1
|
Yang B, Guo X, Shi C, Liu G, Qin X, Chen S, Gan L, Liang D, Shao K, Xu R, Zhong J, Mo Y, Li H, Luo D. Alterations in purine and pyrimidine metabolism associated with latent tuberculosis infection: insights from gut microbiome and metabolomics analyses. mSystems 2024:e0081224. [PMID: 39436103 DOI: 10.1128/msystems.00812-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/24/2024] [Indexed: 10/23/2024] Open
Abstract
Individuals with latent tuberculosis infection (LTBI) account for almost 30% of the population worldwide and have the potential to develop active tuberculosis (ATB). Despite this, the current understanding of the pathogenesis of LTBI is limited. The gut microbiome can be altered in tuberculosis patients, and an understanding of the changes associated with the progression from good health to LTBI to ATB can provide novel perspectives for understanding the pathogenesis of LTBI by identifying microbial and molecular biomarkers associated therewith. In this study, fecal samples from healthy controls (HC), individuals with LTBI and ATB patients were collected for gut microbiome and metabolomics analyses. Compared to HC and LTBI subjects, participants with ATB showed a significant decrease in gut bacterial α-diversity. Additionally, there were significant differences in gut microbial communities and metabolism among the HC, LTBI, and ATB groups. PICRUSt2 analysis revealed that microbiota metabolic pathways involving the degradation of purine and pyrimidine metabolites were upregulated in LTBI and ATB individuals relative to HCs. Metabolomic profiling similarly revealed that purine and pyrimidine metabolite levels were decreased in LTBI and ATB samples relative to those from HCs. Further correlation analyses revealed that the levels of purine and pyrimidine metabolites were negatively correlated with those of gut microbial genera represented by Ruminococcus_gnavus_group (R. gnavus), and the levels of R. gnavus were also positively correlated with adenosine nucleotide degradation II, which is a purine degradation pathway. Moreover, a combined signature including hypoxanthine and xanthine was found to effectively distinguish between LTBI and HC samples (area under the curve [AUC] of training set = 0.796; AUC of testing set = 0.924). Therefore, through gut microbiome and metabolomic analyses, these findings provide valuable clues regarding how alterations in gut purine and pyrimidine metabolism are linked to the pathogenesis of LTBI.IMPORTANCEThis study provides valuable insight into alterations in the gut microbiome and metabolomic profiles in a cohort of adults with LTBI and ATB. Perturbed gut purine and pyrimidine metabolism in LTBI was associated with the compositional alterations of gut microbiota, which may be an impetus for developing novel diagnostic strategies and interventions targeting LTBI.
Collapse
Affiliation(s)
- Boyi Yang
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
- Department of Physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- The First Clinical College, Guangxi Medical University, Nanning, China
| | - Xiaojing Guo
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Chongyu Shi
- Molecular Biology Laboratory of Respiratory Disease, College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Gang Liu
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiaoling Qin
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Shiyi Chen
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Li Gan
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Dongxu Liang
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Kai Shao
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Ruolan Xu
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Jieqing Zhong
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Yujie Mo
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Hai Li
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, China
| | - Dan Luo
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, China
| |
Collapse
|
2
|
Szachniewicz MM, van den Eeden SJF, van Meijgaarden KE, Franken KLMC, van Veen S, Geluk A, Bouwstra JA, Ottenhoff THM. Cationic pH-sensitive liposome-based subunit tuberculosis vaccine induces protection in mice challenged with Mycobacterium tuberculosis. Eur J Pharm Biopharm 2024; 203:114437. [PMID: 39122053 DOI: 10.1016/j.ejpb.2024.114437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Tuberculosis (TB) has been and still is a global emergency for centuries. Prevention of disease through vaccination would have a major impact on disease prevalence, but the only available current vaccine, BCG, has insufficient impact. In this article, a novel subunit vaccine against TB was developed, using the Ag85B-ESAT6-Rv2034 fusion antigen, two adjuvants - CpG and MPLA, and a cationic pH-sensitive liposome as a delivery system, representing a new TB vaccine delivery strategy not previously reported for TB. In vitro in human dendritic cells (DCs), the adjuvanted formulation induced a significant increase in the production of (innate) cytokines and chemokines compared to the liposome without additional adjuvants. In vivo, the new vaccine administrated subcutaneously significantly reduced Mycobacterium tuberculosis (Mtb) bacterial load in the lungs and spleens of mice, significantly outperforming results from mice vaccinated with the antigen mixed with adjuvants without liposomes. In-depth analysis underpinned the vaccine's effectiveness in terms of its capacity to induce polyfunctional CD4+ and CD8+ T-cell responses, both considered essential for controlling Mtb infection. Also noteworthy was the differential abundance of various CD69+ B-cell subpopulations, which included IL17-A-producing B-cells. The vaccine stimulated robust antigen-specific antibody titers, further extending its potential as a novel protective agent against TB.
Collapse
Affiliation(s)
- M M Szachniewicz
- Leiden University Center for Infectious Diseases, Leiden University Medical Center (LUMC), the Netherlands.
| | - S J F van den Eeden
- Leiden University Center for Infectious Diseases, Leiden University Medical Center (LUMC), the Netherlands
| | - K E van Meijgaarden
- Leiden University Center for Infectious Diseases, Leiden University Medical Center (LUMC), the Netherlands
| | - K L M C Franken
- Leiden University Center for Infectious Diseases, Leiden University Medical Center (LUMC), the Netherlands
| | - S van Veen
- Leiden University Center for Infectious Diseases, Leiden University Medical Center (LUMC), the Netherlands
| | - A Geluk
- Leiden University Center for Infectious Diseases, Leiden University Medical Center (LUMC), the Netherlands
| | - J A Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, the Netherlands
| | - T H M Ottenhoff
- Leiden University Center for Infectious Diseases, Leiden University Medical Center (LUMC), the Netherlands
| |
Collapse
|
3
|
Zhao D, Song YH, Li D, Zhang R, Xu JB, Shi K, Li JM, Leng X, Zong Y, Zeng FL, Gong QL, Du R. Mycobacterium tuberculosis Rv3435c regulates inflammatory cytokines and promotes the intracellular survival of recombinant Mycobacteria. Acta Trop 2023; 246:106974. [PMID: 37355194 DOI: 10.1016/j.actatropica.2023.106974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/14/2023] [Accepted: 06/18/2023] [Indexed: 06/26/2023]
Abstract
Mycobacterium tuberculosis is a pathogenic bacterium that is parasitic in macrophages and show high adaptation to the host's immune response. It can also trigger a complex immune response in the host. This relies on proteins encoded by a series of M. tuberculosis-encoded virulence genes. We found that the M. tuberculosis Rv3435c gene is highly conserved among pathogenic mycobacteria, and might be a virulence gene. To explore the gene function of Rv3435c, we used Mycobacterium smegmatis to construct a recombinant mycobacterium expressing Rv3435c heterologously. The results that Rv3435c is a cell wall-related protein that changes bacterial and colony morphology, inhibits the growth rate of recombinant mycobacteria, and enhances their resistance to various stresses. We also found that the fatty acid levels of the recombinant strain changed. Simultaneously, Rv3435c can inhibit the expression and secretion of inflammatory factors and host cell apoptosis, and enhance the survival of recombinant bacteria in macrophages. Experimental data indicated that Rv3435c might play an important role in Mycobacterium tuberculosis infection.
Collapse
Affiliation(s)
- Dan Zhao
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; Ginseng and Antler Products Testing Center of the Ministry of Agricultural PRC, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Yu-Hao Song
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Dong Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Rui Zhang
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Jin-Biao Xu
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Kun Shi
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Jian-Ming Li
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Xue Leng
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Ying Zong
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China
| | - Fan-Li Zeng
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China.
| | - Qing-Long Gong
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China.
| | - Rui Du
- College of Chinese Medicine Materials, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China; Laboratory of Production and Product Application of Sika Deer of Jilin Province, Jilin Agricultural University, Changchun, Jilin Province, 130118, P.R. China.
| |
Collapse
|
4
|
Ahmad S, Ahmed J, Khalifa EH, Khattak FA, Khan AS, Farooq SU, Osman SMA, Salih MM, Ullah N, Khan TA. Novel mutations in genes of the IL-12/IFN-γ axis cause susceptibility to tuberculosis. J Infect Public Health 2023; 16:1368-1378. [PMID: 37437430 DOI: 10.1016/j.jiph.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/15/2023] [Accepted: 06/06/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND The IL-12/23/ISG15-IFN-γ pathway is the main immunological pathway for controlling intra-macrophagic microorganisms such as Mycobacteria, Salmonella, and Leishmania spp. Consequently, upon mutations in genes of the IL-12/23/ISG15-IFN-γ pathway cause increased susceptibility to intra-macrophagic pathogens, particularly to Mycobacteria. Therefore, the purpose of this study was to characterize the mutations in genes of the IL-12/23/ISG15-IFN-γ pathway in severe tuberculosis (TB) patients. METHODS Clinically suspected TB was initially confirmed in four patients (P) (P1, P2, P3, and P4) using the GeneXpert MTB/RIF and culturing techniques. The patients' Peripheral blood mononuclear cells (PBMCs) were then subjected to ELISA to measure Interleukin 12 (IL-12) and interferon gamma (IFN-γ). Flow cytometry was used to detect the surface expressions of IFN-γR1 and IFN-γR2 as well as IL-12Rβ1and IL-12Rβ2 on monocytes and T lymphocytes, respectively.The phosphorylation of signal transducer and activator of transcription 1(STAT1) on monocytes and STAT4 on T lymphocytes were also detected by flow cytometry. Sanger sequencing was used to identify mutations in the IL-12Rβ1, STAT1, NEMO, and CYBB genes. RESULTS P1's PBMCs exhibited reduced IFN-γ production, while P2's and P3's PBMCs exhibited impaired IL-12 induction. Low IL-12Rβ1 surface expression and reduced STAT4 phosphorylation were demonstrated by P1's T lymphocytes, while impaired STAT1 phosphorylation was detected in P2's monocytes. The impaired IκB-α degradation and abolished H2O2 production in monocytes and neutrophils of P3 and P4 were observed, respectively. Sanger sequencing revealed novel nonsense homozygous mutation: c.191 G>A/p.W64 * in exon 3 of the IL-12Rβ1 gene in P1, novel missense homozygous mutation: c.107 A>T/p.Q36L in exon 3 of the STAT1 gene in P2, missense hemizygous mutation:: c.950 A>C/p.Q317P in exon 8 of the NEMO gene in P3, and nonsense hemizygous mutation: c.868 C>T/p.R290X in exon 8 of CYBB gene in P4. CONCLUSION Our findings broaden the clinical and genetic spectra associated with IL-12/23/ISG15-IFN-γ axis anomalies. Additionally, our data suggest that TB patients in Pakistan should be investigated for potential genetic defects due to high prevalence of parental consanguinity and increased incidence of TB in the country.
Collapse
Affiliation(s)
- Sajjad Ahmad
- Institute of Basic Medical Science, Khyber Medical University, Peshawar, KP, Pakistan
| | - Jawad Ahmed
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan
| | - Eman H Khalifa
- Al Baha University Faculty of Applied Medical Sciences, Saudi Arabia
| | - Farhad Ali Khattak
- Research & development Cell, Khyber College of Dentistry (KCD), Peshawar, Pakistan
| | - Anwar Sheed Khan
- Provincial TB Reference laboratory, Hayatabad Medical Complex, Peshawar, PK, Pakistan
| | - Syed Umar Farooq
- Department of oral pathology, Khyber College of Dentistry, Peshawar. Pakistan
| | | | | | - Nadeem Ullah
- Department of Clinical Microbiology, Umeå University, 90185 Umeå, Sweden.
| | - Taj Ali Khan
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan; Division of Infectious Diseases & Global Medicine,Department of Medicine,University of Florida, Gainesville, FL,United States.
| |
Collapse
|
5
|
MASTORINO L, DAPAVO P, TRUNFIO M, AVALLONE G, RUBATTO M, CALCAGNO A, RIBERO S, QUAGLINO P. Risk of Reactivation of Latent Tuberculosis in Psoriasis Patients on Biologic Therapies: A Retrospective Cohort from a Tertiary Care Centre in Northern Italy. Acta Derm Venereol 2022; 102:adv00821. [PMID: 36065745 PMCID: PMC9811291 DOI: 10.2340/actadv.v102.1982] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Psoriatic patients with latent tuberculosis infection and properly treated active tuberculosis need careful management when prescribing modern biological drugs. Although data and guidelines regarding tumour necrosis factor-α inhibitors advise caution and initiation of prophylactic therapy in patients with latent tuberculosis infection, the same indications do not seem to find equal force for interleukin (IL)-23 and IL-17 inhibitors. In order to evaluate the risk of reactivation in patients with latent tuberculosis infection or properly treated active tuberculosis, an observational retrospective study was conducted on the population referred to our centre at Dermatologic Clinic of University of Turin, Italy. In the last 10 years at the clinic 19 psoriatic patients were found to be at risk of tuberculosis reactivation: 10 patients were QuantiFERON- TB-positive at baseline, 2 became positive during treatment, 6 reported prior tuberculous infection, and 1 was QuantiFERON-TB-negative at baseline and developed disseminated tuberculosis during treatment with anti-tumour necrosis factor-α. Overall, 10.5% of this group of patients developed active tuberculosis; however, stratifying by biologic therapy, zero cases were observed among patients treated with anti-IL-17, -23, or -12/23 over a relatively long follow-up (48.1 months) A review of the available literature following our experience confirms the increased risk of tuberculosis reactivation with tumour necrosis factor-α inhibitors. Concerning anti-IL-23 and IL-17 drugs, available data showed high safety in patients at risk of tuberculosis reactivation. Screening of patients who should be taking IL-17 and IL-23 inhibitors is recommended for public health purposes. In case of a positive result with these therapies, consulting with an infectious diseases specialist is suggested in order to weigh up the risks and benefits of prophylactic treatment.
