1
|
Kim H, Shin SJ. Revolutionizing control strategies against Mycobacterium tuberculosis infection through selected targeting of lipid metabolism. Cell Mol Life Sci 2023; 80:291. [PMID: 37704889 PMCID: PMC11072447 DOI: 10.1007/s00018-023-04914-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 04/12/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023]
Abstract
Lipid species play a critical role in the growth and virulence expression of Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB). During Mtb infection, foamy macrophages accumulate lipids in granulomas, providing metabolic adaptation and survival strategies for Mtb against multiple stresses. Host-derived lipid species, including triacylglycerol and cholesterol, can also contribute to the development of drug-tolerant Mtb, leading to reduced efficacy of antibiotics targeting the bacterial cell wall or transcription. Transcriptional and metabolic analyses indicate that lipid metabolism-associated factors of Mtb are highly regulated by antibiotics and ultimately affect treatment outcomes. Despite the well-known association between major antibiotics and lipid metabolites in TB treatment, a comprehensive understanding of how altered lipid metabolites in both host and Mtb influence treatment outcomes in a drug-specific manner is necessary to overcome drug tolerance. The current review explores the controversies and correlations between lipids and drug efficacy in various Mtb infection models and proposes novel approaches to enhance the efficacy of anti-TB drugs. Moreover, the review provides insights into the efficacious control of Mtb infection by elucidating the impact of lipids on drug efficacy. This review aims to improve the effectiveness of current anti-TB drugs and facilitate the development of innovative therapeutic strategies against Mtb infection by making reverse use of Mtb-favoring lipid species.
Collapse
Affiliation(s)
- Hagyu Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
2
|
Naicker N, Rodel H, Perumal R, Ganga Y, Bernstein M, Benede N, Abdool Karim S, Padayacthi N, Sigal A, Naidoo K. Metformin Increases Cell Viability and Regulates Pro-Inflammatory Response to Mtb. Infect Drug Resist 2023; 16:3629-3638. [PMID: 37309381 PMCID: PMC10257915 DOI: 10.2147/idr.s401403] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/28/2023] [Indexed: 06/14/2023] Open
Abstract
Introduction Current TB treatment regimens are pathogen-directed and can be severely compromised by the development of drug resistance. Metformin has been proposed as an adjunctive therapy for TB, however relatively little is known about how metformin modulates the cellular interaction between Mtb and macrophages. We aimed to characterize how metformin modulates Mtb growth within macrophages. Methods We utilized live cell tracking through time-lapse microscopy to better understand the biological effect of metformin in response to Mtb infection. Furthermore, the potent first-line anti-TB drug, isoniazid, was used as a comparator and as a companion drug. Results Metformin caused a 14.2-fold decrease in Mtb growth compared to the untreated control. Metformin combined with isoniazid controlled Mtb growth is slightly better than isoniazid alone. Metformin demonstrated the ability to regulate the cytokine and chemokine response over a 72 hour period, better than isoniazid only. Conclusion We provide novel evidence that metformin controls mycobacterial growth by increasing host cell viability, and a direct and independent pro-inflammatory response to Mtb. Understanding the impact of metformin on Mtb growth within macrophages will advance our current knowledge on metformin as an adjunctive therapy, providing a new host-directed approach to TB treatment.
Collapse
Affiliation(s)
- Nikita Naicker
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Hylton Rodel
- Africa Health Research Institute, Durban, South Africa
| | - Rubeshan Perumal
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Yashica Ganga
- Africa Health Research Institute, Durban, South Africa
| | | | - Ntombi Benede
- Africa Health Research Institute, Durban, South Africa
| | - Salim Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute; University of KwaZulu-Natal, Durban, South Africa
| | - Nesri Padayacthi
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute; University of KwaZulu-Natal, Durban, South Africa
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences; University of KwaZulu-Natal, Durban, South Africa
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute; University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
3
|
Biomarkers Correlated with Tuberculosis Preventive Treatment Response: A Systematic Review and Meta-Analysis. Microorganisms 2023; 11:microorganisms11030743. [PMID: 36985316 PMCID: PMC10057454 DOI: 10.3390/microorganisms11030743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/07/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Background: There is a need to identify alternative biomarkers to predict tuberculosis (TB) preventive treatment response because observing the incidence decline renders a long follow-up period. Methods: We searched PubMed, Embase and Web of Science up to 9 February 2023. The biomarker levels during preventive treatment were quantitatively summarized by means of meta-analysis using the random-effect model. Results: Eleven eligible studies, published during 2006–2022, were included in the meta-analysis, with frequently heterogeneous results. Twenty-six biomarkers or testing methods were identified regarding TB preventive treatment monitoring. The summarized standard mean differences of interferon-γ (INF-γ) were −1.44 (95% CI: −1.85, −1.03) among those who completed preventive treatment (τ2 = 0.21; I2 = 95.2%, p < 0.001) and −0.49 (95% CI: −1.05, 0.06) for those without preventive treatment (τ2 = 0.13; I2 = 82.0%, p < 0.001), respectively. Subgroup analysis showed that the INF-γ level after treatment decreased significantly from baseline among studies with high TB burden (−0.98, 95% CI: −1.21, −0.75) and among those with a history of Bacillus Calmette–Guérin vaccination (−0.87, 95% CI: −1.10, −0.63). Conclusions: Our results suggested that decreased INF-γ was observed among those who completed preventive treatment but not in those without preventive treatment. Further studies are warranted to explore its value in preventive treatment monitoring due to limited available data and extensive between-study heterogeneity.
Collapse
|
4
|
Acen EL, Kateete DP, Worodria W, Olum R, Joloba ML, Bbuye M, Biraro IA. Evaluation of circulating serum cathelicidin levels as a potential biomarker to discriminate between active and latent tuberculosis in Uganda. PLoS One 2022; 17:e0272788. [PMID: 36018845 PMCID: PMC9416991 DOI: 10.1371/journal.pone.0272788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/26/2022] [Indexed: 01/08/2023] Open
Abstract
Background
Tuberculosis remains a major public health problem worldwide accounting for 1.4 million deaths annually. LL-37 is an effector molecule involved in immunity with both antimicrobial and immunomodulatory properties. The purpose of this study was to compare LL-37 circulatory levels among participants with active and latent tuberculosis and to determine its ability to discriminate between the two infectious states.
Methods
A cross-sectional study was performed among 56 active tuberculosis patients, 49 latent tuberculosis individuals, and 43 individuals without tuberculosis infection. The enzyme-linked immunosorbent assay was used to assess LL-37 levels. Data analysis was performed using STATA software and Graph pad Prism version 8. Mann-Whitney U test was used for correlation between variables with two categories and the Kruskal-Wallis test for three or more categories.
Results
The study had more female participants than males, with similar median ages across the three groups, 29.5, 25.0, and 23.0 years respectively. Active tuberculosis patients had significantly higher LL-37 levels compared to those with latent tuberculosis and without tuberculosis. The median/interquartile ranges were 318.8 ng/ml (157.9–547.1), 242.2 ng/ml (136.2–579.3), 170.9 ng/ml (129.3–228.3); p = 0.002 respectively. Higher LL-37 was found in the male participant with median/interquartile range, 424.8 ng/ml (226.2–666.8) compared to the females 237.7 ng/ml (129.6–466.6); p = 0.045. LL-37 had better discriminatory potential between active tuberculosis and no tuberculosis (AUC = 0.71, sensitivity 71.4% specificity = 69.8%) than with latent tuberculosis (AUC = 0.55, sensitivity = 71.4%, specificity = 44.9%). There was moderate differentiation between latent tuberculosis and no tuberculosis (AUC = 0.63, sensitivity = 44.9% specificity = 90.7%).
