1
|
Choi E, Choi HH, Kwon KW, Kim H, Ryu JH, Hong JJ, Shin SJ. Permissive lung neutrophils facilitate tuberculosis immunopathogenesis in male phagocyte NADPH oxidase-deficient mice. PLoS Pathog 2024; 20:e1012500. [PMID: 39178329 PMCID: PMC11376565 DOI: 10.1371/journal.ppat.1012500] [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: 11/14/2023] [Revised: 09/05/2024] [Accepted: 08/12/2024] [Indexed: 08/25/2024] Open
Abstract
NADPH oxidase 2 (NOX2) is an enzyme responsible for generating reactive oxygen species, primarily found in phagocytes. Chronic Granulomatous Disease (CGD), along with bacterial infections such as Mycobacterium tuberculosis (Mtb), is a representative NOX2-deficient X-linked disease characterized by uncontrolled inflammation. However, the precise roles of host-derived factors that induce infection-mediated hyperinflammation in NOX2-deficient condition remain incompletely understood. To address this, we compared Mtb-induced pathogenesis in Nox2-/- and wild type (WT) mice in a sex-dependent manner. Among age- and sex-matched mice subjected to Mtb infection, male Nox2-/- mice exhibited a notable increase in bacterial burden and lung inflammation. This was characterized by significantly elevated pro-inflammatory cytokines such as G-CSF, TNF-α, IL-1α, IL-1β, and IL-6, excessive neutrophil infiltration, and reduced pulmonary lymphocyte levels as tuberculosis (TB) progressed. Notably, lungs of male Nox2-/- mice were predominantly populated with CD11bintLy6GintCXCR2loCD62Llo immature neutrophils which featured mycobacterial permissiveness. By diminishing total lung neutrophils or reducing immature neutrophils, TB immunopathogenesis was notably abrogated in male Nox2-/- mice. Ultimately, we identified G-CSF as the pivotal trigger that exacerbates the generation of immature permissive neutrophils, leading to TB immunopathogenesis in male Nox2-/- mice. In contrast, neutralizing IL-1α and IL-1β, which are previously known factors responsible for TB pathogenesis in Nox2-/- mice, aggravated TB immunopathogenesis. Our study revealed that G-CSF-driven immature and permissive pulmonary neutrophils are the primary cause of TB immunopathogenesis and lung hyperinflammation in male Nox2-/- mice. This highlights the importance of quantitative and qualitative control of pulmonary neutrophils to alleviate TB progression in a phagocyte oxidase-deficient condition.
Collapse
Affiliation(s)
- Eunsol Choi
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Hong-Hee Choi
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Hagyu Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji-Hwan Ryu
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea
- KRIBB School of Bioscience, Korea University of Science & Technology (UST), Daejeon, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
2
|
Mishra A, Khan A, Singh VK, Glyde E, Saikolappan S, Garnica O, Das K, Veerapandian R, Dhandayuthapani S, Jagannath C. The ΔfbpAΔsapM candidate vaccine derived from Mycobacterium tuberculosis H37Rv is markedly immunogenic in macrophages and induces robust immunity to tuberculosis in mice. Front Immunol 2024; 15:1321657. [PMID: 38975346 PMCID: PMC11224292 DOI: 10.3389/fimmu.2024.1321657] [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: 10/14/2023] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Tuberculosis (TB) remains a significant global health challenge, with approximately 1.5 million deaths per year. The Bacillus Calmette-Guérin (BCG) vaccine against TB is used in infants but shows variable protection. Here, we introduce a novel approach using a double gene knockout mutant (DKO) from wild-type Mycobacterium tuberculosis (Mtb) targeting fbpA and sapM genes. DKO exhibited enhanced anti-TB gene expression in mouse antigen-presenting cells, activating autophagy and inflammasomes. This heightened immune response improved ex vivo antigen presentation to T cells. Subcutaneous vaccination with DKO led to increased protection against TB in wild-type C57Bl/6 mice, surpassing the protection observed in caspase 1/11-deficient C57Bl/6 mice and highlighting the critical role of inflammasomes in TB protection. The DKO vaccine also generated stronger and longer-lasting protection than the BCG vaccine in C57Bl/6 mice, expanding both CD62L-CCR7-CD44+/-CD127+ effector T cells and CD62L+CCR7+/-CD44+CD127+ central memory T cells. These immune responses correlated with a substantial ≥ 1.7-log10 reduction in Mtb lung burden. The DKO vaccine represents a promising new approach for TB immunization that mediates protection through autophagy and inflammasome pathways.
Collapse
Affiliation(s)
- Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Vipul Kumar Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Emily Glyde
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| | - Sankaralingam Saikolappan
- Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Omar Garnica
- Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Kishore Das
- Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Raja Veerapandian
- Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Subramanian Dhandayuthapani
- Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX, United States
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, United States
| |
Collapse
|
3
|
Rana HK, Singh AK, Kumar R, Pandey AK. Antitubercular drugs: possible role of natural products acting as antituberculosis medication in overcoming drug resistance and drug-induced hepatotoxicity. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1251-1273. [PMID: 37665346 DOI: 10.1007/s00210-023-02679-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023]
Abstract
Mycobacterium tuberculosis (Mtb) is a pathogenic bacterium which causes tuberculosis (TB). TB control programmes are facing threats from drug resistance. Multidrug-resistant (MDR) and extensively drug-resistant (XDR) Mtb strains need longer and more expensive treatment with many medications resulting in more adverse effects and decreased chances of treatment outcomes. The World Health Organization (WHO) has emphasised the development of not just new individual anti-TB drugs, but also novel medication regimens as an alternative treatment option for the drug-resistant Mtb strains. Many plants, as well as marine creatures (sponge; Haliclona sp.) and fungi, have been continuously used to treat TB in various traditional treatment systems around the world, providing an almost limitless supply of active components. Natural products, in addition to their anti-mycobacterial action, can be used as adjuvant therapy to increase the efficacy of conventional anti-mycobacterial medications, reduce their side effects, and reverse MDR Mtb strain due to Mycobacterium's genetic flexibility and environmental adaptation. Several natural compounds such as quercetin, ursolic acid, berberine, thymoquinone, curcumin, phloretin, and propolis have shown potential anti-mycobacterial efficacy and are still being explored in preclinical and clinical investigations for confirmation of their efficacy and safety as anti-TB medication. However, more high-level randomized clinical trials are desperately required. The current review provides an overview of drug-resistant TB along with the latest anti-TB medications, drug-induced hepatotoxicity and oxidative stress. Further, the role and mechanisms of action of first and second-line anti-TB drugs and new drugs have been highlighted. Finally, the role of natural compounds as anti-TB medication and hepatoprotectants have been described and their mechanisms discussed.
Collapse
Affiliation(s)
- Harvesh Kumar Rana
- Department of Biochemistry, University of Allahabad, Prayagraj (Allahabad), 211002, India
- Department of Zoology, Feroze Gandhi College, Raebareli, 229001, India
| | - Amit Kumar Singh
- Department of Biochemistry, University of Allahabad, Prayagraj (Allahabad), 211002, India
- Department of Botany, BMK Government. Girls College, Balod, Chhattisgarh, 491226, India
| | - Ramesh Kumar
- Department of Biochemistry, University of Allahabad, Prayagraj (Allahabad), 211002, India
- Department of Biochemistry, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Abhay K Pandey
- Department of Biochemistry, University of Allahabad, Prayagraj (Allahabad), 211002, India.
