1
|
Ssekamatte P, Sitenda D, Nabatanzi R, Nakibuule M, Kibirige D, Kyazze AP, Kateete DP, Bagaya BS, Sande OJ, van Crevel R, Cose S, Biraro IA. Isoniazid preventive therapy modulates Mycobacterium tuberculosis-specific T-cell responses in individuals with latent tuberculosis and type 2 diabetes. Sci Rep 2025; 15:10423. [PMID: 40140681 PMCID: PMC11947150 DOI: 10.1038/s41598-025-95386-z] [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: 12/21/2024] [Accepted: 03/20/2025] [Indexed: 03/28/2025] Open
Abstract
Diabetes mellitus (DM) is a significant contributor to tuberculosis (TB) incidence and poor treatment outcomes. This study explored the impact of isoniazid preventive therapy (IPT) on Mycobacterium tuberculosis (Mtb)-specific T-cell memory phenotypes and function among participants with latent TB infection and DM (LTBI-DM) at baseline and after 6 months of IPT; and compared the responses to healthy controls (HC). Peripheral blood mononuclear cells were stimulated with ESAT-6 and CFP-10 peptide pools to analyse CD4+ and CD8+ T-cell responses using flow cytometry. In LTBI-DM participants, effector memory CD4+ and CD8+ T cells were decreased post-IPT, suggesting a shift towards a less-activated state or differentiation into other subsets. CXCR5 expression on both CD4+ and CD8+ T cells was upregulated, while PD-1 expression was downregulated post-IPT, indicating reduced T-cell exhaustion and improved homing capabilities. Lastly, IL-17 A and IL-13 production in CD4+ and CD8+ T cells was increased post-IPT, respectively, which play a role in enhanced Mtb infection control. The post-IPT T-cell alterations were similar to normal HC levels. These findings suggest that IPT modulates and normalises specific T-cell memory phenotypes and functional responses in LTBI-DM participants, potentially contributing to improved long-term immunity and protection against TB. This study highlights the importance of preventive therapy in high-risk populations, and larger studies with more extended follow-up are needed to assess long-lasting IPT effects.
Collapse
Affiliation(s)
- Phillip Ssekamatte
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda.
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda.
| | - Diana Sitenda
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Rose Nabatanzi
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Marjorie Nakibuule
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Davis Kibirige
- Department of Medicine, Uganda Martyrs Hospital Lubaga, Kampala, Uganda
| | - Andrew Peter Kyazze
- Department of Internal Medicine, School of Medicine, 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
| | - Bernard Ssentalo Bagaya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Obondo James Sande
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Medical Centre, Kampala, Uganda
| | - Stephen Cose
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - Irene Andia Biraro
- Medical Research Council/Uganda Virus Research Institute and London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
- Department of Internal Medicine, School of Medicine, College of Health Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
2
|
Zeng Q, Tong Z, Zhong J, Li X, Shen B, Chen H, Ge D. The correlation between immune profiles and pathological changes in pulmonary tuberculosis granulomas revealed by bioinformatic analysis and experimental validation. Tuberculosis (Edinb) 2025; 152:102614. [PMID: 39999566 DOI: 10.1016/j.tube.2025.102614] [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: 10/23/2024] [Revised: 01/10/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025]
Abstract
Most of Mycobacterium tuberculosis(Mtb) infection result in the formation of granulomas, which are often rich in immune cells, with subsequent clinical symptoms. However, the role of the immune system in the formation of tuberculosis granuloma structures has not been fully revealed. Here we first analyzed single-cell transcriptome and microenvironment spatial characteristics to reveal the contribution of immune cells to granuloma expansion with validation by immunofluorescence. We then integrated published peripheral blood transcriptome data for Mtb-infected patients and healthy controls. Immune cell profiles were deconvoluted and results were validated on a local cohort using flow cytometry. At the same time, an in-depth evaluation of the changes in the population and function of multiple peripheral blood immune cells during tuberculosis infection were conducted to define correlation with granuloma area. Finally, we screened 6 cytokines (IL6, IL8, IL10, IFNγ, TNFα, TGFβ) through machine learning bioinformatics and analyzed their correlation with the size of tuberculosis granuloma. Based on these findings, we confirmed that the dynamic variation in proportion of immune cells in peripheral blood and the levels of cytokine profiles are closely related to the occurrence and development of tuberculosis granuloma. This study provides a theoretical basis for the molecular mechanism of tuberculosis granuloma.
Collapse
Affiliation(s)
- Qingqiu Zeng
- Department of Infectious Diseases, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Zhaowei Tong
- Department of Infectious Diseases, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China; Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Huzhou, Zhejiang, 313000, China
| | - Jianfeng Zhong
- Department of Infectious Diseases, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China; Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Huzhou, Zhejiang, 313000, China
| | - Xiaofeng Li
- Department of Infectious Diseases, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Bin Shen
- Department of Infectious Diseases, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Haiyan Chen
- Department of Infectious Diseases, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China
| | - Dating Ge
- Department of Pathology, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, 313000, China.
| |
Collapse
|
3
|
Djibougou DA, Mensah GI, Cissé M, Inoussa T, Sawadogo LT, Combary A, Sanou A, Bonfoh B, Addo KK, Belem AMG, Meda CZ, Dabiré RK, Kaboré A, Diagbouga PS. Intestinal Protozoa, Helminth Infection, and Associated Factors among Tuberculosis Patients and Nontuberculosis Persons in Bobo-Dioulasso City, Burkina Faso. Am J Trop Med Hyg 2024; 111:1265-1272. [PMID: 39317180 PMCID: PMC11619520 DOI: 10.4269/ajtmh.23-0346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 06/13/2024] [Indexed: 09/26/2024] Open
Abstract
We report the frequency and associated factors of tuberculosis (TB) and parasite coinfection from newly diagnosed pulmonary TB patients (TB+) and non-TB participants (TB-) from the Regional Tuberculosis Control Center, households, and health facilities in Bobo-Dioulasso from 2019 to 2021. Biological samples were examined for parasite infection using direct microscopy, concentration techniques, and the immunochromatographic rapid test. Data were analyzed using STATA 14. Of a total of 192 participants involved, 95 were TB+ and 97 were non-TB. There was no statistically significant difference in parasitic infections between the two groups, although it was higher in TB+ than TB- (69.5% [66/95] versus 55.7% [54/97]; P = 0.07). Protozoal infection prevalence was significantly higher in patients with TB+ than in those TB- (61.1% versus 37.1%; P = 0.001). Specifically, Entamoeba spp. and Cryptosporidium spp. followed this pattern with 35.8% versus 19.6% (P = 0.01) and 22.1% versus 8.3% (P = 0.007), respectively. Although higher in TB+ patients, helminthiasis frequency was not significantly different between the two groups (23.2% versus 15.5%; P = 0.2). Helminth species were Schistosomia mansoni (17.9% versus 12.4%), Dicrocoelium dendriticum (3.2% versus 1.0%), Enterobius vermicularis (2.1% versus 2.1%), Wuchereria bancrofti (1.1% versus 0.0%), and Hymenolepis nana (1.1% versus 0.0%). Illiteracy (adjusted odds ratio [aOR]: 2.5; 95% CI: 1.0-6.1), smoking (aOR: 2.4; 95% CI: 1.1-5.3), and handwashing after defecation (aOR: 2.4; 95% CI: 1.2-4.7) were associated with parasites. This study reported a high frequency of parasite coinfection in TB patients. These findings suggest the need for adequate health education for behavioral change and systematic diagnosing of parasites in TB patients for better coinfection management.