Collapse
Affiliation(s)
| | - Paolo DAPAVO
- Dermatology Clinic, Department of Medical Sciences
| | - Mattia TRUNFIO
- Unit of Infectious Diseases, Amedeo di Savoia Hospital, Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | | - Andrea CALCAGNO
- Unit of Infectious Diseases, Amedeo di Savoia Hospital, Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | |
Collapse
|
6
|
Kilinç G, Walburg KV, Franken KLMC, Valkenburg ML, Aubry A, Haks MC, Saris A, Ottenhoff THM. Development of Human Cell-Based In Vitro Infection Models to Determine the Intracellular Survival of Mycobacterium avium. Front Cell Infect Microbiol 2022; 12:872361. [PMID: 35811670 PMCID: PMC9263196 DOI: 10.3389/fcimb.2022.872361] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/24/2022] [Indexed: 11/21/2022] Open
Abstract
The Mycobacterium avium (Mav) complex accounts for more than 80% of all pulmonary diseases caused by non-tuberculous mycobacteria (NTM) infections, which have an alarming increase in prevalence and vary in different regions, currently reaching 0.3–9.8 per 100,000 individuals. Poor clinical outcomes, as a result of increasing microbial drug resistance and low treatment adherence due to drug-toxicities, emphasize the need for more effective treatments. Identification of more effective treatments, however, appears to be difficult, which may be due to the intracellular life of NTM and concomitant altered drug sensitivity that is not taken into account using traditional drug susceptibility testing screenings. We therefore developed human cell-based in vitro Mav infection models using the human MelJuSo cell line as well as primary human macrophages and a fluorescently labeled Mav strain. By testing a range of multiplicity of infection (MOI) and using flow cytometry and colony-forming unit (CFU) analysis, we found that an MOI of 10 was the most suitable for Mav infection in primary human macrophages, whereas an MOI of 50 was required to achieve similar results in MelJuSo cells. Moreover, by monitoring intracellular bacterial loads over time, the macrophages were shown to be capable of controlling the infection, while MelJuSo cells failed to do so. When comparing the MGIT system with the classical CFU counting assay to determine intracellular bacterial loads, MGIT appeared as a less labor-intensive, more precise, and more objective alternative. Next, using our macrophage Mav infection models, the drug efficacy of the first-line drug rifampicin and the more recently discovered bedaquiline on intracellular bacteria was compared to the activity on extracellular bacteria. The efficacy of the antibiotics inhibiting bacterial growth was significantly lower against intracellular bacteria compared to extracellular bacteria. This finding emphasizes the crucial role of the host cell during infection and drug susceptibility and highlights the usefulness of the models. Taken together, the human cell-based Mav infection models are reliable tools to determine the intracellular loads of Mav, which will enable researchers to investigate host–pathogen interactions and to evaluate the efficacy of (host-directed) therapeutic strategies against Mav.
Collapse
Affiliation(s)
- Gül Kilinç
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Kimberley V. Walburg
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Kees L. M. C. Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Merel L. Valkenburg
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Alexandra Aubry
- Sorbonne Université, INSERM, Centre d’Immunologie et des Maladies Infectieuses, U1135, AP-HP, Hôpital Pitié-Salpêtrière, Centre National de Référence des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, Paris, France
| | - Mariëlle C. Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Anno Saris
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Anno Saris, ; orcid.org/0000-0003-0493-9501
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
7
|
Sholeye AR, Williams AA, Loots DT, Tutu van Furth AM, van der Kuip M, Mason S. Tuberculous Granuloma: Emerging Insights From Proteomics and Metabolomics. Front Neurol 2022; 13:804838. [PMID: 35386409 PMCID: PMC8978302 DOI: 10.3389/fneur.2022.804838] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/24/2022] [Indexed: 12/24/2022] Open
Abstract
Mycobacterium tuberculosis infection, which claims hundreds of thousands of lives each year, is typically characterized by the formation of tuberculous granulomas — the histopathological hallmark of tuberculosis (TB). Our knowledge of granulomas, which comprise a biologically diverse body of pro- and anti-inflammatory cells from the host immune responses, is based mainly upon examination of lungs, in both human and animal studies, but little on their counterparts from other organs of the TB patient such as the brain. The biological heterogeneity of TB granulomas has led to their diverse, relatively uncoordinated, categorization, which is summarized here. However, there is a pressing need to elucidate more fully the phenotype of the granulomas from infected patients. Newly emerging studies at the protein (proteomics) and metabolite (metabolomics) levels have the potential to achieve this. In this review we summarize the diverse nature of TB granulomas based upon the literature, and amplify these accounts by reporting on the relatively few, emerging proteomics and metabolomics studies on TB granulomas. Metabolites (for example, trimethylamine-oxide) and proteins (such as the peptide PKAp) associated with TB granulomas, and knowledge of their localizations, help us to understand the resultant phenotype. Nevertheless, more multidisciplinary ‘omics studies, especially in human subjects, are required to contribute toward ushering in a new era of understanding of TB granulomas – both at the site of infection, and on a systemic level.
Collapse
Affiliation(s)
- Abisola Regina Sholeye
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Aurelia A. Williams
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Du Toit Loots
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - A. Marceline Tutu van Furth
- Department of Pediatric Infectious Diseases and Immunology, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Emma Children's Hospital, Amsterdam, Netherlands
| | - Martijn van der Kuip
- Department of Pediatric Infectious Diseases and Immunology, Pediatric Infectious Diseases and Immunology, Amsterdam University Medical Center, Emma Children's Hospital, Amsterdam, Netherlands
| | - Shayne Mason
- Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
- *Correspondence: Shayne Mason
| |
Collapse
|
8
|
Druszczyńska M, Godkowicz M, Kulesza J, Wawrocki S, Fol M. Cytokine Receptors-Regulators of Antimycobacterial Immune Response. Int J Mol Sci 2022; 23:1112. [PMID: 35163035 PMCID: PMC8835057 DOI: 10.3390/ijms23031112] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 12/18/2022] Open
Abstract
Cytokine receptors are critical regulators of the antimycobacterial immune response, playing a key role in initiating and coordinating the recruitment and activation of immune cells during infection. They recognize and bind specific cytokines and are involved in inducing intracellular signal transduction pathways that regulate a diverse range of biological functions, including proliferation, differentiation, metabolism and cell growth. Due to mutations in cytokine receptor genes, defective signaling may contribute to increased susceptibility to mycobacteria, allowing the pathogens to avoid killing and immune surveillance. This paper provides an overview of cytokine receptors important for the innate and adaptive immune responses against mycobacteria and discusses the implications of receptor gene defects for the course of mycobacterial infection.
Collapse
Affiliation(s)
- Magdalena Druszczyńska
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (M.G.); (S.W.); (M.F.)
| | - Magdalena Godkowicz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (M.G.); (S.W.); (M.F.)
- Lodz Institutes of the Polish Academy of Sciences, The Bio-Med-Chem Doctoral School, University of Lodz, 90-237 Lodz, Poland
| | - Jakub Kulesza
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, Kniaziewicza 1/5, 91-347 Lodz, Poland;
| | - Sebastian Wawrocki
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (M.G.); (S.W.); (M.F.)
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, 7265 Davos, Switzerland
| | - Marek Fol
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland; (M.G.); (S.W.); (M.F.)
| |
Collapse
|
9
|
Effect of breastfeeding on children's health and its relationship to NRAMP1 expression: A cross-sectional study. Ann Med Surg (Lond) 2021; 71:103017. [PMID: 34840765 PMCID: PMC8606844 DOI: 10.1016/j.amsu.2021.103017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 11/24/2022] Open
Abstract
Toddlers with exclusive breastfeeding can increase immunity in preventing infectious diseases such as Upper Respiratory Tract Infections (ARI). The body's resistance to disease is controlled by Natural Resistance-Associated Macrophage Protein 1 (NRAMP1). NRAMP1 contributes to the pathophysiology of several intercellular infections, including ARI. The purpose of this study was to determine the effect of breastfeeding on children's health and its relationship to NRAMP1 expression. A cross-sectional study was conducted on 124 toddlers in October 2020–June 2021. Toddlers were selected purposively from three Community Health Centers in Central Jakarta. The first group (n = 62) was exclusive breastfeeding and the second group (n = 62) was not exclusive breastfeeding. The characteristics of mothers and toddlers were collected using a structured questionnaire supported by medical record data. Meanwhile, NRAMP1 expression and NRAMP1 protein levels were obtained from the examination of blood samples. Examination of NRAMP1 gene mRNA expression by real-time PCR method and serum NRAMP1 protein levels by ELISA method. Data were analyzed using t-test, ANOVA, and multiple linear regression. The results obtained that the average mRNA expression of NRAMP1 gene and protein levels of NRAMP1 in infants who were not exclusive breastfeeding were 6.88 fold change (FC) and 315.02 pg/ml compared to those who received exclusive breastfeeding of 11.36 FC and 1087.74 pg./ml. Parity, immunization history, exclusive breastfeeding, and frequency of ARI were significantly associated with NRAMP1 gene mRNA expression and NRAMP1 protein levels (P < 0.05). Maternal and under-five age, gender, and nutritional status were not significantly related (P > 0.05). Exclusive breastfeeding was the dominant factor influencing NRAMP1 gene mRNA expression (OR: 4268) and NRAMP1 protein content (OR: 737,362). Antibodies obtained from exclusive breastfeeding in synergy with the NRAMP1 gene form the body's immunity in infants suffering from ARI. Breast milk contains antibodies that work in synergy with the NRAMP1 gene mRNA expression and NRAMP1 protein levels to form body immunity in children with ARI. Exclusive breastfeeding significantly increased the NRAMP1 gene mRNA expression and NRAMP1 protein levels. Immunization can significantly increase NRAMP1 gene mRNA expression and NRAMP1 protein levels. Exclusive breastfeeding can increase NRAMP1 gene mRNA expression, and NRAMP1 protein levels improved children's health.
Collapse
|
10
|
Kushnareva Y, Mathews IT, Andreyev AY, Altay G, Lindestam Arlehamn CS, Pandurangan V, Nilsson R, Jain M, Sette A, Peters B, Sharma S. Functional Analysis of Immune Signature Genes in Th1* Memory Cells Links ISOC1 and Pyrimidine Metabolism to IFN-γ and IL-17 Production. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:1181-1193. [PMID: 33547171 PMCID: PMC7946769 DOI: 10.4049/jimmunol.2000672] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
CCR6+CXCR3+CCR4-CD4+ memory T cells, termed Th1*, are important for long-term immunity to Mycobacterium tuberculosis and the pathogenesis of autoimmune diseases. Th1* cells express a unique set of lineage-specific transcription factors characteristic of both Th1 and Th17 cells and display distinct gene expression profiles compared with other CD4+ T cell subsets. To examine molecules and signaling pathways important for the effector function of Th1* cells, we performed loss-of-function screening of genes selectively enriched in the Th1* subset. The genetic screen yielded candidates whose depletion significantly impaired TCR-induced IFN-γ production. These included genes previously linked to IFN-γ or M. tuberculosis susceptibility and novel candidates, such as ISOC1, encoding a metabolic enzyme of unknown function in mammalian cells. ISOC1-depleted T cells, which produced less IFN-γ and IL-17, displayed defects in oxidative phosphorylation and glycolysis and impairment of pyrimidine metabolic pathway. Supplementation with extracellular pyrimidines rescued both bioenergetics and IFN-γ production in ISOC1-deficient T cells, indicating that pyrimidine metabolism is a key driver of effector functions in CD4+ T cells and Th1* cells. Results provide new insights into the immune-stimulatory function of ISOC1 as well as the particular metabolic requirements of human memory T cells, providing a novel resource for understanding long-term T cell-driven responses.