| |
Collapse
|
4
|
Kwon KW, Kang TG, Lee A, Jin SM, Lim YT, Shin SJ, Ha SJ. Protective Efficacy and Immunogenicity of Rv0351/Rv3628 Subunit Vaccine Formulated in Different Adjuvants Against Mycobacterium tuberculosis Infection. Immune Netw 2023; 23:e16. [PMID: 37179749 PMCID: PMC10166659 DOI: 10.4110/in.2023.23.e16] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 05/15/2023] Open
Abstract
Bacillus Calmette-Guerin (BCG) vaccine is the only licensed vaccine for tuberculosis (TB) prevention. Previously, our group demonstrated the vaccine potential of Rv0351 and Rv3628 against Mycobacterium tuberculosis (Mtb) infection by directing Th1-biased CD4+ T cells co-expressing IFN-γ, TNF-α, and IL-2 in the lungs. Here, we assessed immunogenicity and vaccine potential of the combined Ags (Rv0351/Rv3628) formulated in different adjuvants as subunit booster in BCG-primed mice against hypervirulent clinical Mtb strain K (Mtb K). Compared to BCG-only or subunit-only vaccine, BCG prime and subunit boost regimen exhibited significantly enhanced Th1 response. Next, we evaluated the immunogenicity to the combined Ags when formulated with four different types of monophosphoryl lipid A (MPL)-based adjuvants: 1) dimethyldioctadecylammonium bromide (DDA), MPL, and trehalose dicorynomycolate (TDM) in liposome form (DMT), 2) MPL and Poly I:C in liposome form (MP), 3) MPL, Poly I:C, and QS21 in liposome form (MPQ), and 4) MPL and Poly I:C in squalene emulsion form (MPS). MPQ and MPS displayed greater adjuvancity in Th1 induction than DMT or MP did. Especially, BCG prime and subunit-MPS boost regimen significantly reduced the bacterial loads and pulmonary inflammation against Mtb K infection when compared to BCG-only vaccine at a chronic stage of TB disease. Collectively, our findings highlighted the importance of adjuvant components and formulation to induce the enhanced protection with an optimal Th1 response.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Tae Gun Kang
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Ara Lee
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| | - Seung Mo Jin
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT), Department of Nano Engineering, School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea
| | - Sung Jae Shin
- Department of Microbiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Korea
- Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul 03722, Korea
- Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for Biosystems, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
5
|
Witt KD. Role of MHC class I pathways in Mycobacterium tuberculosis antigen presentation. Front Cell Infect Microbiol 2023; 13:1107884. [PMID: 37009503 PMCID: PMC10050577 DOI: 10.3389/fcimb.2023.1107884] [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: 11/25/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
MHC class I antigen processing is an underappreciated area of nonviral host–pathogen interactions, bridging both immunology and cell biology, where the pathogen’s natural life cycle involves little presence in the cytoplasm. The effective response to MHC-I foreign antigen presentation is not only cell death but also phenotypic changes in other cells and stimulation of the memory cells ready for the next antigen reoccurrence. This review looks at the MHC-I antigen processing pathway and potential alternative sources of the antigens, focusing on Mycobacterium tuberculosis (Mtb) as an intracellular pathogen that co-evolved with humans and developed an array of decoy strategies to survive in a hostile environment by manipulating host immunity to its own advantage. As that happens via the selective antigen presentation process, reinforcement of the effective antigen recognition on MHC-I molecules may stimulate subsets of effector cells that act earlier and more locally. Vaccines against tuberculosis (TB) could potentially eliminate this disease, yet their development has been slow, and success is limited in the context of this global disease’s spread. This review’s conclusions set out potential directions for MHC-I-focused approaches for the next generation of vaccines.
Collapse
Affiliation(s)
- Karolina D. Witt
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- *Correspondence: Karolina D. Witt,
| |
Collapse
|
6
|
Costa MFDS, Pereira-Dutra F, Deboosere N, Jouny S, Song OR, Iack G, Souza AL, Roma EH, Delorme V, Bozza PT, Brodin P. Mycobacterium tuberculosis induces delayed lipid droplet accumulation in dendritic cells depending on bacterial viability and virulence. Mol Microbiol 2023; 119:224-236. [PMID: 36579614 DOI: 10.1111/mmi.15023] [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: 11/30/2021] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 12/30/2022]
Abstract
Tuberculosis remains a global health threat with high morbidity. Dendritic cells (DCs) participate in the acute and chronic inflammatory responses to Mycobacterium tuberculosis (Mtb) by directing the adaptive immune response and are present in lung granulomas. In macrophages, the interaction of lipid droplets (LDs) with mycobacteria-containing phagosomes is central to host-pathogen interactions. However, the data available for DCs are still a matter of debate. Here, we reported that bone marrow-derived DCs (BMDCs) were susceptible to Mtb infection and replication at similar rate to macrophages. Unlike macrophages, the analysis of gene expression showed that Mtb infection induced a delayed increase in lipid droplet-related genes and proinflammatory response. Hence, LD accumulation has been observed by high-content imaging in late periods. Infection of BMDCs with killed H37Rv demonstrated that LD accumulation depends on Mtb viability. Moreover, infection with the attenuated strains H37Ra and Mycobacterium bovis-BCG induced only an early transient increase in LDs, whereas virulent Mtb also induced delayed LD accumulation. In addition, infection with the BCG strain with the reintroduced virulence RD1 locus induced higher LD accumulation and bacterial replication when compared to parental BCG. Collectively, our data suggest that delayed LD accumulation in DCs is dependent on mycobacterial viability and virulence.
Collapse
Affiliation(s)
- Maria Fernanda de Souza Costa
- Instituto de Biologia, Departamento de Imunobiologia, Universidade Federal Fluminense, Niteroi, Brazil.,Center for Technological Development in Health, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Filipe Pereira-Dutra
- Immunopharmacology Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Nathalie Deboosere
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Samuel Jouny
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Ok-Ryul Song
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Guilherme Iack
- Instituto de Biologia, Departamento de Imunobiologia, Universidade Federal Fluminense, Niteroi, Brazil.,Immunopharmacology Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Andreia Lamoglia Souza
- Fundação Oswaldo Cruz, Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Eric Henrique Roma
- Fundação Oswaldo Cruz, Laboratory of Immunology and Immunogenetics in Infectious Diseases at Evandro Chagas National Institute of Infectious Diseases, Rio de Janeiro, Brazil
| | - Vincent Delorme
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| | - Patricia T Bozza
- Immunopharmacology Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Priscille Brodin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
7
|
Saelee C, Hanthamrongwit J, Soe PT, Khaenam P, Inthasin N, Ekpo P, Chootong P, Leepiyasakulchai C. Toll-like receptor-mediated innate immune responses by recognition of the recombinant dormancy-associated Mycobacterium tuberculosis proteins Rv2659c and Rv1738. PLoS One 2022; 17:e0273517. [PMID: 36048884 PMCID: PMC9436120 DOI: 10.1371/journal.pone.0273517] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 08/09/2022] [Indexed: 02/08/2023] Open
Abstract
Tuberculosis (TB) caused by Mycobacterium tuberculosis (Mtb) poses a major threat to the global public health. Importantly, latent tuberculosis infection (LTBI) still impedes the elimination of TB incidence since it has a substantial risk to develop active disease. A multi-stage subunit vaccine comprising active and latency antigens of Mtb has been raised as the promising vaccine to trigger immune protection against all stages of TB. Therefore, the discovery of new antigens that could trigger broad immune response is essential. While current development of TB vaccine mainly focuses on protective immunity mediated by adaptive immune response, the knowledge on triggering the innate immune response by antigens is still limited. We showed that recombinant dormancy-associated Mtb proteins Rv2659c and Rv1738 were recognized by human innate immune recognition molecules, Toll-like receptors (TLRs) 2 and 4 by using HEK-Blue™ hTLR2/hTLR4 systems. We further demonstrated that these two proteins activated phosphorylated NF-κB p65 (Ser536) in the human CD14+ blood cells. We also investigated that these two proteins significantly induced level of pro- and anti-inflammatory cytokines (IL-1β, IL-6, IL-8, IL-10 and TNF-α) which were mediated through TLR2 and TLR4 pathways in human peripheral blood mononuclear cells (hPBMCs). These findings suggest that proteins Rv2659c and Rv1738 stimulated innate immune response targeting TLR2 and TLR4 to produce inflammatory cytokines, and their benefits would be valuable for the development of an effective prophylactic tuberculosis vaccine.
Collapse
Affiliation(s)
- Chutiphon Saelee
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| | - Jariya Hanthamrongwit
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| | - Phyu Thwe Soe
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
- Department of Medical Laboratory Technology, University of Medical Technology, Mandalay, Myanmar
| | - Prasong Khaenam
- Faculty of Medical Technology, Center of Standardization and Product Validation, Mahidol University, Bangkok, Thailand
| | - Naharuthai Inthasin
- Faculty of Medicine Siriraj Hospital, Department of Immunology, Mahidol University, Bangkok, Thailand
| | - Pattama Ekpo
- Faculty of Medicine Siriraj Hospital, Department of Immunology, Mahidol University, Bangkok, Thailand
| | - Patchanee Chootong
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| | - Chaniya Leepiyasakulchai
- Faculty of Medical Technology, Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
8
|
Pattanaik KP, Sengupta S, Jit BP, Kotak R, Sonawane A. Host-Mycobacteria conflict: Immune responses of the host vs. the mycobacteria TLR2 and TLR4 ligands and concomitant host-directed therapy. Microbiol Res 2022; 264:127153. [DOI: 10.1016/j.micres.2022.127153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/21/2022] [Accepted: 07/29/2022] [Indexed: 12/15/2022]
|
9
|
Hoseinpur R, Hasani A, Baradaran B, Abdolalizadeh J, Amini Y, Salehi R, Samadi Kafil H, Azizian K, Memar MY, Gholizadeh P, Hasani A. Chitosan nanoparticles containing fusion protein (Hspx–PPE44–EsxV) and resiquimod adjuvant (HPERC) as a novel booster vaccine for Mycobacterium tuberculosis. J Biomater Appl 2022; 37:40-47. [DOI: 10.1177/08853282221079105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
This study attempted to explore the immunogenicity of chitosan nanoparticles containing fusion protein (Hspx–PPE44–EsxV; HPE) and resiquimod adjuvant (HPERC) in BALB/c mice. HPE was initially expressed in E. coli BL21 cells. HPE and resiquimod adjuvant were then encapsulated in chitosan nanoparticles (HPERC). One group of mice were subcutaneously vaccinated on days 0, 14, and 28 with HPERC, and the other group was primed with bacilli Calmette-Guérin (BCG) on day 0 and then boosted with HPERC on days 14 and 28. Two weeks after the last injection, IFN-γ, IL-4, and IL-17 in spleen cell culture supernatants, and IgG2a and IgG1 titers in sera were measured. HPERC size was 130.84 ± 12.08 nm ( n = 5). Zeta potential of HPERC was 29 ± 4 mv. The highest IFN-γ concentration was detected in BCG-primed mice that were boosted with HPERC. In addition, IL-17 production was significantly increased in all groups compared with that of control, except in those that received nanoparticle (NP), adjuvant (ADJ), NP/ADJ, and fusion protein (Hspx–PPE44–EsxV) (HPE). Comparison of IFN-γ and IL-4 concentration determined that Th1 was activated in BCG-primed and HPERC-boosted group in comparison to the other groups. No significant difference in concentration of IL-4 was observed between groups receiving HPERC and BCG-primed and HPERC-boosted group in comparison to group BCG. Concentrations of IgG2a and IgG1 also increased compared to the control group and the rate of IgG2a was higher compared to IgG1. Chitosan containing HPERC vaccine could induce a high level of specific cytokines in mice. The group of mice which first received BCG and then HPERC as booster vaccine could produce significant amounts of IFN-γ, IL-17, and IgG2a.