Conclusion
Significantly higher LL-37 levels were observed among active tuberculosis patients than those without tuberculosis infection and were, therefore able to discriminate between active tuberculosis and other tuberculosis infectious states, especially with no tuberculosis. Further assessment of this biomarker as a screening tool to exclude tuberculosis is required.
Collapse
Affiliation(s)
- Ester Lilian Acen
- Department of Physiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
- * E-mail:
| | - David Patrick Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - William Worodria
- Pulmonary Division, Department of Medicine, Mulago National Referral Hospital, Kampala, Uganda
| | - Ronald Olum
- Department of Medicine, School of Medicine, College of Health Sciences Unit, Makerere University, Kampala, Uganda
| | - Moses L. Joloba
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Mudarshiru Bbuye
- Makerere Lung Institute College of Health Sciences, Makerere University, Kampala, Uganda
| | - Irene Andia Biraro
- Department of Medicine, School of Medicine, College of Health Sciences Unit, Makerere University, Kampala, Uganda
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| |
Collapse
|
5
|
Valinetz ED, Matemo D, Gersh JK, Joudeh LL, Mendelsohn SC, Scriba TJ, Hatherill M, Kinuthia J, Wald A, Cangelosi GA, Barnabas RV, Hawn TR, Horne DJ. Isoniazid preventive therapy and tuberculosis transcriptional signatures in people with HIV. AIDS 2022; 36:1363-1371. [PMID: 35608118 PMCID: PMC9329226 DOI: 10.1097/qad.0000000000003262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To examine the association between isoniazid preventive therapy (IPT) or nontuberculous mycobacteria (NTM) sputum culture positivity and tuberculosis (TB) transcriptional signatures in people with HIV. DESIGN Cross-sectional study. METHODS We enrolled adults living with HIV who were IPT-naive or had completed IPT more than 6 months prior at HIV care clinics in western Kenya. We calculated TB signatures using gene expression data from qRT-PCR. We used multivariable linear regression to analyze the association between prior receipt of IPT or NTM sputum culture positivity with a transcriptional TB risk score, RISK6 (range 0-1). In secondary analyses, we explored the association between IPT or NTM positivity and four other TB transcriptional signatures. RESULTS Among 381 participants, 99.7% were receiving antiretroviral therapy and 86.6% had received IPT (completed median of 1.1 years prior). RISK6 scores were lower (mean difference 0.10; 95% confidence interval (CI): 0.06-0.15; P < 0.001) among participants who received IPT than those who did not. In a model that adjusted for age, sex, duration of ART, and plasma HIV RNA, the RISK6 score was 52.8% lower in those with a history of IPT ( P < 0.001). No significant association between year of IPT receipt and RISK6 scores was detected. There was no association between NTM sputum culture positivity and RISK6 scores. CONCLUSION In people with HIV, IPT was associated with significantly lower RISK6 scores compared with persons who did not receive IPT. These data support investigations of its performance as a TB preventive therapy response biomarker.
Collapse
Affiliation(s)
- Ethan D Valinetz
- Department of Medicine, University of Washington, Seattle, Washington
- Division of Infectious Disease, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Daniel Matemo
- Department of Research and Programs, Kenyatta National Hospital, Nairobi
- School of Public Health and Community Development Maseno University, Kisumu, Kenya
| | - Jill K Gersh
- Department of Medicine, University of Washington, Seattle, Washington
| | - Lara L Joudeh
- Department of Medicine, University of Washington, Seattle, Washington
| | - Simon C Mendelsohn
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, and Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - John Kinuthia
- Department of Research and Programs, Kenyatta National Hospital, Nairobi
- Department of Global Health
| | - Anna Wald
- Department of Medicine, University of Washington, Seattle, Washington
- Department of Epidemiology
- Department of Lab Medicine & Pathology, University of Washington
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Ruanne V Barnabas
- Department of Medicine, University of Washington, Seattle, Washington
- Department of Global Health
- Department of Epidemiology
| | - Thomas R Hawn
- Department of Medicine, University of Washington, Seattle, Washington
| | - David J Horne
- Department of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
6
|
Bouzeyen R, Javid B. Therapeutic Vaccines for Tuberculosis: An Overview. Front Immunol 2022; 13:878471. [PMID: 35812462 PMCID: PMC9263712 DOI: 10.3389/fimmu.2022.878471] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis is the world’s deadliest bacterial infection, resulting in more than 1.4 million deaths annually. The emergence of drug-resistance to first-line antibiotic therapy poses a threat to successful treatment, and novel therapeutic options are required, particularly for drug-resistant tuberculosis. One modality emerging for TB treatment is therapeutic vaccination. As opposed to preventative vaccination – the aim of which is to prevent getting infected by M. tuberculosis or developing active tuberculosis, the purpose of therapeutic vaccination is as adjunctive treatment of TB or to prevent relapse following cure. Several candidate therapeutic vaccines, using killed whole-cell or live attenuated mycobacteria, mycobacterial fragments and viral vectored vaccines are in current clinical trials. Other modes of passive immunization, including monoclonal antibodies directed against M. tuberculosis antigens are in various pre-clinical stages of development. Here, we will discuss these various therapeutics and their proposed mechanisms of action. Although the full clinical utility of therapeutic vaccination for the treatment of tuberculosis is yet to be established, they hold potential as useful adjunct therapies.
Collapse
|
7
|
Acen EL, Biraro IA, Bbuye M, Kateete DP, Joloba ML, Worodria W. Hypovitaminosis D among newly diagnosed pulmonary TB patients and their household contacts in Uganda. Sci Rep 2022; 12:5296. [PMID: 35351933 PMCID: PMC8964708 DOI: 10.1038/s41598-022-09375-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/22/2022] [Indexed: 11/29/2022] Open
Abstract
An estimated one billion people globally live with hypovitaminosis D. Studies have indicated that vitamin D deficiency is a risk factor for active tuberculosis (TB) disease. The aim of this study was to determine the association between vitamin D deficiency and TB status among patients with active TB, latent TB infection (LTBI) and those without TB infection. In a cross-sectional study of active TB patients, LTBI, QuantiFERON GOLD testpositive and (QFN+TST+) household contact and controls QuantiFERON GOLD testnegative (QFN-TST-) samples vitamin D levels were compared. Vitamin D status was determined by measurement of total vitamin D levels with 56 samples of active TB patients, 17 with LTBI, and 22 without TB infection using electrochemiluminescence. The median interquartile range (IQR) age of the study participants was 28 (20-35) years, and the majority (63%) were females. The median (IQR) vitamin D levels were 18 ng/ml (14-24). All groups had vitamin D hypovitaminosis with significantly lower levels among active TB patients (17 ng/ml, 13, 2) than among LTBI individuals (23 ng/ml 16-29) and those without TB infection (22 ng/ml, 17-28).
