1
|
Guallar-Garrido S, Soldati T. Exploring host-pathogen interactions in the Dictyostelium discoideum-Mycobacterium marinum infection model of tuberculosis. Dis Model Mech 2024; 17:dmm050698. [PMID: 39037280 PMCID: PMC11552500 DOI: 10.1242/dmm.050698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Mycobacterium tuberculosis is a pathogenic mycobacterium that causes tuberculosis. Tuberculosis is a significant global health concern that poses numerous clinical challenges, particularly in terms of finding effective treatments for patients. Throughout evolution, host immune cells have developed cell-autonomous defence strategies to restrain and eliminate mycobacteria. Concurrently, mycobacteria have evolved an array of virulence factors to counteract these host defences, resulting in a dynamic interaction between host and pathogen. Here, we review recent findings, including those arising from the use of the amoeba Dictyostelium discoideum as a model to investigate key mycobacterial infection pathways. D. discoideum serves as a scalable and genetically tractable model for human phagocytes, providing valuable insights into the intricate mechanisms of host-pathogen interactions. We also highlight certain similarities between M. tuberculosis and Mycobacterium marinum, and the use of M. marinum to more safely investigate mycobacteria in D. discoideum.
Collapse
Affiliation(s)
- Sandra Guallar-Garrido
- Department of Biochemistry, Faculty of Science, University of Geneva, 30 quai Ernest-Ansermet, Science II, 1211 Geneva-4, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, 30 quai Ernest-Ansermet, Science II, 1211 Geneva-4, Switzerland
| |
Collapse
|
2
|
Khairullah AR, Moses IB, Kusala MKJ, Tyasningsih W, Ayuti SR, Rantam FA, Fauziah I, Silaen OSM, Puspitasari Y, Aryaloka S, Raharjo HM, Hasib A, Yanestria SM, Nurhidayah N. Unveiling insights into bovine tuberculosis: A comprehensive review. Open Vet J 2024; 14:1330-1344. [PMID: 39055751 PMCID: PMC11268907 DOI: 10.5455/ovj.2024.v14.i6.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/17/2024] [Indexed: 07/27/2024] Open
Abstract
The frequent zoonotic disease known as "bovine tuberculosis" is brought on by the Mycobacterium bovis bacteria, which can infect both people and animals. The aim of this review article is to provide an explanation of the etiology, history, epidemiology, pathogenesis, clinical symptoms, diagnosis, transmission, risk factors, public health importance, economic impact, treatment, and control of bovine tuberculosis. Primarily, bovine tuberculosis affects cattle, but other animals may also be affected. Bovine tuberculosis is present throughout the world, with the exception of Antarctica. Cattle that contract bovine tuberculosis might suffer from a persistent, crippling illness. In the early stages of the disease, there are no symptoms. The tuberculin test is the primary method for detecting bovine tuberculosis in cows. Depending on its localized site in the infected animal, M. bovis can be found in respiratory secretions, milk, urine, feces, vaginal secretions, semen, feces, and exudates from lesions (such as lymph node drainage and some skin lesions). This illness generally lowers cattle productivity and could have a negative financial impact on the livestock business, particularly the dairy industry. The most effective first-line anti-tuberculosis chemotherapy consists of isoniazid, ethambutol, rifampin, and streptomycin. Second-line drugs used against bovine tuberculosis include ethionamide, capreomycin, thioacetazone, and cycloserine. To successfully control and eradicate bovine tuberculosis, developed nations have implemented routine testing and culling of infected animals under national mandatory programs.