Collapse
Affiliation(s)
- Rasul Hoseinpur
- Immunology Research Center (IRC)Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alka Hasani
- Immunology Research Center (IRC)Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center (IRC)Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Allied Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Amini
- Department of Microbiology and Virology, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Roya Salehi
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Samadi Kafil
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khalil Azizian
- Department of Microbiology, Faculty of Medicine, Kurdistan University of Medical Science, Sanandaj, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pourya Gholizadeh
- Department of Medical Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center and Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Akbar Hasani
- Department of Clinical Biochemistry and Applied Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Wu J, Yang H, Xu JC, Hu Z, Gu WF, Chen ZY, Xia JX, Lowrie DB, Lu SH, Fan XY. Mycobacterium tuberculosis Rv3628 isan effective adjuvant via activationof dendritic cells for cancer immunotherapy. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:288-302. [PMID: 34786473 PMCID: PMC8571481 DOI: 10.1016/j.omto.2021.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 09/24/2021] [Accepted: 10/07/2021] [Indexed: 12/30/2022]
Abstract
Tumor antigens (Ags) are weakly immunogenic and elicit inadequate immune responses, thus induction of antigen-specific immune activation via the maturation of dendritic cells (DCs) is a strategy used for cancer immunotherapy. In this study, we examined the effect of Rv3628 from Mycobacterium tuberculosis (Mtb) on activation of DCs and anti-tumor immunity in vivo. Intravenous injection of mice with Rv3628 promoted DC activation of spleen and lymph nodes. More importantly, Rv3628 also induced activation of DCs and enhanced Ag presentation in tumor-bearing mice. In mice bearing ovalbumin (OVA)-expressing tumors, combination treatment with Rv3628 and OVA peptide promoted OVA-specific T cell activation and accumulation of interferon (IFN)-γ and tumor necrosis factor (TNF)-α-producing OT-I and OT-II cells in tumor-draining lymph nodes. Moreover, three different tumor Ags in three different tumor models showed enhanced anti-tumor activity with Rv3628 as adjuvant, including inhibition of growth of OVA-expressing B16 melanoma, CT26 carcinoma, and B16 melanoma tumors, and a synergistic effect with anti-programmed cell death protein 1 (PD-1) antibody treatment. Additionally, potential application against human tumors was indicated by similar activation of human peripheral blood DCs by Rv3628. Taken together, these data demonstrate that Rv3628 could be an effective adjuvant in tumor immunotherapy via enhanced capacity of DC activation and Ag presentation.
Collapse
Affiliation(s)
- Juan Wu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China.,TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai 201508, China
| | - Heng Yang
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Jin-Chuan Xu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Zhidong Hu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China.,TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai 201508, China
| | - Wen-Fei Gu
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhen-Yan Chen
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Jing-Xian Xia
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Douglas B Lowrie
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China
| | - Shui-Hua Lu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China.,TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai 201508, China
| | - Xiao-Yong Fan
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai 201508, China.,TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai 201508, China.,School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou 325035, China
| |
Collapse
|
11
|
Han S, Ma W, Jiang D, Sutherlin L, Zhang J, Lu Y, Huo N, Chen Z, Engle JW, Wang Y, Xu X, Kang L, Cai W, Wang L. Intracellular signaling pathway in dendritic cells and antigen transport pathway in vivo mediated by an OVA@DDAB/PLGA nano-vaccine. J Nanobiotechnology 2021; 19:394. [PMID: 34838057 PMCID: PMC8626881 DOI: 10.1186/s12951-021-01116-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/02/2021] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Poly(D, L-lactic-co-glycolic acid) (PLGA) nanoparticles have potential applications as a vaccine adjuvant and delivery system due to its unique advantages as biodegradability and biocompatibility. EXPERIMENTAL We fabricated cationic solid lipid nanoparticles using PLGA and dimethyl-dioctadecyl-ammonium bromide (DDAB), followed by loading of model antigen OVA (antigen ovalbumin, OVA257-264) to form an OVA@DDAB/PLGA nano-vaccine. And we investigated the intracellular signaling pathway in dendritic cells in vitro and antigen transport pathway and immune response in vivo mediated by an OVA@DDAB/PLGA nano-vaccine. RESULTS In vitro experiments revealed that the antigen uptake of BMDCs after nanovaccine incubation was two times higher than pure OVA or OVA@Al at 12 h. The BMDCs were well activated by p38 MAPK signaling pathway. Furthermore, the nano-vaccine induced antigen escape from lysosome into cytoplasm with 10 times increased cross-presentation activity than those of OVA or OVA@Al. Regarding the transport of antigen into draining lymph nodes (LNs), the nano-vaccine could rapidly transfer antigen to LNs by passive lymphatic drainage and active DC transport. The antigen+ cells in inguinal/popliteal LNs for the nano-vaccine were increased over two folds comparing to OVA@Al and OVA at 12 h. Moreover, the antigen of nano-vaccine stayed in LNs for over 7 days, germinal center formation over two folds higher than those of OVA@Al and OVA. After immunization, the nano-vaccine induced a much higher ratio of IgG2c/IgG1 than OVA@Al. It also effectively activated CD4+ T, CD8+ T and B cells for immune memory with a strong cellular response. CONCLUSION These results indicated that DDAB/PLGA NP was a potent platform to improve vaccine immunogenicity by p38 signaling pathway in BMDCs, enhancing transport of antigens to LNs, and higher immunity response.
Collapse
Affiliation(s)
- Shulan Han
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, People's Republic of China
| | - Wenyan Ma
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
- Tianjin University of Science and Technology, Tianjin, 300222, People's Republic of China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Logan Sutherlin
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Jing Zhang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Yu Lu
- Institute of Veterinary Immunology &Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, 210014, People's Republic of China
| | - Nan Huo
- Department of Genetic Engineering Laboratory, Beijing Institute of Biotechnology, Beijing, 100850, People's Republic of China
| | - Zhao Chen
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China
| | - Jonathan W Engle
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, 53705, USA
| | - Yanping Wang
- Tianjin University of Science and Technology, Tianjin, 300222, People's Republic of China.
| | - Xiaojie Xu
- Department of Genetic Engineering Laboratory, Beijing Institute of Biotechnology, Beijing, 100850, People's Republic of China.
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, People's Republic of China.
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin - Madison, Madison, WI, 53705, USA.
| | - Lianyan Wang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, People's Republic of China.
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China.
| |
Collapse
|
12
|
Jia Z, Gong W, Liang Y, Wu X, Zhao W. Prediction and analyses of HLA-II restricted Mycobacterium tuberculosis CD4 + T cell epitopes in the Chinese population. Biotechnol Appl Biochem 2021; 69:1002-1014. [PMID: 33886144 DOI: 10.1002/bab.2171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/12/2021] [Indexed: 11/07/2022]
Abstract
The Bacillus Calmette-Guérin (BCG) vaccine has been used to prevent tuberculosis (TB), but it cannot prevent adults against TB. The Mycobacterium tuberculosis Beijing strain is the most popular strain in China, but no vaccine is designed for the Beijing strain. It is vital to design a multiepitopes-based vaccine against the Beijing strain for the Chinese population. The bioinformatics tools were used to predict CD4+ T-cell epitopes in five protective antigens based on the Chinese population-specific alleles. The antigenicity, allergenicity, toxicity, IFN-γ level, population coverage, and three-dimensional structure were predicted using Vaxijen, AllerTOP, ToxinPred, IFN-γ epitope server, IEDB, and I-TASSER, respectively. One-hundred one promiscuous epitopes were obtained from Rv1813c, Rv2608, Rv3131, and Rv3628 proteins. After screening with antigenicity, allergenicity, toxicity, and IFN-γ level, seven epitopes from Rv2608 and Rv3131 proteins were selected to be vaccine candidates. Further study determined their three-dimensional structure and the coverage in the Chinese population as high as 99%. Our study predicted seven CD4+ T-cell dominant epitopes from the proteins Rv2608 and Rv3131 of M. tuberculosis Beijing strain for the first time, which may provide a basis for improving the design of multiepitopes-based vaccines for TB.