Collapse
Affiliation(s)
- Diakourga Arthur Djibougou
- Université Nazi BONI, Bobo-Dioulasso, Burkina Faso
- Centre MURAZ/Institut National de Santé Publique, Bobo-Dioulasso, Burkina Faso
- Institut de Recherche en Sciences de la Santé/Centre National de Recherche Scientifique et Technologique, Ouagadougou, Burkina Faso
| | - Gloria Ivy Mensah
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Mamoudou Cissé
- Université Nazi BONI, Bobo-Dioulasso, Burkina Faso
- Centre MURAZ/Institut National de Santé Publique, Bobo-Dioulasso, Burkina Faso
| | - Toé Inoussa
- Université Nazi BONI, Bobo-Dioulasso, Burkina Faso
| | - Leon Tinnoga Sawadogo
- Université Nazi BONI, Bobo-Dioulasso, Burkina Faso
- Programme National Tuberculose, Ministry of Health, Ouagadougou, Burkina Faso
| | - Adjima Combary
- Programme National Tuberculose, Ministry of Health, Ouagadougou, Burkina Faso
| | - Adama Sanou
- Université Nazi BONI, Bobo-Dioulasso, Burkina Faso
- Centre MURAZ/Institut National de Santé Publique, Bobo-Dioulasso, Burkina Faso
| | - Bassirou Bonfoh
- Centre Suisse de Recherches Scientifique de Côte d’Ivoire, Abidjan, Côte d’Ivoire
| | - Kennedy Kwasi Addo
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | | | - Clément Ziemlé Meda
- Université Nazi BONI, Bobo-Dioulasso, Burkina Faso
- Centre MURAZ/Institut National de Santé Publique, Bobo-Dioulasso, Burkina Faso
| | - Roch Konbobr Dabiré
- Centre MURAZ/Institut National de Santé Publique, Bobo-Dioulasso, Burkina Faso
- Institut de Recherche en Sciences de la Santé/Centre National de Recherche Scientifique et Technologique, Ouagadougou, Burkina Faso
| | - Achille Kaboré
- Family Health International 360 (FHI 360), Washington, District of Columbia
| | - Potiandi Serge Diagbouga
- Institut de Recherche en Sciences de la Santé/Centre National de Recherche Scientifique et Technologique, Ouagadougou, Burkina Faso
- Etudes Formation et Recherches Développement en Santé (EFORDS), Ouagadougou, Burkina Faso
| |
Collapse
|
4
|
Baroncini L, Muller CKS, Kadzioch NP, Wolfensberger R, Russenberger D, Bredl S, Mlambo T, Speck RF. Pro-inflammatory macrophages suppress HIV replication in humanized mice and ex vivo co-cultures. Front Immunol 2024; 15:1439328. [PMID: 39575258 PMCID: PMC11578737 DOI: 10.3389/fimmu.2024.1439328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/04/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction Very little is known about the role of macrophages as immune mediators during natural HIV infection. Humanized mice are an extremely valuable in vivo model for studying HIV pathogenesis. However, the presence of murine mononuclear phagocytes in these models represents a significant limitation for studying their human counterpart. Therefore, we have developed a novel humanized mouse model that allows selective depletion of human myeloid cells at a time point of our choosing. Methods We genetically engineered human hematopoietic stem and progenitor cells (HSPCs) to express an inducible caspase-9 (iCas9) suicide system under a synthetic myeloid promoter. Using these HSPCs, we generated humanized mice. iCasp9 induction in vivo resulted in selective human myeloid cell death in this inducible human myeloid depletion (iHMD) mouse model. In addition, we co-cultured monocyte-derived macrophages with ex vivo HIV-infected PBMCs to further mechanistically investigate the effect of macrophages on HIV replication using flow cytometry, cytokine analysis, and RNA sequencing of both macrophages and CD4+ T cells. Results HIV infection induced a pro-inflammatory phenotype in HIV-infected humanized NSG mice during the early and late stages of HIV infection. Myeloid cell depletion in HIV-infected iHMD-NSG mice resulted in a rapid increase in HIV RNA replication, which was accompanied by a loss of pro-inflammatory cytokines. Co-culture of macrophages with ex vivo HIV-infected PBMCs reproduced their anti-HIV effects observed in vivo. Transcriptomic data showed macrophages upregulate antiviral cytokines and chemokines in co-culture, while inducing CD4+ T cells to upregulate HIV restriction factors and downregulate pathways involved in protein expression and cell replication. Discussion This study describes a novel role of macrophages as effector cells, both ex vivo and in vivo, acting against HIV replication and limiting disease progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Roberto F. Speck
- Department of Infectious Diseases and Hospital Epidemiology, University of Zurich,
University Hospital of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Djibougou DA, Mensah GI, Kaboré A, Toé I, Sawadogo LT, Lompo PF, Kone AMM, Hien H, Meda CZ, Combary A, Bonfoh B, Addo KK, Belem AMG, Dabiré RK, Hoffmann J, Perreau M, Diagbouga PS. Immunological and Haematological Relevance of Helminths and Mycobacterium tuberculosis Complex Coinfection among Newly Diagnosed Pulmonary Tuberculosis Patients from Bobo-Dioulasso, Burkina Faso. Biomedicines 2024; 12:1472. [PMID: 39062045 PMCID: PMC11274831 DOI: 10.3390/biomedicines12071472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
The effect of helminthiasis on host immunity is a neglected area of research, particularly in tuberculosis (TB) infection. This study aimed to evaluate the effect of helminthiasis on immunological and haematological parameters in newly diagnosed TB patients in Bobo-Dioulasso. After all biological analyses, we formed three subpopulations: group 1 (n = 82), as control, were participants without helminthic or Mycobacterium tuberculosis complex infection (Mtb-/Helm-), group 2 (n = 73) were TB patients without helminthic infection (Mtb+/Helm-), and group 3 (n = 22) were TB patients with helminthic infection (Mtb+/Helm+). The proportion of helminth coinfection was 23.16% (22/95) in TB patients, and Schistosoma mansoni infection was found in 77.3% (17/22) cases of helminthiasis observed in this study. A low CD4 T cell count and a low CD4:CD8 ratio were significantly associated with concomitant infection with helminths and the Mtb complex (Mtb+/Helm+) compared to the other groups (p < 0.05). However, there was no statistically significant difference in the CD8 median among the three participating groups (p > 0.05). Lymphopenia, monocytosis, thrombocytosis, and hypochromic microcytic anaemia were the haematological defects observed in the Mtb+/Helm+ and Mtb+/Helm- patients. Exploring these types of immune-haematological biomarkers would be a valuable aid in diagnosing and a better follow-up and monitoring of the tuberculosis-helminthiasis coinfection.
Collapse
Affiliation(s)
- Diakourga Arthur Djibougou
- Doctoral School of Natural Sciences and Agronomy, Université Nazi BONI, Bobo-Dioulasso 1091, Burkina Faso; (I.T.); (L.T.S.); (C.Z.M.); (A.M.-G.B.)
- Infectious Diseases Program, Centre MURAZ, Institut National de Santé Publique, Bobo-Dioulasso 1091, Burkina Faso; (A.M.M.K.); (H.H.); (R.K.D.)
- Institut de Recherche en Sciences de la Santé, CNRST, Bobo-Dioulasso 545, Burkina Faso
| | - Gloria Ivy Mensah
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra 00233, Ghana; (G.I.M.); (K.K.A.)
| | - Achille Kaboré
- Infectious Disease and Health Systems (IDHS), FHI 360, Washington, DC 20037, USA;
| | - Inoussa Toé
- Doctoral School of Natural Sciences and Agronomy, Université Nazi BONI, Bobo-Dioulasso 1091, Burkina Faso; (I.T.); (L.T.S.); (C.Z.M.); (A.M.-G.B.)
- Institut de Recherche en Sciences de la Santé, CNRST, Bobo-Dioulasso 545, Burkina Faso
| | - Leon Tinnoga Sawadogo
- Doctoral School of Natural Sciences and Agronomy, Université Nazi BONI, Bobo-Dioulasso 1091, Burkina Faso; (I.T.); (L.T.S.); (C.Z.M.); (A.M.-G.B.)
- National Tuberculosis Programme, Ministry of Health and Public Hygiene, Ouagadougou 01 P.O. Box 690, Burkina Faso;
| | - Palpouguini Felix Lompo
- Etudes Formation et Recherches Développement en Santé (EFORDS), Ouagadougou 10 P.O. Box 13064, Burkina Faso;
| | - Amariane M. M. Kone
- Infectious Diseases Program, Centre MURAZ, Institut National de Santé Publique, Bobo-Dioulasso 1091, Burkina Faso; (A.M.M.K.); (H.H.); (R.K.D.)
| | - Hervé Hien
- Infectious Diseases Program, Centre MURAZ, Institut National de Santé Publique, Bobo-Dioulasso 1091, Burkina Faso; (A.M.M.K.); (H.H.); (R.K.D.)
- Institut de Recherche en Sciences de la Santé, CNRST, Bobo-Dioulasso 545, Burkina Faso
| | - Clement Ziemlé Meda
- Doctoral School of Natural Sciences and Agronomy, Université Nazi BONI, Bobo-Dioulasso 1091, Burkina Faso; (I.T.); (L.T.S.); (C.Z.M.); (A.M.-G.B.)
- Infectious Diseases Program, Centre MURAZ, Institut National de Santé Publique, Bobo-Dioulasso 1091, Burkina Faso; (A.M.M.K.); (H.H.); (R.K.D.)
| | - Adjima Combary
- National Tuberculosis Programme, Ministry of Health and Public Hygiene, Ouagadougou 01 P.O. Box 690, Burkina Faso;
| | - Bassirou Bonfoh
- Centre Suisse de Recherches Scientifique de Côte d’Ivoire, Adiopodoumé 01 P.O. Box 1303, Côte d’Ivoire;
| | - Kennedy Kwasi Addo
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Legon, Accra 00233, Ghana; (G.I.M.); (K.K.A.)
| | - Adrien Marie-Gaston Belem
- Doctoral School of Natural Sciences and Agronomy, Université Nazi BONI, Bobo-Dioulasso 1091, Burkina Faso; (I.T.); (L.T.S.); (C.Z.M.); (A.M.-G.B.)
| | - Roch Konbobr Dabiré
- Infectious Diseases Program, Centre MURAZ, Institut National de Santé Publique, Bobo-Dioulasso 1091, Burkina Faso; (A.M.M.K.); (H.H.); (R.K.D.)
- Institut de Recherche en Sciences de la Santé, CNRST, Bobo-Dioulasso 545, Burkina Faso
| | - Jonathan Hoffmann
- Département Médical et Scientifique, Fondation Mérieux, 17 rue Bourgelat, 69002 Lyon, France;
| | - Matthieu Perreau
- Faculty of Biology and Medicine, Université de Lausanne, 1010 Lausanne, Switzerland;
| | - Potiandi Serge Diagbouga
- Institut de Recherche en Sciences de la Santé, CNRST, Bobo-Dioulasso 545, Burkina Faso
- Etudes Formation et Recherches Développement en Santé (EFORDS), Ouagadougou 10 P.O. Box 13064, Burkina Faso;
| |
Collapse
|
6
|
Li Q, Xin T, Liu Z, Wang Q, Ma L. Construction of ceRNA regulatory networks for active pulmonary tuberculosis. Sci Rep 2024; 14:10595. [PMID: 38719908 PMCID: PMC11079045 DOI: 10.1038/s41598-024-61451-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
Delayed diagnosis in patients with pulmonary tuberculosis (PTB) often leads to serious public health problems. High throughput sequencing was used to determine the expression levels of lncRNAs, mRNAs, and miRNAs in the lesions and adjacent health lung tissues of patients with PTB. Their differential expression profiles between the two groups were compared, and 146 DElncRs, 447 DEmRs, and 29 DEmiRs were obtained between lesions and adjacent health tissues in patients with PTB. Enrichment analysis for mRNAs showed that they were mainly involved in Th1, Th2, and Th17 cell differentiation. The lncRNAs, mRNAs with target relationship with miRNAs were predicted respectively, and correlation analysis was performed. The ceRNA regulatory network was obtained by comparing with the differentially expressed transcripts (DElncRs, DEmRs, DEmiRs), then 2 lncRNAs mediated ceRNA networks were established. The expression of genes within the network was verified by quantitative real-time PCR (qRT-PCR). Flow cytometric analysis revealed that the proportion of Th1 cells and Th17 cells was lower in PTB than in controls, while the proportion of Th2 cells increased. Our results provide rich transcriptome data for a deeper investigation of PTB. The ceRNA regulatory network we obtained may be instructive for the diagnosis and treatment of PTB.