Collapse
Affiliation(s)
| | - Ian T Mathews
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Alexander Y Andreyev
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093
- The Scripps Research Institute, La Jolla, CA 92037; and
| | - Gokmen Altay
- La Jolla Institute for Immunology, La Jolla, CA 92037
| | | | | | | | - Mohit Jain
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Alessandro Sette
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Bjoern Peters
- La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Sonia Sharma
- La Jolla Institute for Immunology, La Jolla, CA 92037;
| |
Collapse
|
11
|
Kilinç G, Saris A, Ottenhoff THM, Haks MC. Host-directed therapy to combat mycobacterial infections. Immunol Rev 2021; 301:62-83. [PMID: 33565103 PMCID: PMC8248113 DOI: 10.1111/imr.12951] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 12/27/2020] [Indexed: 12/27/2022]
Abstract
Upon infection, mycobacteria, such as Mycobacterium tuberculosis (Mtb) and nontuberculous mycobacteria (NTM), are recognized by host innate immune cells, triggering a series of intracellular processes that promote mycobacterial killing. Mycobacteria, however, have developed multiple counter‐strategies to persist and survive inside host cells. By manipulating host effector mechanisms, including phagosome maturation, vacuolar escape, autophagy, antigen presentation, and metabolic pathways, pathogenic mycobacteria are able to establish long‐lasting infection. Counteracting these mycobacteria‐induced host modifying mechanisms can be accomplished by host‐directed therapeutic (HDT) strategies. HDTs offer several major advantages compared to conventional antibiotics: (a) HDTs can be effective against both drug‐resistant and drug‐susceptible bacteria, as well as potentially dormant mycobacteria; (b) HDTs are less likely to induce bacterial drug resistance; and (c) HDTs could synergize with, or shorten antibiotic treatment by targeting different pathways. In this review, we will explore host‐pathogen interactions that have been identified for Mtb for which potential HDTs impacting both innate and adaptive immunity are available, and outline those worthy of future research. We will also discuss possibilities to target NTM infection by HDT, although current knowledge regarding host‐pathogen interactions for NTM is limited compared to Mtb. Finally, we speculate that combinatorial HDT strategies can potentially synergize to achieve optimal mycobacterial host immune control.
Collapse
Affiliation(s)
- Gül Kilinç
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Anno Saris
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
12
|
Harada M, Furuhashi K, Karayama M, Suzuki Y, Hozumi H, Enomoto N, Fujisawa T, Nakamura Y, Inui N, Suda T. Subcutaneous injection of interferon gamma therapy could be useful for anti-IFN-γ autoantibody associated disseminated nontuberculous mycobacterial infection. J Infect Chemother 2020; 27:373-378. [PMID: 33071037 DOI: 10.1016/j.jiac.2020.09.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 11/29/2022]
Abstract
One of the human natural defense systems protects against nontuberculous mycobacterial (NTM) infection by IFN-γ producing T lymphocyte cells. Most disseminated NTM infections usually occur in severe immune-compromised patients, such as HIV infection or after organ transplant patients. However, there have been several reports of non-compromised patients with disseminated NTM infection, including antibiotic resistance cases and the presence of a neutralizing antibody against IFN-γ. We elucidated the anti-IFN-γ neutralizing antibody in a 65 year-old Japanese man whose legs were paralyzed because of multiple abscesses in vertebral bodies. Although his vertebral bodies were released due to an operation and antibiotics were administered, this treatment efficacy was poor. Patient's plasma demanded not only IFN-γ expression in peripheral blood mononuclear cells (PBMC) obtained from healthy controls, but also recombinant human IFN-γ expression. Furthermore, IFN-γ receptor expression was increased, compared to the healthy control. Finally, anti-IFN-γ antibody was detected in his plasma. These results suggested that anti-IFN-γ antibody induced an incurable NTM infection. IFN-γ was subcutaneously administrated with antibiotics, and then the abscesses diminished and his general condition was successfully improved. This therapy might be useful against severe NTM infections.
Collapse
Affiliation(s)
- Masanori Harada
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan; Department of Respiratory Medicine, Fujieda Municipal General Hospital, Fujieda, Japan.
| | - Kazuki Furuhashi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan; Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Masato Karayama
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Yuzo Suzuki
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Hironao Hozumi
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Tomoyuki Fujisawa
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Yutaro Nakamura
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Naoki Inui
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| | - Takafumi Suda
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan.
| |
Collapse
|
13
|
Moreira JD, Koch BEV, van Veen S, Walburg KV, Vrieling F, Mara Pinto Dabés Guimarães T, Meijer AH, Spaink HP, Ottenhoff THM, Haks MC, Heemskerk MT. Functional Inhibition of Host Histone Deacetylases (HDACs) Enhances in vitro and in vivo Anti-mycobacterial Activity in Human Macrophages and in Zebrafish. Front Immunol 2020; 11:36. [PMID: 32117228 PMCID: PMC7008710 DOI: 10.3389/fimmu.2020.00036] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/08/2020] [Indexed: 12/27/2022] Open
Abstract
The rapid and persistent increase of drug-resistant Mycobacterium tuberculosis (Mtb) infections poses increasing global problems in combatting tuberculosis (TB), prompting for the development of alternative strategies including host-directed therapy (HDT). Since Mtb is an intracellular pathogen with a remarkable ability to manipulate host intracellular signaling pathways to escape from host defense, pharmacological reprogramming of the immune system represents a novel, potentially powerful therapeutic strategy that should be effective also against drug-resistant Mtb. Here, we found that host-pathogen interactions in Mtb-infected primary human macrophages affected host epigenetic features by modifying histone deacetylase (HDAC) transcriptomic levels. In addition, broad spectrum inhibition of HDACs enhanced the antimicrobial response of both pro-inflammatory macrophages (Mϕ1) and anti-inflammatory macrophages (Mϕ2), while selective inhibition of class IIa HDACs mainly decreased bacterial outgrowth in Mϕ2. Moreover, chemical inhibition of HDAC activity during differentiation polarized macrophages into a more bactericidal phenotype with a concomitant decrease in the secretion levels of inflammatory cytokines. Importantly, in vivo chemical inhibition of HDAC activity in Mycobacterium marinum-infected zebrafish embryos, a well-characterized animal model for tuberculosis, significantly reduced mycobacterial burden, validating our in vitro findings in primary human macrophages. Collectively, these data identify HDACs as druggable host targets for HDT against intracellular Mtb.
Collapse
Affiliation(s)
- Jôsimar D Moreira
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.,Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Bjørn E V Koch
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Kimberley V Walburg
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Frank Vrieling
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Tânia Mara Pinto Dabés Guimarães
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Herman P Spaink
- Institute of Biology Leiden, Leiden University, Leiden, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Matthias T Heemskerk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
14
|
Kroesen VM, Madacki J, Frigui W, Sayes F, Brosch R. Mycobacterial virulence: impact on immunogenicity and vaccine research. F1000Res 2019; 8. [PMID: 32047597 PMCID: PMC6979476 DOI: 10.12688/f1000research.20572.1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2019] [Indexed: 12/17/2022] Open
Abstract
The borderline between virulence and efficacy in live attenuated vaccine strains is often blurred and this is also the case for the Bacillus Calmette–Guérin (BCG), the only currently licensed anti-tuberculosis vaccine used on a large, global scale, which was obtained almost 100 years ago. While BCG is more than 99% identical at the genome level to
Mycobacterium tuberculosis, the causative pathogen of human tuberculosis, some important differences in virulence factors cause naturally irreversible attenuation and safety of this vaccine in the immunocompetent host. Some of these virulence factors are involved in persistence capacities of the vaccine strains and also represent strong immunogens, responsible for inducing different host signaling pathways, which have to be taken into consideration for the development of revised and new vaccine strains. Here we discuss a number of selected mycobacterial features in relation to their biological functions and potential impact on virulence and vaccine efficacy.
Collapse
Affiliation(s)
- Vera M Kroesen
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France.,Faculty VI, University of Oldenburg, Oldenburg, Germany
| | - Jan Madacki
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France
| | - Wafa Frigui
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France
| | - Fadel Sayes
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France
| | - Roland Brosch
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut Pasteur, Paris, France
| |
Collapse
|
15
|
Dijkman K, Vervenne RAW, Sombroek CC, Boot C, Hofman SO, van Meijgaarden KE, Ottenhoff THM, Kocken CHM, Haanstra KG, Vierboom MPM, Verreck FAW. Disparate Tuberculosis Disease Development in Macaque Species Is Associated With Innate Immunity. Front Immunol 2019; 10:2479. [PMID: 31736945 PMCID: PMC6838139 DOI: 10.3389/fimmu.2019.02479] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
While tuberculosis continues to afflict mankind, the immunological mechanisms underlying TB disease development are still incompletely understood. Advanced preclinical models for TB research include both rhesus and cynomolgus macaques (Macaca mulatta and Macaca fascicularis, respectively), with rhesus typically being more susceptible to acute progressive TB disease than cynomolgus macaques. To determine which immune mechanisms are responsible for this dissimilar disease development, we profiled a broad range of innate and adaptive responses, both local and peripheral, following experimental pulmonary Mycobacterium tuberculosis (Mtb) infection of both species. While T-cell and antibody responses appeared indistinguishable, we identified anti-inflammatory skewing of peripheral monocytes in rhesus and a more prominent local pro-inflammatory cytokine release profile in cynomolgus macaques associated with divergent TB disease outcome. Importantly, these differences were detectable both before and early after infection. This work shows that inflammatory and innate immune status prior to and at early stages after infection, critically affects outcome of TB infection.
Collapse
Affiliation(s)
- Karin Dijkman
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Richard A W Vervenne
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Claudia C Sombroek
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Charelle Boot
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Sam O Hofman
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | | | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Centre, Leiden, Netherlands
| | - Clemens H M Kocken
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Krista G Haanstra
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Michel P M Vierboom
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Frank A W Verreck
- TB Research Group, Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| |
Collapse
|
16
|
Ruan C, Li J, Niu J, Li P, Huang Y, Li X, Duan W, Yan S, Zhen J, Xie J. Mycobacterium tuberculosis Rv0426c promotes recombinant mycobacteria intracellular survival via manipulating host inflammatory cytokines and suppressing cell apoptosis. INFECTION GENETICS AND EVOLUTION 2019; 77:104070. [PMID: 31614213 DOI: 10.1016/j.meegid.2019.104070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 01/19/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) is still a leading cause of death worldwide. M. tuberculosis has evolved multipronged strategies to subvert host immune defenses and establish an immunologically privileged niche in macrophages. Rv0426c has been predicted to be an effector involved in the Mtb-host interactions. To investigate the potential role played by Rv0426c, we constructed recombinant M. smegmatis strains with heterologous expression of Rv0426c. We observed that Rv0426c recombinants became more susceptible to various stresses by increasing cell wall permeability, however with elevated early survival rate within macrophages. This was accompanied by decreased levels of pro-inflammatory cytokines and host cell apoptosis. The data suggested that Rv0426c was a new player involved in the interactions between Mtb and macrophages.
Collapse
Affiliation(s)
- Cao Ruan
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Jiang Li
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Jingjing Niu
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Ping Li
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Yu Huang
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Xue Li
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Wei Duan
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Shuangquan Yan
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Junfeng Zhen
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China.
| |
Collapse
|
17
|
Kashihara E, Fujita K, Uchida N, Yamamoto Y, Mio T, Koyama H. Case Report: Disseminated Mycobacterium kansasii Disease in a Patient with Anti-Interferon-Gamma Antibody. Am J Trop Med Hyg 2019; 101:1066-1069. [PMID: 31549614 DOI: 10.4269/ajtmh.19-0047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Disseminated nontuberculous mycobacterial (NTM) infections usually occur in severely immunosuppressed patients. These infections may also occur in previously immunocompetent patients with acquired anti-interferon-gamma antibodies (anti-IFN-γ Abs). A previously healthy 33-year-old man presented with a 3-week history of cough and fever. Chest computed tomography showed air-space consolidation in the middle lobe of the right lung and enlargement of the supraclavicular, mediastinal, and hilar lymph nodes. Tissue samples obtained via mediastinoscopy showed granuloma formation with acid-fast bacteria; cultures from the tissue revealed Mycobacterium kansasii. Accordingly, a diagnosis of disseminated M. kansasii disease was made. The positive control tested negative in the QuantiFERON-TB Gold In-tube test, suggesting the presence of anti-IFN-γ Abs. The ELISA test for anti-IFN-γ Abs demonstrated an increased titer. Antimycobacterial drug treatments were initiated after diagnosis. His symptoms improved over 2 months, and he remains well on outpatient management. Disseminated M. kansasii disease is a very rare condition suggestive of immunosuppression. Testing for anti-IFN-γ antibodies might be important in all cases of disseminated M. kansasii disease.