Collapse
Affiliation(s)
- Zaixing Jia
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, 8th Medical Center, Chinese PLA General Hospital, Haidian District, Beijing, China.,Hebei North University, Zhangjiakou, Hebei, China
| | - Wenping Gong
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, 8th Medical Center, Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Yan Liang
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, 8th Medical Center, Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Xueqiong Wu
- Tuberculosis Prevention and Control Key Laboratory/Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, 8th Medical Center, Chinese PLA General Hospital, Haidian District, Beijing, China
| | - Weiguo Zhao
- Department of Respiration, 8th Medical Center, Chinese PLA General Hospital, Haidian District, Beijing, China
| |
Collapse
|
13
|
Park J, Kim H, Kwon KW, Choi HH, Kang SM, Hong JJ, Shin SJ. Toll-like receptor 4 signaling-mediated responses are critically engaged in optimal host protection against highly virulent Mycobacterium tuberculosis K infection. Virulence 2021; 11:430-445. [PMID: 32403973 PMCID: PMC7239029 DOI: 10.1080/21505594.2020.1766401] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Toll-like receptors (TLRs) play critical roles in the innate recognition of Mycobacterium tuberculosis (Mtb) by host immune cells. However, controversy has arisen regarding the role of TLR4 in determining the outcomes of Mtb infection. To address this controversy, the function of TLR4 in the induction of an optimal protective immune response against the highly virulent Mtb K-infection was comparatively investigated in C3 H/HeJ (TLR4-deficient mutant) and C3 H/HeN (TLR4-competent wild-type) mice. Interestingly, following Mtb infection, C3 H/HeJ mice showed a more severe disease phenotype than C3 H/HeN mice, exhibiting reduced weight and a marked increase in bacterial burden along with necrotic lung inflammation. Analysis of the immune cell composition revealed significantly increased neutrophils in the lung and significant production of IL-10 accompanied by the impairment of the protective Th1 response in C3 H/HeJ mice. Reducing the neutrophil numbers by treating C3 H/HeJ mice with an anti-Ly6 G monoclonal antibody (mAb) and blocking IL-10 signaling with an anti-IL-10 receptor mAb reduced the excessive lung inflammation and bacterial burden in C3 H/HeJ mice. Therefore, abundant IL-10 signaling and neutrophils have detrimental effects in TLR4-deficient mice during Mtb infection. However, the blockade of IL-10 signaling produced an increase in the CD11bhiLy6 Ghi neutrophil population, but the phenotypes of these neutrophils were different from those of the CD11bintLy6 Gint neutrophils from mice with controlled infections. Collectively, these results show that TLR4 positively contributes to the generation of an optimal protective immunity against Mtb infection. Furthermore, investigating the TLR4-mediated response will provide insight for the development of effective control measures against tuberculosis.
Collapse
Affiliation(s)
- Jaehun Park
- 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
| | - Hongmin 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
| | - Hong-Hee Choi
- 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
| | - Soon Myung Kang
- 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
| | - Jung Joo Hong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 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
|
14
|
Kim M, Choe Y, Lee H, Jeon MG, Park JH, Noh HS, Cheon YH, Park HJ, Park J, Shin SJ, Lee K, Lee SI. Blockade of translationally controlled tumor protein attenuated the aggressiveness of fibroblast-like synoviocytes and ameliorated collagen-induced arthritis. Exp Mol Med 2021; 53:67-80. [PMID: 33408335 PMCID: PMC8080778 DOI: 10.1038/s12276-020-00546-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/15/2020] [Accepted: 11/05/2020] [Indexed: 01/29/2023] Open
Abstract
Histamine releasing factor/translationally controlled tumor protein (HRF/TCTP) stimulates cancer progression and allergic responses, but the role of HRF/TCTP in rheumatoid arthritis (RA) remains undefined. In this study, we explored the pathogenic significance of HRF/TCTP and evaluated the therapeutic effects of HRF/TCTP blockade in RA. HRF/TCTP transgenic (TG) and knockdown (KD) mice with collagen-induced arthritis (CIA) were used to determine the experimental phenotypes of RA. HRF/TCTP levels in the sera of RA patients were measured and compared to those from patients with osteoarthritis (OA), ankylosing spondylitis, Behçet's disease, and healthy controls. HRF/TCTP expression was also assessed in the synovium and fibroblast-like synoviocytes (FLSs) obtained from RA or OA patients. Finally, we assessed the effects of HRF/TCTP and dimerized HRF/TCTP-binding peptide-2 (dTBP2), an HRF/TCTP inhibitor, in RA-FLSs and CIA mice. Our clinical, radiological, histological, and biochemical analyses indicate that inflammatory responses and joint destruction were increased in HRF/TCTP TG mice and decreased in KD mice compared to wild-type littermates. HRF/TCTP levels in the sera, synovial fluid, synovium, and FLSs were higher in patients with RA than in control groups. Serum levels of HRF/TCTP correlated well with RA disease activity. The tumor-like aggressiveness of RA-FLSs was exacerbated by HRF/TCTP stimulation and ameliorated by dTBP2 treatment. dTBP2 exerted protective and therapeutic effects in CIA mice and had no detrimental effects in a murine tuberculosis model. Our results indicate that HRF/TCTP is a novel biomarker and therapeutic target for the diagnosis and treatment of RA.
Collapse
Affiliation(s)
- Mingyo Kim
- grid.256681.e0000 0001 0661 1492Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, 52727 Republic of Korea
| | - Yongho Choe
- grid.256681.e0000 0001 0661 1492Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, 52727 Republic of Korea
| | - Heewon Lee
- grid.255649.90000 0001 2171 7754Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Min-Gyu Jeon
- grid.256681.e0000 0001 0661 1492Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, 52727 Republic of Korea
| | - Jin-Ho Park
- grid.256681.e0000 0001 0661 1492Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, 52727 Republic of Korea
| | - Hae Sook Noh
- grid.256681.e0000 0001 0661 1492Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, 52727 Republic of Korea
| | - Yun-Hong Cheon
- grid.256681.e0000 0001 0661 1492Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, 52727 Republic of Korea
| | - Hee Jin Park
- grid.256681.e0000 0001 0661 1492Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, 52727 Republic of Korea
| | - Jaehun Park
- grid.15444.300000 0004 0470 5454Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Sung Jae Shin
- grid.15444.300000 0004 0470 5454Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722 Republic of Korea
| | - Kyunglim Lee
- grid.255649.90000 0001 2171 7754Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, 03760 Republic of Korea
| | - Sang-Il Lee
- grid.256681.e0000 0001 0661 1492Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, 52727 Republic of Korea
| |
Collapse
|
15
|
Fusion of Dendritic Cells Activating Rv2299c Protein Enhances the Protective Immunity of Ag85B-ESAT6 Vaccine Candidate against Tuberculosis. Pathogens 2020; 9:pathogens9110865. [PMID: 33105734 PMCID: PMC7690420 DOI: 10.3390/pathogens9110865] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022] Open
Abstract
In Mycobacterium tuberculosis infection, naïve T cells that encounter mycobacterial antigens through dendritic cells (DCs) induce various CD4+ T-cell responses; therefore, appropriate DC activation is the key for protective immunity against tuberculosis. We previously found that Rv2299c-matured DCs induce Th1 differentiation with bactericidal activity. In this study, to prove that Rv2299c could enhance the protective immunity of other vaccine candidates comprising T-cell-stimulating antigens, Ag85B-ESAT6, a well-known vaccine candidate, was selected as a fusion partner of Rv2299c. Recombinant Rv2299c-Ag85B-ESAT6 protein induced DC maturation and activation. Furthermore, fusion of Rv2299c enhanced the protective efficacy of the Ag85B-ESAT6 vaccine in a mouse model and significantly higher production of TNF-α and IL-2 was detected in the lungs, spleen, and lymph nodes of the group immunized with the Rv2299c-fused protein than with Ag85B-ESAT6. In addition, fusion of Rv2299c enhanced the Ag85B-ESAT6-mediated expansion of multifunctional CD4+ T cells. These data suggested that the DC-activating protein Rv2299c may potentiate the protective immunity of the vaccine candidate comprising T cell antigens.