Collapse
Affiliation(s)
- Ester Lilian Acen
- Department of Physiology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda.
| | - Irene Andia Biraro
- Department of Internal Medicine, School of Medicine, College of Health Sciences Unit Makerere University, Kampala, Uganda
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Mudarshiru Bbuye
- Makerere Lung Institute College of Health Sciences Makerere University, Kampala, Uganda
| | - David Patrick Kateete
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Moses L Joloba
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - William Worodria
- Pulmonary Division, Department of Medicine, Mulago National Referral Hospital, Kampala, Uganda
| |
Collapse
|
8
|
Park HE, Lee W, Shin MK, Shin SJ. Understanding the Reciprocal Interplay Between Antibiotics and Host Immune System: How Can We Improve the Anti-Mycobacterial Activity of Current Drugs to Better Control Tuberculosis? Front Immunol 2021; 12:703060. [PMID: 34262571 PMCID: PMC8273550 DOI: 10.3389/fimmu.2021.703060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, remains a global health threat despite recent advances and insights into host-pathogen interactions and the identification of diverse pathways that may be novel therapeutic targets for TB treatment. In addition, the emergence and spread of multidrug-resistant Mtb strains led to a low success rate of TB treatments. Thus, novel strategies involving the host immune system that boost the effectiveness of existing antibiotics have been recently suggested to better control TB. However, the lack of comprehensive understanding of the immunomodulatory effects of anti-TB drugs, including first-line drugs and newly introduced antibiotics, on bystander and effector immune cells curtailed the development of effective therapeutic strategies to combat Mtb infection. In this review, we focus on the influence of host immune-mediated stresses, such as lysosomal activation, metabolic changes, oxidative stress, mitochondrial damage, and immune mediators, on the activities of anti-TB drugs. In addition, we discuss how anti-TB drugs facilitate the generation of Mtb populations that are resistant to host immune response or disrupt host immunity. Thus, further understanding the interplay between anti-TB drugs and host immune responses may enhance effective host antimicrobial activities and prevent Mtb tolerance to antibiotic and immune attacks. Finally, this review highlights novel adjunctive therapeutic approaches against Mtb infection for better disease outcomes, shorter treatment duration, and improved treatment efficacy based on reciprocal interactions between current TB antibiotics and host immune cells.
Collapse
Affiliation(s)
- Hyun-Eui Park
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Graduate School of Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
Foster M, Hill PC, Setiabudiawan TP, Koeken VACM, Alisjahbana B, van Crevel R. BCG-induced protection against Mycobacterium tuberculosis infection: Evidence, mechanisms, and implications for next-generation vaccines. Immunol Rev 2021; 301:122-144. [PMID: 33709421 PMCID: PMC8252066 DOI: 10.1111/imr.12965] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/20/2022]
Abstract
The tuberculosis (TB) vaccine Bacillus Calmette-Guérin (BCG) was introduced 100 years ago, but as it provides insufficient protection against TB disease, especially in adults, new vaccines are being developed and evaluated. The discovery that BCG protects humans from becoming infected with Mycobacterium tuberculosis (Mtb) and not just from progressing to TB disease provides justification for considering Mtb infection as an endpoint in vaccine trials. Such trials would require fewer participants than those with disease as an endpoint. In this review, we first define Mtb infection and disease phenotypes that can be used for mechanistic studies and/or endpoints for vaccine trials. Secondly, we review the evidence for BCG-induced protection against Mtb infection from observational and BCG re-vaccination studies, and discuss limitations and variation of this protection. Thirdly, we review possible underlying mechanisms for BCG efficacy against Mtb infection, including alternative T cell responses, antibody-mediated protection, and innate immune mechanisms, with a specific focus on BCG-induced trained immunity, which involves epigenetic and metabolic reprogramming of innate immune cells. Finally, we discuss the implications for further studies of BCG efficacy against Mtb infection, including for mechanistic research, and their relevance to the design and evaluation of new TB vaccines.
Collapse
Affiliation(s)
- Mitchell Foster
- Department of Microbiology and ImmunologyUniversity of OtagoDunedinNew Zealand
| | - Philip C. Hill
- Centre for International HealthUniversity of OtagoDunedinNew Zealand
| | - Todia Pediatama Setiabudiawan
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI)Radboud University Medical CenterNijmegenThe Netherlands
| | - Valerie A. C. M. Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI)Radboud University Medical CenterNijmegenThe Netherlands
- Department of Computational Biology for Individualised Infection MedicineCentre for Individualised Infection Medicine (CiiM) & TWINCOREJoint Ventures between The Helmholtz‐Centre for Infection Research (HZI) and The Hannover Medical School (MHH)HannoverGermany
| | - Bachti Alisjahbana
- Tuberculosis Working GroupFaculty of MedicineUniversitas PadjadjaranBandungIndonesia
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI)Radboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
10
|
Lin D, Liu Q, Wang W, Li Y, Li Y, Lin B, Ye Z, Huang J, Yu X, Chen Y, Mei Y, Huang M, Yang W, Zhou J, Liu X, Zeng J. Aberrant expression of miR-16, B12 and CD272 in peripheral blood mononuclear cells from patients with active tuberculosis. Am J Transl Res 2020; 12:6076-6091. [PMID: 33194015 PMCID: PMC7653578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Tuberculosis (TB) immunity is affected by complex immune regulation processes, which involve various immune cells, immune molecules, and cytokines. Here, we evaluated the expression of B12, CD272 and miR-16 in peripheral blood mononuclear cells (PBMC) of patients with active pulmonary tuberculosis. The results showed that monocytes expressing CD272 or B12 were down-regulated in patients with tuberculosis. The expression of B12 and CD272 in T cells and monocytes is related to tuberculosis. In TB patients, the up-regulation of miR-16 was negatively correlated with B12 mRNA expression, miR-16 was mainly expressed in CD14+ monocytes, and CD272 mRNA was mainly expressed in CD19+ B cells. It is worth noting that the overexpression of miR-16 inhibits the expression of CD272 and B12 in monocytes of TB patients. After BCG stimulation, miR-16 expression of CD14+ monocytes was up-regulated and B12 mRNA and CD272 mRNA expressions were down-regulated in TB patients. Finally, we found that miR-16 may participate in the TB immunization process through targeted regulation of B12 expression. These studies indicate that the expression of B12, CD272 and miR-16 in PBMC may be related to tuberculosis.