Collapse
Affiliation(s)
- Qifeng Li
- Xinjiang Institute of Pediatrics, Children's Hospital of Xinjiang Uygur Autonomous Region, NO. 393, Aletai Road, Shayibake District, Urumqi, 830054, Xinjiang, China.
| | - Tao Xin
- Department of Pediatrics, The Eighth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830049, China
| | - Zhigang Liu
- Department of Thoracic Surgery, The Eighth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830049, China
| | - Quan Wang
- Department of Clinical Laboratory, The Eighth Affiliated Hospital of Xinjiang Medical University, Urumqi, 830049, China
| | - Lanhong Ma
- Department of Pediatrics, Children's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 830054, China
| |
Collapse
|
7
|
Karbalaei M, Mosavat A, Soleimanpour S, Farsiani H, Ghazvini K, Amini AA, Sankian M, Rezaee SA. Production and Evaluation of Ag85B:HspX:hFcγ1 Immunogenicity as an Fc Fusion Recombinant Multi-Stage Vaccine Candidate Against Mycobacterium tuberculosis. Curr Microbiol 2024; 81:127. [PMID: 38575759 DOI: 10.1007/s00284-024-03655-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 02/29/2024] [Indexed: 04/06/2024]
Abstract
An urgent need is to introduce an effective vaccine against Mycobacterium tuberculosis (M.tb) infection. In the present study, a multi-stage M.tb immunodominant Fcγ1 fusion protein (Ag85B:HspX:hFcγ1) was designed and produced, and the immunogenicity of purified protein was evaluated. This recombinant fusion protein was produced in the Pichia pastoris expression system. The HiTrap-rPA column affinity chromatography purified and confirmed the fusion protein using ELISA and Western blotting methods. The co-localisation assay was used to confirm its proper folding and function. IFN-γ, IL-12, IL-4, and TGF-β expression in C57BL/6 mice then evaluated the immunogenicity of the construct in the presence and absence of BCG. After expression optimisation, medium-scale production and the Western blotting test confirmed suitable production of Ag85B:HspX:hFcγ1. The co-localisation results on antigen-presenting cells (APCs) showed that Ag85B:HspX:hFcγ1 properly folded and bound to hFcγRI. This strong co-localisation with its receptor can confirm inducing proper Th1 responses. The in vivo immunisation assay showed no difference in the expression of IL-4 but a substantial increase in the expression of IFN-γ and IL-12 (P ≤ 0.02) and a moderate increase in TGF-β (P = 0.05). In vivo immunisation assay revealed that Th1-inducing pathways have been stimulated, as IFN-γ and IL-12 strongly, and TGF-β expression moderately increased in Ag85B:HspX:hFcγ1 group and Ag85B:HspX:hFcγ1+BCG. Furthermore, the production of IFN-γ from splenocytes in the Ag85B:HspX:hFcγ1 group was enormously higher than in other treatments. Therefore, this Fc fusion protein can make a selective multi-stage delivery system for inducing appropriate Th1 responses and is used as a subunit vaccine alone or in combination with others.
Collapse
Affiliation(s)
- Mohsen Karbalaei
- Department of Microbiology and Virology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Arman Mosavat
- Blood Borne Infections Research Center, Academic Center for Education, Culture, and Research (ACECR), Razavi Khorasan, Mashhad, Iran
| | - Saman Soleimanpour
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Farsiani
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kiarash Ghazvini
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Ali Amini
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mojtaba Sankian
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Abdolrahim Rezaee
- Inflammation and Inflammatory Diseases Division, Faculty of Medicine, Immunology Research Center, Mashhad University of Medical Sciences, Azadi-Square, Medical Campus, Mashhad, 9177948564, Iran.
| |
Collapse
|
8
|
Yandrapally S, Agarwal A, Chatterjee A, Sarkar S, Mohareer K, Banerjee S. Mycobacterium tuberculosis EspR modulates Th1-Th2 shift by transcriptionally regulating IL-4, steering increased mycobacterial persistence and HIV propagation during co-infection. Front Immunol 2023; 14:1276817. [PMID: 37928551 PMCID: PMC10621737 DOI: 10.3389/fimmu.2023.1276817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) and HIV are known to mutually support each other during co-infection by multiple mechanisms. This synergistic influence could be either by direct interactions or indirectly through secreted host or pathogen factors that work in trans. Mtb secretes several virulence factors to modulate the host cellular environment for its persistence and escaping cell-intrinsic immune responses. We hypothesized that secreted Mtb transcription factors that target the host nucleus can directly interact with host DNA element(s) or HIV LTR during co-infection, thereby modulating immune gene expression, or driving HIV transcription, helping the synergistic existence of Mtb and HIV. Here, we show that the Mtb-secreted protein, EspR, a transcription regulator, increased mycobacterial persistence and HIV propagation during co-infection. Mechanistically, EspR localizes to the nucleus of the host cells during infection, binds to its putative cognate motif on the promoter region of the host IL-4 gene, activating IL-4 gene expression, causing high IL-4 titers that induce a Th2-type microenvironment, shifting the macrophage polarization to an M2 state as evident from CD206 dominant population over CD64. This compromised the clearance of the intracellular mycobacteria and enhanced HIV propagation. It was interesting to note that EspR did not bind to HIV LTR, although its transient expression increased viral propagation. This is the first report of an Mtb transcription factor directly regulating a host cytokine gene. This augments our understanding of the evolution of Mtb immune evasion strategies and unveils how Mtb aggravates comorbidities, such as HIV co-infection, by modulating the immune microenvironment.
Collapse
|
9
|
da Silva Graça Amoras E, de Morais TG, do Nascimento Ferreira R, Gomes STM, de Sousa FDM, de Paula Souza I, Ishak R, Vallinoto ACR, Queiroz MAF. Association of Cytokine Gene Polymorphisms and Their Impact on Active and Latent Tuberculosis in Brazil's Amazon Region. Biomolecules 2023; 13:1541. [PMID: 37892223 PMCID: PMC10605732 DOI: 10.3390/biom13101541] [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: 08/29/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Some genetic variations in cytokine genes can alter their expression and influence the evolution of Mycobacterium tuberculosis (Mtb) infection. This study aimed to investigate the association of polymorphisms in cytokine genes and variability in plasma levels of cytokines with the development of tuberculosis (TB) and latent tuberculosis infection (LTBI). Blood samples from 245 patients with TB, 80 with LTBI, and healthy controls (n = 100) were included. Genotyping of the IFNG +874A/T, IL6 -174G/C, IL4 -590C/T, and IL10 -1082A/G polymorphisms was performed by real-time PCR, and cytokine levels were determined by flow cytometry. Higher frequencies of genotypes AA (IFNG +874A/T), GG (IL6 -174G/C), TT (IL4 -590C/T), and GG (IL10 -1082A/G) were associated with an increased risk of TB compared to that of LTBI (p = 0.0027; p = 0.0557; p = 0.0286; p = 0.0361, respectively) and the control (p = <0.0001, p = 0.0021; p = 0.01655; p = 0.0132, respectively). In combination, the A allele for IFNG +874A/T and the T allele for IL4 -590C/T were associated with a higher chance of TB (p = 0.0080; OR = 2.753 and p < 0.0001; OR = 3.273, respectively). The TB group had lower levels of IFN-γ and higher concentrations of IL-6, IL-4, and IL-10. Cytokine levels were different between the genotypes based on the polymorphisms investigated (p < 0.05). The genotype and wild-type allele for IFNG +874A/T and the genotype and polymorphic allele for IL4 -590C/T appear to be more relevant in the context of Mtb infection, which has been associated with the development of TB among individuals infected by the bacillus and with susceptibility to active infection but not with susceptibility to latent infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maria Alice Freitas Queiroz
- Virus Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.d.S.G.A.); (T.G.d.M.); (R.d.N.F.); (S.T.M.G.); (F.D.M.d.S.); (I.d.P.S.); (R.I.); (A.C.R.V.)
| |
Collapse
|
10
|
Shaukat SN, Eugenin E, Nasir F, Khanani R, Kazmi SU. Identification of immune biomarkers in recent active pulmonary tuberculosis. Sci Rep 2023; 13:11481. [PMID: 37460564 DOI: 10.1038/s41598-023-38372-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
Tuberculosis (TB) has remained an unsolved problem and a major public health issue, particularly in developing countries. Pakistan is one of the countries with the highest tuberculosis infection rates globally. However, methods or biomarkers to detect early signs of TB infection are limited. Here, we characterized the mRNA profiles of immune responses in unstimulated Peripheral blood mononuclear cells obtained from treatment naïve patients with early signs of active pulmonary tuberculosis without previous history of clinical TB. We identified a unique mRNA profile in active TB compared to uninfected controls, including cytokines such as IL-27, IL-15, IL-2RA, IL-24, and TGFβ, transcription factors such as STAT1 and NFATC1 and immune markers/receptors such as TLR4, IRF1, CD80, CD28, and PTGDR2 from an overall 84 different transcripts analyzed. Among 12 significant differentially expressed transcripts, we identified five gene signatures which included three upregulated IL-27, STAT1, TLR4 and two downregulated IL-24 and CD80 that best discriminate between active pulmonary TB and uninfected controls with AUC ranging from 0.9 to 1. Our data identified a molecular immune signature associated with the early stages of active pulmonary tuberculosis and it could be further investigated as a potential biomarker of pulmonary TB.
Collapse
Affiliation(s)
- Sobia Naz Shaukat
- Immunology and Infectious Diseases Research Laboratory (IIDRL), Department of Microbiology, Karachi University, Karachi, Pakistan.
- Department of Biological and Biomedical Sciences, Aga Khan University Hospital, Stadium Road, P.O. Box 3500, Karachi, 74800, Pakistan.
| | - Eliseo Eugenin
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Faizan Nasir
- Department of Immunology, Dadabhoy Institute of Higher Education, Karachi, Pakistan
| | - Rafiq Khanani
- Dow University of Health Sciences, Ojha Campus, Karachi, Pakistan
| | - Shahana Urooj Kazmi
- Immunology and Infectious Diseases Research Laboratory (IIDRL), Department of Microbiology, Karachi University, Karachi, Pakistan
| |
Collapse
|
11
|
Sousa FDMD, Souza IDP, Amoras EDSG, Lima SS, Cayres-Vallinoto IMV, Ishak R, Vallinoto ACR, Queiroz MAF. Low levels of TNFA gene expression seem to favor the development of pulmonary tuberculosis in a population from the Brazilian Amazon. Immunobiology 2023; 228:152333. [PMID: 36630812 DOI: 10.1016/j.imbio.2023.152333] [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: 09/03/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
TNF-α is a Th1 cytokine profile active in the control of Mycobacterium tuberculosis infection, IL-10 is associated with persistence of bacterial infection. The aim of the study was to investigate the association of TNFA -308G/A and IL10 -819C/T polymorphisms and TNFA and IL10 gene expression levels with pulmonary and extrapulmonary tuberculosis (n = 200) and control (n = 200). The individuals were submitted to genotyping and quantification of gene expression performed by real-time quantitative polymerase chain reaction (qPCR). No association was observed between the frequencies of polymorphisms evaluated and pulmonary tuberculosis. The frequency of polymorphic genotypes for TNFA -308G/A were associated with the extrapulmonary tuberculosis (p = 0.0445). The levels of TNFA expression were lower in the pulmonary tuberculosis group than in the control (p = 0.0009). There was a positive correlation between the levels of TNFA and IL10 in patients with pulmonary tuberculosis (r = 0.560; p = 0.0103). Reduced levels of TNFA expression may promote the formation of an anti-inflammatory microenvironment, favoring the persistence of the bacillus in the host, contributing to the establishment of pulmonary tuberculosis.
