Maria Teresa Maggiorella
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Stefano Buttò
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Orietta Picconi
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Antonella Tripiciano
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Cecilia Sgadari
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Paolo Monini
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Aurelio Cafaro
- National HIV/AIDS Research Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| |
Collapse
|
5
|
Palma C, La Rocca C, Gigantino V, Aquino G, Piccaro G, Di Silvestre D, Brambilla F, Rossi R, Bonacina F, Lepore MT, Audano M, Mitro N, Botti G, Bruzzaniti S, Fusco C, Procaccini C, De Rosa V, Galgani M, Alviggi C, Puca A, Grassi F, Rezzonico-Jost T, Norata GD, Mauri P, Netea MG, de Candia P, Matarese G. Caloric Restriction Promotes Immunometabolic Reprogramming Leading to Protection from Tuberculosis. Cell Metab 2021; 33:300-318.e12. [PMID: 33421383 DOI: 10.1016/j.cmet.2020.12.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 11/13/2020] [Accepted: 12/16/2020] [Indexed: 12/17/2022]
Abstract
There is a strong relationship between metabolic state and susceptibility to Mycobacterium tuberculosis (MTB) infection, with energy metabolism setting the basis for an exaggerated immuno-inflammatory response, which concurs with MTB pathogenesis. Herein, we show that controlled caloric restriction (CR), not leading to malnutrition, protects susceptible DBA/2 mice against pulmonary MTB infection by reducing bacterial load, lung immunopathology, and generation of foam cells, an MTB reservoir in lung granulomas. Mechanistically, CR induced a metabolic shift toward glycolysis, and decreased both fatty acid oxidation and mTOR activity associated with induction of autophagy in immune cells. An integrated multi-omics approach revealed a specific CR-induced metabolomic, transcriptomic, and proteomic signature leading to reduced lung damage and protective remodeling of lung interstitial tightness able to limit MTB spreading. Our data propose CR as a feasible immunometabolic manipulation to control MTB infection, and this approach offers an unexpected strategy to boost immunity against MTB.
Collapse
Affiliation(s)
- Carla Palma
- Dipartimento Malattie Infettive, Istituto Superiore di Sanità, 00161 Roma, Italy.
| | - Claudia La Rocca
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy
| | - Vincenzo Gigantino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, 80131 Naples, Italy
| | - Gabriella Aquino
- Pathology Unit, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, 80131 Naples, Italy
| | - Giovanni Piccaro
- Dipartimento Malattie Infettive, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Dario Di Silvestre
- Proteomics and Metabolomics Unit, Institute for Biomedical Technologies, Consiglio Nazionale delle Ricerche (ITB-CNR), 20090 Segrate, Milano, Italy
| | - Francesca Brambilla
- Proteomics and Metabolomics Unit, Institute for Biomedical Technologies, Consiglio Nazionale delle Ricerche (ITB-CNR), 20090 Segrate, Milano, Italy
| | - Rossana Rossi
- Proteomics and Metabolomics Unit, Institute for Biomedical Technologies, Consiglio Nazionale delle Ricerche (ITB-CNR), 20090 Segrate, Milano, Italy
| | - Fabrizia Bonacina
- Department of Excellence in Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Maria Teresa Lepore
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy
| | - Matteo Audano
- Department of Excellence in Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Nico Mitro
- Department of Excellence in Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy
| | - Gerardo Botti
- Scientific Directorate, Istituto Nazionale Tumori, Fondazione G. Pascale, IRCCS, 80131 Naples, Italy
| | - Sara Bruzzaniti
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy; Dipartimento di Biologia, Università degli Studi di Napoli "Federico II", 80126 Napoli, Italy
| | - Clorinda Fusco
- Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Claudio Procaccini
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy; Unità di Neuroimmunologia, IRCCS-Fondazione Santa Lucia, 00143 Roma, Italy
| | - Veronica De Rosa
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy; Unità di Neuroimmunologia, IRCCS-Fondazione Santa Lucia, 00143 Roma, Italy
| | - Mario Galgani
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy; Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy
| | - Carlo Alviggi
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy; Department of Neuroscience, Reproductive Science, and Odontostomatology, University of Naples, Federico II, Naples, Italy
| | - Annibale Puca
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi-Salerno, Italy; IRCCS MultiMedica, 20138 Milano, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Tanja Rezzonico-Jost
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6500 Bellinzona, Switzerland
| | - Giuseppe Danilo Norata
- Department of Excellence in Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy; Center for the Study of Atherosclerosis, Società Italiana Studio Aterosclerosi, Bassini Hospital, 20092 Cinisello Balsamo, Milano, Italy
| | - Pierluigi Mauri
- Proteomics and Metabolomics Unit, Institute for Biomedical Technologies, Consiglio Nazionale delle Ricerche (ITB-CNR), 20090 Segrate, Milano, Italy; Istituto di Scienze della Vita, Scuola Superiore Sant'Anna, 56127 Pisa, Italy
| | - Mihai G Netea
- Radboud Center for Infectious Diseases and Department of Internal Medicine, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | | | - Giuseppe Matarese
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Napoli, Italy; Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli "Federico II", 80131 Napoli, Italy.