Collapse
Affiliation(s)
- Dongzi Lin
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
- Department of Laboratory Medicine, Foshan Forth People’s HospitalFoshan 528041, Guangdong, China
| | - Qiankun Liu
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Wei Wang
- Department of Laboratory Medicine, Foshan Forth People’s HospitalFoshan 528041, Guangdong, China
| | - Yanyun Li
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Yumei Li
- Department of Laboratory Medicine, Dongguan Sixth People’s HospitalDongguan 523008, Guangdong, China
| | - Bihua Lin
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Ziyu Ye
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Juan Huang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Xiaolin Yu
- Department of Laboratory Medicine, Dongguan Sixth People’s HospitalDongguan 523008, Guangdong, China
| | - Yinwen Chen
- Department of Laboratory Medicine, Dongguan Sixth People’s HospitalDongguan 523008, Guangdong, China
| | - Yuezhi Mei
- Department of Laboratory Medicine, Dongguan Sixth People’s HospitalDongguan 523008, Guangdong, China
| | - Minyuan Huang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Weiqin Yang
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Jie Zhou
- Department of Laboratory Medicine, Foshan Forth People’s HospitalFoshan 528041, Guangdong, China
| | - Xinguang Liu
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| | - Jincheng Zeng
- Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical UniversityDongguan 523808, Guangdong, China
| |
Collapse
|
11
|
Nakanjako D, Zalwango F, Wairagala P, Luboga F, Andia Biraro I, Bukirwa VD, Mboowa MG, Cose S, Seeley J, Elliott A. Career development for infection and immunity research in Uganda: a decade of experience from the Makerere University - Uganda Virus Research Institute research and training programme. AAS Open Res 2020; 3:26. [PMID: 32734140 PMCID: PMC7372530 DOI: 10.12688/aasopenres.13066.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2020] [Indexed: 11/23/2022] Open
Abstract
Background: The Makerere University/Uganda Virus Research Institute (UVRI) Centre of Excellence for Infection & Immunity Research and Training (MUII) is a collaborative programme supporting excellence in Infection and Immunity (I&I) research in Uganda. Set up in 2008, MUII aims to produce internationally competitive Ugandan and East African I&I research leaders, and develop human and infrastructural resources to support research and training excellence. We undertook an internal evaluation of MUII’s achievements, challenges and lessons learned between 08-2008 and 12-2019, to inform programmes seeking to build Africa’s health research expertise. Methods: Quantitative data were abstracted from programme annual reports. Qualitative data were obtained in 03-04/2019: a cross-sectional evaluation was undertaken among a purposefully selected representative sample of 27 trainees and two programme staff. Qualitative data was analysed according to pre-determined themes of achievements, challenges, lessons learned and recommendations for improvement. Results: By 12-2019, MUII had supported 68 fellowships at master’s-level and above (50% female: 23 Masters, 27 PhD, 15 post-doctoral, three group-leaders) and over 1,000 internships. Fellows reported career advancement, mentorship by experts, and improved research skills and outputs. Fellows have published over 300 papers, secured grants worth over £20m, established over 40 international collaborations, and taken on research and academic leadership positions in the country. Key lessons were: i) Efficient administration provides a conducive environment for high quality research; ii) Institutions need supportive policies for procurement, including provisions for purchases of specific biological research reagents from international manufacturers; iii) Strong international and multi-disciplinary collaboration provides a critical mass of expertise to mentor researchers in development; and iv) Mentorship catalyses young scientists to progress from graduate trainees to productive academic researchers, relevant to society’s most pressing health challenges. Conclusions: Sustainable academic productivity can be achieved through efficient operational support, global collaboration and mentorship to provide solutions to Africa’s health challenges.
Collapse
Affiliation(s)
- Damalie Nakanjako
- Makerere University-Uganda Virus Research Institute Infection and Immunity (MUII), Uganda Virus Research Institute, Entebbe, Uganda.,Department of Medicine, School of Medicine, Makerere University, College of Health Sciences, Kampala, Uganda
| | - Flavia Zalwango
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit,, Uganda Virus Research Institute, Entebbe, Uganda
| | - Pamela Wairagala
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit,, Uganda Virus Research Institute, Entebbe, Uganda
| | - Fiona Luboga
- Makerere University-Uganda Virus Research Institute Infection and Immunity (MUII), Uganda Virus Research Institute, Entebbe, Uganda
| | - Irene Andia Biraro
- Makerere University-Uganda Virus Research Institute Infection and Immunity (MUII), Uganda Virus Research Institute, Entebbe, Uganda.,Department of Medicine, School of Medicine, Makerere University, College of Health Sciences, Kampala, Uganda
| | - Victoria Diana Bukirwa
- Makerere University-Uganda Virus Research Institute Infection and Immunity (MUII), Uganda Virus Research Institute, Entebbe, Uganda
| | - Mary Gorrethy Mboowa
- Makerere University-Uganda Virus Research Institute Infection and Immunity (MUII), Uganda Virus Research Institute, Entebbe, Uganda
| | - Steve Cose
- Makerere University-Uganda Virus Research Institute Infection and Immunity (MUII), Uganda Virus Research Institute, Entebbe, Uganda.,Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit,, Uganda Virus Research Institute, Entebbe, Uganda
| | - Janet Seeley
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit,, Uganda Virus Research Institute, Entebbe, Uganda.,Global Health and Development Department, London School of Hygiene and Tropical Medicine, London, UK
| | - Alison Elliott
- Makerere University-Uganda Virus Research Institute Infection and Immunity (MUII), Uganda Virus Research Institute, Entebbe, Uganda.,Medical Research Council/Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine Uganda Research Unit,, Uganda Virus Research Institute, Entebbe, Uganda
| |
Collapse
|
12
|
Negi S, Pahari S, Bashir H, Agrewala JN. Intestinal microbiota disruption limits the isoniazid mediated clearance of Mycobacterium tuberculosis in mice. Eur J Immunol 2020; 50:1976-1987. [PMID: 32673409 DOI: 10.1002/eji.202048556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 07/05/2020] [Accepted: 07/14/2020] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB) continues to remain a global threat due to the emergence of drug-resistant Mycobacterium tuberculosis (Mtb) strains and toxicity associated with TB drugs. Intestinal microbiota has been reported to affect the host response to immunotherapy and drugs. However, how it affects the potency of first-line TB drug isoniazid (INH) is largely unknown. Here, we examined the impact of gut microbial dysbiosis on INH efficiency to kill Mtb. In this study, we employed in vivo mouse model, pretreated with broad-spectrum antibiotics (Abx) cocktail to disrupt their intestinal microbial population prior to Mtb infection and subsequent INH therapy. We demonstrated that microbiota disruption results in the impairment of INH-mediated Mtb clearance, and aggravated TB-associated tissue pathology. Further, it suppressed the innate immunity and reduced CD4 T-cell response against Mtb. Interestingly, a distinct shift of gut microbial profile was noted with abundance of Enterococcus and reduction of Lactobacillus and Bifidobacterium population. Our results show that the intestinal microbiota is crucial determinant in efficacy of INH to kill Mtb and impacts the host immune response against infection. This work provides an intriguing insight into the potential links between host gut microbiota and potency of INH.
Collapse
Affiliation(s)
- Shikha Negi
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India.,Present address: Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Susanta Pahari
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India.,Immunology Division, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Hilal Bashir
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Division, CSIR-Institute of Microbial Technology, Chandigarh, India.,Centre for Biomedical Engineering, Indian Institute of Technology-Ropar, Rupnagar, Punjab, India
| |
Collapse
|
13
|
Nakanjako D, Zalwango F, Wairagala P, Luboga F, Andia Biraro I, Bukirwa VD, Mboowa MG, Cose S, Seeley J, Elliott A. Career development for infection and immunity research in Uganda: a decade of experience from the Makerere University – Uganda Virus Research Institute research and training programme. AAS Open Res 2020; 3:26. [DOI: 10.12688/aasopenres.13066.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2020] [Indexed: 11/20/2022] Open
Abstract
Background: The Makerere University/Uganda Virus Research Institute (UVRI) Centre of Excellence for Infection & Immunity Research and Training (MUII) is a collaborative programme supporting excellence in Infection and Immunity (I&I) research in Uganda. Set up in 2008, MUII aims to produce internationally competitive Ugandan and East African I&I research leaders, and develop human and infrastructural resources to support research and training excellence. We undertook an internal evaluation of MUII’s achievements, challenges and lessons learned between August 2008 and December 2019, to inform programmes seeking to build Africa’s health research expertise. Methods: Quantitative data were abstracted from programme annual reports. Qualitative data were obtained in March and April 2019: a cross-sectional evaluation was undertaken among a purposefully selected representative sample of 27 trainees and two programme staff. Qualitative data was analysed according to pre-determined themes of achievements, challenges, lessons learned and recommendations for improvement. Results: By December 2019, MUII had supported 68 fellowships at master’s-level and above (50% female: 23 Masters, 27 PhD, 15 post-doctoral, three group-leader fellows) and over 1,000 internships. Fellows reported career advancement, mentorship by experts, and improved research skills and outputs. Fellows have published over 300 papers, secured grants worth over £20m, established over 40 international collaborations, and taken on research and academic leadership positions in the country. Key lessons for success include the following: efficient administration provides an enabling environment; institutions need supportive policies for procurement, including provisions for purchases of specific biological research reagents from international manufacturers; strong international, multi-disciplinary collaboration provides a critical mass of expertise to mentor researchers in development; and mentorship catalyses young scientists to progress from graduate trainees to productive academic researchers, relevant to society’s most pressing health challenges. Conclusions: Sustainable academic productivity can be achieved through efficient operational support, global collaboration and mentorship to provide solutions to Africa’s health challenges.