Collapse
Affiliation(s)
- Aswin Rafif Khairullah
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Ikechukwu Benjamin Moses
- Department of Applied Microbiology, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Wiwiek Tyasningsih
- Division of Veterinary Microbiology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Siti Rani Ayuti
- Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Fedik Abdul Rantam
- Division of Veterinary Microbiology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ima Fauziah
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Otto Sahat Martua Silaen
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Yulianna Puspitasari
- Division of Veterinary Microbiology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Suhita Aryaloka
- Master Program of Veterinary Agribusiness, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Hartanto Mulyo Raharjo
- Division of Veterinary Microbiology, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Abdullah Hasib
- School of Agriculture and Food Sustainability, The University of Queensland, Gatton, Australia
| | | | - Nanis Nurhidayah
- Research Center for Veterinary Science, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| |
Collapse
|
3
|
Borborema MEDA, Miranda DEDO, de Lucena TMC, de Lorena VMB, Rabello MCDS, de Azevêdo Silva J. Steroid immune responsive gene regulation in Mycobacterium tuberculosis infection in vitro. Tuberculosis (Edinb) 2024; 146:102497. [PMID: 38408402 DOI: 10.1016/j.tube.2024.102497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/06/2024] [Accepted: 02/19/2024] [Indexed: 02/28/2024]
Abstract
Tuberculosis (TB) is an infectious disease displaying a multifactorial pathology. The immunomodulatory role attributed to steroid hormones, such as vitamin D3 (VD3) and 17β-estradiol (E2), highlighted the importance of these hormones against Mycobacterium tuberculosis (Mtb) infection. In order to understand their influence upon gene expression of immune and inflammatory responsive genes against Mtb we tested it in vitro using peripheral blood mononuclear cells (PBMCs). Cells were pretreated with VD3 (50 ng/mL) or E2 (100 nM/mL) and co-cultured with H37Rv Mtb or stimulated with lipopolysaccharide from Escherichia coli (LPS). After 24 h and 72 h of co-culture the Mtb viability in macrophages test was performed, as well the total RNA isolation for gene expression analysis by RT-qPCR of the following target genes: NLRP3, DC-SIGN, IL-1β, and IL-10. We also measured IL-10, TNF, IFN-γ, IL-4, IL-6, and IL-2 supernatant levels. As the main results, we found that VD3 and E2 downregulated the expression of inflammatory genes NLRP3, IL-1β, and IL-10 expression in Mtb co-cultured cells. Finally, VD3 treatment increased the release of the cytokine IFN-γ in Mtb-infected cells, while E2 treatment inhibited the release of IL-10, TNF, IFN-γ, and IL-6. Therefore, we report an immunogenetic influence of VD3 and E2 upon Mtb co-culture.
Collapse
Affiliation(s)
- Maria Eduarda de Albuquerque Borborema
- Laboratory of Human Genetics and Molecular Biology, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Débora Elienai de Oliveira Miranda
- Laboratory of Human Genetics and Molecular Biology, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Thays Maria Costa de Lucena
- Laboratory of Human Genetics and Molecular Biology, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | | | - Jaqueline de Azevêdo Silva
- Laboratory of Human Genetics and Molecular Biology, Genetics Department, Federal University of Pernambuco, Recife, Pernambuco, Brazil; Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| |
Collapse
|
4
|
Tamene W, Wassie L, Marconi VC, Abebe M, Kebede A, Sack U, Howe R. Protein Expression of TLR2, TLR4, and TLR9 on Monocytes in TB, HIV, and TB/HIV. J Immunol Res 2024; 2024:9399524. [PMID: 38660059 PMCID: PMC11042910 DOI: 10.1155/2024/9399524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/08/2024] [Accepted: 04/06/2024] [Indexed: 04/26/2024] Open
Abstract
Toll-like receptors (TLRs) have a critical role in recognizing pathogenic patterns and initiating immune responses against TB and HIV. Previously, studies described the gene expression of TLRs in patients with TB and HIV. Here, we demonstrated TLRs protein expressions and their association with clinical status and plasma markers in TB, HIV, and TB/HIV coinfection. The phenotyping of TLR2, TLR4, and TLR9 on CD14+ monocytes and their subsets were determined by multicolor flow cytometry. Host plasma biomarkers and microbial indices were measured using Luminex Multiplex assay and standard of care tools, respectively. TLR2 expression significantly enhanced in TB, slightly increased in HIV but slightly reduced in TB/HIV coinfection compared to apparently health controls (HC). On the other hand, TLR4 expression was significantly increased in TB, HIV, and TB/HIV compared to HC. Expression of TLR4 was equally enhanced on classical and intermediate monocytes while higher TLR2 expression on intermediate than classical monocytes. TLR4 had a positive correlation pattern with plasma biomarkers while TLR2 had an inverse correlation pattern. TLR4 is associated with disease severity while TLR2 is with the immune-competent status of patients. Our findings demonstrated that the pattern of TLR expression is disease as well as monocyte subset specific and distinct factors drive these differences.