Collapse
Affiliation(s)
- Francisca Dayse Martins de Sousa
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66.075-110, Brazil; Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Iury de Paula Souza
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66.075-110, Brazil; Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém, Pará, Brazil
| | - Ednelza da Silva Graça Amoras
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66.075-110, Brazil
| | - Sandra Souza Lima
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66.075-110, Brazil
| | | | - Ricardo Ishak
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66.075-110, Brazil
| | | | - Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará (UFPA), Belém 66.075-110, Brazil.
| |
Collapse
|
12
|
Noto A, Joo V, Mancarella A, Suffiotti M, Pellaton C, Fenwick C, Perreau M, Pantaleo G. CXCL12 and CXCL13 Cytokine Serum Levels Are Associated with the Magnitude and the Quality of SARS-CoV-2 Humoral Responses. Viruses 2022; 14:2665. [PMID: 36560669 PMCID: PMC9785906 DOI: 10.3390/v14122665] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022] Open
Abstract
A better understanding of the immunological markers associated with long-lasting immune responses to SARS-CoV-2 infection is of paramount importance. In the present study, we characterized SARS-CoV-2-specific humoral responses in hospitalized (ICU and non-ICU) and non-hospitalized individuals at six months post-onset of symptoms (POS) (N = 95). We showed that the proportion of individuals with detectable anti-SARS-CoV-2 IgG or neutralizing (NAb) responses and the titers of antibodies were significantly reduced in non-hospitalized individuals, compared to ICU- or non-ICU-hospitalized individuals at 6 months POS. Interestingly, SARS-CoV-2-specific memory B cells persist at 6 months POS in both ICU and non-ICU patients and were enriched in cells harboring an activated and/or exhausted phenotype. The frequency/phenotype of SARS-CoV-2-specific memory B cells and the magnitude of IgG or NAb responses at 6 months POS correlated with the serum immune signature detected at patient admission. In particular, the serum levels of CXCL13, IL-1RA, and G-CSF directly correlated with the frequency of Spike-specific B cells and the magnitude of Spike-specific IgG or NAb, while the serum levels of CXCL12 showed an antagonizing effect. Our results indicate that the balance between CXCL12 and CXCL13 is an early marker associated with the magnitude and the quality of the SARS-CoV-2 humoral memory.
Collapse
Affiliation(s)
- Alessandra Noto
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Victor Joo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Antonio Mancarella
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Madeleine Suffiotti
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Celine Pellaton
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
- Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
13
|
Immunological Interactions between Intestinal Helminth Infections and Tuberculosis. Diagnostics (Basel) 2022; 12:diagnostics12112676. [PMID: 36359526 PMCID: PMC9689268 DOI: 10.3390/diagnostics12112676] [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: 09/21/2022] [Revised: 10/13/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
Helminth infections are among the neglected tropical diseases affecting billions of people globally, predominantly in developing countries. Helminths’ effects are augmented by coincident tuberculosis disease, which infects a third of the world’s population. The role of helminth infections on the pathogenesis and pathology of active tuberculosis (T.B.) remains controversial. Parasite-induced suppression of the efficacy of Bacille Calmette-Guerin (BCG) has been widely reported in helminth-endemic areas worldwide. T.B. immune response is predominantly proinflammatory T-helper type 1 (Th1)-dependent. On the other hand, helminth infections induce an opposing anti-inflammatory Th2 and Th3 immune-regulatory response. This review summarizes the literature focusing on host immune response profiles during single-helminth, T.B. and dual infections. It also aims to necessitate investigations into the complexity of immunity in helminth/T.B. coinfected patients since the research data are limited and contradictory. Helminths overlap geographically with T.B., particularly in Sub-Saharan Africa. Each disease elicits a response which may skew the immune responses. However, these effects are helminth species-dependent, where some parasites have no impact on the immune responses to concurrent T.B. The implications for the complex immunological interactions that occur during coinfection are highlighted to inform government treatment policies and encourage the development of high-efficacy T.B. vaccines in areas where helminths are prevalent.
Collapse
|
14
|
Epidemiological and Cytokine Profile of Patients with Pulmonary and Extrapulmonary Tuberculosis in a Population of the Brazilian Amazon. Microorganisms 2022; 10:microorganisms10102075. [PMID: 36296351 PMCID: PMC9609616 DOI: 10.3390/microorganisms10102075] [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] [Received: 09/07/2022] [Revised: 10/13/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Several factors are associated with the development of different clinical forms of tuberculosis (TB). The present study evaluated epidemiological variables and cytokine levels in samples from 89 patients with TB (75 with pulmonary TB and 14 with extrapulmonary TB) and 45 controls. Cytokines were measured by flow cytometry (Human Th1/Th2/Th17 Cytometric Bead Array kit). The TB group had a higher frequency of individuals who were 39 years of age or older, married, with primary education or illiterate and had a lower family income (p < 0.05). All individuals with extrapulmonary TB reported that they were not working, and the main reasons were related to disease symptoms or treatment. The levels of IFN-γ (OR = 4.06) and IL-4 (OR = 2.62) were more likely to be elevated in the TB group (p = 0.05), and IFN-γ levels were lower in patients with extrapulmonary TB compared to those with pulmonary TB (OR = 0.11; p = 0.0050). The ROC curve was applied to investigate the diagnostic accuracy of IFN-γ levels between the different clinical forms of tuberculosis, resulting in high AUC (0.8661; p < 0.0001), sensitivity (93.85%) and specificity median (65.90%), suggesting that IFN-γ levels are useful to differentiate pulmonary TB from extrapulmonary TB. The dysregulation of pro- and anti-inflammatory cytokine levels represent a risk for the development of TB and contribute to the pathogenesis of the disease, especially variation in IFN-γ levels, which may determine protection or risk for extrapulmonary TB.
Collapse
|
15
|
Ali ZA, Mankhi AA, Ad'hiah AH. Interleukin-37 gene polymorphism and susceptibility to pulmonary tuberculosis among Iraqi patients. Indian J Tuberc 2022; 69:191-200. [PMID: 35379401 DOI: 10.1016/j.ijtb.2021.08.003] [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: 04/17/2021] [Revised: 07/10/2021] [Accepted: 08/05/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Control of tuberculosis (TB) depends on a balance between host's immune factors and bacterial evasion strategies. Interleukin-37 (IL-37) is among the immunomodulatory factors that have been proposed to influence susceptibility to tuberculosis. METHODS A case-control study was conducted on 105 patients with pulmonary TB (37 active, 41 multi-drug resistant and 27 relapse) and 79 healthy controls to determine serum levels and single nucleotide polymorphisms (SNPs) of IL-37. The IL-37 level was assessed with an enzyme-linked immunosorbent kit, while DNA-sequencing was used to detect SNPs in the promoter region of IL37 gene. RESULTS Median level of IL-37 was markedly increased in serum of TB patients compared to controls (325.0 vs. 169.1 pg/mL; p < 0.001). This increase was universally determined in subgroups of patients distributed according to gender, age groups, and clinical type of disease, while no significant differences were found between the subgroups in patients or controls. Analysis of receiver operating characteristic curve confirmed these findings and IL-37 occupied a very good area under the curve, which was 0.816 (95% CI = 0.744-0.888; p < 0.001). At a cut-off value of 185.6 pg/mL, the sensitivity and specificity of IL-37 were 81.0 and 82.3%, respectively. Of the nine detected SNPs (rs2466449 G/A, rs2466450 A/G, rs2723168 G/A, rs3811042 G/A, rs3811045 T/C, rs3811046 G/T, rs3811047 A/G, rs3811048 G/A and rs200782323 G/A), only rs3811048 showed a significant association with TB; the G allele showed a significantly decreased frequency in TB patients compared to controls (25.2 vs. 44.9%; OR = 0.41; p < 0.001). It was possible to assign five haplotypes, and three showed significant differences between patients and controls. Frequency of haplotype A-A-G-A-C-T-G-A-G (0.331 vs. 0.213; OR = 2.10; p = 0.015) was significantly increased in TB patients compared to controls. On the contrary, frequencies of haplotypes A-A-G-A-C-T-G-G-G (0.029 vs. 0.116; OR = 0.24; p = 0.01) and A-A-G-G-T-G-A-G-G (0.140 vs. 0.275; OR = 0.45; p = 0.015) were significantly decreased in patients. CONCLUSIONS IL-37 was up-regulated in the serum of TB patients irrespective of their gender, age or clinical type of disease. SNPs in the promoter region of IL37 gene were proposed to be associated with susceptibility to TB.
Collapse
Affiliation(s)
- Zainab A Ali
- Biotechnology Department, College of Science, University of Baghdad, Baghdad, Iraq
| | - Ahmed A Mankhi
- National Specialized Center for Chest and Respiratory Diseases, Ministry of Health and Environment, Baghdad, Iraq
| | - Ali H Ad'hiah
- Tropical-Biological Research Unit, College of Science, University of Baghdad, Baghdad, Iraq.
| |
Collapse
|
16
|
Sun X, Liu K, Zhao Y, Zhang T. High miRNA-378 expression has high diagnostic values for pulmonary tuberculosis and predicts adverse outcomes. BMC Mol Cell Biol 2022; 23:14. [PMID: 35305574 PMCID: PMC8934448 DOI: 10.1186/s12860-022-00413-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/28/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Pulmonary tuberculosis (TB) is a chronic infectious disease. microRNA (miR)-378 is involved in TB diagnosis. This study explored the effects of miR-378 on TB patients. METHODS A total of 126 TB patients were selected, including 63 active TB and 63 latent TB, with 62 healthy subjects as controls. Serum miR-378 expression was detected. The diagnostic value of miR-378 in TB was analyzed using the ROC curve. Immune inflammatory factor levels were detected and their correlations with miR-378 expression were analyzed. The drug resistance of active TB patients was recorded after standard treatment. miR-378 expression in drug-resistant TB patients was detected. The effects of miR-378 on adverse outcome incidence were analyzed. RESULTS miR-378 expression was highly expressed in TB and the expression was higher in the active group than the latent group. Serum miR-378 expression > 1.490 had high sensitivity and specificity in TB diagnosis. miR-378 expression was correlated with TB clinical indexes. IL-4, IL-6, and IL-1β levels were highly expressed, while IFN-γ, TNF-α, and IL-12 levels were lowly expressed in TB patients. Serum miR-378 level in the active group was positively correlated with serum IL-4, IL-6, and IL-1β, and negatively correlated with serum IFN-γ, TNF-α, and IL-12 concentrations. miR-378 expression was downregulated in the TB treated, single (SDR TB) and multi-drug resistance (MDR TB) groups, the miR-378 expression in SDR TB and MDR TB groups was higher than the TB treated group and lower in the SDR TB group than the MDR TB group. High miR-378 expression predicted higher adverse outcome incidence. CONCLUSIONS High miR-378 expression assisted TB diagnosis and predicted adverse outcomes.