Collapse
|
14
|
Kwan PKW, Periaswamy B, De Sessions PF, Lin W, Molton JS, Naftalin CM, Naim ANM, Hibberd ML, Paton NI. A blood RNA transcript signature for TB exposure in household contacts. BMC Infect Dis 2020; 20:403. [PMID: 32517725 PMCID: PMC7282166 DOI: 10.1186/s12879-020-05116-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 05/24/2020] [Indexed: 11/10/2022] Open
Abstract
Background Current tools for diagnosing latent TB infection (LTBI) detect immunological memory of past exposure but are unable to determine whether exposure is recent. We sought to identify a whole-blood transcriptome signature of recent TB exposure. Methods We studied household contacts of TB patients; healthy volunteers without recent history of TB exposure; and patients with active TB. We performed whole-blood RNA sequencing (in all), an interferon gamma release assay (IGRA; in contacts and healthy controls) and PET/MRI lung scans (in contacts only). We evaluated differentially-expressed genes in household contacts (log2 fold change ≥1 versus healthy controls; false-discovery rate < 0.05); compared these to differentially-expressed genes seen in the active TB group; and assessed the association of a composite gene expression score to independent exposure/treatment/immunological variables. Results There were 186 differentially-expressed genes in household contacts (n = 26, age 22–66, 46% male) compared with healthy controls (n = 5, age 29–38, 100% male). Of these genes, 141 (76%) were also differentially expressed in active TB (n = 14, age 27–69, 71% male). The exposure signature included genes from inflammatory response, type I interferon signalling and neutrophil-mediated immunity pathways; and genes such as BATF2 and SCARF1 known to be associated with incipient TB. The composite gene-expression score was higher in IGRA-positive contacts (P = 0.04) but not related to time from exposure, isoniazid prophylaxis, or abnormalities on PET/MRI (all P > 0.19). Conclusions Transcriptomics can detect TB exposure and, with further development, may be an approach of value for epidemiological research and targeting public health interventions.
Collapse
Affiliation(s)
- Philip Kam Weng Kwan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 10, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Balamurugan Periaswamy
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Paola Florez De Sessions
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Wenwei Lin
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 10, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - James S Molton
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 10, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Claire M Naftalin
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 10, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Ahmad Nazri Mohamed Naim
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore
| | - Martin L Hibberd
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore.,London School of Hygiene & Tropical Medicine, London, UK.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicholas I Paton
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, NUHS Tower Block Level 10, 1E Kent Ridge Road, Singapore, 119228, Singapore. .,London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
15
|
Giraud-Gatineau A, Coya JM, Maure A, Biton A, Thomson M, Bernard EM, Marrec J, Gutierrez MG, Larrouy-Maumus G, Brosch R, Gicquel B, Tailleux L. The antibiotic bedaquiline activates host macrophage innate immune resistance to bacterial infection. eLife 2020; 9:e55692. [PMID: 32369020 PMCID: PMC7200153 DOI: 10.7554/elife.55692] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/04/2020] [Indexed: 12/12/2022] Open
Abstract
Antibiotics are widely used in the treatment of bacterial infections. Although known for their microbicidal activity, antibiotics may also interfere with the host's immune system. Here, we analyzed the effects of bedaquiline (BDQ), an inhibitor of the mycobacterial ATP synthase, on human macrophages. Genome-wide gene expression analysis revealed that BDQ reprogramed cells into potent bactericidal phagocytes. We found that 579 and 1,495 genes were respectively differentially expressed in naive- and M. tuberculosis-infected macrophages incubated with the drug, with an over-representation of lysosome-associated genes. BDQ treatment triggered a variety of antimicrobial defense mechanisms, including phagosome-lysosome fusion, and autophagy. These effects were associated with activation of transcription factor EB, involved in the transcription of lysosomal genes, resulting in enhanced intracellular killing of different bacterial species that were naturally insensitive to BDQ. Thus, BDQ could be used as a host-directed therapy against a wide range of bacterial infections.
Collapse
Affiliation(s)
- Alexandre Giraud-Gatineau
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut PasteurParisFrance
- Université Paris Diderot, Sorbonne Paris Cité, Cellule PasteurParisFrance
| | | | - Alexandra Maure
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut PasteurParisFrance
- Université Paris Diderot, Sorbonne Paris Cité, Cellule PasteurParisFrance
| | - Anne Biton
- Bioinformatics and Biostatistics, Department of Computational Biology, USR 3756 CNRS, Institut PasteurParisFrance
| | - Michael Thomson
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College LondonLondonUnited Kingdom
| | - Elliott M Bernard
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Jade Marrec
- Mycobacterial Genetics Unit, Institut PasteurParisFrance
| | - Maximiliano G Gutierrez
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Gérald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection, Department of Life Sciences, Faculty of Natural Sciences, Imperial College LondonLondonUnited Kingdom
| | - Roland Brosch
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut PasteurParisFrance
| | - Brigitte Gicquel
- Mycobacterial Genetics Unit, Institut PasteurParisFrance
- Department of Tuberculosis Control and Prevention, Shenzhen Nanshan Center for Chronic Disease ControlShenzhenChina
| | - Ludovic Tailleux
- Unit for Integrated Mycobacterial Pathogenomics, CNRS UMR 3525, Institut PasteurParisFrance
- Mycobacterial Genetics Unit, Institut PasteurParisFrance
| |
Collapse
|
16
|
Nalwoga A, Webb EL, Chihota B, Miley W, Walusimbi B, Nassuuna J, Sanya RE, Nkurunungi G, Labo N, Elliott AM, Cose S, Whitby D, Newton R. Kaposi's sarcoma-associated herpesvirus seropositivity is associated with parasite infections in Ugandan fishing communities on Lake Victoria islands. PLoS Negl Trop Dis 2019; 13:e0007776. [PMID: 31618208 PMCID: PMC6816576 DOI: 10.1371/journal.pntd.0007776] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 10/28/2019] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
We investigated the impact of helminths and malaria infection on Kaposi's sarcoma associated herpesvirus (KSHV) seropositivity, using samples and data collected from a cluster-randomised trial of intensive versus standard anthelminthic treatment. The trial was carried out in 2012 to 2016 among fishing communities on Lake Victoria islands in Uganda. Plasma samples from 2881 participants from two household surveys, the baseline (1310 participants) and the final (1571 participants) surveys were tested for KSHV IgG antibody responses to K8.1 and ORF73 recombinant proteins using ELISA. The baseline survey was carried out before the trial intervention while the final survey was carried out after three years of the trial intervention. Additionally, a subset sample of 372 participants from the final survey was tested for IgE, IgG and IgG4 antibody concentrations to S. mansoni adults worm antigen (SWA) and S. mansoni egg antigen (SEA) using ELISA. Infection by helminths (S. mansoni, N. americanus, T. trichiura and S. stercoralis) was diagnosed using real-time PCR, urine circulating cathodic antigen (CCA) and stool microscopy (Kato-Katz method) while malaria infection was diagnosed using microscopy. We analysed the relationship between helminth and malaria infections and KSHV seropositivity using regression modelling, allowing for survey design. At baseline, 56% of the participants were male while 48% of the participants were male in the final survey. The most prevalent helminth infection was S. mansoni (at baseline 52% and 34% in the final survey by microscopy, 86% by CCA and 50% by PCR in the final survey). KSHV seropositivity was 66% (baseline) and 56% (final survey) among those 1-12 years and >80% in those 13+ years in both surveys; malaria parasitaemia prevalence was 7% (baseline) and 4% (final survey). At baseline, individuals infected with S. mansoni (detected by microscopy) were more likely to be KSHV seropositive (aOR = 1.86 (1.16, 2.99) p = 0.012) and had higher anti-K8.1 antibody levels (acoefficient = 0.03 (0.01, 0.06) p = 0.02). In the final survey, S. mansoni (by microscopy, adjusted Odds Ratio (aOR = 1.43 (1.04-1.95), p = 0.028) and malaria parasitaemia (aOR = 3.49 (1.08-11.28), p = 0.038) were positively associated with KSHV seropositivity. Additionally, KSHV seropositive participants had higher S. mansoni-specific IgE and IgG antibody concentrations in plasma. Furthermore, HIV infected individuals on cART were less likely to be KSHV seropositive compared to HIV negative individuals (aOR = 0.46 (0.30, 0.71) p = 0.002). Schistosoma species skew the immune response towards Th2 and regulatory responses, which could impact on KSHV reactivation if co-infected with both organisms.