Collapse
|
16
|
Lee KI, Choi S, Choi HG, Kebede SG, Dang TB, Back YW, Park HS, Kim HJ. Recombinant Rv3261 protein of Mycobacterium tuberculosis induces apoptosis through a mitochondrion-dependent pathway in macrophages and inhibits intracellular bacterial growth. Cell Immunol 2020; 354:104145. [DOI: 10.1016/j.cellimm.2020.104145] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 02/08/2023]
|
17
|
Choi HH, Kwon KW, Han SJ, Kang SM, Choi E, Kim A, Cho SN, Shin SJ. PPE39 of the Mycobacterium tuberculosis strain Beijing/K induces Th1-cell polarization through dendritic cell maturation. J Cell Sci 2019; 132:jcs.228700. [PMID: 31371491 DOI: 10.1242/jcs.228700] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 07/17/2019] [Indexed: 12/22/2022] Open
Abstract
In a previous study, we have identified MTBK_24820, the complete protein form of PPE39 in the hypervirulent Mycobacterium tuberculosis (Mtb) strain Beijing/K by using comparative genomic analysis. PPE39 exhibited vaccine potential against Mtb challenge in a murine model. Thus, in this present study, we characterize PPE39-induced immunological features by investigating the interaction of PPE39 with dendritic cells (DCs). PPE39-treated DCs display reduced dextran uptake and enhanced MHC-I, MHC-II, CD80 and CD86 expression, indicating that this PPE protein induces phenotypic DC maturation. In addition, PPE39-treated DCs produce TNF-α, IL-6 and IL-12p70 to a similar and/or greater extent than lipopolysaccharide-treated DCs in a dose-dependent manner. The activating effect of PPE39 on DCs was mediated by TLR4 through downstream MAPK and NF-κB signaling pathways. Moreover, PPE39-treated DCs promoted naïve CD4+ T-cell proliferation accompanied by remarkable increases of IFN-γ and IL-2 secretion levels, and an increase in the Th1-related transcription factor T-bet but not in Th2-associated expression of GATA-3, suggesting that PPE39 induces Th1-type T-cell responses through DC activation. Collectively, the results indicate that the complete form of PPE39 is a so-far-unknown TLR4 agonist that induces Th1-cell biased immune responses by interacting with DCs.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Hong-Hee Choi
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Seung Jung Han
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Soon Myung Kang
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Eunsol Choi
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Ahreum Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sang-Nae Cho
- Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, South Korea .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, South Korea.,Institute for Immunology and Immunological Disease, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
18
|
Back YW, Choi S, Choi HG, Shin KW, Son YJ, Paik TH, Kim HJ. Cell wall skeleton of Mycobacterium bovis BCG enhances the vaccine potential of antigen 85B against tuberculosis by inducing Th1 and Th17 responses. PLoS One 2019; 14:e0213536. [PMID: 30849108 PMCID: PMC6407753 DOI: 10.1371/journal.pone.0213536] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 02/22/2019] [Indexed: 11/18/2022] Open
Abstract
A safe and effective adjuvant is necessary to induce reliable protective efficacy of the protein-based vaccines against tuberculosis (TB). Mycobacterial components, such as synthetic cord factor and arabinogalactan, have been used as one of the adjuvant components. Mycobacterium bovis bacillus Calmette- Guérin cell-wall skeleton (BCG-CWS) has been used as an effective immune-stimulator. However, it is not proven whether BCG-CWS can be an effective adjuvant for the subunit protein vaccine of TB. In this study, we demonstrated that the BCG-CWS effectively coupled with Ag85B and enhanced the conjugated Ag85B activity on the maturation of dendritic cells (DCs). Ag85B-BCG-CWS-matured DCs induced significant Th1 and Th17 responses when compared to BCG-CWS or Ag85B alone. In addition, significant Ag85B-specific Th1 and Th17 responses were induced in Ag85B-BCG-CWS-immunized mice before infection with M. tuberculosis and maintained after infection. Moreover, Ag85B-BCG-CWS showed significant protective effect comparable to live BCG at 6 weeks after infection and maintained its protective efficacy at 32 weeks post-challenge, whereas live BCG did not. These results suggest that the BCG-CWS may be an effective adjuvant candidate for a protein-based vaccine against TB.
Collapse
Affiliation(s)
- Yong Woo Back
- Department of Microbiology, and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seunga Choi
- Department of Microbiology, and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Han-Gyu Choi
- Department of Microbiology, and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Ki-Won Shin
- Department of Microbiology, and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yeo-Jin Son
- Department of Microbiology, and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Tae-Hyun Paik
- Department of Microbiology, and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hwa-Jung Kim
- Department of Microbiology, and Department of Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
19
|
Kwon KW, Choi HH, Han SJ, Kim JS, Kim WS, Kim H, Kim LH, Kang SM, Park J, Shin SJ. Vaccine efficacy of a Mycobacterium tuberculosis Beijing-specific proline-glutamic acid (PE) antigen against highly virulent outbreak isolates. FASEB J 2019; 33:6483-6496. [PMID: 30753099 DOI: 10.1096/fj.201802604r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacillus Calmette-Guerin vaccine confers insufficient pulmonary protection against tuberculosis (TB), particularly the Mycobacterium tuberculosis (Mtb) Beijing strain infection. Identification of vaccine antigens (Ags) by considering Mtb genetic diversity is crucial for the development of improved TB vaccine. MTBK_20640, a new Beijing genotype-specific proline-glutamic acid-family Ag, was identified by comparative genomic analysis. Its immunologic features were characterized by evaluating interactions with dendritic cells (DCs), and immunogenicity and vaccine efficacy were determined against highly virulent Mtb Beijing outbreak Korean Beijing (K) strain and HN878 strain in murine infection model. MTBK_20640 induced DCs via TLR2 and downstream MAPK and NF-κB signaling pathways, effectively promoting naive CD4-positive (CD4+) T-cell proliferation and IFN-γ production. Different IFN-γ response was observed in mice infected with Mtb K or reference H37Rv strain. Significant induction of T helper type 1 cell-polarized Ag-specific multifunctional CD4+ T cells and a marked Ag-specific IgG2c response were observed in mice immunized with MTBK_20640/glucopyranosyl lipid adjuvant-stable emulsion. The immunization conferred long-term protection against 2 Mtb Beijing outbreak strains, as evidenced by a significant reduction in colony-forming units in the lung and spleen and reduced lung inflammation. MTBK_20640 vaccination conferred long-term protection against highly virulent Mtb Beijing strains. MTBK_20640 may be developed into a novel Ag component in multisubunit TB vaccines in the future.-Kwon, K. W., Choi, H.-H., Han, S. J., Kim, J.-S., Kim, W. S., Kim, H., Kim, L.-H., Kang, S. M., Park, J., Shin, S. J. Vaccine efficacy of a Mycobacterium tuberculosis Beijing-specific proline-glutamic acid (PE) antigen against highly virulent outbreak isolates.
Collapse
Affiliation(s)
- Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hong-Hee Choi
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Jung Han
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong-Seok Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (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
| | - Woo Sik Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (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
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Lee-Han Kim
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Soon Myung Kang
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jaehun Park
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Disease, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
20
|
He J, Fu W, Zhao S, Zhang C, Sun T, Jiang T. Lack of MSMEG_6281, a peptidoglycan amidase, affects cell wall integrity and virulence of Mycobacterium smegmatis. Microb Pathog 2019; 128:405-413. [PMID: 30685363 DOI: 10.1016/j.micpath.2019.01.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 01/29/2023]
Abstract
Mycolyl-arabinogalactan-peptidoglycan (mAGP) is the major content of the mycobacterium cell wall structure and essential for mycobacterial survival. Peptidoglycan (PG) plays an important role in maintenance of cell division, cell wall integrity and pathogenesis. Mycobacterium smegmatis MSMEG_6281, a peptidoglycan amidase, is vital for mycobacterial cell division. However, the effects of MSMEG_6281on cell wall integrity and mycobacterial virulence remain unknown. In the current study, we demonstrate that MSMEG_6281gene knockout in M.smegmatis alters the microbiological characteristics. Our results revealed that MSMEG_6281gene knockout bacteria (M. sm-ΔM_6281) lost their acid-fastness, increased their sensitivity to lipophilic compounds and presented an abnormal morphology. Our results revealed that MSMEG_6281was related to maintaining the cell wall integrity. Furthermore, we investigated the effects of MSMEG_6281 inactivation on mycobacterial virulence using mice models infected by different M.smegmatis strains. MSMEG_6281 inactivation in the M sm-ΔM_6281 infected group caused less mycobacterial colonization, reduced pathological signs, decreased the anti-microbial enzymes production including iNOS and β-defensins in mouse lungs. Moreover, IL-1β and TLR2 expression were significantly down-regulated, while the production of IFN-γ and TNF-α was up-regulated. These findings indicated the diversity of host immune responses induced by different strains of M.smegmatis, suggesting that MSMEG_6281 inactivation impact mycobacterial virulence. In conclusion, the MSMEG_6281 protein plays important roles in maintaining cell wall integrity and mycobacterial virulence.