Collapse
Affiliation(s)
- Xiaolu Sun
- Department of Tuberculosis Treatmen and Prevention, Shaanxi Provincial Institute for Tuberculosis Control and Prevention, 4# of Xingqing South Road, Xi'an, 710048, Shaanxi, China.
| | - Kai Liu
- Department of Orthopedics, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhao
- Department of Tuberculosis Treatmen and Prevention, Shaanxi Provincial Institute for Tuberculosis Control and Prevention, 4# of Xingqing South Road, Xi'an, 710048, Shaanxi, China
| | - Tianhua Zhang
- Department of Tuberculosis Treatmen and Prevention, Shaanxi Provincial Institute for Tuberculosis Control and Prevention, 4# of Xingqing South Road, Xi'an, 710048, Shaanxi, China
| |
Collapse
|
17
|
Lundahl MLE, Mitermite M, Ryan DG, Case S, Williams NC, Yang M, Lynch RI, Lagan E, Lebre FM, Gorman AL, Stojkovic B, Bracken AP, Frezza C, Sheedy FJ, Scanlan EM, O'Neill LAJ, Gordon SV, Lavelle EC. Macrophage innate training induced by IL-4 and IL-13 activation enhances OXPHOS driven anti-mycobacterial responses. eLife 2022; 11:74690. [PMID: 36173104 PMCID: PMC9555863 DOI: 10.7554/elife.74690] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 09/28/2022] [Indexed: 01/05/2023] Open
Abstract
Macrophages are a highly adaptive population of innate immune cells. Polarization with IFNγ and LPS into the 'classically activated' M1 macrophage enhances pro-inflammatory and microbicidal responses, important for eradicating bacteria such as Mycobacterium tuberculosis. By contrast, 'alternatively activated' M2 macrophages, polarized with IL-4, oppose bactericidal mechanisms and allow mycobacterial growth. These activation states are accompanied by distinct metabolic profiles, where M1 macrophages favor near exclusive use of glycolysis, whereas M2 macrophages up-regulate oxidative phosphorylation (OXPHOS). Here, we demonstrate that activation with IL-4 and IL-13 counterintuitively induces protective innate memory against mycobacterial challenge. In human and murine models, prior activation with IL-4/13 enhances pro-inflammatory cytokine secretion in response to a secondary stimulation with mycobacterial ligands. In our murine model, enhanced killing capacity is also demonstrated. Despite this switch in phenotype, IL-4/13 trained murine macrophages do not demonstrate M1-typical metabolism, instead retaining heightened use of OXPHOS. Moreover, inhibition of OXPHOS with oligomycin, 2-deoxy glucose or BPTES all impeded heightened pro-inflammatory cytokine responses from IL-4/13 trained macrophages. Lastly, this work identifies that IL-10 attenuates protective IL-4/13 training, impeding pro-inflammatory and bactericidal mechanisms. In summary, this work provides new and unexpected insight into alternative macrophage activation states in the context of mycobacterial infection.
Collapse
Affiliation(s)
- Mimmi LE Lundahl
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland,School of Chemistry, Scanlan Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Morgane Mitermite
- School of Veterinary Medicine, UCD Veterinary Sciences Centre, University College DublinDublinIreland
| | - Dylan Gerard Ryan
- School of Biochemistry and Immunology, Inflammation Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland,Hutchison/MRC Research centre, MRC Cancer Unit, University of CambridgeCambridgeUnited Kingdom
| | - Sarah Case
- School of Biochemistry and Immunology, Macrophage Homeostasis Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Niamh C Williams
- School of Biochemistry and Immunology, Inflammation Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Ming Yang
- Hutchison/MRC Research centre, MRC Cancer Unit, University of CambridgeCambridgeUnited Kingdom
| | - Roisin I Lynch
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Eimear Lagan
- School of Genetics and Microbiology, Department of Genetics, Trinity College DublinDublinIreland
| | - Filipa M Lebre
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Aoife L Gorman
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Bojan Stojkovic
- School of Veterinary Medicine, UCD Veterinary Sciences Centre, University College DublinDublinIreland
| | - Adrian P Bracken
- School of Genetics and Microbiology, Department of Genetics, Trinity College DublinDublinIreland
| | - Christian Frezza
- Hutchison/MRC Research centre, MRC Cancer Unit, University of CambridgeCambridgeUnited Kingdom
| | - Frederick J Sheedy
- School of Biochemistry and Immunology, Macrophage Homeostasis Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Eoin M Scanlan
- School of Chemistry, Scanlan Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Luke AJ O'Neill
- School of Biochemistry and Immunology, Inflammation Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| | - Stephen V Gordon
- School of Veterinary Medicine, UCD Veterinary Sciences Centre, University College DublinDublinIreland
| | - Ed C Lavelle
- School of Biochemistry and Immunology, Adjuvant Research Group, Trinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| |
Collapse
|
18
|
Perreau M, Suffiotti M, Marques-Vidal P, Wiedemann A, Levy Y, Laouénan C, Ghosn J, Fenwick C, Comte D, Roger T, Regina J, Vollenweider P, Waeber G, Oddo M, Calandra T, Pantaleo G. The cytokines HGF and CXCL13 predict the severity and the mortality in COVID-19 patients. Nat Commun 2021; 12:4888. [PMID: 34373466 PMCID: PMC8352963 DOI: 10.1038/s41467-021-25191-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/15/2021] [Indexed: 12/12/2022] Open
Abstract
The objective of the present study was to identify biological signatures of severe coronavirus disease 2019 (COVID-19) predictive of admission in the intensive care unit (ICU). Over 170 immunological markers were investigated in a 'discovery' cohort (n = 98 patients) of the Lausanne University Hospital (LUH-1). Here we report that 13 out of 49 cytokines were significantly associated with ICU admission in the three cohorts (P < 0.05 to P < 0.001), while cellular immunological markers lacked power in discriminating between ICU and non-ICU patients. The cytokine results were confirmed in two 'validation' cohorts, i.e. the French COVID-19 Study (FCS; n = 62) and a second LUH-2 cohort (n = 47). The combination of hepatocyte growth factor (HGF) and C-X-C motif chemokine ligand 13 (CXCL13) was the best predictor of ICU admission (positive and negative predictive values ranging from 81.8% to 93.1% and 85.2% to 94.4% in the 3 cohorts) and occurrence of death during patient follow-up (8.8 fold higher likelihood of death when both cytokines were increased). Of note, HGF is a pleiotropic cytokine with anti-inflammatory properties playing a fundamental role in lung tissue repair, and CXCL13, a pro-inflammatory chemokine associated with pulmonary fibrosis and regulating the maturation of B cell response. Up-regulation of HGF reflects the most powerful counter-regulatory mechanism of the host immune response to antagonize the pro-inflammatory cytokines including CXCL13 and to prevent lung fibrosis in COVID-19 patients.
Collapse
Affiliation(s)
- Matthieu Perreau
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Madeleine Suffiotti
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Pedro Marques-Vidal
- Service of Internal Medicine, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Aurelie Wiedemann
- Vaccine Research Institute, Université Paris-Est, Faculté de Médecine, INSERM U955, Créteil, France.,Assistance Publique-Hôpitaux de Paris, Groupe Henri-Mondor Albert-Chenevier, Service d'Immunologie Clinique, Créteil, France
| | - Yves Levy
- Vaccine Research Institute, Université Paris-Est, Faculté de Médecine, INSERM U955, Créteil, France.,Assistance Publique-Hôpitaux de Paris, Groupe Henri-Mondor Albert-Chenevier, Service d'Immunologie Clinique, Créteil, France
| | - Cédric Laouénan
- AP-HP, Hôpital Bichat, Département Épidémiologie Biostatistiques et Recherche Clinique, INSERM, Centre d'Investigation clinique-Epidémiologie Clinique 1425, Paris, France.,Université de Paris, INSERM, IAME UMR 1137, Paris, France
| | - Jade Ghosn
- AP-HP, Hôpital Bichat, Service de Maladies Infectieuses et Tropicales, Paris, France
| | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Denis Comte
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Thierry Roger
- Service of Infectious Diseases, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Jean Regina
- Service of Infectious Diseases, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Peter Vollenweider
- Service of Internal Medicine, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Gerard Waeber
- Service of Internal Medicine, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Mauro Oddo
- Service of Intensive Care, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Thierry Calandra
- Service of Infectious Diseases, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland. .,Vaccine Research Institute, Université Paris-Est, Faculté de Médecine, INSERM U955, Créteil, France. .,Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
19
|
Jianfang W, Hui W, Le K. LINC00870 regulates Th1/Th2 via the JAK/STAT pathway in peripheral blood mononuclear cells infected with Mycobacterium tuberculosis. Int Immunopharmacol 2021; 102:107188. [PMID: 34407915 DOI: 10.1016/j.intimp.2020.107188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 10/23/2020] [Accepted: 11/05/2020] [Indexed: 12/27/2022]
Abstract
Long, noncoding RNAs reportedly play vital roles in tuberculosis (TB). For example, upregulation of LINC00870 has been observed in tuberculosis, though its role and underlying mechanism remains unclear. In this study, we investigated the expression and effect of LINC00870 in Mycobacterium tuberculosis (MTB) infection by comparing MTB-infected peripheral blood mononuclear cells (PBMCs) with controls. The results showed LINC00870 was significantly increased in MTB infected PBMCs. Additionally, LINC00870 overexpression inhibited Th1-secreted cytokines while promoted Th2-secreted cytokine in MTB-infected PBMCs. Furthermore, LINC00870 promoted p-STAT5 and p-JAK2 protein expression, thus activating JAK/STAT signaling in MTB-infected PBMCs. Sputum and plasma samples were obtained from TB, latent tuberculosis infection (LTBI) patients and healthy controls. The qRT-PCR results showed higher levels of LINC00870 in the sputum and plasma from TB and LTBI patients compared to healthy controls. In addition, LINC00870 were decreased in both TB and LTBI patients after three month of therapy, respectively. The results showed a correlation between LINC00870 inhibition and Th1/Th2, as well as JAK/STAT signaling pathway in PBMCs from active TB patients. In conclusion, higher levels of LINC00870 in tuberculosis might be associated with Th1/Th2-related immune responses by activating JAK/STAT signaling. LINC00870 thus may be a novel biomarker for diagnosing and treating tuberculosis.