Collapse
Affiliation(s)
- Angela Nalwoga
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Emily L. Webb
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Belinda Chihota
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Wendell Miley
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | | | | | - Richard E. Sanya
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- College of Health Sciences, Makerere University, Kampala, Uganda
| | - Gyaviira Nkurunungi
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Nazzarena Labo
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Alison M. Elliott
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Stephen Cose
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Robert Newton
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- University of York, York; United Kingdom
| |
Collapse
|
17
|
BCG vaccination following latent TB treatment: Possible implications for different settings. Int J Infect Dis 2019; 80S:S17-S19. [DOI: 10.1016/j.ijid.2019.02.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 11/18/2022] Open
|
18
|
Huaman MA, Ticona E, Miranda G, Kryscio RJ, Mugruza R, Aranda E, Rondan PL, Henson D, Ticona C, Sterling TR, Fichtenbaum CJ, Garvy BA. The Relationship Between Latent Tuberculosis Infection and Acute Myocardial Infarction. Clin Infect Dis 2018; 66:886-892. [PMID: 29069328 PMCID: PMC5850031 DOI: 10.1093/cid/cix910] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/19/2017] [Indexed: 01/04/2023] Open
Abstract
Background Tuberculosis has been associated with an increased risk of cardiovascular disease (CVD), including acute myocardial infarction (AMI). We investigated whether latent tuberculosis infection (LTBI) is associated with AMI. Methods We conducted a case-control study in 2 large national public hospital networks in Lima, Peru, between July 2015 and March 2017. Case patients were patients with a first time diagnosis of type 1 (spontaneous) AMI. Controls were patients without a history of AMI. We excluded patients with known human immunodeficiency virus infection, tuberculosis disease, or prior LTBI treatment. We used the QuantiFERON-TB Gold In-Tube assay to identify LTBI. We used logistic regression modeling to estimate the odds ratio (OR) of LTBI in AMI case patients versus non-AMI controls. Results We enrolled 105 AMI case patients and 110 non-AMI controls during the study period. Overall, the median age was 62 years (interquartile range, 56-70 years); 69% of patients were male; 64% had hypertension, 40% dyslipidemia, and 39% diabetes mellitus; 30% used tobacco; and 24% were obese. AMI case patients were more likely than controls to be male (80% vs 59%; P < .01) and tobacco users (41% vs 20%; P < .01). LTBI was more frequent in AMI case patients than in controls (64% vs 49% [P = .03]; OR, 1.86; 95% confidence interval [CI], 1.08-3.22). After adjustment for age, sex, hypertension, dyslipidemia, diabetes mellitus, tobacco use, obesity, and family history of coronary artery disease, LTBI remained independently associated with AMI (adjusted OR, 1.90; 95% CI, 1.05-3.45). Conclusions LTBI was independently associated with AMI. Our results suggest a potentially important role of LTBI in CVD.
Collapse
Affiliation(s)
- Moises A Huaman
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati College of Medicine, Ohio
- Department of Medicine, Division of Infectious Diseases, University of Kentucky College of Medicine, Lexington
| | - Eduardo Ticona
- Department of Infectious Diseases and Tropical Medicine, Hospital Nacional Dos de Mayo
- Department of Internal Medicine, Universidad Nacional Mayor de San Marcos
| | - Gustavo Miranda
- Department of Cardiology, Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
| | - Richard J Kryscio
- Departments of Biostatistics and Statistics, University of Kentucky Colleges of Public Health and Arts & Sciences, Lexington
| | - Raquel Mugruza
- Department of Infectious Diseases and Tropical Medicine, Hospital Nacional Dos de Mayo
| | - Ernesto Aranda
- Department of Cardiology, Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
- Department of Internal Medicine, Division of Infectious Diseases, Wake Forest University School of Medicine, Winston-Salem, North Carolina
| | - Paola L Rondan
- Department of Infectious Diseases and Tropical Medicine, Hospital Nacional Dos de Mayo
| | - David Henson
- Department of Medicine, Division of Infectious Diseases, University of Kentucky College of Medicine, Lexington
| | - Cesar Ticona
- Department of Infectious Diseases and Tropical Medicine, Hospital Nacional Dos de Mayo
| | - Timothy R Sterling
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Carl J Fichtenbaum
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati College of Medicine, Ohio
| | - Beth A Garvy
- Department of Medicine, Division of Infectious Diseases, University of Kentucky College of Medicine, Lexington
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington
| |
Collapse
|
19
|
Use of QuantiFERON®-TB Gold in-tube culture supernatants for measurement of antibody responses. PLoS One 2017; 12:e0188396. [PMID: 29161328 PMCID: PMC5697869 DOI: 10.1371/journal.pone.0188396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/06/2017] [Indexed: 11/19/2022] Open
Abstract
QuantiFERON®-TB Gold in-tube (QFT-GIT) supernatants may be important samples for use in assessment of anti-tuberculosis (TB) antibodies when only limited volumes of blood can be collected and when a combination of antibody and cytokine measurements are required. These analytes, when used together, may also have the potential to differentiate active pulmonary TB (APTB) from latent TB infection (LTBI). However, few studies have explored the use of QFT-GIT supernatants for investigations of antibody responses. This study determined the correlation and agreement between anti-CFP-10 and anti-ESAT-6 antibody concentrations in QFT-GIT nil supernatant and serum pairs from 68 TB household contacts. We also explored the ability of Mycobacterium tuberculosis (M.tb) specific antibodies, or ratios of antibody to interferon gamma (IFN-γ) in QFT-GIT supernatants, to differentiate 97 APTB cases from 58 individuals with LTBI. Sputum smear microscopy was used to define APTB, whereas the QFT-GIT and tuberculin skin test were used to define LTBI. There were strong and statistically significant correlations between anti-CFP-10 and anti-ESAT-6 antibodies in unstimulated QFT-GIT supernatants and sera (r = 0.89; p<0.0001 for both), and no significant differences in antibody concentration between them. Anti-CFP-10 & anti-ESAT-6 antibodies differentiated APTB from LTBI with sensitivities of 88.7% & 71.1% and specificities of 41.4% & 51.7% respectively. Anti-CFP-10 antibody/M.tb specific IFN-γ and anti-ESAT-6 antibody/M.tb specific IFN-γ ratios had sensitivities of 48.5% & 54.6% and specificities of 89.7% and 75.9% respectively. We conclude that QFT-GIT nil supernatants may be used in the place of sera when measuring antibody responses, reducing blood volumes needed for such investigations. Antibodies in QFT-GIT nil supernatants on their own discriminate APTB from LTBI with high sensitivity but have poor specificity, whereas the reverse is true when antibodies are used in combination with M.tb specific cytokines. Further antibody and antibody/cytokine combinations need to be explored to achieve better diagnostic accuracy.