Collapse
Affiliation(s)
- Wegene Tamene
- HIV and TB Research Directorate, Ethiopian Public Health Institute (EPHI), Addis Ababa, Ethiopia
- Mycobacterial Disease Research Directorate, Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Liya Wassie
- Mycobacterial Disease Research Directorate, Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Vincent C. Marconi
- School of Medicine, Rollins School of Public Health and the Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Meseret Abebe
- Mycobacterial Disease Research Directorate, Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| | - Amha Kebede
- HIV and TB Research Directorate, Ethiopian Public Health Institute (EPHI), Addis Ababa, Ethiopia
| | - Ulrich Sack
- Institute of Clinical Immunology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Rawleigh Howe
- Mycobacterial Disease Research Directorate, Armauer Hansen Research Institute (AHRI), Addis Ababa, Ethiopia
| |
Collapse
|
5
|
Borborema MEA, da Silva Santos AF, de Lucena TMC, Crovella S, da Silva Rabello MC, de Azevêdo Silva J. Pathogen recognition pathway gene variants and inflammasome sensors gene expression in tuberculosis patients under treatment. Mol Biol Rep 2024; 51:161. [PMID: 38252221 DOI: 10.1007/s11033-023-09155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Several epidemiological studies have suggested that genetic variations in encoding pattern recognition receptors (PRRs) genes such as Toll Like Receptors (TLRs) and their signaling products, may influence the susceptibility, severity and outcome of tuberculosis (TB). After sensing a pathogen, the cell responds producing an inflammatory response, to restrain the pathogen's successful course of infection. Herein we assessed single nucleotide polymorphisms (SNP) and gene expression from pathogen recognition and inflammasome pathways in Brazilian TB patients. METHODS AND RESULTS For genetic association analysis we included MYD88 and TLR4, PRRs sensing proteins. Allele distribution for MYD88 rs6853 (A > G) and TLR4 rs7873784 (C > G) presented conserved among the tested samples with statistically differential distribution in TB patients versus controls. However, when testing according to sample ethnicity (African or Caucasian-derived individuals) we identified that the rs6853 G/G genotype was associated with a lower susceptibility to TB in Caucasian population. Meanwhile, the rs7873784 G/G genotype was associated with a higher TB susceptibility in Afro-descendant ethnicity individuals. We also aimed to verify MYD88 and the inflammasome genes NLRP1 and NLRC4 expression in order to connect to active TB and/or clinical aspects. CONCLUSIONS We identified that inflammasome gene expression in TB patients under treatment display a similar pattern as in healthy controls, indicating that TB treatment impairs NLRP1 inflammasome activation.