Collapse
Affiliation(s)
- Wang Jianfang
- Clinical Laboratory, Zhumadian Central Hospital, Zhumadian, Henan 463000, China
| | - Wang Hui
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, China.
| | - Kang Le
- Department of Neonatology, Zhumadian Central Hospital, Zhumadian, Henan 463000, China
| |
Collapse
|
20
|
Abdelwahab WM, Riffey A, Buhl C, Johnson C, Ryter K, Evans JT, Burkhart DJ. Co-adsorption of synthetic Mincle agonists and antigen to silica nanoparticles for enhanced vaccine activity: A formulation approach to co-delivery. Int J Pharm 2020; 593:120119. [PMID: 33249249 DOI: 10.1016/j.ijpharm.2020.120119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/12/2020] [Accepted: 11/22/2020] [Indexed: 01/14/2023]
Abstract
To date there is no clinically approved adjuvant to drive a protective T-helper cell 17 (Th17) immune response against Mycobacterium tuberculosis (Mtb). Trehalose Dimycolate (TDM) is a glycolipid molecule found in the cell wall of Mtb and similar species. Our team has discovered novel synthetic TDM derivatives that target Mincle receptors and when presented on the surface of amine functionalized silica nanoparticles (A-SNPs) adopt the requisite supramolecular structure for Mincle receptor agonism. Here we describe the preparation and characterization methods for these critical silica nanoparticles (SNPs) co-loaded with Mincle agonists (MAs) and a model antigen. In this work, A-SNPs with a particle diameter of 246 ± 11 nm were prepared and examined for co-adsorption of two synthetic MAs along with ovalbumin (OVA). Due to the insolubility of the studied MAs in aqueous environment, aggregation of the MAs made separation of the adjuvant-loaded A-SNPs from the free-form MAs via centrifugation very challenging. To facilitate separation, we synthesized modified SNPs with comparable amine surface functionalization to the original A-SNPs, but with a superparamagnetic iron oxide core (M-A-SNPs), to allow for magnetic separation. We also substituted Alexa Fluor 488-labeled ovalbumin (AF 488 OVA) for the un-tagged OVA to improve the sensitivity of our quantitation method. A RP-HPLC method was developed to simultaneously determine the amount of adsorption of both the Mincle adjuvant and the model antigen to the A-SNPs. AF488 OVA demonstrated higher than 90% adsorption, with or without the co-adsorption of MAs. Likewise, MAs exhibited higher than 80% adsorption in the presence or absence of antigen. The developed formulations were tested in vitro using murine RAW cells and human peripheral blood mononuclear cells, exhibiting good cytokine induction in both cell lines. Results from these studies indicate that A-SNPs could be used as a customizable presentation platform to co-deliver antigens along with different MAs of varying structural features and biophysical properties.
Collapse
Affiliation(s)
- Walid M Abdelwahab
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Alexander Riffey
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Cassie Buhl
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Craig Johnson
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Kendal Ryter
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Chemistry, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - Jay T Evans
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States
| | - David J Burkhart
- Center for Translational Medicine, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States; Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, United States.
| |
Collapse
|
21
|
McLaughlin TA, Khayumbi J, Ongalo J, Tonui J, Campbell A, Allana S, Gurrion Ouma S, Odhiambo FH, Gandhi NR, Day CL. CD4 T Cells in Mycobacterium tuberculosis and Schistosoma mansoni Co-infected Individuals Maintain Functional TH1 Responses. Front Immunol 2020; 11:127. [PMID: 32117277 PMCID: PMC7020828 DOI: 10.3389/fimmu.2020.00127] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/17/2020] [Indexed: 12/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) is a serious public health concern, infecting a quarter of the world and leading to 10 million cases of tuberculosis (TB) disease and 1. 5 million deaths annually. An effective type 1 CD4 T cell (TH1) immune response is necessary to control Mtb infection and defining factors that modulate Mtb-specific TH1 immunity is important to better define immune correlates of protection in Mtb infection. Helminths stimulate type 2 (TH2) immune responses, which antagonize TH1 cells. As such, we sought to evaluate whether co-infection with the parasitic helminth Schistosoma mansoni (SM) modifies CD4 T cell lineage profiles in a cohort of HIV-uninfected adults in Kisumu, Kenya. Individuals were categorized into six groups by Mtb and SM infection status: healthy controls (HC), latent Mtb infection (LTBI) and active tuberculosis (TB), with or without concomitant SM infection. We utilized flow cytometry to evaluate the TH1/TH2 functional and phenotypic lineage state of total CD4 T cells, as well as CD4 T cells specific for the Mtb antigens CFP-10 and ESAT-6. Total CD4 T cell lineage profiles were similar between SM+ and SM− individuals in all Mtb infection groups. Furthermore, in both LTBI and TB groups, SM infection did not impair Mtb-specific TH1 cytokine production. In fact, SM+ LTBI individuals had higher frequencies of IFNγ+ Mtb-specific CD4 T cells than SM− LTBI individuals. Mtb-specific CD4 T cells were characterized by expression of both classical TH1 markers, CXCR3 and T-bet, and TH2 markers, CCR4, and GATA3. The expression of these markers was similar between SM+ and SM− individuals with LTBI. However, SM+ individuals with active TB had significantly higher frequencies of GATA3+ CCR4+ TH1 cytokine+ Mtb-specific CD4 T cells, compared with SM− TB individuals. Together, these data indicate that Mtb-specific TH1 cytokine production capacity is maintained in SM-infected individuals, and that Mtb-specific TH1 cytokine+ CD4 T cells can express both TH1 and TH2 markers. In high pathogen burden settings where co-infection is common and reoccurring, plasticity of antigen-specific CD4 T cell responses may be important in preserving Mtb-specific TH1 responses.
Collapse
Affiliation(s)
| | - Jeremiah Khayumbi
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Joshua Ongalo
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Joan Tonui
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Angela Campbell
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Salim Allana
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Samuel Gurrion Ouma
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | | | - Neel R Gandhi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Cheryl L Day
- Emory Vaccine Center, Emory University, Atlanta, GA, United States.,Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
22
|
Keikha M, Soleimanpour S, Eslami M, Yousefi B, Karbalaei M. The mystery of tuberculosis pathogenesis from the perspective of T regulatory cells. Meta Gene 2020; 23:100632. [DOI: 10.1016/j.mgene.2019.100632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
23
|
Yong YK, Tan HY, Saeidi A, Wong WF, Vignesh R, Velu V, Eri R, Larsson M, Shankar EM. Immune Biomarkers for Diagnosis and Treatment Monitoring of Tuberculosis: Current Developments and Future Prospects. Front Microbiol 2019; 10:2789. [PMID: 31921004 PMCID: PMC6930807 DOI: 10.3389/fmicb.2019.02789] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 11/18/2019] [Indexed: 12/22/2022] Open
Abstract
Tuberculosis (TB) treatment monitoring is paramount to clinical decision-making and the host biomarkers appears to play a significant role. The currently available diagnostic technology for TB detection is inadequate. Although GeneXpert detects total DNA present in the sample regardless live or dead bacilli present in clinical samples, all the commercial tests available thus far have low sensitivity. Humoral responses against Mycobacterium tuberculosis (Mtb) antigens are generally low, which precludes the use of serological tests for TB diagnosis, prognosis, and treatment monitoring. Mtb-specific CD4+ T cells correlate with Mtb antigen/bacilli burden and hence might serve as good biomarkers for monitoring treatment progress. Omics-based techniques are capable of providing a more holistic picture for disease mechanisms and are more accurate in predicting TB disease outcomes. The current review aims to discuss some of the recent advances on TB biomarkers, particularly host biomarkers that have the potential to diagnose and differentiate active TB and LTBI as well as their use in disease prognosis and treatment monitoring.
Collapse
Affiliation(s)
- Yean K Yong
- Laboratory Center, Xiamen University Malaysia, Sepang, Malaysia
| | - Hong Y Tan
- Laboratory Center, Xiamen University Malaysia, Sepang, Malaysia.,Department of Traditional Chinese Medicine, Xiamen University Malaysia, Sepang, Malaysia
| | - Alireza Saeidi
- Department of Pediatrics, Emory Vaccine Center, Atlanta, GA, United States
| | - Won F Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Vijayakumar Velu
- Department of Microbiology and Immunology, Emory Vaccine Center, Atlanta, GA, United States
| | - Rajaraman Eri
- School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Marie Larsson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linkoping University, Linkoping, Sweden
| | - Esaki M Shankar
- Division of Infection Biology and Medical Microbiology, Department of Life Sciences, Central University of Tamil Nadu (CUTN), Thiruvarur, India
| |
Collapse
|
24
|
Lim HS, Lee SI, Park S. Association between Tuberculosis Case and CD44Gene Polymorphism. KOREAN JOURNAL OF CLINICAL LABORATORY SCIENCE 2019. [DOI: 10.15324/kjcls.2019.51.3.323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Hee-Seon Lim
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan, Korea
| | - Sang-In Lee
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan, Korea
| | - Sangjung Park
- Department of Biomedical Laboratory Science, College of Life and Health Sciences, Hoseo University, Asan, Korea
| |
Collapse
|
25
|
Whittaker E, López-Varela E, Broderick C, Seddon JA. Examining the Complex Relationship Between Tuberculosis and Other Infectious Diseases in Children. Front Pediatr 2019; 7:233. [PMID: 31294001 PMCID: PMC6603259 DOI: 10.3389/fped.2019.00233] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Millions of children are exposed to tuberculosis (TB) each year, many of which become infected with Mycobacterium tuberculosis. Most children can immunologically contain or eradicate the organism without pathology developing. However, in a minority, the organism overcomes the immunological constraints, proliferates and causes TB disease. Each year a million children develop TB disease, with a quarter dying. While it is known that young children and those with immunodeficiencies are at increased risk of progression from TB infection to TB disease, our understanding of risk factors for this transition is limited. The most immunologically disruptive process that can happen during childhood is infection with another pathogen and yet the impact of co-infections on TB risk is poorly investigated. Many diseases have overlapping geographical distributions to TB and affect similar patient populations. It is therefore likely that infection with viruses, bacteria, fungi and protozoa may impact on the risk of developing TB disease following exposure and infection, although disentangling correlation and causation is challenging. As vaccinations also disrupt immunological pathways, these may also impact on TB risk. In this article we describe the pediatric immune response to M. tuberculosis and then review the existing evidence of the impact of co-infection with other pathogens, as well as vaccination, on the host response to M. tuberculosis. We focus on the impact of other organisms on the risk of TB disease in children, in particularly evaluating if co-infections drive host immune responses in an age-dependent way. We finally propose priorities for future research in this field. An improved understanding of the impact of co-infections on TB could assist in TB control strategies, vaccine development (for TB vaccines or vaccines for other organisms), TB treatment approaches and TB diagnostics.