Collapse
Affiliation(s)
- Eriko Kashihara
- Division of General Medicine, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Kohei Fujita
- Division of Respiratory Medicine, Center for Respiratory Diseases, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Naohiro Uchida
- Division of Respiratory Medicine, Center for Respiratory Diseases, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Yuki Yamamoto
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Division of Respiratory Medicine, Center for Respiratory Diseases, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Tadashi Mio
- Division of Respiratory Medicine, Center for Respiratory Diseases, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Hiroshi Koyama
- Division of General Medicine, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| |
Collapse
|
18
|
Rv2626c and Rv2032 activate TH1 response and downregulate regulatory T cells in peripheral blood mononuclear cells of tuberculosis patients. Comp Immunol Microbiol Infect Dis 2019; 62:46-53. [DOI: 10.1016/j.cimid.2018.11.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/19/2022]
|
19
|
Liu Q, Gao Y, Zhang B, Sun F, Yang Q, Liu Y, Wu J, Chen K, Weng X, Zhang W, Huang R, Shao L. Cytokine profiles in cerebrospinal fluid of patients with meningitis at a tertiary general hospital in China. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2018; 53:216-224. [PMID: 30448438 DOI: 10.1016/j.jmii.2018.08.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/18/2018] [Accepted: 08/18/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND There has been a great deal of evidence indicating that cytokines participate in meningeal inflammation. Different cytokine profiles may be presented in central nervous system (CNS) infection due to different pathogens. We have attempted to investigate cytokine profiles in cerebrospinal fluid (CSF) of patients with CNS infection. METHODS Forty-three patients with CNS infection including tuberculous meningitis, purulent meningitis and cryptococcal meningitis were enrolled and 11 patients with normal CSF were enrolled as control group. The concentrations of Th1-, Th2- and Th17-type cytokines in CSF were detected using multiplex cytokine assay. Furthermore, the correlation between CSF cytokines and CSF parameters in CNS infection was analyzed. RESULTS The CSF levels of IL-1β, IL-4, IL-6, IL-10, IL-17, IL-23, IL-33, IFN-γ, TNF-α and sCD40L among the patients with CNS infection were all higher than control group (all P < 0.05). A remarkable elevation of CSF IL-6 in the patients with CNS infection was observed with the least overlap of the CSF concentrations compared to controls. Moreover, CSF IL-6 levels were strongly negatively correlated with CSF glucose and the CSF/blood glucose ratio (r = -0.4375, P = 0.0042; r = -0.4991, P = 0.0009). CONCLUSIONS The excessive activation of immune response characterized by elevated levels of CSF Th1-, Th2- and Th17-type cytokines has been observed during CNS infection. Furthermore, we observed negative correlations between CSF IL-6 levels and CSF glucose and CSF/blood glucose ratio in CNS infection. And we suggested that combined CSF IL-6 levels with CSF glucose may serve as a novel biomarker pool for the differential of CNS infection.
Collapse
Affiliation(s)
- Qianqian Liu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yan Gao
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Bingyan Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Feng Sun
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qingluan Yang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuanyuan Liu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Wu
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Kun Chen
- Department of Clinical Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xinhua Weng
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wenhong Zhang
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China; Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, Shanghai Medical College, Institutes of Biomedical Science, Fudan University, Shanghai 200032, China
| | - Ruofan Huang
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Lingyun Shao
- Department of Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
20
|
Gong Z, Kuang Z, Li H, Li C, Ali MK, Huang F, Li P, Li Q, Huang X, Ren S, Li J, Xie J. Regulation of host cell pyroptosis and cytokines production by Mycobacterium tuberculosis effector PPE60 requires LUBAC mediated NF-κB signaling. Cell Immunol 2018; 335:41-50. [PMID: 30415762 DOI: 10.1016/j.cellimm.2018.10.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/13/2018] [Accepted: 10/30/2018] [Indexed: 10/28/2022]
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis infection, remains a global public health threat. The success of M. tuberculosis largely contributes to its manipulation of host cell fate. The role of M. tuberculosis PE/PPE family effectors in the host destiny was intensively explored. In this study, the role of PPE60 (Rv3478) was characterized by using Rv3478 recombinant M. smegmatis. PPE60 can promote host cell pyroptosis via caspases/NLRP3/gasdermin. The production of pro-inflammatory cytokines, such as IL-1β, IL-6, IL-12p40 and TNF-α was altered by PPE60. We found that LUBAC was involved in PPE60-elicited NF-κB signaling by using Linear Ubiquitin Chain Assembly Complex (LUBAC)-specific inhibitor gliotoxin. The PPE60 recombinant M. smegmatis survival rate within macrophages is increased, as well as elevated resistance to stresses such as low pH, surface stresses and antibiotics exposure. For a first time it is firstly reported that M. tuberculosis effector PPE60 can modulate the host cell fate via LUBAC-mediated NF-κB signaling.
Collapse
Affiliation(s)
- Zhen Gong
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Zhongmei Kuang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Hui Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China; Institute of Chengdu Medical College, School of Laboratory Medicine, No 783 Xindu Avenue, Chengdu, Sichuan 610083, China
| | - Chunyan Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Md Kaisar Ali
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Fujing Huang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Ping Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Qiming Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Xue Huang
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Sai Ren
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Jiang Li
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China
| | - Jianping Xie
- Institute of Modern Biopharmaceuticals, State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Southwest University, Chongqing, China.
| |
Collapse
|
21
|
Fletcher HA. Systems approaches to correlates of protection and progression to TB disease. Semin Immunol 2018; 39:81-87. [PMID: 30316693 DOI: 10.1016/j.smim.2018.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/01/2018] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) is the leading cause of death due to a single infectious disease and an effective vaccine would substantially accelerate global efforts to control TB. An immune correlate of protection (CoP) from TB disease could aid vaccine optimization and licensure. This paper summarises opportunities for identifying CoP and highlights results from correlates of risk studies. Although we don't have CoP, there are ongoing efficacy trials with both disease and infection endpoints which provide opportunities for such an analysis. Transcriptomics has successfully identified robust CoR, with transcripts found in the Type I IFN pathway. Correlates of lower risk include BCG antigen specific IFN-γ and natural killer cells. Collating evidence from multiple studies using a range of systems approaches supports a role for IFN-γ in protection from TB disease. In addition, the cells that express the IFN-γ receptor are also important in protective immunity. Protection is a culmination not only of the amount of IFN-γ produced by T cells and NK cells but by the ability of IFN-γ receptor expressing monocytes to respond to IFN-γ. To better understand IFN-γ as a correlate we need to understand host-factors such as age, sex, co-infection, nutritional status and stress which may alter or impair the ability of cells to respond to IFN-γ. These studies highlight recent advances in our understanding of the immune mechanisms of TB disease risk and show the importance of whole systems approaches to correlates of risk analysis. CoP may be useful tools for specific vaccine products in specific populations, but a well-designed CoR analysis can identify novel immune mechanisms and provide insights critical for the development of new and better TB vaccines.
Collapse
Affiliation(s)
- Helen A Fletcher
- TB Centre, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
22
|
Lai HC, Chang CJ, Lin CS, Wu TR, Hsu YJ, Wu TS, Lu JJ, Martel J, Ojcius DM, Ku CL, Young JD, Lu CC. NK Cell-Derived IFN-γ Protects against Nontuberculous Mycobacterial Lung Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:1478-1490. [PMID: 30061197 DOI: 10.4049/jimmunol.1800123] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 07/05/2018] [Indexed: 01/09/2023]
Abstract
In developed countries, pulmonary nontuberculous mycobacteria (NTM) infections are more prevalent than Mycobacterium tuberculosis infections. Given the differences in the pathogenesis of NTM and M. tuberculosis infections, separate studies are needed to investigate the pathological effects of NTM pathogens. Our previous study showed that anti-IFN-γ autoantibodies are detected in NTM-infected patients. However, the role of NK cells and especially NK cell-derived IFN-γ in this context has not been studied in detail. In the current study, we show that NK1.1 cell depletion increases bacterial load and mortality in a mouse model of pulmonary NTM infection. NK1.1 cell depletion exacerbates NTM-induced pathogenesis by reducing macrophage phagocytosis, dendritic cell development, cytokine production, and lung granuloma formation. Similar pathological phenomena are observed in IFN-γ-deficient (IFN-γ-/-) mice following NTM infection, and adoptive transfer of wild-type NK cells into IFN-γ-/- mice considerably reduces NTM pathogenesis. Injection of rIFN-γ also prevents NTM-induced pathogenesis in IFN-γ-/- mice. We observed that NK cells represent the main producers of IFN-γ in the lungs and production starts as soon as 1 d postinfection. Accordingly, injection of rIFN-γ into IFN-γ-/- mice 1 d (but not 2 wk) postinfection significantly improves immunity against NTM infection. NK cells also stimulate mycobacterial killing and IL-12 production by macrophages. Our results therefore indicate that IFN-γ production by NK cells plays an important role in activating and enhancing innate and adaptive immune responses at early stages of pulmonary NTM infection.
Collapse
Affiliation(s)
- Hsin-Chih Lai
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taoyuan 33303, Taiwan.,Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Gueishan, Taoyuan 33303, Taiwan
| | - Chih-Jung Chang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Chuan-Sheng Lin
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Microbiota Research Center, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Research Center for Emerging Viral Infections, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Tsung-Ru Wu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Ya-Jing Hsu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - Ting-Shu Wu
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - Jang-Jih Lu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Department of Biomedical Sciences, Arthur A. Dugoni School of Dentistry, University of the Pacific, San Francisco, CA 94103
| | - Cheng-Lung Ku
- Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Division of Infectious Diseases, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Laboratory of Human Immunology and Infectious Diseases, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan
| | - John D Young
- Center for Molecular and Clinical Immunology, Chang Gung University, Gueishan, Taoyuan 33302, Taiwan.,Chang Gung Immunology Consortium, Linkou Chang Gung Memorial Hospital, Gueishan, Taoyuan 33305, Taiwan.,Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, NY 10021; and
| | - Chia-Chen Lu
- Department of Respiratory Therapy, Fu Jen Catholic University, Xinzhuang, New Taipei City 24205, Taiwan
| |
Collapse
|
23
|
Coppola M, Ottenhoff TH. Genome wide approaches discover novel Mycobacterium tuberculosis antigens as correlates of infection, disease, immunity and targets for vaccination. Semin Immunol 2018; 39:88-101. [PMID: 30327124 DOI: 10.1016/j.smim.2018.07.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 07/02/2018] [Indexed: 01/15/2023]
Abstract
Every day approximately six thousand people die of Tuberculosis (TB). Its causative agent, Mycobacterium tuberculosis (Mtb), is an ancient pathogen that through its evolution developed complex mechanisms to evade immune surveillance and acquire the ability to establish persistent infection in its hosts. Currently, it is estimated that one-fourth of the human population is latently infected with Mtb and among those infected 3-10% are at risk of developing active TB disease during their lifetime. The currently available diagnostics are not able to detect this risk group for prophylactic treatment to prevent transmission. Anti-TB drugs are available but only as long regimens with considerable side effects, which could both be reduced if adequate tests were available to monitor the response of TB to treatment. New vaccines are also urgently needed to substitute or boost Bacille Calmette-Guérin (BCG), the only approved TB vaccine: although BCG prevents disseminated TB in infants, it fails to impact the incidence of pulmonary TB in adults, and therefore has little effect on TB transmission. To achieve TB eradication, the discovery of Mtb antigens that effectively correlate with the human response to infection, with the curative host response following TB treatment, and with natural as well as vaccine induced protection will be critical. Over the last decade, many new Mtb antigens have been found and proposed as TB biomarkers and vaccine candidates, but only a very small number of these is being used in commercial diagnostic tests or is being assessed as candidate TB vaccine antigens in human clinical trials, aiming to prevent infection, disease or disease recurrence following treatment. Most of these antigens were discovered decades ago, before the complete Mtb genome sequence became available, and thus did not harness the latest insights from post-genomic antigen discovery strategies and genome wide approaches. These have, for example, revealed critical phase variation in Mtb replication and accompanying gene -and therefore antigen- expression patterns. In this review, we present a brief overview of past methodologies, and subsequently focus on the most important recent Mtb antigen discovery studies which have mined the Mtb antigenome through "unbiased" genome wide approaches. We compare the results for these approaches -as far as we know for the first time-, highlight Mtb antigens that have been identified independently by different strategies and present a comprehensive overview of the Mtb antigens thus discovered.