| |
Collapse
|
6
|
Wang J, Hussain T, Zhang K, Liao Y, Yao J, Song Y, Sabir N, Cheng G, Dong H, Li M, Ni J, Mangi MH, Zhao D, Zhou X. Inhibition of type I interferon signaling abrogates early Mycobacterium bovis infection. BMC Infect Dis 2019; 19:1031. [PMID: 31801478 PMCID: PMC6894119 DOI: 10.1186/s12879-019-4654-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/22/2019] [Indexed: 12/17/2022] Open
Abstract
Background Mycobacterium bovis (M. bovis) is the principal causative agent of bovine tuberculosis; however, it may also cause serious infection in human being. Type I IFN is a key factor in reducing viral multiplication and modulating host immune response against viral infection. However, the regulatory pathways of Type I IFN signaling during M. bovis infection are not yet fully explored. Here, we investigate the role of Type I IFN signaling in the pathogenesis of M. bovis infection in mice. Methods C57BL/6 mice were treated with IFNAR1-blocking antibody or Isotype control 24 h before M. bovis infection. After 21 and 84 days of infection, mice were sacrificed and the role of Type I IFN signaling in the pathogenesis of M. bovis was investigated. ELISA and qRT-PCR were performed to detect the expression of Type I IFNs and related genes. Lung lesions induced by M. bovis were assessed by histopathological examination. Viable bacterial count was determined by CFU assay. Results We observed an abundant expression of Type I IFNs in the serum and lung tissues of M. bovis infected mice. In vivo blockade of Type I IFN signaling reduced the recruitment of neutrophils to the lung tissue, mediated the activation of macrophages leading to an increased pro-inflammatory profile and regulated the inflammatory cytokine production. However, no impact was observed on T cell activation and recruitment in the early acute phase of infection. Additionally, blocking of type I IFN signaling reduced bacterial burden in the infected mice as compared to untreated infected mice. Conclusions Altogether, our results reveal that Type I IFN mediates a balance between M. bovis-mediated inflammatory reaction and host defense mechanism. Thus, modulating Type I IFN signaling could be exploited as a therapeutic strategy against a large repertoire of inflammatory disorders including tuberculosis.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China.,Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS), Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China
| | - Tariq Hussain
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Kai Zhang
- School of Agriculture, Ningxia University, Ningxia, China
| | - Yi Liao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiao Yao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yinjuan Song
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Naveed Sabir
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Guangyu Cheng
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haodi Dong
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Miaoxuan Li
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jiamin Ni
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Mazhar Hussain Mangi
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Deming Zhao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
7
|
Riccomi A, Piccaro G, Christensen D, Palma C, Andersen P, Vendetti S. Parenteral Vaccination With a Tuberculosis Subunit Vaccine in Presence of Retinoic Acid Provides Early but Transient Protection to M. Tuberculosis Infection. Front Immunol 2019; 10:934. [PMID: 31130946 PMCID: PMC6509564 DOI: 10.3389/fimmu.2019.00934] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
Most microbes invading through mucosal surfaces cause disease and therefore strategies to induce mucosal immune responses are strongly needed. Vitamin A metabolites, such as retinoic acid (RA), play crucial roles in programming T and B cells to home to mucosal compartments, therefore we evaluated the capacity of RA to elicit mucosal immune responses against tuberculosis (TB) after parenteral vaccination. We found that mice immunized through subcutaneous injections with the TB subunit vaccine (CAF01+H56) in presence of RA show enhanced mucosal H56-specific IgA responses and enhanced Ag-specific CD4+ T lymphocytes homing to the lung as compared with control mice. Immunization with CAF01+H56 in presence of RA resulted in lower bacterial loads in the lungs of mice 14 days after challenge with virulent Mycobacterium tuberculosis (Mtb) as compared to mice immunized in the absence of RA or vaccinated with BCG. Higher amounts of IFNγ and IL-17 pro-inflammatory cytokines were found in lung homogenates of mice immunized with CAF01+H56 and RA 24 h after Mtb infection. However, 6 weeks after infection the protection was comparable in vaccinated mice with or without RA even though treatment with RA during immunization is able to better contain the inflammatory response by the host. Furthermore, at later stage of the infection a higher percentage of Mtb specific CD4+PD1+ T lymphocytes were found in the lungs of mice immunized with CAF01+H56 and RA. These data show that an enhanced mucosal immune response is generated during parenteral vaccination in presence of RA. Furthermore, RA treatment contained the bacterial growth at an early stage of the infection and limited the inflammatory response in the lung at later time points.
Collapse
Affiliation(s)
- Antonella Riccomi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Dennis Christensen
- Department of Infectious Diseases Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Carla Palma
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Peter Andersen
- Department of Infectious Diseases Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Silvia Vendetti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|