Collapse
Affiliation(s)
- Elizabeth Whittaker
- Department of Paediatrics, Imperial College London, London, United Kingdom
- Department of Paediatric Infectious Diseases, Imperial College Healthcare NHS Trust, St. Mary's Campus, London, United Kingdom
| | - Elisa López-Varela
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Claire Broderick
- Department of Paediatrics, Imperial College London, London, United Kingdom
| | - James A. Seddon
- Department of Paediatrics, Imperial College London, London, United Kingdom
- Department of Paediatric Infectious Diseases, Imperial College Healthcare NHS Trust, St. Mary's Campus, London, United Kingdom
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
26
|
Lyadova I, Nikitina I. Cell Differentiation Degree as a Factor Determining the Role for Different T-Helper Populations in Tuberculosis Protection. Front Immunol 2019; 10:972. [PMID: 31134070 PMCID: PMC6517507 DOI: 10.3389/fimmu.2019.00972] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/16/2019] [Indexed: 12/15/2022] Open
Abstract
Efficient tuberculosis (TB) control depends on early TB prediction and prevention. Solution to these tasks requires knowledge of TB protection correlates (TB CoPs), i.e., laboratory markers that are mechanistically involved in the protection and which allow to determine how well an individual is protected against TB or how efficient the candidate TB vaccine is. The search for TB CoPs has been largely focused on different T-helper populations, however, the data are controversial, and no reliable CoPs are still known. Here we discuss the role of different T-helper populations in TB protection focusing predominantly on Th17, “non-classical” Th1 (Th1*) and “classical” Th1 (cTh1) populations. We analyze how these populations differ besides their effector activity and suggest the hypothesis that: (i) links the protective potential of Th17, Th1*, and cTh1 to their differentiation degree and plasticity; (ii) implies different roles of these populations in response to vaccination, latent TB infection (LTBI), and active TB. One of the clinically relevant outcomes of this hypothesis is that over-stimulating T cells during vaccination and biasing T cell response toward the preferential generation of Th1 are not beneficial. The review sheds new light on the problem of TB CoPs and will help develop better strategies for TB control.
Collapse
Affiliation(s)
- Irina Lyadova
- Laboratory of Cellular and Molecular Mechanisms of Histogenesis, Koltsov Institute of Developmental Biology, Moscow, Russia.,Laboratory of Biotechnology, Department of Immunology, Central Tuberculosis Research Institute, Moscow, Russia
| | - Irina Nikitina
- Laboratory of Cellular and Molecular Mechanisms of Histogenesis, Koltsov Institute of Developmental Biology, Moscow, Russia.,Laboratory of Biotechnology, Department of Immunology, Central Tuberculosis Research Institute, Moscow, Russia
| |
Collapse
|
27
|
HIV Infection Functionally Impairs Mycobacterium tuberculosis-Specific CD4 and CD8 T-Cell Responses. J Virol 2019; 93:JVI.01728-18. [PMID: 30541853 PMCID: PMC6384080 DOI: 10.1128/jvi.01728-18] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/28/2018] [Indexed: 12/29/2022] Open
Abstract
Human immunodeficiency virus (HIV) infection is the major risk factor predisposing for Mycobacterium tuberculosis progression from latent tuberculosis infection (LTBI) to tuberculosis disease (TB). Since long-term-treated aviremic HIV-infected individuals remained at higher risk of developing TB than HIV-uninfected individuals, we hypothesized that progression from LTBI to pulmonary TB (PTB) might be due not only to CD4 T-cell depletion but also to M. tuberculosis-specific CD4 T-cell functional impairment. To test this hypothesis, M. tuberculosis-specific T-cell frequencies and cytokine profiles were investigated in untreated Tanzanian individuals suffering from LTBI (n = 20) or PTB (n = 67) and compared to those of untreated M. tuberculosis/HIV-coinfected individuals suffering from LTBI (n = 15) or PTB (n = 10). We showed that HIV infection significantly reduced the proportion of Th2 (interleukin 4 [IL-4]/IL-5/IL-13) producing M. tuberculosis-specific CD4 T cells and IL-2-producing M. tuberculosis-specific CD4 and CD8 T cells in individuals with LTBI or PTB (P < 0.05). Interestingly, the loss of IL-2 production was associated with a significant increase of PD-1 expression on M. tuberculosis-specific CD4 and CD8 T cells (P < 0.05), while the loss of Th2 cytokine production was associated with a significant reduction of Gata-3 expression in memory CD4 T cells (P < 0.05). Finally, we showed that the serum levels of IL-1α, IL-6, C-reactive protein (CRP), IL-23, and IP-10 were significantly reduced in M. tuberculosis/HIV-coinfected individuals with PTB compared to those in HIV-negative individuals with PTB (P < 0.05), suggesting that HIV infection significantly suppresses M. tuberculosis-induced systemic proinflammatory cytokine responses. Taken together, this study suggests that in addition to depleting M. tuberculosis-specific CD4 T cells, HIV infection significantly impairs functionally favorable M. tuberculosis-specific CD4 T-cell responses in Tanzanian individuals with LTBI or PTB.IMPORTANCE Mycobacterium tuberculosis and human immunodeficiency virus (HIV) infections are coendemic in several regions of the world, and M. tuberculosis/HIV-coinfected individuals are more susceptible to progression to tuberculosis disease. We therefore hypothesized that HIV infection would potentially impair M. tuberculosis-specific protective immunity in individuals suffering from latent tuberculosis infection (LTBI) or active pulmonary tuberculosis (PTB). In this study, we demonstrated that M. tuberculosis/HIV-coinfected individuals have fewer circulating M. tuberculosis-specific CD4 T cells and that those that remained were functionally impaired in both LTBI and PTB settings. In addition, we showed that HIV infection significantly interferes with M. tuberculosis-induced systemic proinflammatory cytokine/chemokine responses. Taken together, these data suggest that HIV infection impairs functionally favorable M. tuberculosis-specific immunity.
Collapse
|
28
|
Basingnaa A, Antwi-Baffour S, Nkansah DO, Afutu E, Owusu E. Plasma Levels of Cytokines (IL-10, IFN-γ and TNF-α) in Multidrug Resistant Tuberculosis and Drug Responsive Tuberculosis Patients in Ghana. Diseases 2018; 7:E2. [PMID: 30583589 PMCID: PMC6473728 DOI: 10.3390/diseases7010002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 02/06/2023] Open
Abstract
The emergence of multidrug-resistant tuberculosis (MDR⁻TB) and more recently, extensively drug-resistant (XDR) TB has intensified the need for studies aimed at identifying factors associated with TB drug resistance. This study determined the differences in plasma concentrations of pro-inflammatory (IFN-γ and TNF-α) and anti-inflammatory (IL-10) cytokines in MDR-TB and drug-susceptible (DS) TB patients, in addition to some socio-economic factors. Plasma levels of IL-10, IFN-γ and TNF-α were measured in 83 participants (comprising 49 MDR-TB and 34 DS-TB patients) using sandwich ELISA. Levels of the three cytokines were elevated in MDR-TB patients compared to DS-TB patients. The mean level of IL-10 (7.8 ± 3.61 ρg/mL) measured in MDR-TB cases was relatively higher than those of TNF-α and IFN-γ, and statistically significant (p = 0.0022) when compared to the level of IL-10 (4.8 ± 4.94 ρg/mL) in the DS-TB cases. There were statistically significant associations between MDR-TB and factors such as education level (X² = 9.895, p = 0.043), employment status (X² = 19.404, p = 0.001) and alcoholism (X² = 3.971, p = 0.046). This study adds to the knowledge that IFN-γ, TNF-α and IL-10 play a role in the host response to Mycobacterium tuberculosis (MTB). Alcohol intake can be considered as an important MDR-TB risk factor.
Collapse
Affiliation(s)
- Anthony Basingnaa
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana.
- Ghana Health Service, PMB, Ministries, Accra, Ghana.
| | - Samuel Antwi-Baffour
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana.
| | - Dinah Obenewaa Nkansah
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana.
| | - Emmanuel Afutu
- Department of Medical Microbiology, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana.
| | - Enid Owusu
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana.
| |
Collapse
|
29
|
Divangahi M, Khan N, Kaufmann E. Beyond Killing Mycobacterium tuberculosis: Disease Tolerance. Front Immunol 2018; 9:2976. [PMID: 30619333 PMCID: PMC6305711 DOI: 10.3389/fimmu.2018.02976] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 12/04/2018] [Indexed: 12/30/2022] Open
Abstract
Host defense strategies against infectious diseases are comprised of both host resistance and disease tolerance. Resistance is the ability of the host to prevent invasion or to eliminate the pathogen, while disease tolerance is defined by limiting the collateral tissue damage caused by the pathogen and/or the immune response without exerting direct effects on pathogen growth. Our incomplete understanding of host immunity against tuberculosis (TB) is predominately rooted in our bias toward investigating host resistance. Thus, we must refocus our efforts to understand the entire spectrum of immunity against M. tuberculosis to control TB.