Collapse
Affiliation(s)
- Mariateresa Coppola
- Dept. Infectious Diseases, LUMC, PO Box 9600, 2300RC Leiden, The Netherlands.
| | - Tom Hm Ottenhoff
- Dept. Infectious Diseases, LUMC, PO Box 9600, 2300RC Leiden, The Netherlands
| |
Collapse
|
24
|
Abstract
Protective immunity in tuberculosis (TB) is subject of debate in the TB research community, as this is key to fully understand TB pathogenesis and to develop new promising tools for TB diagnosis and prognosis as well as a more efficient TB vaccine. IFN-γ producing CD4+ T cells are key in TB control, but may not be sufficient to provide protection. Additional subsets have been identified that contribute to protection such as multifunctional and cytolytic T-cell subsets, including classical and nonclassical T cells as well as novel innate immune cell subsets resulting from trained immunity. However, to define protective immune responses against TB, the complexity of balancing TB immunity also has to be considered. In this review, insights into effector cell immunity and how this is modulated by regulatory cells, associated comorbidities and the host microbiome, is discussed. We systematically map how different suppressive immune cell subsets may affect effector cell responses at the local site of infection. We also dissect how common comorbidities such as HIV, helminths and diabetes may bias protective TB immunity towards pathogenic and regulatory responses. Finally, also the composition and diversity of the microbiome in the lung and gut could affect host TB immunity. Understanding these various aspects of the immunological balance in the human host is fundamental to prevent TB infection and disease.
Collapse
Affiliation(s)
- Susanna Brighenti
- Karolinska Institutet, Department of Medicine, Center for Infectious Medicine (CIM), Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Simone A. Joosten
- Leiden University Medical Center, Department of Infectious Diseases, Leiden, The Netherlands
| |
Collapse
|
25
|
Richardus R, van Hooij A, van den Eeden SJF, Wilson L, Alam K, Richardus JH, Geluk A. BCG and Adverse Events in the Context of Leprosy. Front Immunol 2018; 9:629. [PMID: 29670618 PMCID: PMC5893643 DOI: 10.3389/fimmu.2018.00629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/13/2018] [Indexed: 11/21/2022] Open
Abstract
Background Notwithstanding its beneficial immunoprophylactic outcomes regarding leprosy and childhood TB, BCG vaccination may cause adverse events, particularly of the skin. However, this local hyper-immune reactivity cannot be predicted before vaccination, nor is its association with protection against leprosy known. In this study we investigated the occurrence of adverse events after BCG (re)vaccination in contacts of leprosy patients and analyzed whether the concomitant systemic anti-mycobacterial immunity was associated with these skin manifestations. Methods Within a randomized controlled BCG vaccination trial in Bangladesh, 14,828 contacts of newly diagnosed leprosy patients received BCG vaccination between 2012 and 2017 and were examined for adverse events 8 to 12 weeks post-vaccination. From a selection of vaccinated contacts, venous blood was obtained at follow-up examination and stimulated with Mycobacterium leprae (M. leprae) antigens in overnight whole-blood assays (WBA). M. leprae phenolic glycolipid-I-specific antibodies and 32 cytokines were determined in WBAs of 13 individuals with and 13 individuals without adverse events after vaccination. Results Out of the 14,828 contacts who received BCG vaccination, 50 (0.34%) presented with adverse events, mainly (80%) consisting of skin ulcers. Based on the presence of BCG scars, 30 of these contacts (60%) had received BCG in this study as a booster vaccination. Similar to the pathological T-cell immunity observed for tuberculoid leprosy patients, contacts with adverse events at the site of BCG vaccination showed elevated IFN-γ levels in response to M. leprae-specific proteins in WBA. However, decreased levels of sCD40L in serum and GRO (CXCL1) in response to M. leprae simultaneously indicated less T-cell regulation in these individuals, potentially causing uncontrolled T-cell immunity damaging the skin. Conclusion Skin complications after BCG vaccination present surrogate markers for protective immunity against leprosy, but also indicate a higher risk of developing tuberculoid leprosy. Clinical Trial Registration Netherlands Trial Register: NTR3087.
Collapse
Affiliation(s)
- Renate Richardus
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands.,Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Anouk van Hooij
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Louis Wilson
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Korshed Alam
- Rural Health Program, The Leprosy Mission International Bangladesh, Nilphamari, Bangladesh
| | - Jan Hendrik Richardus
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
26
|
Joosten SA, van Meijgaarden KE, Arend SM, Prins C, Oftung F, Korsvold GE, Kik SV, Arts RJ, van Crevel R, Netea MG, Ottenhoff TH. Mycobacterial growth inhibition is associated with trained innate immunity. J Clin Invest 2018; 128:1837-1851. [PMID: 29461976 DOI: 10.1172/jci97508] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/13/2018] [Indexed: 12/29/2022] Open
Abstract
The lack of defined correlates of protection hampers development of vaccines against tuberculosis (TB). In vitro mycobacterial outgrowth assays are thought to better capture the complexity of the human host/Mycobacterium tuberculosis (Mtb) interaction. Here, we used a mycobacterial growth inhibition assay (MGIA) based on peripheral blood mononuclear cells to investigate the capacity to control outgrowth of bacille Calmette-Guérin (BCG). Interestingly, strong control of BCG outgrowth was observed almost exclusively in individuals with recent exposure to Mtb, but not in (long-term) latent TB infection, and only modestly in BCG vaccinees. Mechanistically, control of mycobacterial outgrowth strongly correlated with the presence of a CD14dim monocyte population, but also required the presence of T cells. The nonclassical monocytes produced CXCL10, and CXCR3 receptor blockade inhibited the capacity to control BCG outgrowth. Expression of CXCR3 splice variants was altered in recently Mtb-exposed individuals. Cytokines previously associated with trained immunity were detected in MGIA supernatants, and CXCL9, CXCL10, and CXCL11 represent new markers of trained immunity. These data indicate that CXCR3 ligands are associated with trained immunity and are critical factors in controlling mycobacterial outgrowth. In conclusion, control of mycobacterial outgrowth early after exposure to Mtb is the result of trained immunity mediated by a CXCL10-producing nonclassical CD14dim monocyte subset.
Collapse
Affiliation(s)
- Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Sandra M Arend
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Corine Prins
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Fredrik Oftung
- Division of Infection Control and Environmental Health, Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Gro Ellen Korsvold
- Division of Infection Control and Environmental Health, Department of Infectious Disease Immunology, Norwegian Institute of Public Health, Oslo, Norway
| | - Sandra V Kik
- KNCV Tuberculosis Foundation, The Hague, Netherlands
| | - Rob Jw Arts
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Tom Hm Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
27
|
Coppola M, van den Eeden SJF, Robbins N, Wilson L, Franken KLMC, Adams LB, Gillis TP, Ottenhoff THM, Geluk A. Vaccines for Leprosy and Tuberculosis: Opportunities for Shared Research, Development, and Application. Front Immunol 2018. [PMID: 29535713 PMCID: PMC5834475 DOI: 10.3389/fimmu.2018.00308] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tuberculosis (TB) and leprosy still represent significant public health challenges, especially in low- and lower middle-income countries. Both poverty-related mycobacterial diseases require better tools to improve disease control. For leprosy, there has been an increased emphasis on developing tools for improved detection of infection and early diagnosis of disease. For TB, there has been a similar emphasis on such diagnostic tests, while increased research efforts have also focused on the development of new vaccines. Bacille Calmette–Guérin (BCG), the only available TB vaccine, provides insufficient and inconsistent protection to pulmonary TB in adults. The impact of BCG on leprosy, however, is significant, and the introduction of new TB vaccines that might replace BCG could, therefore, have serious impact also on leprosy. Given the similarities in antigenic makeup between the pathogens Mycobacterium tuberculosis (Mtb) and M. leprae, it is well possible, however, that new TB vaccines could cross-protect against leprosy. New TB subunit vaccines currently evaluated in human phase I and II studies indeed often contain antigens with homologs in M. leprae. In this review, we discuss pre-clinical studies and clinical trials of subunit or whole mycobacterial vaccines for TB and leprosy and reflect on the development of vaccines that could provide protection against both diseases. Furthermore, we provide the first preclinical evidence of such cross-protection by Mtb antigen 85B (Ag85B)-early secretory antigenic target (ESAT6) fusion recombinant proteins in in vivo mouse models of Mtb and M. leprae infection. We propose that preclinical integration and harmonization of TB and leprosy research should be considered and included in global strategies with respect to cross-protective vaccine research and development.
Collapse
Affiliation(s)
- Mariateresa Coppola
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | | | - Naoko Robbins
- The National Hansen's Disease Programs, Baton Rouge, LA, United States
| | - Louis Wilson
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Linda B Adams
- The National Hansen's Disease Programs, Baton Rouge, LA, United States
| | - Tom P Gillis
- The National Hansen's Disease Programs, Baton Rouge, LA, United States
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
28
|
Tugal-Tutkun I, Pavesio C, De Cordoue A, Bernard-Poenaru O, Gül A. Use of Gevokizumab in Patients with Behçet’s Disease Uveitis: An International, Randomized, Double-Masked, Placebo-Controlled Study and Open-Label Extension Study. Ocul Immunol Inflamm 2018; 26:1023-1033. [DOI: 10.1080/09273948.2017.1421233] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ilknur Tugal-Tutkun
- Department of Ophthalmology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Carlos Pavesio
- Moorfields Eye Hospital and Biomedical Research Center, NHS Foundation Trust, London, UK
| | - Agnès De Cordoue
- Institut de Recherches Internationales Servier, Suresnes, France
| | | | - Ahmet Gül
- Department of Internal Medicine, Division of Rheumatology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
29
|
Susceptibility to mycobacterial disease due to mutations in IL-12Rβ1 in three Iranian patients. Immunogenetics 2017; 70:373-379. [PMID: 29256176 PMCID: PMC5943370 DOI: 10.1007/s00251-017-1041-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 10/26/2017] [Indexed: 10/31/2022]
Abstract
In the last decade, autosomal recessive interleukin-12 receptor β1 (IL-12Rβ1) deficiency, the most common cause of Mendelian susceptibility to mycobacterial disease (MSMD), has been diagnosed in a few children and adults with severe tuberculosis in Iran. Here, we report three cases referred to the Immunology, Asthma and Allergy ward at the National Research Institute of Tuberculosis and Lung Diseases (NRITLD) at Masih Daneshvari Hospital from 2012 to 2017 with Mycobacterium tuberculosis and non-tuberculous mycobacteria infections due to defects in IL-12Rβ1 but with different clinical manifestations. All three were homozygous for either an IL-12Rβ1 missense or nonsense mutation that caused the IL-12Rβ1 protein not to be expressed on the cell membrane and completely abolished the cellular response to recombinant IL-12. Our findings suggest that the presence of IL-12Rβ1 deficiency should be determined in children with mycobacterial infections at least in countries with a high prevalence of parental consanguinity and in areas endemic for TB like Iran.
Collapse
|
30
|
Ahmed A, Rakshit S, Vyakarnam A. HIV-TB co-infection: mechanisms that drive reactivation of Mycobacterium tuberculosis in HIV infection. Oral Dis 2017; 22 Suppl 1:53-60. [PMID: 27109273 DOI: 10.1111/odi.12390] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
HIV infection predisposes the host to tuberculosis by impairing the hosts' immune system principally by killing and altering CD4 T-cell function. How HIV infection disrupts CD4 T-cell function, which specifically compromises host immunity to Mycobacterium tuberculosis, is poorly understood and is a critical roadblock in developing better vaccine- or immune-based strategies to control and monitor TB in HIV-infected subjects. This review considers key pathways that are altered in HIV-infected subjects that impair anti-TB immunity.