Collapse
|
20
|
Wajja A, Kizito D, Nassanga B, Nalwoga A, Kabagenyi J, Kimuda S, Galiwango R, Mutonyi G, Vermaak S, Satti I, Verweij J, Tukahebwa E, Cose S, Levin J, Kaleebu P, Elliott AM, McShane H. The effect of current Schistosoma mansoni infection on the immunogenicity of a candidate TB vaccine, MVA85A, in BCG-vaccinated adolescents: An open-label trial. PLoS Negl Trop Dis 2017; 11:e0005440. [PMID: 28472067 PMCID: PMC5417418 DOI: 10.1371/journal.pntd.0005440] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/27/2017] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Helminth infection may affect vaccine immunogenicity and efficacy. Adolescents, a target population for tuberculosis booster vaccines, often have a high helminth burden. We investigated effects of Schistosoma mansoni (Sm) on the immunogenicity and safety of MVA85A, a model candidate tuberculosis vaccine, in BCG-vaccinated Ugandan adolescents. METHODS In this phase II open label trial we enrolled 36 healthy, previously BCG-vaccinated adolescents, 18 with no helminth infection detected, 18 with Sm only. The primary outcome was immunogenicity measured by Ag85A-specific interferon gamma ELISpot assay. Tuberculosis and schistosome-specific responses were also assessed by whole-blood stimulation and multiplex cytokine assay, and by antibody ELISAs. RESULTS Ag85A-specific cellular responses increased significantly following immunisation but with no differences between the two groups. Sm infection was associated with higher pre-immunisation Ag85A-specific IgG4 but with no change in antibody levels following immunisation. There were no serious adverse events. Most reactogenicity events were of mild or moderate severity and resolved quickly. CONCLUSIONS The significant Ag85A-specific T cell responses and lack of difference between Sm-infected and uninfected participants is encouraging for tuberculosis vaccine development. The implications of pre-existing Ag85A-specific IgG4 antibodies for protective immunity against tuberculosis among those infected with Sm are not known. MVA85A was safe in this population. TRIAL REGISTRATION ClinicalTrials.gov NCT02178748.
Collapse
Affiliation(s)
- Anne Wajja
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Dennison Kizito
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Beatrice Nassanga
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Angela Nalwoga
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Joyce Kabagenyi
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Simon Kimuda
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Ronald Galiwango
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Gertrude Mutonyi
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Samantha Vermaak
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Iman Satti
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Jaco Verweij
- Laboratory for Medical Microbiology and Immunology & Laboratory for Clinical Pathology, St. Elisabeth Hospital, Tilburg, The Netherlands
| | | | - Stephen Cose
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Jonathan Levin
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
- School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Pontiano Kaleebu
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
| | - Alison M. Elliott
- Co-infection Studies Program, MRC/UVRI Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Helen McShane
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Pease C, Hutton B, Yazdi F, Wolfe D, Hamel C, Quach P, Skidmore B, Moher D, Alvarez GG. Efficacy and completion rates of rifapentine and isoniazid (3HP) compared to other treatment regimens for latent tuberculosis infection: a systematic review with network meta-analyses. BMC Infect Dis 2017; 17:265. [PMID: 28399802 PMCID: PMC5387294 DOI: 10.1186/s12879-017-2377-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 04/01/2017] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND We conducted a systematic review and network meta-analysis (NMA) to examine the efficacy and completion rates of treatments for latent tuberculosis infection (LTBI). While a previous review found newer, short-duration regimens to be effective, several included studies did not confirm LTBI, and analyses did not account for variable follow-up or assess completion. METHODS We searched MEDLINE, Embase, CENTRAL, PubMed, and additional sources to identify RCTs in patients with confirmed LTBI that involved a regimen of interest and reported on efficacy or completion. Regimens of interest included isoniazid (INH) with rifapentine once weekly for 12 weeks (INH/RPT-3), 6 and 9 months of daily INH (INH-6; INH-9), 3-4 months daily INH plus rifampicin (INH/RFMP 3-4), and 4 months daily rifampicin alone (RFMP-4). NMAs were performed to compare regimens for both endpoints. RESULTS Sixteen RCTs (n = 44,149) and 14 RCTs (n = 44,128) were included in analyses of efficacy and completion. Studies were published between 1968 and 2015, and there was diversity in patient age and comorbidities. All regimens of interest except INH-9 showed significant benefits in preventing active TB compared to placebo. Comparisons between active regimens did not reveal significant differences. While definitions of regimen completion varied across studies, regimens of 3-4 months were associated with a greater likelihood of adequate completion. CONCLUSIONS Most of the active regimens showed an ability to reduce the risk of active TB relative to no treatment, however important differences between active regimens were not found. Shorter rifamycin-based regimens may offer comparable benefits to longer INH regimens. Regimens of 3-4 months duration are more likely to be completed than longer regimens.