Collapse
Affiliation(s)
- Maria Eduarda Albuquerque Borborema
- Laboratory of Human Genetics and Molecular Biology, Department of Genetics, Center for Biosciences, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Recife, PE, 50670-901 - CEP, Brazil
- Keizo Asami Institute, Federal University of Pernambuco, Recife, PE, 50740-465 - CEP, Brazil
| | - Ariane Fernandes da Silva Santos
- Laboratory of Human Genetics and Molecular Biology, Department of Genetics, Center for Biosciences, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Recife, PE, 50670-901 - CEP, Brazil
- Keizo Asami Institute, Federal University of Pernambuco, Recife, PE, 50740-465 - CEP, Brazil
| | - Thays Maria Costa de Lucena
- Laboratory of Human Genetics and Molecular Biology, Department of Genetics, Center for Biosciences, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Recife, PE, 50670-901 - CEP, Brazil
- Keizo Asami Institute, Federal University of Pernambuco, Recife, PE, 50740-465 - CEP, Brazil
| | - Sergio Crovella
- Keizo Asami Institute, Federal University of Pernambuco, Recife, PE, 50740-465 - CEP, Brazil
| | | | - Jaqueline de Azevêdo Silva
- Laboratory of Human Genetics and Molecular Biology, Department of Genetics, Center for Biosciences, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Recife, PE, 50670-901 - CEP, Brazil.
- Keizo Asami Institute, Federal University of Pernambuco, Recife, PE, 50740-465 - CEP, Brazil.
| |
Collapse
|
6
|
Krishnan V, Nath S, Nair P, Das B. Mycobacterium tuberculosis and its clever approaches to escape the deadly macrophage. World J Microbiol Biotechnol 2023; 39:300. [PMID: 37667129 DOI: 10.1007/s11274-023-03735-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/19/2023] [Indexed: 09/06/2023]
Abstract
Mycobacterium tuberculosis (Mt.b), a deadly disease causer, is a facultative parasite. This microorganism has developed several methods to defend itself, once internalized within specialised vacuoles in the macrophages. A wide array of receptors like the complement receptor mannose receptors, scavenger receptor assists the entry of the microbe within the phagocytic macrophages. However, Mt.b is clever enough to protect itself from the hostile environment of the macrophage thereby prevailing within it. The microbe can efficiently inhibit processes like phagosome-lysosome fusion, acidification of phagosomes, release of proinflammatory cytokines and stop crucial events like apoptosis. Additionally, it also adopts resistance to killing by reactive oxygen intermediates and reactive nitrogen intermediates. There are multiple genes both in host and the pathogen which are involved in this successful survival of Mt.b. The regulation of phagolysosome fusion is mediated by proteins such as Coronin, TlyA, SapM, PnkG, EsxH. The microbe has certain mechanisms to even acquire iron from the host cell, to withstand iron deprivation as a mode of host's defence mechanism. This review focuses on the various defensive adaptations acquired by Mt.b for fighting against the deprived conditions existing within the macrophages and their capability of proliferating successfully within it, thereby resulting in a diseased condition.
Collapse
Affiliation(s)
- Vinaya Krishnan
- Department of Biotechnology, Mount Carmel College Autonomous, Bengaluru, 560052, India
| | | | - Preetha Nair
- Department of Biotechnology, Mount Carmel College Autonomous, Bengaluru, 560052, India
| | - Bannhi Das
- Department of Biotechnology, Mount Carmel College Autonomous, Bengaluru, 560052, India.