Collapse
Affiliation(s)
- A Ahmed
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - S Rakshit
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - A Vyakarnam
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India.,Department of Infectious Diseases, King's College London, London, UK
| |
Collapse
|
31
|
Mycobacterium tuberculosis PPE44 (Rv2770c) is involved in response to multiple stresses and promotes the macrophage expression of IL-12 p40 and IL-6 via the p38, ERK, and NF-κB signaling axis. Int Immunopharmacol 2017; 50:319-329. [DOI: 10.1016/j.intimp.2017.06.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/11/2017] [Accepted: 06/26/2017] [Indexed: 11/19/2022]
|
32
|
Coppola M, Arroyo L, van Meijgaarden KE, Franken KL, Geluk A, Barrera LF, Ottenhoff THM. Differences in IgG responses against infection phase related Mycobacterium tuberculosis (Mtb) specific antigens in individuals exposed or not to Mtb correlate with control of TB infection and progression. Tuberculosis (Edinb) 2017; 106:25-32. [PMID: 28802401 DOI: 10.1016/j.tube.2017.06.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/02/2017] [Accepted: 06/04/2017] [Indexed: 11/26/2022]
Abstract
Tuberculosis (TB) occurs in only 3-10% of Mycobacterium tuberculosis (Mtb) infected individuals, suggesting that natural immunity can contain Mtb infection, although this remains poorly understood. Next to T-cells, a potentially protective role for B-cells and antibodies has emerged recently. However, the Mtb antigens involved remain ill-defined. Here, we investigated in a TB-endemic setting IgG levels against 15 Mtb antigens, representing various phases of Mtb infection and known to be potent human T-cell antigens. IgG levels against ESAT6/CFP10, Rv0440, Rv0867c, Rv1737c, Rv2029c, Rv2215, Rv2389c, Rv3616c and Mtb purified protein derivative (PPD) were higher in TB patients than in endemic and non-endemic controls. The only exception was Rv1733c that was preferentially recognized by antibodies from endemic controls compared to TB patients and non-endemic controls, suggesting a potential correlation with control of TB infection and progression. In patients, IgG levels against Ag85B and Rv2029c correlated with Mtb loads, while immunoglobulins against Rv0440 differed between genders. Our results support the potential role of certain Mtb antigen-(Rv1733c) specific antibodies in the control of TB infection and progression, while other Mtb antigen-specific antibodies correlate with TB disease activity and bacillary loads. The findings for Rv1733c agree with previous T-cell results and have implications for including antibody-mediated immunity in designing new strategies to control TB.
Collapse
Affiliation(s)
- Mariateresa Coppola
- Dept. of Infectious Diseases, Leiden University Medical Center, PO Box 9600, 2300, RC Leiden, The Netherlands.
| | - Leonar Arroyo
- Grupo de Inmunología Cellular e Inmunogenética (GICIG), Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Krista E van Meijgaarden
- Dept. of Infectious Diseases, Leiden University Medical Center, PO Box 9600, 2300, RC Leiden, The Netherlands
| | - Kees Lmc Franken
- Dept. of Infectious Diseases, Leiden University Medical Center, PO Box 9600, 2300, RC Leiden, The Netherlands
| | - Annemieke Geluk
- Dept. of Infectious Diseases, Leiden University Medical Center, PO Box 9600, 2300, RC Leiden, The Netherlands
| | - Luis F Barrera
- Grupo de Inmunología Cellular e Inmunogenética (GICIG), Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Tom H M Ottenhoff
- Dept. of Infectious Diseases, Leiden University Medical Center, PO Box 9600, 2300, RC Leiden, The Netherlands
| |
Collapse
|
33
|
Lee TL, Agrawal R, Tan JYL, Ong KH, Wong CS, Ho SL. Disseminated nontuberculous mycobacterial infection with multifocal retinitis and vasculitis in an immunocompromised patient with anti-IFN-ɣ autoantibodies. J Ophthalmic Inflamm Infect 2016; 6:39. [PMID: 27771870 PMCID: PMC5075326 DOI: 10.1186/s12348-016-0106-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 09/27/2016] [Indexed: 11/16/2022] Open
Abstract
Background Nontuberculous mycobacteria (NTM) are found ubiquitously in the environment. Since exposure to NTM is universal, infection likely represents underlying host susceptibility factors. Anti-IFN-ɣ autoantibodies have been described previously in patients with NTM. Up to 88 % of patients with disseminated NTM or other opportunistic infections have high-titer anti-IFN-ɣ autoantibodies, compared with 2 % of patients with TB and healthy controls. Findings We report a unique presentation of a patient with anti-IFN-ɣ autoantibodies with disseminated NTM infection who presents with panuveitis with multifocal retinitis and vasculitis. Treatment with systemic anti tubercular therapy resulted in complete clinical resolution with good visual recovery. Conclusions Patients with anti-IFN-ɣ autoantibodies present with a novel syndrome that links autoimmunity and immunodeficiency. This case emphasizes the importance of testing for anti-IFN-ɣ autoantibodies in patients with disseminated mycobacterial infection.
Collapse
Affiliation(s)
- Tian Loon Lee
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | - Rupesh Agrawal
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, 308433, Singapore
| | - Jackie Yu-Ling Tan
- National Healthcare Group, Department of General Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Kiat Hoe Ong
- National Healthcare Group, Department of Haematology, Tan Tock Seng Hospital, Singapore, Singapore
| | - Chen Seong Wong
- National Healthcare Group, Department of Infectious Disease, Tan Tock Seng Hospital, Singapore, Singapore
| | - Su Ling Ho
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, 308433, Singapore.
| |
Collapse
|
34
|
Su H, Kong C, Zhu L, Huang Q, Luo L, Wang H, Xu Y. PPE26 induces TLR2-dependent activation of macrophages and drives Th1-type T-cell immunity by triggering the cross-talk of multiple pathways involved in the host response. Oncotarget 2016; 6:38517-37. [PMID: 26439698 PMCID: PMC4770718 DOI: 10.18632/oncotarget.5956] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 09/12/2015] [Indexed: 12/27/2022] Open
Abstract
The pathophysiological functions and the underlying molecular basis of PE /PPE proteins of M. tuberculosis remain largely unknown. In this study, we focused on the link between PPE26 and host response. We demonstrated that PPE26 can induce extensive inflammatory responses in macrophages through triggering the cross-talk of multiple pathways involved in the host response, as revealed by iTRAQ-based subcellular quantitative proteomics. We observed that PPE26 is able to specifically bind to TLR2 leading to the subsequent activation of MAPKs and NF-κB signaling. PPE26 functionally stimulates macrophage activation by augmenting pro-inflammatory cytokine production (TNF-α, IL-6 and IL-12 p40) and the expression of cell surface markers (CD80, CD86, MHC class I and II). We observed that PPE26-treated macrophages effectively polarizes naïve CD4+ T cells to up-regulate CXCR3 expression, and to secrete IFN-γ and IL-2, indicating PPE26 contributes to the Th1 polarization during the immune response. Importantly, rBCG::PPE26 induces stronger antigen-specific TNF-α and IFN-γ activity, and higher levels of the Th1 cytokines TNF-α and IFN-γ comparable to BCG. Moreover, PPE26 effectively induces the reciprocal expansion of effector/memory CD4+/CD8+ CD44highCD62Llow T cells in the spleens of mice immunized with this strain. These results suggest that PPE26 may be a TLR2 agonist that stimulates innate immunity and adaptive immunity, indicating that PPE26 is a potential antigen for the rational design of an efficient vaccine against M. tuberculosis.
Collapse
Affiliation(s)
- Haibo Su
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Cong Kong
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Lin Zhu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Qi Huang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Liulin Luo
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China.,Department of Clinical Laboratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Honghai Wang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| | - Ying Xu
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Science, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Multifunctional T Cell Response to DosR and Rpf Antigens Is Associated with Protection in Long-Term Mycobacterium tuberculosis-Infected Individuals in Colombia. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:813-824. [PMID: 27489136 DOI: 10.1128/cvi.00217-16] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/27/2016] [Indexed: 01/03/2023]
Abstract
Multifunctional T cells have been shown to be protective in chronic viral infections. In mycobacterial infections, however, evidence for a protective role of multifunctional T cells remains inconclusive. Short-term cultures of peripheral blood mononuclear cells stimulated with the Mycobacterium tuberculosis RD1 antigens 6-kDa early secretory antigenic target (ESAT6) and 10-kDa culture filtrate antigen (CFP10), which are induced in the early infection phase, have been mainly used to assess T cell multifunctionality, although long-term culture assays have been proposed to be more sensitive than short-term assays for assessment of memory T cells, which are essential for long-term immunity. Here we used a long-term culture assay system to study the T cell immune responses to the M. tuberculosis latency-associated DosR antigens and reactivation-associated Rpf antigens, compared to ESAT6 and CFP10, in patients with pulmonary tuberculosis (PTB) and household contacts of PTB patients with long-term latent tuberculosis infection (ltLTBI), in a community in which M. tuberculosis is endemic. Our results showed that the DosR antigens Rv1737c (narK2) and Rv2029c (pfkB) and the Rv2389c (rpfD) antigen of M. tuberculosis induced higher frequencies of CD4+ or CD8+ mono- or bifunctional (but not multifunctional) T cells producing interferon gamma (IFN-γ) and/or tumor necrosis alpha (TNF-α) in ltLTBI, compared to PTB. Moreover, the frequencies of CD4+ and/or CD8+ T cells with a CD45RO+ CD27+ phenotype were higher in ltLTBI than in PTB. Thus, the immune responses to selected DosR and Rpf antigens may be associated with long-term latency, correlating with protection from M. tuberculosis reactivation in ltLTBI. Further study of the functional and memory phenotypes may contribute to further discrimination between the different states of M. tuberculosis infections.
Collapse
|
36
|
Opportunities for the development of novel therapies based on host-microbial interactions. Pharmacol Res 2016; 112:68-83. [PMID: 27107789 DOI: 10.1016/j.phrs.2016.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/07/2016] [Accepted: 04/08/2016] [Indexed: 12/21/2022]
Abstract
Immune responses are fundamental for protecting against most infectious agents. However, there is now much evidence to suggest that the pathogenesis and tissue damage after infection are not usually related to the direct action of the replication of microorganisms, but instead to altered immune responses triggered after the contact with the pathogen. This review article discusses several mechanisms necessary for the host to protect against microbial infection and focuses in aspects that cause altered inflammation and drive immunopathology. These basic findings can ultimately reveal pathways amenable to host-directed therapy in adjunct to antimicrobial therapy for future improved control measures for many infectious diseases. Therefore, modulating the effects of inflammatory pathways may represent a new therapy during infection outcome and disease.
Collapse
|
37
|
de Oliveira FM, Trentini MM, Junqueira-Kipnis AP, Kipnis A. The mc2-CMX vaccine induces an enhanced immune response against Mycobacterium tuberculosis compared to Bacillus Calmette-Guérin but with similar lung inflammatory effects. Mem Inst Oswaldo Cruz 2016; 111:223-31. [PMID: 27074251 PMCID: PMC4830111 DOI: 10.1590/0074-02760150411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 02/24/2016] [Indexed: 01/15/2023] Open
Abstract
Although the attenuated Mycobacterium bovis Bacillus Calmette-Guérin (BCG) vaccine has been used since 1921, tuberculosis (TB) control still proceeds at a slow pace. The main reason is the variable efficacy of BCG protection against TB among adults, which ranges from 0-80%. Subsequently, the mc2-CMX vaccine was developed with promising results. Nonetheless, this recombinant vaccine needs to be compared to the standard BCG vaccine. The objective of this study was to evaluate the immune response induced by mc2-CMX and compare it to the response generated by BCG. BALB/c mice were immunised with both vaccines and challenged with Mycobacterium tuberculosis (Mtb). The immune and inflammatory responses were evaluated by ELISA, flow cytometry, and histopathology. Mice vaccinated with mc2-CMX and challenged with Mtb induced an increase in the IgG1 and IgG2 levels against CMX as well as recalled specific CD4+ T-cells that produced T-helper 1 cytokines in the lungs and spleen compared with BCG vaccinated and challenged mice. Both vaccines reduced the lung inflammatory pathology induced by the Mtb infection. The mc2-CMX vaccine induces a humoral and cellular response that is superior to BCG and is efficiently recalled after challenge with Mtb, although both vaccines induced similar inflammatory reductions.