Collapse
Affiliation(s)
| | - Brian Hutton
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6 Canada
- Ottawa University School of Epidemiology, Public Health and Preventive Medicine, Ottawa, Canada
| | - Fatemeh Yazdi
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6 Canada
| | - Dianna Wolfe
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6 Canada
| | - Candyce Hamel
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6 Canada
| | - Pauline Quach
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6 Canada
| | - Becky Skidmore
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6 Canada
| | - David Moher
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6 Canada
- Ottawa University School of Epidemiology, Public Health and Preventive Medicine, Ottawa, Canada
| | - Gonzalo G. Alvarez
- Department of Medicine, The Ottawa Hospital, Ottawa, Canada
- Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6 Canada
- Ottawa University Faculty of Medicine, Ottawa, Canada
| |
Collapse
|
22
|
Delogu G, Vanini V, Cuzzi G, Chiacchio T, De Maio F, Battah B, Pinnetti C, Sampaolesi A, Antinori A, Goletti D. Lack of Response to HBHA in HIV-Infected Patients with Latent Tuberculosis Infection. Scand J Immunol 2017; 84:344-352. [PMID: 27636597 DOI: 10.1111/sji.12493] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 09/09/2016] [Indexed: 12/20/2022]
Abstract
Heparin-binding haemagglutinin (HBHA) has been proposed as an immunological biomarker for discriminating active tuberculosis (TB) from latent TB infection (LTBI) and to identify those at higher risk of progressing to active disease. Few data are available in immune-compromised patients, which are those with increased risk of TB reactivation. The aim of this stusy was to evaluate the immune response to HBHA in HIV-infected subjects with LTBI (HIV-LTBI) or active TB (HIV-TB) in comparison with the immune response to additional Mycobacterium tuberculosis (Mtb) or HIV and CMV antigens. The responses are evaluated in relation to TB status and in the LTBI subjects with the progression to active TB within 2 years. Forty-one HIV-infected antiretroviral-naïve subjects were prospectively enrolled: 18 were HIV-TB and 23 HIV-LTBI. Whole blood was in vitro stimulated overnight with several antigens and mitogen. Interferon-γ response in the harvested plasma was evaluated by ELISA. Despite that CD4 cell count was significantly different between HIV-LTBI and HIV-TB, no differences were observed in response to Mtb- or HIV-specific antigens. Differently, low responses to HBHA were observed in both HIV-LTBI and HIV-TB subjects. Importantly, none of the six HIV-LTBI responding to HBHA developed TB, while two of 17 non-HBHA responders developed active disease. HIV-TB-coinfected subjects, regardless of their TB status, showed low responses to HBHA despite maintaining detectable responses to other antigens; moreover, among the HIV-LTBI, the lack of HBHA responses indicated an increased risk to develop active TB. These results, although preliminary, suggest that a positive response to HBHA in HIV-LTBI correlates with Mtb containment.
Collapse
Affiliation(s)
- G Delogu
- Epidemiology Department, National Institute for Infectious Diseases, IRCCS "Lazzaro Spallanzani", Rome, Italy.,Institute of Microbiology, Catholic University of Sacred Heart, Rome, Italy
| | - V Vanini
- Translational Research Unit, Epidemiology Department, National Institute for Infectious Diseases, IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - G Cuzzi
- Translational Research Unit, Epidemiology Department, National Institute for Infectious Diseases, IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - T Chiacchio
- Translational Research Unit, Epidemiology Department, National Institute for Infectious Diseases, IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - F De Maio
- Institute of Microbiology, Catholic University of Sacred Heart, Rome, Italy
| | - B Battah
- Institute of Microbiology, Catholic University of Sacred Heart, Rome, Italy
| | - C Pinnetti
- Clinical Department, National Institute for Infectious Diseases, IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - A Sampaolesi
- Clinical Department, National Institute for Infectious Diseases, IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - A Antinori
- Clinical Department, National Institute for Infectious Diseases, IRCCS "Lazzaro Spallanzani", Rome, Italy
| | - D Goletti
- Translational Research Unit, Epidemiology Department, National Institute for Infectious Diseases, IRCCS "Lazzaro Spallanzani", Rome, Italy
| |
Collapse
|
23
|
Michelsen SW, Soborg B, Agger EM, Diaz LJ, Hoff ST, Koch A, Sorensen HCF, Andersen P, Wohlfahrt J, Melbye M. Host immunity to Mycobacterium tuberculosis and risk of tuberculosis: A longitudinal study among Greenlanders. Vaccine 2016; 34:5975-5983. [DOI: 10.1016/j.vaccine.2016.09.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022]
|
24
|
Changes in IL-2 and IL-10 during Chronic Administration of Isoniazid, Nevirapine, and Paracetamol in Rats. Adv Pharmacol Sci 2016; 2016:3094783. [PMID: 27990159 PMCID: PMC5136381 DOI: 10.1155/2016/3094783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/10/2016] [Accepted: 10/23/2016] [Indexed: 11/17/2022] Open
Abstract
The aim of this study was to illustrate the initial subclinical drug-induced liver injury and the associated adaptive immune response by monitoring for the changes in plasma IL-2, IL-10, and some cytochrome P450 activity during chronic administration of nevirapine (NVP), isoniazid (INH), and paracetamol (PAR) in rats without clinical hepatotoxicity. Male Sprague-Dawley (SD) rats were divided into four groups (saline (S), NVP, INH, and PAR) of 25 animals each. The drugs were administered daily for 42 days at therapeutic doses (NVP 200 mg/kg, PAR 500 mg/kg, and INH 20 mg/kg) to the respective groups by oral gavage and five rats per group were sacrificed weekly. All the three drugs induced a subclinical liver injury in the first 2-3 weeks followed by healing, indicating adaption. The liver injury was pathologically similar and was associated with immune stimulation and increased cytochrome P450 activity. NVP- and PAR-induced liver injury lasted up to 14 days while that for INH lasted for 28 days. NVP-induced liver injury was associated with increased IL-2, CD4 count, and CYP3A2 activity, followed by increased IL-10 during the healing phase. In conclusion, the initial drug-induced subclinical liver injury, its spontaneous healing, and the associated adaptive immune response have been demonstrated.
Collapse
|
25
|
Mattos AMM, Chaves AS, Franken KLMC, Figueiredo BBM, Ferreira AP, Ottenhoff THM, Teixeira HC. Detection of IgG1 antibodies against Mycobacterium tuberculosis DosR and Rpf antigens in tuberculosis patients before and after chemotherapy. Tuberculosis (Edinb) 2015; 96:65-70. [PMID: 26786656 DOI: 10.1016/j.tube.2015.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 12/20/2022]
Abstract
Diagnosis of tuberculosis (TB) remains challenging. Serum IgG1 antibodies against Mycobacterium tuberculosis active growth phase antigens (ESAT-6/CFP-10, Rv0717 and Rv3353), DosR regulon-encoded proteins (Rv1733, Rv1737, Rv2628 and Rv2029), and resuscitation-promoting factors (Rv0867 and Rv2389) were evaluated in TB patients using ELISA. Active TB patients showed elevated levels of IgG1 antibodies against ESAT-6/CFP-10, Rv0717, Rv3353, Rv1733, Rv2628, Rv2029 and Rv0867 in comparison to healthy controls (p < 0.001). These levels remained high after the initiation of treatment, while responses to Rv0717 and Rv1733 peaked early during treatment. IgG1 responses to ESAT-6/CFP-10, Rv3353, Rv2628, Rv2029 and Rv0867 declined to control levels after the completion of 6 months chemotherapy. ROC analysis confirmed the good diagnostic performance of Rv0717, Rv1733, Rv3353, Rv2628, Rv2029 and Rv0867antigens. These data suggest that detecting IgG1 antibodies against M. tuberculosis antigens, including DosR and Rpf proteins, may represent an additional tool in the diagnosis of tuberculosis.
Collapse
Affiliation(s)
- Ana Márcia Menezes Mattos
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz De Fora, 36036-900, Juiz de Fora, Minas Gerais, Brazil
| | - Alexandre Silva Chaves
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz De Fora, 36036-900, Juiz de Fora, Minas Gerais, Brazil
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Bárbara Bruna Muniz Figueiredo
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz De Fora, 36036-900, Juiz de Fora, Minas Gerais, Brazil
| | - Ana Paula Ferreira
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz De Fora, 36036-900, Juiz de Fora, Minas Gerais, Brazil
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Henrique Couto Teixeira
- Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz De Fora, 36036-900, Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|