Collapse
Affiliation(s)
- Jiajia He
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Weizhe Fu
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Shijia Zhao
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Cuili Zhang
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Tieying Sun
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Tao Jiang
- Department of Biotechnology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
21
|
Fereig RM, Abdelbaky HH, Kuroda Y, Nishikawa Y. Critical role of TLR2 in triggering protective immunity with cyclophilin entrapped in oligomannose-coated liposomes against Neospora caninum infection in mice. Vaccine 2019; 37:937-944. [PMID: 30660401 DOI: 10.1016/j.vaccine.2019.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/25/2018] [Accepted: 01/03/2019] [Indexed: 10/27/2022]
Abstract
Neospora caninum is an intracellular protozoan parasite closely related to Toxoplasma gondii. N. caninum is thought to be a major cause of abortion in cattle worldwide. Given the current situation of drastic economic losses and a lack of efficient control strategies against such parasites, the challenge to develop potent vaccine candidates and technologies remains. We investigated the immune stimulating activity of N. caninum cyclophilin (NcCyp) with and without a formulation with oligomannose-coated-liposomes (OML) as the potential adjuvant. NcCyp-OML activated NF-κB in RAW 264.7 cells and triggered interleukin (IL)-12p40 production from murine peritoneal macrophages. In BALB/c mice, immunization with NcCyp-OML was associated with the production of specific antibodies (IgG1 and IgG2a). The specific antibody (IgG1) against naked NcCyp was also observed after the challenge infection, but it was significantly lower than those of NcCyp-OML. Moreover, significant cellular immune responses were induced, including spleen cell proliferation and interferon-gamma production. The immunization of mice with NcCyp-OML, and to lesser extent with naked NcCyp, induced significant protection against challenge with a lethal dose of N. caninum compared with the PBS control group. This protection was associated with a higher survival rate, slight changes in body weight, and lower clinical score of mice. In addition, the significant protective efficacy of NcCyp-OML was confirmed in another mouse strain, male C57BL/6 mice. The current study revealed the marked contribution of Toll-like receptor 2 (TLR2) to the protective immunity triggered by NcCyp-OML because higher numbers of TLR2-/- mice succumbed to a lethal dose of N. caninum compared with C57BL/6 mice. Moreover, prominent spleen cell proliferation and IFN-γ production was induced in NcCyp-OML-immunized mice by a TLR2-dependent mechanism.
Collapse
Affiliation(s)
- Ragab M Fereig
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan; Department of Animal Medicine, Faculty of Veterinary Medicine, South Valley University, Qena City, Qena 83523, Egypt
| | - Hanan H Abdelbaky
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan
| | - Yasuhiro Kuroda
- Department of Applied Biochemistry, Tokai University, Kita-kaname, Hiratsuka, Kanagawa 259-1292, Japan.
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-cho, Obihiro, Hokkaido 080-8555, Japan.
| |
Collapse
|
22
|
Kwon KW, Kim SJ, Kim H, Kim WS, Kang SM, Choi E, Ha SJ, Yoon JH, Shin SJ. IL-15 Generates IFN-γ-producing Cells Reciprocally Expressing Lymphoid-Myeloid Markers during Dendritic Cell Differentiation. Int J Biol Sci 2019; 15:464-480. [PMID: 30745835 PMCID: PMC6367559 DOI: 10.7150/ijbs.25743] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 12/02/2018] [Indexed: 11/23/2022] Open
Abstract
Recently, interest in IL-15-differentiated cells has increased; however, the phenotypic definition of IL-15-differentiated bone marrow-derived cells (IL-15-DBMCs) is still under debate, particularly the generation of IFN-γ-producing innate cells such as premature NK (pre-mNK) cells, natural killer dendritic cells (NKDCs), interferon-producing killer dendritic cells (IKDCs), and type 1 innate lymphoid cells (ILC1s), all of which are IL-15-dependent. Here, we revisited the immunophenotypic characteristics of IFN-γ-producing IL-15-DBMCs and their functional role in the control of intracellular Mycobacterium tuberculosis (Mtb) infection. When comparing the cytokine levels between bone marrow-derived dendritic cells (BMDCs) and IL-15-DBMCs upon stimulation with various TLR agonists, only the CD11cint population of IL-15-DBMCs produced significant levels of IFN-γ, decreased levels of MHC-II, and increased levels of B220. Neither BMDCs nor IL-15-DBMCs were found to express DX5 or NK1.1, which are representative markers for the NK cell lineage and IKDCs. When the CD11cintB220+ population of IL-15-DBMCs was enriched, the Thy1.2+Sca-1+ population showed a marked increase in IFN-γ production. In addition, while depletion of the B220+ and Thy1.2+ populations of IL-15-DBMCs, but not the CD19+ population, inhibited IFN-γ production, enrichment of these cell populations increased IFN-γ. Ultimately, co-culture of sorted IFN-γ-producing B220+Thy1.2+ IL-15-DBMCs with Mtb-infected macrophages resulted in control of the intracellular growth of Mtb via the IFN-γ-nitric oxide axis in a donor cell number-dependent manner. Taken together, the results indicate that IFN-γ-producing IL-15-DBMCs could be redefined as CD11cintB220+Thy1.2+Sca-1+ cells, which phenotypically resemble both IKDCs and ILC1s, and may have therapeutic potential for controlling infectious intracellular bacteria such as Mtb.
Collapse
Affiliation(s)
- 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
| | - So Jeong 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
| | - Hongmin 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
| | - 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
| | - Soon Myung Kang
- 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
| | - Eunsol Choi
- 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
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, South Korea
| | - Joo-Heon Yoon
- The Airway Mucus Institute, and Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, 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
|
23
|
Bezerra-Santos M, do Vale-Simon M, Barreto AS, Cazzaniga RA, de Oliveira DT, Barrios MR, Ferreira AR, Santos-Bio NC, Reed SG, de Almeida RP, Corrêa CB, Duthie MS, de Jesus AR. Mycobacterium leprae Recombinant Antigen Induces High Expression of Multifunction T Lymphocytes and Is Promising as a Specific Vaccine for Leprosy. Front Immunol 2018; 9:2920. [PMID: 30631322 PMCID: PMC6315144 DOI: 10.3389/fimmu.2018.02920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/28/2018] [Indexed: 11/13/2022] Open
Abstract
Leprosy is a chronic disease caused by M. leprae infection that can cause severe neurological complications and physical disabilities. A leprosy-specific vaccine would be an important component within control programs but is still lacking. Given that multifunctional CD4 T cells [i.e., those capable of simultaneously secreting combinations of interferon (IFN)-γ, interleukin (IL)-2, and tumor necrosis factor (TNF)] have now been implicated in the protective response to several infections, we tested the hypothesis if a recombinant M. leprae antigen-specific multifunctional T cells differed between leprosy patients and their healthy contacts. We used whole blood assays and peripheral blood mononuclear cells to characterize the antigen-specific T cell responses of 39 paucibacillary (PB) and 17 multibacillary (MB) leprosy patients and 31 healthy household contacts (HHC). Cells were incubated with either crude mycobacterial extracts (M. leprae cell sonicate–MLCS) and purified protein derivative (PPD) or recombinant ML2028 protein, the homolog of M. tuberculosis Ag85B. Multiplex assay revealed antigen-specific production of IFN-γ and IL-2 from cells of HHC and PB, confirming a Th1 bias within these individuals. Multiparameter flow cytometry then revealed that the population of multifunctional ML2028-specific T cells observed in HHC was larger than that observed in PB patients. Taken together, our data suggest that these multifunctional antigen-specific T cells provide a more effective response against M. leprae infection that prevents the development of leprosy. These data further our understanding of M. leprae infection/leprosy and are instructive for vaccine development.
Collapse
Affiliation(s)
- Márcio Bezerra-Santos
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil.,Department of Health Education, Federal University of Sergipe, Lagarto, Brazil
| | - Marise do Vale-Simon
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil
| | - Aline Silva Barreto
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil
| | | | | | - Mônica Rueda Barrios
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil
| | - Alex Ricardo Ferreira
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil.,Infectious Disease Research Institute, Seattle, WA, United States
| | - Nanci C Santos-Bio
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil.,Infectious Disease Research Institute, Seattle, WA, United States
| | - Steven G Reed
- Instituto de Investigação em Imunologia, INCT-CNPq, São Paulo, Brazil
| | - Roque Pacheco de Almeida
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil.,Infectious Disease Research Institute, Seattle, WA, United States.,Department of Medicine, Federal University of Sergipe, Aracaju, Brazil
| | - Cristiane Bani Corrêa
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil
| | - Malcolm S Duthie
- Instituto de Investigação em Imunologia, INCT-CNPq, São Paulo, Brazil
| | - Amélia Ribeiro de Jesus
- Laboratory of Immunology and Molecular Biology, Federal University of Sergipe, Aracaju, Brazil.,Infectious Disease Research Institute, Seattle, WA, United States.,Department of Medicine, Federal University of Sergipe, Aracaju, Brazil
| |
Collapse
|
24
|
Luo X, Zhang X, Gan L, Zhou C, Zhao T, Zeng T, Liu S, Xiao Y, Yu J, Zhao F. The outer membrane protein Tp92 of Treponema pallidum induces human mononuclear cell death and IL-8 secretion. J Cell Mol Med 2018; 22:6039-6054. [PMID: 30596396 PMCID: PMC6237608 DOI: 10.1111/jcmm.13879] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
Treponema pallidum is the pathogen that causes syphilis, a sexually transmitted disease; however, the pathogenic mechanism of this organism remains unclear. Tp92 is the only T. pallidum outer membrane protein that has structural features similar to the outer membrane proteins of other Gram-negative bacteria, but the exact functions of this protein remain unknown. In the present study, we demonstrated that the recombinant Tp92 protein can induce human mononuclear cell death. Tp92 mediated the human monocytic cell line derived from an acute monicytic leukemia patient (THP-1) cell death by recognizing CD14 and/or TLR2 on cell surfaces. After the stimulation of THP-1 cells by the Tp92 protein, Tp92 may induce atypical pyroptosis of THP-1 cells via the pro-caspase-1 pathway. Meanwhile, this protein caused the apoptosis of THP-1 cells via the receptor-interacting protein kinase 1/caspase-8/aspase-3 pathway. Tp92 reduced the number of monocytes among peripheral blood mononuclear cells. Interestingly, further research showed that Tp92 failed to increase the tumour necrosis factor-α, interleukin (IL)-1β, IL-6, IL-10, IL-18 and monocyte chemotactic protein 1 (MCP)-1 levels but slightly elevated the IL-8 levels via the Nuclear Factor (NF)-κB pathway in THP-1 cells. The data suggest that Tp92 recognizes CD14 and TLR2, transfers the signal to a downstream pathway, and activates NF-κB to mediate the production of IL-8. This mechanism may help T. pallidum escape recognition and elimination by the host innate immune system.