Collapse
Affiliation(s)
- Fábio Muniz de Oliveira
- Universidade Federal de Goiás, Instituto de Patologia Tropical e Saúde
Pública, Laboratório de Bacteriologia Molecular, Goiânia, GO, Brasil
| | - Monalisa Martins Trentini
- Universidade Federal de Goiás, Instituto de Patologia Tropical e Saúde
Pública, Laboratório de Imunopatologia das Doenças Infecciosas, Goiânia, GO,
Brasil
| | - Ana Paula Junqueira-Kipnis
- Universidade Federal de Goiás, Instituto de Patologia Tropical e Saúde
Pública, Laboratório de Imunopatologia das Doenças Infecciosas, Goiânia, GO,
Brasil
| | - André Kipnis
- Universidade Federal de Goiás, Instituto de Patologia Tropical e Saúde
Pública, Laboratório de Bacteriologia Molecular, Goiânia, GO, Brasil
| |
Collapse
|
38
|
Martin C, Paibomesai M, Emam S, Gallienne J, Hine B, Thompson-Crispi K, Mallard B. Short communication: Cytokine profiles from blood mononuclear cells of dairy cows classified with divergent immune response phenotypes. J Dairy Sci 2016; 99:2364-2371. [DOI: 10.3168/jds.2015-9449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 10/23/2015] [Indexed: 12/16/2022]
|
39
|
Jia H, Liu S, Wu J, Hou S, Xin T, Guo X, Yuan W, Gao X, Zhang G, Li M, Qu H, Zhu H. Recombinant TB9.8 of Mycobacterium bovis Triggers the Production of IL-12 p40 and IL-6 in RAW264.7 Macrophages via Activation of the p38, ERK, and NF-κB Signaling Pathways. Inflammation 2016; 38:1337-46. [PMID: 25577342 DOI: 10.1007/s10753-014-0105-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The TB9.8 of Mycobacterium bovis can induce strong antigen-specific T-cell responses in proliferation assays and IFN-γ assays. However, whether and how TB9.8 activates innate immune cells remain unclear. Therefore, recombinant protein TB9.8 (rTB9.8)-induced proinflammatory cytokine profile by RAW264.7 cells was investigated and the related signaling pathway was studied. Stimulation with rTB9.8 triggered RAW264.7 cells to produce IL-6 and IL-12 p40. In addition, rTB9.8 activated the mitogen-activated protein kinase (MAPK) cascade in RAW264.7 cells by inducing the phosphorylation of extracellular signal-regulated kinase (ERK) and p38 kinase (p38) and also promoted nuclear translocation of phosphorylated p38 and ERK1/2. Furthermore, rTB9.8 activated nuclear factor κB (NF-κB) signaling pathway by inducing p65 translocation into the nucleus and the phosphorylation of IκBα in the cytosol. Blocking assays showed that specific inhibitors of ERK1/2, p38, and IκBα can significantly reduce the expression of IL-6 and IL-12 p40, which demonstrated that rTB9.8-mediated cytokine production is dependent on the activation of these kinases. Thus, this study demonstrates that rTB9.8 can activate RAW264.7 and trigger IL-6 and IL-12 p40 production via the ERK, p38, and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Hong Jia
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, 100193, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Wang YS, He X, Du Y, Su J, Gao M, Ma Y, Hua S, Quan F, Liu J, Zhang Y. Transgenic cattle produced by nuclear transfer of fetal fibroblasts carrying Ipr1 gene at a specific locus. Theriogenology 2015; 84:608-16. [DOI: 10.1016/j.theriogenology.2015.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 04/14/2015] [Accepted: 04/17/2015] [Indexed: 01/08/2023]
|
41
|
Teimourpour R, Sadeghian A, Meshkat Z, Esmaelizad M, Sankian M, Jabbari AR. Construction of a DNA Vaccine Encoding Mtb32C and HBHA Genes of Mycobacterium tuberculosis. Jundishapur J Microbiol 2015; 8:e21556. [PMID: 26464766 PMCID: PMC4600342 DOI: 10.5812/jjm.21556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/14/2014] [Accepted: 10/01/2014] [Indexed: 11/23/2022] Open
Abstract
Background: Tuberculosis (TB) is a contagious disease caused by Mycobacterium tuberculosis. Development of a new vaccine for tuberculosis requires immunogenic antigens capable of inducing suitable and long-lasting T cell immunity. The emergence of multidrugs and extensively drug-resistant strains of M. tuberculosis has made it a global public health concern. Objectives: DNA vaccine is a straightforward method to introduce antigens to the host. In the present study, two immunodominant mycobacterial antigens (Mtb32C and HBHA) were isolated and cloned into pcDNA3.1 (+) to design and construct a new DNA vaccine. This vector is capable of producing new potent chimeric protein. Materials and Methods: Mtb32C (Rv0125) and heparin-binding haemagglutinin adhesion (HBHA) genes were amplified using polymerase chain reaction (PCR) of M. tuberculosis H37Rv genome and ligated into pcDNA3.1 (+). Colony-PCR and restriction enzyme analysis were used to confirm the accuracy of the cloning procedure. Results: In the current study, recombinant pcDNA3.1 (+) vector containing Mtb32C and HBHA genes was successfully constructed. The desired size of DNA fragment was observed using single and double digestion methods. Conclusions: Mtb32C and HBHA genes were successfully isolated from H37Rv genome and cloned into an eukaryotic expression vector. This vector can be considered as a vaccine to evaluate immune responses in animal models.
Collapse
Affiliation(s)
- Roghayeh Teimourpour
- Department of Microbiology and Virology, Antimicrobial Resistance Research Center, Bu Ali Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran
| | - Ali Sadeghian
- Department of Microbiology and Virology, Antimicrobial Resistance Research Center, Bu Ali Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran
| | - Zahra Meshkat
- Department of Microbiology and Virology, Antimicrobial Resistance Research Center, Bu Ali Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran
- Corresponding author: Zahra Meshkat, Department of Microbiology and Virology, Antimicrobial Resistance Research Center, Bu Ali Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, IR Iran. Tel: +98-5138002313, Fax: +98-5138002287, E-mail:
| | - Majid Esmaelizad
- Genomics and Genetic Engineering Department, Razi Vaccine and Serum Research Institute, Karaj, IR Iran
| | - Mojtaba Sankian
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, IR Iran
| | - Ahmad-Reza Jabbari
- Genomics and Genetic Engineering Department, Razi Vaccine and Serum Research Institute, Karaj, IR Iran
| |
Collapse
|
42
|
The rLrp of Mycobacterium tuberculosis inhibits proinflammatory cytokine production and downregulates APC function in mouse macrophages via a TLR2-mediated PI3K/Akt pathway activation-dependent mechanism. Cell Mol Immunol 2015; 13:729-746. [PMID: 26166760 DOI: 10.1038/cmi.2015.58] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 05/25/2015] [Accepted: 05/27/2015] [Indexed: 12/13/2022] Open
Abstract
We demonstrate that Mycobacterium tuberculosis recombinant leucine-responsive regulatory protein (rLrp) inhibits lipopolysaccharide (LPS)-induced tumor necrosis factor alpha (TNF-α), interleukin-6, and interleukin-12 production and blocks the nuclear translocation of subunits of the nuclear-receptor transcription factor NF-κB (Nuclear factor-kappa B). Moreover, rLrp attenuated LPS-induced DNA binding and NF-κB transcriptional activity, which was accompanied by the degradation of inhibitory IκBα and a consequent decrease in the nuclear translocation of the NF-κB p65 subunit. RLrp interfered with the LPS-induced clustering of TNF receptor-associated factor 6 and with interleukin-1 receptor-associated kinase 1 binding to TAK1. Furthermore, rLrp did not attenuate proinflammatory cytokines or the expression of CD86 and major histocompatibility complex class-II induced by interferon-gamma in the macrophages of Toll-like receptor 2 deletion (TLR2-/-) mice and in protein kinase b (Akt)-depleted mouse cells, indicating that the inhibitory effects of rLrp were dependent on TLR2-mediated activation of the phosphatidylinositol 3-OH kinase (PI3K)/Akt pathway. RLrp could also activate the PI3K/Akt pathway by stimulating the rapid phosphorylation of PI3K, Akt, and glycogen synthase kinase 3 beta in macrophages. In addition, 19 amino acid residues in the N-terminus of rLrp were determined to be important and required for the inhibitory effects mediated by TLR2. The inhibitory function of these 19 amino acids of rLrp raises the possibility that mimetic inhibitory peptides could be used to restrict innate immune responses in situations in which prolonged TLR signaling has deleterious effects. Our study offers new insight into the inhibitory mechanisms by which the TLR2-mediated PI3K/Akt pathway ensures the transient expression of potent inflammatory mediators.
Collapse
|
43
|
Mycobacterium bovis BCG Vaccination Induces Divergent Proinflammatory or Regulatory T Cell Responses in Adults. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:778-88. [PMID: 25947145 DOI: 10.1128/cvi.00162-15] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 04/30/2015] [Indexed: 12/31/2022]
Abstract
Mycobacterium bovis bacillus Calmette-Guérin (BCG), the only currently available vaccine against tuberculosis, induces variable protection in adults. Immune correlates of protection are lacking, and analyses on cytokine-producing T cell subsets in protected versus unprotected cohorts have yielded inconsistent results. We studied the primary T cell response, both proinflammatory and regulatory T cell responses, induced by BCG vaccination in adults. Twelve healthy adult volunteers who were tuberculin skin test (TST) negative, QuantiFERON test (QFT) negative, and BCG naive were vaccinated with BCG and followed up prospectively. BCG vaccination induced an unexpectedly dichotomous immune response in this small, BCG-naive, young-adult cohort: BCG vaccination induced either gamma interferon-positive (IFN-γ(+)) interleukin 2-positive (IL-2(+)) tumor necrosis factor α-positive (TNF-α(+)) polyfunctional CD4(+) T cells concurrent with CD4(+) IL-17A(+) and CD8(+) IFN-γ(+) T cells or, in contrast, virtually absent cytokine responses with induction of CD8(+) regulatory T cells. Significant induction of polyfunctional CD4(+) IFN-γ(+) IL-2(+) TNF-α(+) T cells and IFN-γ production by peripheral blood mononuclear cells (PBMCs) was confined to individuals with strong immunization-induced local skin inflammation and increased serum C-reactive protein (CRP). Conversely, in individuals with mild inflammation, regulatory-like CD8(+) T cells were uniquely induced. Thus, BCG vaccination either induced a broad proinflammatory T cell response with local inflammatory reactogenicity or, in contrast, a predominant CD8(+) regulatory T cell response with mild local inflammation, poor cytokine induction, and absent polyfunctional CD4(+) T cells. Further detailed fine mapping of the heterogeneous host response to BCG vaccination using classical and nonclassical immune markers will enhance our understanding of the mechanisms and determinants that underlie the induction of apparently opposite immune responses and how these impact the ability of BCG to induce protective immunity to TB.
Collapse
|
44
|
Walzl G, Haks MC, Joosten SA, Kleynhans L, Ronacher K, Ottenhoff THM. Clinical immunology and multiplex biomarkers of human tuberculosis. Cold Spring Harb Perspect Med 2014; 5:cshperspect.a018515. [PMID: 25475107 DOI: 10.1101/cshperspect.a018515] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The discovery of tuberculosis (TB) biomarkers is an important goal in current TB research, because the availability of such markers would have significant impact on TB prevention and treatment. Correlates of protection would greatly facilitate vaccine development and evaluation, whereas correlates of TB disease risk would facilitate early diagnosis and help installing early or preventive treatment. Currently, no such markers are available. This review describes several strategies that are currently being pursued to identify TB biomarkers and places these in a clinical context. The approaches discussed include both targeted and untargeted hypothesis-free strategies. Among the first are the measurements of specific biomarkers in antigen-stimulated peripheral blood, in serum or plasma, and detailed immune cell phenotyping. Among the latter are proteomic, genomic, and transcriptomic (mRNA, miRNA) approaches. Recent and promising developments are described.