Collapse
Affiliation(s)
- Maziar Divangahi
- Meakins-Christie Laboratories, Departments of Medicine, Microbiology and Immunology, Pathology McGill University, McGill International TB Centre, McGill University Health Centre, Montreal, QC, Canada
| | - Nargis Khan
- Meakins-Christie Laboratories, Departments of Medicine, Microbiology and Immunology, Pathology McGill University, McGill International TB Centre, McGill University Health Centre, Montreal, QC, Canada
| | - Eva Kaufmann
- Meakins-Christie Laboratories, Departments of Medicine, Microbiology and Immunology, Pathology McGill University, McGill International TB Centre, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
30
|
O'Shea MK, Fletcher TE, Muller J, Tanner R, Matsumiya M, Bailey JW, Jones J, Smith SG, Koh G, Horsnell WG, Beeching NJ, Dunbar J, Wilson D, Cunningham AF, McShane H. Human Hookworm Infection Enhances Mycobacterial Growth Inhibition and Associates With Reduced Risk of Tuberculosis Infection. Front Immunol 2018; 9:2893. [PMID: 30619265 PMCID: PMC6302045 DOI: 10.3389/fimmu.2018.02893] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 11/26/2018] [Indexed: 12/22/2022] Open
Abstract
Soil-transmitted helminths and Mycobacterium tuberculosis frequently coincide geographically and it is hypothesized that gastrointestinal helminth infection may exacerbate tuberculosis (TB) disease by suppression of Th1 and Th17 responses. However, few studies have focused on latent TB infection (LTBI), which predominates globally. We performed a large observational study of healthy adults migrating from Nepal to the UK (n = 645). Individuals were screened for LTBI and gastrointestinal parasite infections. A significant negative association between hookworm and LTBI-positivity was seen (OR = 0.221; p = 0.039). Hookworm infection treatment did not affect LTBI conversions. Blood from individuals with hookworm had a significantly greater ability to control virulent mycobacterial growth in vitro than from those without, which was lost following hookworm treatment. There was a significant negative relationship between mycobacterial growth and eosinophil counts. Eosinophil-associated differential gene expression characterized the whole blood transcriptome of hookworm infection and correlated with improved mycobacterial control. These data provide a potential alternative explanation for the reduced prevalence of LTBI among individuals with hookworm infection, and possibly an anti-mycobacterial role for helminth-induced eosinophils.
Collapse
Affiliation(s)
- Matthew K. O'Shea
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
- Royal Centre for Defence Medicine, Joint Medical Command, Birmingham, United Kingdom
| | - Thomas E. Fletcher
- Royal Centre for Defence Medicine, Joint Medical Command, Birmingham, United Kingdom
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Julius Muller
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Rachel Tanner
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Magali Matsumiya
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - J. Wendi Bailey
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jayne Jones
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Steven G. Smith
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Gavin Koh
- Department of Infectious Diseases, Northwick Park Hospital, London, United Kingdom
| | - William G. Horsnell
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, United Kingdom
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Nicholas J. Beeching
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - James Dunbar
- Royal Centre for Defence Medicine, Joint Medical Command, Birmingham, United Kingdom
- Department of Infectious Diseases, The Friarage Hospital, Northallerton, United Kingdom
| | - Duncan Wilson
- Royal Centre for Defence Medicine, Joint Medical Command, Birmingham, United Kingdom
| | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Helen McShane
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
31
|
Kim WS, Kim JS, Kim HM, Kwon KW, Eum SY, Shin SJ. Comparison of immunogenicity and vaccine efficacy between heat-shock proteins, HSP70 and GrpE, in the DnaK operon of Mycobacterium tuberculosis. Sci Rep 2018; 8:14411. [PMID: 30258084 PMCID: PMC6158166 DOI: 10.1038/s41598-018-32799-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/13/2018] [Indexed: 12/17/2022] Open
Abstract
Antigens (Ags) in Mycobacterium tuberculosis (Mtb) that are constitutively expressed, overexpressed during growth, essential for survival, and highly conserved may be good vaccine targets if they induce the appropriate anti-Mtb Th1 immune response. In this context, stress response-related antigens of Mtb might serve as attractive targets for vaccine development as they are rapidly expressed and are up-regulated during Mtb infection in vivo. Our group recently demonstrated that GrpE, encoded by rv0351 as a cofactor of heat-shock protein 70 (HSP70) in the DnaK operon, is a novel immune activator that interacts with DCs to generate Th1-biased memory T cells in an antigen-specific manner. In this study, GrpE was evaluated as a subunit vaccine in comparison with the well-known HSP70 against the hyper-virulent Mtb Beijing K-strain. Both HSP70- and GrpE-specific effector/memory T cells expanded to a similar extent as those stimulated with ESAT-6 in the lung and spleen of Mtb-infected mice, but GrpE only produced a similar level of IFN-γ to that produced by ESAT-6 stimulation during the late phase and the early phase of Mtb K infection, indicating that GrpE is highly-well recognised by the host immune system as a T cell antigen. Mice immunised with the GrpE subunit vaccine displayed enhanced antigen-specific IFN-γ and serum IgG2c responses along with antigen-specific effector/memory T cell expansion in the lungs. In addition, GrpE-immunisation markedly induced multifunctional Th1-type CD4+ T cells co-expressing IFN-γ, TNF-α, and IL-2 in the lungs of Mtb K-infected mice, whereas HSP70-immunisation induced mixed Th1/Th2 immune responses. GrpE-immunisation conferred a more significant protective effect than that of HSP70-immunisation in terms of bacterial reduction and improved inflammation, accompanied by the remarkable persistence of GrpE-specific multifunctional CD4+ T cells. These results suggest that GrpE is an excellent vaccine antigen component for the development of a multi-antigenic Mtb subunit vaccine by generating Th1-biased memory T cells with multifunctional capacity, and confers durable protection against the highly virulent Mtb K.
Collapse
Affiliation(s)
- Woo Sik Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, South Korea
| | - Jong-Seok Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Myunggok Medical Research Institute, College of Medicine, Konyang University, Daejeon, South Korea
| | - Hong Min Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Seok-Yong Eum
- Division of Immunopathology and Cellular Immunology, International Tuberculosis Research Center, Changwon, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
32
|
Shamaei M, Mortaz E, Pourabdollah M, Garssen J, Tabarsi P, Velayati A, Adcock IM. Evidence for M2 macrophages in granulomas from pulmonary sarcoidosis: A new aspect of macrophage heterogeneity. Hum Immunol 2017; 79:63-69. [PMID: 29107084 DOI: 10.1016/j.humimm.2017.10.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/11/2017] [Accepted: 10/24/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Sarcoidosis is a granulomatous disease of unknown etiology. Macrophages play a key role in granuloma formation with the T cells, having a significant impact on macrophage polarization (M1 and M2) and the cellular composition of the granuloma. This study evaluates macrophage polarization in granulomas in pulmonary sarcoidosis. MATERIALS AND METHODS Tissue specimens from the Department of Pathology biobank at the Masih Daneshvari Hospital were obtained. Paraffin sections from 10 sarcoidosis patients were compared with those from 12 cases of tuberculosis using immunohistochemical staining. These sections consisted of mediastinal lymph nodes and transbronchial lung biopsy (TBLB) for sarcoidosis patients versus pleural tissue, neck, axillary lymph nodes and TBLB for tuberculosis patients. The sections were stained for T-cells (CD4+, CD8+) and mature B lymphocytes (CD22+). CD14+ and CD68+ staining was used as a marker of M1 macrophages and CD163+ as a marker for M2 macrophages. RESULTS Immunohistochemical staining revealed a 4/1 ratio of CD4+/CD8+ T-cells in sarcoidosis granuloma sections and a 3/1 ratio in tuberculosis sections. There was no significance difference in single CD4+, CD8+, CD22+, CD14+ and CD68+ staining between sarcoidosis and tuberculosis sections. CD163 expression was significantly increased in sarcoidosis sections compared with those from tuberculosis subjects. CONCLUSION Enhanced CD163+ staining indicates a shift towards M2 macrophage subsets in granulomas from sarcoidosis patients. Further research is required to determine the functional role of M2 macrophages in the immunopathogenesis of sarcoidosis.
Collapse
Affiliation(s)
- Masoud Shamaei
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Esmaeil Mortaz
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran; Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Mihan Pourabdollah
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands; Nutricia Research Centre for Specialized Nutrition, Utrecht, Netherlands
| | - Payam Tabarsi
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aliakbar Velayati
- Mycobacteriology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ian M Adcock
- Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, UK; Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
33
|
Pastille E, Frede A, McSorley HJ, Gräb J, Adamczyk A, Kollenda S, Hansen W, Epple M, Buer J, Maizels RM, Klopfleisch R, Westendorf AM. Intestinal helminth infection drives carcinogenesis in colitis-associated colon cancer. PLoS Pathog 2017; 13:e1006649. [PMID: 28938014 PMCID: PMC5627963 DOI: 10.1371/journal.ppat.1006649] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 10/04/2017] [Accepted: 09/15/2017] [Indexed: 12/26/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract, strongly associated with an increased risk of colorectal cancer development. Parasitic infections caused by helminths have been shown to modulate the host’s immune response by releasing immunomodulatory molecules and inducing regulatory T cells (Tregs). This immunosuppressive state provoked in the host has been considered as a novel and promising approach to treat IBD patients and alleviate acute intestinal inflammation. On the contrary, specific parasite infections are well known to be directly linked to carcinogenesis. Whether a helminth infection interferes with the development of colitis-associated colon cancer (CAC) is not yet known. In the present study, we demonstrate that the treatment of mice with the intestinal helminth Heligmosomoides polygyrus at the onset of tumor progression in a mouse model of CAC does not alter tumor growth and distribution. In contrast, H. polygyrus infection in the early inflammatory phase of CAC strengthens the inflammatory response and significantly boosts tumor development. Here, H. polygyrus infection was accompanied by long-lasting alterations in the colonic immune cell compartment, with reduced frequencies of colonic CD8+ effector T cells. Moreover, H. polygyrus infection in the course of dextran sulfate sodium (DSS) mediated colitis significantly exacerbates intestinal inflammation by amplifying the release of colonic IL-6 and CXCL1. Thus, our findings indicate that the therapeutic application of helminths during CAC might have tumor-promoting effects and therefore should be well-considered. Evidence from epidemiological studies indicates an inverse correlation between the incidence of certain immune-mediated diseases, including inflammatory bowel diseases, and exposure to helminths. As a consequence, helminth parasites were tested for treating IBD patients, resulting in clinical amelioration of the disease due to the induction of an immunosuppressive microenvironment. However, some infection–related cancers can be attributed to helminth infection, probably due to the generation of a microenvironment that might be conductive to the initiation and development of cancer. In the present study, we aimed to unravel the apparently controversial function of helminths in a mouse model of colitis-associated colon cancer. We show that helminth infection in the onset of colitis and colitis-associated colon cancer does not ameliorate colonic inflammation but activates intestinal immune cells that further facilitate tumor development. Therefore, a better understanding of mechanisms by which helminths modulate host immune responses in the gut should be defined precisely before application of helminths in autoimmune diseases like IBD.
Collapse
Affiliation(s)
- Eva Pastille
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Annika Frede
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Henry J. McSorley
- Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jessica Gräb
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Alexandra Adamczyk
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sebastian Kollenda
- Institute for Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Duisburg, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Matthias Epple
- Institute for Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Duisburg, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Rick M. Maizels
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universitaet Berlin, Berlin, Germany
| | - Astrid M. Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- * E-mail:
| |
Collapse
|