| |
Collapse
|
7
|
Xu J, Qin C, Xie J, Wang J, He Y, Tan J, Shi X. Transcriptome analysis of Chinese sucker (Myxocyprinus asiaticus) head kidney and discovery of key immune-related genes to cold stress after swimming fatigue. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 47:101104. [PMID: 37390763 DOI: 10.1016/j.cbd.2023.101104] [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: 03/02/2023] [Revised: 06/11/2023] [Accepted: 06/13/2023] [Indexed: 07/02/2023]
Abstract
For Chinese sucker (Myxocyprinus asiaticus), passing through a dam with fast flow and cold water are always unavoidable, and this process can cause stress, disease or even death. In this study, comparative transcriptome analysis was conducted to investigate the potential immune mechanism in head kidney of M. asiaticus with swimming fatigue stress and cold stress after fatigue. In general, a total of 181,781 unigenes were generated, and 38,545 differentially expressed genes (DEGs) were identified. In these DEGs, 22,593, 7286 and 8666 DEGs were identified among groups of fatigue vs. cold, control vs. cold, and control vs. fatigue, respectively. Enrichment analysis revealed these DEGs were involved in coagulation cascades and complement, natural killer cell mediated cytotoxicity, antigen processing and presentation, Toll-like receptor signaling pathways, and chemokine signaling pathway. Notably, immune genes including heat shock protein 4a (HSP4a), HSP70 and HSP90α genes were significantly up-regulated in fishes with cold stress after fatigue. Differently, more immune genes in control vs. cold compared with that in control vs. fatigue were significantly down-regulated expression, such as claudin-15-like, Toll-like receptor 13, antimicrobial peptide (hepcidin), immunoglobulin, CXCR4 chemokine receptor, T-cell receptor, complement factor B/C2-A3, and interleukin 8. In this study, the number of DEGs in the head kidney was less than that our previous study in the spleen, which we speculated was more sensitive to changes in water temperature than the head kidney. In summary, lots of immune-related genes in the head kidney were down-regulated under cold stress after fatigue, suggesting that M. asiaticus might have experienced severe immunosuppression in the process of passing through the dam.
Collapse
Affiliation(s)
- Jing Xu
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang 641000, China
| | - Chuanjie Qin
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang 641000, China.
| | - Jiang Xie
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang 641000, China
| | - Jun Wang
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang 641000, China
| | - Yang He
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang 641000, China
| | - Junjun Tan
- Hubei International Science and Technology Cooperation Base of Fish Passage, China Three Gorges University, Yichang 443002, China
| | - Xiaotao Shi
- Hubei International Science and Technology Cooperation Base of Fish Passage, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
8
|
Kottom TJ, Carmona EM, Schaefbauer K, Limper AH. CLEC4A and CLEC12B C-type lectin receptors mediate interactions with Pneumocystis cell wall components. J Med Microbiol 2023; 72. [PMID: 37294293 DOI: 10.1099/jmm.0.001714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
Introduction. C-type lectin receptors (CLRs) are prominently expressed on myeloid cells where they perform multiple functions including serving as pattern recognition receptors (PRRs) to drive innate as well as adaptive immunity to pathogens. Depending on the presence of a tyrosine-based signalling motif, CLR-microbial pathogen engagement may result in either anti- or pro-inflammatory signalling.Impact statement. In this manuscript, we report our laboratory study of two novel CLRs that recognize Pneumocystis murina cell wall homogenates (CWH) and a purified Pneumocystis carinii cell wall fraction (CWF).Aim. To study the potential of newly generated hFc-CLR fusions on binding to Pneumocystis murina CWHs and P. carinii CWFs and subsequent downstream inflammatory signalling analysis.Methods. Newly generated hFc-CLR fusion CLEC4A and CLEC12B were screened against P. murina CWHs and P. carinii CWFs preparations via modified ELISA. Immunofluorescence assay (IFA) was utilized to visualize hFc-CLR fusion binding against intact fixed fungal life forms to verify results. Quantitative PCR (q-PCR) analysis of lung mRNA from the mouse immunosuppressed Pneumocystis pneumonia (PCP) model versus uninfected mice was employed to detect possible changes in the respective Clec4a and Clec12b transcripts. Lastly, siRNA technology of both CLRs was conducted to determine effects on downstream inflammatory events in mouse macrophages stimulated in the presence of P. carinii CWFs.Results. We determined that both CLEC4A and CLEC12B hFc-CLRs displayed significant binding with P. murina CWHs and P. carinii CWFs. Binding events showed significant binding to both curdlan and laminarin, both polysaccharides containing β-(1,3) glucans as well as N-acetylglucosamine (GlcNAc) residues and modest yet non-significant binding to the negative control carbohydrate dextran. IFA with both CLR hFc-fusions against whole P. murina life forms corroborated these findings. Lastly, we surveyed the mRNA expression profiles of both CLRs tested above in the mouse immunosuppressed Pneumocystis pneumonia (PCP) model and determined that both CLRs were significantly up regulated during infection. Lastly, siRNA of both CLRs in the mouse RAW macrophage cell line was conducted and results demonstrated that silencing of Clec4a resulted in no significant changes in TNF-alpha generation in P. carinii CWF stimulated macrophages. On the contrary, silencing of Clec12b CLR resulted in significant decreases in TNF-alpha in RAW cells stimulated with the same CWF.Conclusion. The data presented here provide new members of the CLRs family recognizing Pneumocystis. Future studies using CLEC4A and/or CLEC12B deficient mice in the PCP mouse model should provide further insights into the host immunological response to Pneumocystis.