Collapse
Affiliation(s)
- Gerhard Walzl
- DST/NRF Centre of Excellence for Biomedical TB Research/MRC Centre for Molecular and Cellular Biology, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Mariëlle C Haks
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Simone A Joosten
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Léanie Kleynhans
- DST/NRF Centre of Excellence for Biomedical TB Research/MRC Centre for Molecular and Cellular Biology, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Katharina Ronacher
- DST/NRF Centre of Excellence for Biomedical TB Research/MRC Centre for Molecular and Cellular Biology, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
45
|
Namazova-Baranova LS, Alekseeva AA, Kharit SM, Kozhevnikova TN, Taranushenko TE, Tuzankina IA, Scarci F. Efficacy and safety of pidotimod in the prevention of recurrent respiratory infections in children: a multicentre study. Int J Immunopathol Pharmacol 2014; 27:413-9. [PMID: 25280032 DOI: 10.1177/039463201402700311] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Acute respiratory infections (ARI) still represent a big challenge for paediatricians, especially in those children defined as "ailed" as they are more susceptible to such kinds of disease. In this paediatric population, the immune system is still under-developed with an evident alteration in cytokine levels. A clinical study was carried out in 5 sites in Russia with the intention to enroll children particularly susceptible to contract respiratory infections (defined as "ailing"), assigning them to a treatment group with pidotimod in comparison with a control group, treating them for 30 days and observing the reduction in the number of ARI episodes throughout the follow-up period (6 months). Moreover, changes in serum immunological markers were evaluated at baseline and 30 days after treatment discontinuation. One hundred and fifty-seven ailing children were enrolled and assigned to two arms: a main pidotimod treatment group or a control group. The percentage of incidence of ARIs in the observation period at three different time points was statistically significant (p < 0.05). At the end of the follow-up period (after 6 months), ARIs had developed in 72 children (92.3%) in the main group and in 79 patients (100%) in the control group. Concerning changes of the immunological markers, the treatment group showed a better profile of normalization compared to the control group. The 30-day pidotimod therapy course led to improvement/reduction in the rate of acute respiratory infection recurrence in ailing children within a 3-month period, with a quick elimination of symptoms and signs of infection and, as a result, a faster recovery. The normalisation of the content of the pro-inflammatory cytokine interleukin-8 confirmed the immune-modulatory effect of the investigational drug, underlying its prophylactic effect.
Collapse
Affiliation(s)
- L S Namazova-Baranova
- Scientific Centre of Children's Health under the Russian Academy of Medical Sciences, Moscow, Russia
| | - A A Alekseeva
- Scientific Centre of Children's Health under the Russian Academy of Medical Sciences, Moscow, Russia
| | - S M Kharit
- Department of Prevention of Infectious Diseases, Public Health Committee in Vaccinal Prevention, Saint Petersburg, Russia
| | - T N Kozhevnikova
- Public Healthcare Institution DGB n. 3, Center for Respiratory Pathology of Children, Tula, Russia
| | - T E Taranushenko
- Krasnoyarsk State Medical University of Russia, Krasnoyarsk, Russia
| | - I A Tuzankina
- Institute of Immunology and Physiology, Uralian Department of the Russian Academy of Sciences, Ekaterinburg, Russia
| | - F Scarci
- Scientific Department, Polichem SA, Lugano-Pazzallo, Switzerland
| |
Collapse
|
46
|
Cantarini L, Lopalco G, Caso F, Costa L, Iannone F, Lapadula G, Anelli MG, Franceschini R, Menicacci C, Galeazzi M, Selmi C, Rigante D. Effectiveness and tuberculosis-related safety profile of interleukin-1 blocking agents in the management of Behçet's disease. Autoimmun Rev 2014; 14:1-9. [PMID: 25151975 DOI: 10.1016/j.autrev.2014.08.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 08/09/2014] [Indexed: 12/19/2022]
Abstract
Behçet's disease (BD) is a multi-systemic disorder of unknown etiology characterized by relapsing oral-genital ulcers, uveitis, and involvement of the articular, gastrointestinal, neurologic, and vascular systems. Although the primum movens of this condition remains unknown, a tangled plot combining autoimmune and autoinflammatory pathways has been hypothesized to explain its start and recurrence. In-depth analysis of BD pathogenetic mechanisms, involving dysfunction of multiple proinflammatory molecules, has opened new modalities of treatment: different agents targeting interleukin-1 have been studied in recent years to manage the most difficult and multi-resistant cases of BD. Growing experience with anakinra, canakinumab and gevokizumab is discussed in this review, highlighting the relative efficacy of each drug upon the protean BD clinical manifestations. Safety and tolerability of interleukin-1 antagonists in different doses have been confirmed by numerous observational studies on both large and small cohorts of patients with BD. In particular, the potential for Mycobacterium tuberculosis reactivation and tuberculosis development appears to be significantly lower with interleukin-1 blockers compared to tumor necrosis factor-α inhibitors, thus increasing the beneficial profile of this approach.
Collapse
Affiliation(s)
- Luca Cantarini
- Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy.
| | - Giuseppe Lopalco
- Interdisciplinary Department of Medicine, Rheumatology Unit, Policlinic Hospital, University of Bari, Bari, Italy
| | - Francesco Caso
- Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy; Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Padua, Padua, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Florenzo Iannone
- Interdisciplinary Department of Medicine, Rheumatology Unit, Policlinic Hospital, University of Bari, Bari, Italy
| | - Giovanni Lapadula
- Interdisciplinary Department of Medicine, Rheumatology Unit, Policlinic Hospital, University of Bari, Bari, Italy
| | - Maria Grazia Anelli
- Interdisciplinary Department of Medicine, Rheumatology Unit, Policlinic Hospital, University of Bari, Bari, Italy
| | | | - Cristina Menicacci
- Ophthalmology and Neurosurgery Department, University of Siena, Siena, Italy
| | - Mauro Galeazzi
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Padua, Padua, Italy
| | - Carlo Selmi
- Division of Rheumatology, Allergy, and Clinical Immunology, University of CA, Davis, USA; Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Donato Rigante
- Institute of Pediatrics, Università Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
47
|
Perdikogianni C, Galanakis E. Non-tuberculous mycobacterial cervical lymphadenitis in the immunocompetent child: diagnostic and treatment approach. Expert Rev Anti Infect Ther 2014; 12:959-65. [DOI: 10.1586/14787210.2014.920691] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
48
|
Mihret A, Loxton AG, Bekele Y, Kaufmann SHE, Kidd M, Haks MC, Ottenhoff THM, Aseffa A, Howe R, Walzl G. Combination of gene expression patterns in whole blood discriminate between tuberculosis infection states. BMC Infect Dis 2014; 14:257. [PMID: 24885723 PMCID: PMC4041060 DOI: 10.1186/1471-2334-14-257] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 05/08/2014] [Indexed: 12/19/2022] Open
Abstract
Background Genetic factors are involved in susceptibility or protection to tuberculosis (TB). Apart from gene polymorphisms and mutations, changes in levels of gene expression, induced by non-genetic factors, may also determine whether individuals progress to active TB. Methods We analysed the expression level of 45 genes in a total of 47 individuals (23 healthy household contacts and 24 new smear-positive pulmonary TB patients) in Addis Ababa using a dual colour multiplex ligation-dependent probe amplification (dcRT-MLPA) technique to assess gene expression profiles that may be used to distinguish TB cases and their contacts and also latently infected (LTBI) and uninfected household contacts. Results The gene expression level of BLR1, Bcl2, IL4d2, IL7R, FCGR1A, MARCO, MMP9, CCL19, and LTF had significant discriminatory power between sputum smear-positive TB cases and household contacts, with AUCs of 0.84, 0.81, 0.79, 0.79, 0.78, 0.76, 0.75, 0.75 and 0.68 respectively. The combination of Bcl2, BLR1, FCGR1A, IL4d2 and MARCO identified 91.66% of active TB cases and 95.65% of household contacts without active TB. The expression of CCL19, TGFB1, and Foxp3 showed significant difference between LTBI and uninfected contacts, with AUCs of 0.85, 0.82, and 0.75, respectively, whereas the combination of BPI, CCL19, FoxP3, FPR1 and TGFB1 identified 90.9% of QFT- and 91.6% of QFT+ household contacts. Conclusions Expression of single and especially combinations of host genes can accurately differentiate between active TB cases and healthy individuals as well as between LTBI and uninfected contacts.
Collapse
Affiliation(s)
- Adane Mihret
- Armauer Hansen Research Institute, Addis Ababa, Ethiopia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
García Jacobo RE, Serrano CJ, Enciso Moreno JA, Gaspar Ramírez O, Trujillo Ochoa JL, Uresti Rivera EE, Portales Pérez DP, González-Amaro R, García Hernández MH. Analysis of Th1, Th17 and regulatory T cells in tuberculosis case contacts. Cell Immunol 2014; 289:167-73. [PMID: 24841855 DOI: 10.1016/j.cellimm.2014.03.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 02/20/2014] [Accepted: 03/24/2014] [Indexed: 11/27/2022]
Abstract
We have hypothesized that individuals infected with Mycobacteriumtuberculosis that exhibit different patterns of immune reactivity in serial interferon (IFN)-γ release assays (IGRA's) correspond to different status within the immune spectrum of latent tuberculosis (TB). Accordingly, we analyzed the possible association between the consistent results (negative or positive) in an IGRA test and relevant immune parameters, mainly the levels of Th1 and Th17 lymphocytes and T regulatory (Treg) cells in the peripheral blood of TB case contacts. We found that individuals with a persistently positive IGRA showed increased levels of Th1 and Th17 lymphocytes upon in vitro stimulation with MTB antigens. In addition, a significant increase in the proportion of CD4+CTLA-4+ and CD4+Foxp3+ cells was detected in assays with blood samples from these individuals. Our data support that different immune phenotypes can be identified into the spectrum of latent TB, by combining different parameters of immune reactivity against MTB.
Collapse
Affiliation(s)
- R E García Jacobo
- Unidad de Investigación Médica-Zacatecas, Instituto Mexicano del Seguro Social, IMSS, Mexico
| | - C J Serrano
- Unidad de Investigación Médica-Zacatecas, Instituto Mexicano del Seguro Social, IMSS, Mexico
| | - J A Enciso Moreno
- Unidad de Investigación Médica-Zacatecas, Instituto Mexicano del Seguro Social, IMSS, Mexico
| | - O Gaspar Ramírez
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. Unidad Noreste., Monterrey, Nuevo León, Mexico
| | - J L Trujillo Ochoa
- Unidad de Investigación Médica-Zacatecas, Instituto Mexicano del Seguro Social, IMSS, Mexico
| | - E E Uresti Rivera
- Departamento de Inmunología y Biología Celular y Molecular, Facultad de Ciencias Químicas, UASLP, San Luis Potosí, SLP, Mexico
| | - D P Portales Pérez
- Departamento de Inmunología y Biología Celular y Molecular, Facultad de Ciencias Químicas, UASLP, San Luis Potosí, SLP, Mexico
| | - R González-Amaro
- Departamento de Inmunología, Facultad de Medicina, UASLP, San Luis Potosí, SLP, Mexico
| | - M H García Hernández
- Unidad de Investigación Médica-Zacatecas, Instituto Mexicano del Seguro Social, IMSS, Mexico.
| |
Collapse
|
50
|
Alves Da Silva D, Cavalcanti MAR, Muniz De Oliveira F, Trentini MM, Junqueira-Kipnis AP, Kipnis A. Immunogenicity of a recombinant Mycobacterium smegmatis vaccine expressing the fusion protein CMX in cattle from Goiás State, Brazil. J Vet Med Sci 2014; 76:977-84. [PMID: 24681608 PMCID: PMC4143659 DOI: 10.1292/jvms.13-0338] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study aimed to evaluate the immunogenicity of a recombinant Mycobacterium smegmatis vaccine expressing the CMX fusion protein composed of immunodominant epitopes Ag85C, MPT51 and HspX of Mycobacterium tuberculosis, which are important mycobacteria virulence factors. A group of Nelore heifers that were 10 to 12 months of age and negative for the tuberculin skin test (TST) were immunized with four doses of the recombinant vaccine mc(2)-CMX (M. smegmatis-Ag85C-MPT51-HspX) during a period of one year. Before each immunization, blood was collected to obtain sera for antibody analysis. Serological analysis demonstrated that mc(2)-CMX was able to induce a humoral response with increased levels of specific IgG antibodies against CMX, despite minimum antibody levels being detected for individual Ag85C, MPT51 or HspX recombinant antigens. However, there was no significant increase in specific CD4(+) IFN-γ-positive T cells. Lymphadenomegaly was observed in superficial cervical lymph nodes adjacent to the site of vaccination among mc(2)-CMX-vaccinated bovines, and the histopathological analysis demonstrated follicular hyperplasia without inflammatory infiltrate or granuloma formation. Animals remained negative for the TST until the end of the experiments, showing no cross-reactivity with the recombinant vaccine and tuberculin proteins. We discuss the potential of mc(2)-CMX to induce an immune response in cattle.
Collapse
Affiliation(s)
- Duanne Alves Da Silva
- Tropical Pathology and Public Health Institute, Federal University of Goiás, Rua 235 esquina com 1a Avenida, Setor Universitário, Goiânia, Goiás, CEP 74605-050, Brazil
| | | | | | | | | | | |
Collapse
|