Collapse
MESH Headings
- Antigens, Surface/genetics
- Bacterial Proteins/genetics
- Caspase 1/genetics
- Cell Death/genetics
- Cell Line, Tumor
- Cytokines/genetics
- Host-Pathogen Interactions/genetics
- Humans
- Interleukin-8/genetics
- Leukemia, Monocytic, Acute/genetics
- Leukemia, Monocytic, Acute/microbiology
- Leukemia, Monocytic, Acute/pathology
- Leukocytes, Mononuclear/microbiology
- Leukocytes, Mononuclear/pathology
- Lipopolysaccharide Receptors/genetics
- NF-kappa B/genetics
- Recombinant Proteins/genetics
- Signal Transduction/genetics
- Syphilis/genetics
- Syphilis/microbiology
- Syphilis/pathology
- Toll-Like Receptor 2/genetics
- Treponema pallidum/genetics
- Treponema pallidum/pathogenicity
Collapse
Affiliation(s)
- Xi Luo
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
| | - Xiaohong Zhang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
- Department of Histology and EmbryologySchool of MedicineUniversity of South ChinaHengyangChina
| | - Lin Gan
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
| | - Chenglong Zhou
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
| | - Tie Zhao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
| | - Tiebing Zeng
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
| | - Shuangquan Liu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
- Department of Clinical LaboratoryThe First Affiliated Hospital of University of South ChinaHengyangChina
| | - Yongjian Xiao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
- Department of Clinical LaboratoryThe Second Affiliated Hospital of University of South ChinaHengyangChina
| | - Jian Yu
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
| | - Feijun Zhao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan ProvinceCollaborative Innovation Center for New Molecular Drug ResearchUniversity of South ChinaHengyangChina
| |
Collapse
|
25
|
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
|
26
|
Zhang R, Sun Q, Chen Y, Sun X, Gu Y, Zhao Z, Cheng Y, Zhao L, Huang J, Zhan B, Zhu X. Ts-Hsp70 induces protective immunity against Trichinella spiralis infection in mouse by activating dendritic cells through TLR2 and TLR4. PLoS Negl Trop Dis 2018; 12:e0006502. [PMID: 29775453 PMCID: PMC5979045 DOI: 10.1371/journal.pntd.0006502] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/31/2018] [Accepted: 05/05/2018] [Indexed: 02/03/2023] Open
Abstract
Background Trichinellosis is a serious food-borne parasitic zoonosis worldwide. In the effort to develop vaccine against Trichinella infection, we have identified Trichinella spiralis Heat shock protein 70 (Ts-Hsp70) elicits partial protective immunity against T. spiralis infection via activating dendritic cells (DCs) in our previous study. This study aims to investigate whether DCs were activated by Ts-Hsp70 through TLR2 and/or TLR4 pathways. Methods and findings After blocking with anti-TLR2 and TLR4 antibodies, the binding of Ts-Hsp70 to DCs was significantly reduced. The reduced binding effects were also found in TLR2 and TLR4 knockout (TLR2-/- and TLR4-/-) DCs. The expression of TLR2 and TLR4 on DCs was upregulated after treatment with Ts-Hsp70 in vitro. These results suggest that Ts-Hsp70 is able to directly bind to TLR2 and TLR4 on the surface of mouse bone morrow-derived DCs. In addition, the expression of the co-stimulatory molecules (CD80, CD83) on Ts-Hsp70-induced DCs was reduced in TLR2-/- and TLR4-/- mice. More evidence showed that Ts-Hsp70 reduced its activation on TLR2/4 knockout DCs to subsequently activate the naïve T-cells. Furthermore, Ts-Hsp70 elicited protective immunity against T. spiralis infection was reduced in TLR2-/- and TLR4-/- mice correlating with the reduced humoral and cellular immune responses. Conclusion This study demonstrates that Ts-Hsp70 activates DCs through TLR2 and TLR4, and TLR2 and TLR4 play important roles in Ts-Hsp70-induced DCs activation and immune responses. Trichinellosis is a serious food-borne parasitic zoonosis caused by tissue-dwelling nematode Trichinella spiralis. Vaccine development is needed as an alternative approach to control the infection in domestic livestock or in humans. Ts-Hsp70 has been identified to elicit partial protective immunity against Trichinella spiralis infection via activating dendritic cells (DCs) in our previous study. This study aims to investigate the pathway(s) through which the Ts-Hsp70 activates DCs. Our results identified that Ts-Hsp70 could bind to DCs which was inhibited by blocking TLR2 and TLR4 with antibodies or TLR2 and TLR4 knockout. Ts-Hsp70 stimulated the expression of TLR2 and TLR4 and the co-stimulatory CD80, CD83 and CD86 on the surface of DCs which was reduced in TLR2 or TLR4 knockout mice. With TLR2 or TLR4 knockout, DCs were less stimulated by Ts-Hsp70 and subsequently reduce the activation of naïve T-cells. The protective immunity induced by Ts-Hsp70 against T. spiralis infection was also reduced in TLR2 or TLR4 knockout mice. The results conclude that Ts-Hsp70 activates DCs through activating TLR2 and TLR4 and TLR2 and TLR4 play important roles in Ts-Hsp70-induced protective immunity against Trichinella infection.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Research Centre of Microbiome, Capital Medical University, Beijing, China
| | - Qing Sun
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yi Chen
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Research Centre of Microbiome, Capital Medical University, Beijing, China
| | - Ximeng Sun
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Research Centre of Microbiome, Capital Medical University, Beijing, China
| | - Yuan Gu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhang Zhao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Research Centre of Microbiome, Capital Medical University, Beijing, China
| | - Yuli Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Limei Zhao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Research Centre of Microbiome, Capital Medical University, Beijing, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bin Zhan
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Research Centre of Microbiome, Capital Medical University, Beijing, China
- * E-mail:
| |
Collapse
|
27
|
Zhang J, Zhao Z, Zhong H, Wu L, Zhou W, Peng W, Hu X, Song J, Liu T, Wu Q, Bai H, Zhou Y, Chen X, Chen J, Lu X, Ying B. Importance of common TLR2 genetic variants on clinical phenotypes and risk in tuberculosis disease in a Western Chinese population. INFECTION GENETICS AND EVOLUTION 2018; 60:173-180. [PMID: 29486365 DOI: 10.1016/j.meegid.2018.02.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Abundant studies have suggested that TLR2 genetic variants involve in susceptibility to TB infection. We tried to verified the hypothesis that TLR2 genetic loci effect on the susceptibility to TB in the Western Chinese population. METHODS A total 1109 individuals (634 TB patients and 475 healthy controls) were genotyped for rs3804099, rs3804100 and rs76112010 by using a custom-by-design 2x48-Plex SNP scan TM Kit. The statistical analysis between candidate 3 SNPs and risk and phenotypes of TB were conducted in this study. Significant SNPs were further interrogated in relation to TB susceptibility to TB infection and clinical phenotypes. RESULTS None of the three genetic loci (rs3804099, rs3804100 and rs76112010) showed statistically significant differences between all TB cases and healthy controls in genotype, allele frequencies and genetic models (all p > 0.05). Statistical comparisons of retreatment TB cases and healthy controls or primary cases revealed that rs3804099 was significantly associated with the increased risk of developing TB in Western Chinese population. For genotypes frequencies, the subgroups of retreatment TB group versus healthy control group analysis and retreatment TB group versus primary TB group analysis results showed the p = 0.041 and p = 0.002 respectively. For recessive model, the subgroup of retreatment TB group versus healthy control group and retreatment TB group versus primary TB group analyses showed the p = 0.028 and P = 0.002 after Bonferroni correction respectively. Furthermore, analysis of the genotypes of rs76112010 in relation to clinical phenotypes of active TB using the dominant model demonstrated that it was strongly correlated with different hematological parameters (Erythrocyte P = 0.043, Hemoglobin P = 0.047, Hematocrit P = 0.027). CONCLUSION Our study presented the significant associations of rs3804099 with TB susceptibility in the retreatment TB subgroup analysis. Our study proposed that common TLR2 genetic variants may influence TB development and disease phenotypes in Western Chinese population.
Collapse
Affiliation(s)
- Jingwei Zhang
- Department of Laboratory Medicine, Chengdu Second People's Hospital, Chengdu 610017, PR China
| | - Zhenzhen Zhao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Huiyu Zhong
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lijuan Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wenjing Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wu Peng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xuejiao Hu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jiajia Song
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Tangyuheng Liu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qian Wu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Hao Bai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yi Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xuerong Chen
- Division of Pulmonary Disease, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jie Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xiaojun Lu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
28
|
Choi HG, Choi S, Back YW, Paik S, Park HS, Kim WS, Kim H, Cha SB, Choi CH, Shin SJ, Kim HJ. Rv2299c, a novel dendritic cell-activating antigen of Mycobacterium tuberculosis, fused-ESAT-6 subunit vaccine confers improved and durable protection against the hypervirulent strain HN878 in mice. Oncotarget 2017; 8:19947-19967. [PMID: 28193909 PMCID: PMC5386736 DOI: 10.18632/oncotarget.15256] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 12/16/2016] [Indexed: 12/29/2022] Open
Abstract
Understanding functional interactions between DCs and antigens is necessary for achieving an optimal and desired immune response during vaccine development. Here, we identified and characterized protein Rv2299c (heat-shock protein 90 family), which effectively induced DC maturation. The Rv2299c-maturated DCs showed increased expression of surface molecules and production of proinflammatory cytokines. Rv2299c induced these effects by binding to TLR4 and stimulating the downstream MyD88-, MAPK- and NF-κB-dependent signaling pathways. The Rv2299c-maturated DCs also showed an induced Th1 cell response with bactericidal activity and expansion of effector/memory T cells. The Rv2299c-ESAT-6 fused protein had greater immunoreactivity than ESAT-6. Furthermore, boosting BCG with the fused protein significantly reduced hypervirulent Mycobacterium tuberculosis HN878 burdens post-challenge. The pathological study of the lung from the challenged mice assured the efficacy of the fused protein. The fused protein boosting also induced Rv2299c-ESAT-6-specific multifunctional CD4+ T-cell response in the lungs of the challenged mice. Our findings suggest that Rv2299c is an excellent candidate for the rational design of an effective multiantigenic TB vaccine.
Collapse
Affiliation(s)
- Han-Gyu Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seunga Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yong Woo Back
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seungwha Paik
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hye-Soo Park
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Woo Sik Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Bin Cha
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chul Hee Choi
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hwa-Jung Kim
- Department of Microbiology, and Medical Science, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
29
|
Izzo AA. Tuberculosis vaccines - perspectives from the NIH/NIAID Mycobacteria vaccine testing program. Curr Opin Immunol 2017; 47:78-84. [PMID: 28750280 PMCID: PMC5626602 DOI: 10.1016/j.coi.2017.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/10/2017] [Indexed: 12/14/2022]
Abstract
The development of novel vaccine candidates against infections with Mycobacterium tuberculosis has highlighted our limited understanding of immune mechanisms required to kill M. tuberculosis. The induction of a Th1 immunity is vital, but new studies are required to identify other mechanisms that may be necessary. Novel vaccines formulations that invoke effector cells such as innate lymphoid cells may provide an environment that promote effector mechanisms including T cell and B cell mediated immunity. Identifying pathways associated with killing this highly successful infectious agent has become critical to achieving the goal of reducing the global tuberculosis burden.
Collapse
Affiliation(s)
- Angelo A Izzo
- Colorado State University, Department of Microbiology, Immunology & Pathology, 1682 Campus Delivery, Fort Collins, CO 80523, United States.
| |
Collapse
|
30
|
Faridgohar M, Nikoueinejad H. New findings of Toll-like receptors involved in Mycobacterium tuberculosis infection. Pathog Glob Health 2017; 111:256-264. [PMID: 28715935 DOI: 10.1080/20477724.2017.1351080] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tuberculosis (TB), an important issue in the present age, affects millions of people each year. The infectious agent of TB, Mycobacterium tuberculosis (Mtb), interacts with the immune system which prevents the development of this bacterium as much as possible. In fact, the receptors on the surface of immune cells identify the bacteria, one of which is Toll-like receptors (TLRs). Different TLRs including 2, 4, 9 and 8 play critical roles in tuberculosis infection. In this paper, we focused on the role of TLRs which interact with different components of Mtb and, consequently, prevent the entrance and influence of bacteria on the body.
Collapse
Affiliation(s)
- Majid Faridgohar
- a Molecular Biology Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Hassan Nikoueinejad
- b Nephrology and Urology Research Center , Baqiyatallah University of Medical Sciences , Tehran , Iran
| |
Collapse
|
31
|
Novel vaccine potential of Rv3131, a DosR regulon-encoded putative nitroreductase, against hyper-virulent Mycobacterium tuberculosis strain K. Sci Rep 2017; 7:44151. [PMID: 28272457 PMCID: PMC5341159 DOI: 10.1038/srep44151] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 02/03/2017] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence indicates that latency-associated Mycobacterium tuberculosis (Mtb)-specific antigens from the dormancy survival regulator regulon (DosR) may be promising novel vaccine target antigens for the development of an improved tuberculosis vaccine. After transcriptional profiling of DosR-related genes in the hyper-virulent Beijing Mtb strain K and the reference Mtb strain H37Rv, we selected Rv3131, a hypothetical nitroreductase, as a vaccine antigen and evaluated its vaccine efficacy against Mtb K. Mtb K exhibited stable and constitutive up-regulation of rv3131 relative to Mtb H37Rv under three different growth conditions (at least 2-fold induction) including exponential growth in normal culture conditions, hypoxia, and inside macrophages. Mice immunised with Rv3131 formulated in GLA-SE, a well-defined TLR4 adjuvant, displayed enhanced Rv3131-specific IFN-γ and serum IgG2c responses along with effector/memory T cell expansion and remarkable generation of Rv3131-specific multifunctional CD4+ T cells co-producing TNF-α, IFN-γ and IL-2 in both spleen and lung. Following challenge with Mtb K, the Rv3131/GLA-SE-immunised group exhibited a significant reduction in bacterial number and less extensive lung inflammation accompanied by the obvious persistence of Rv3131-specific multifunctional CD4+ T cells. These results suggest that Rv3131 could be an excellent candidate for potential use in a multi-antigenic Mtb subunit vaccine, especially against Mtb Beijing strains.
Collapse
|
32
|
Jeon D, Jeong MC, Jnawali HN, Kwak C, Ryoo S, Jung ID, Kim Y. Phloretin Exerts Anti-Tuberculosis Activity and Suppresses Lung Inflammation. Molecules 2017; 22:molecules22010183. [PMID: 28117761 PMCID: PMC6155841 DOI: 10.3390/molecules22010183] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/17/2017] [Accepted: 01/18/2017] [Indexed: 12/12/2022] Open
Abstract
An increase in the prevalence of the drug-resistant Mycobacteria tuberculosis necessitates developing new types of anti-tuberculosis drugs. Here, we found that phloretin, a naturally-occurring flavonoid, has anti-mycobacterial effects on H37Rv, multi-drug-, and extensively drug-resistant clinical isolates, with minimum inhibitory concentrations of 182 and 364 μM, respectively. Since Mycobacteria cause lung inflammation that contributes to tuberculosis pathogenesis, anti-inflammatory effects of phloretin in interferon-γ-stimulated MRC-5 human lung fibroblasts and lipopolysaccharide (LPS)-stimulated dendritic cells were investigated. The release of interleukin (IL)-1β, IL-12, and tumor necrosis factor (TNF)-α was inhibited by phloretin. The mRNA levels of IL-1β, IL-6, IL-12, TNF-α, and matrix metalloproteinase-1, as well as p38 mitogen-activated protein kinase and extracellular signal-regulated kinase phosphorylation, were suppressed. A mouse in vivo study of LPS-stimulated lung inflammation showed that phloretin effectively suppressed the levels of TNF-α, IL-1β, and IL-6 in lung tissue with low cytotoxicity. Phloretin was found to bind M. tuberculosis β-ketoacyl acyl carrier protein synthase III (mtKASIII) with high affinity (7.221 × 107 M−1); a binding model showed hydrogen bonding of A-ring 2′-hydroxy and B-ring 4-hydroxy groups of phloretin with Asn261 and Cys122 of mtKASIII, implying that mtKASIII can be a potential target protein. Therefore, phloretin can be a useful dietary natural product with anti-tuberculosis benefits.
Collapse
Affiliation(s)
- Dasom Jeon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Min-Cheol Jeong
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Hum Nath Jnawali
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Chulhee Kwak
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Sungwon Ryoo
- Korean National Tuberculosis Association, Seoul 06763, Korea.
| | - In Duk Jung
- Department of Immunology, School of Medicine, Konkuk University, Seoul 05029, Korea.
| | - Yangmee Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|