Collapse
Affiliation(s)
- Theodore J Kottom
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, Minnesota, 55905, USA
| | - Eva M Carmona
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, Minnesota, 55905, USA
| | - Kyle Schaefbauer
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, Minnesota, 55905, USA
| | - Andrew H Limper
- Thoracic Diseases Research Unit, Departments of Medicine and Biochemistry, Mayo Clinic College of Medicine, Rochester, Minnesota, 55905, USA
| |
Collapse
|
9
|
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
|
10
|
Karakaş Ö, Dogan İ, Güven SC, Erel Ş, Erten S. Evaluation of Tuberculosis Risk under Interleukin-1 Inhibitor Agents
in Patients with Autoinflammatory Diseases: Experience from a Region with
Moderate Tuberculosis Prevalence. AKTUEL RHEUMATOL 2023. [DOI: 10.1055/a-2015-2683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Abstract
Background We aimed to investigate the rate of TBC reactivation in
patients with autoinflammatory conditions who had been followed up under
treatment with IL-1 antagonists in our clinic. We further aimed to evaluate
whether latent TBC screening and isoniazid prophylaxis before IL-1 inhibitor
treatment is beneficial in the prevention of reactivation.
Patients and Methods This study was designed as a cross-sectional and
observational study. It received the approval of the local ethics committee and
was therefore performed in accordance with the ethical standards laid down in
the 1964 Declaration of Helsinki and its later amendments. Adult patients who
had been followed up in our clinic under treatment with IL-1 antagonists were
evaluated with regard to whether they had a TBC infection before and after these
treatments and whether they had been screened for latent TBC infection before
IL-1 antagonist treatment and received prophylaxis. Clinical and laboratory data
were obtained from hospital records and via telephone interviews after the
patients had given their verbal consent to participate.
Results Fifty-nine patients under IL-1 antagonists were consecutively
enrolled. 58 were under follow-up with the diagnosis of familial Mediterranean
fever and one patient with a diagnosis of adult-onset Still’s disease.
35 patients (59.3%) were treated with anakinra and 24 (40.7%)
were treated with canakinumab. 12 (50%) of the canakinumab users had
previously received anakinra treatment and were switched to canakinumab due to
side-effects or ineffectiveness. The number of patients who underwent latent
tuberculosisc infection (LTBI) screening before treatment was 37
(62.7%); the number of patients who received prophylaxis was 13
(22%). None of the patients receiving IL-1 antagonists developed active
tuberculosis.
Conclusion Blocking the IL-1 pathway may be safe regarding the activation
of LTBI or development of a new tuberculosis infection even in locations with a
high geographical risk.
Collapse
Affiliation(s)
| | - İsmail Dogan
- Rheumatology, Ankara Yildirim Beyazit University Faculty of Medicine,
Ankara, Turkey
| | | | - Şule Erel
- İnternal medicine, Ankara City Hospital, ankara,
Turkey
| | - Sukran Erten
- Rheumatology, Yildirim Beyazit University Faculty of Medicine, Ankara,
Turkey
| |
Collapse
|
11
|
Borborema MEDA, de Lucena TMC, Silva JDA. Vitamin D and estrogen steroid hormones and their immunogenetic roles in Infectious respiratory (TB and COVID-19) diseases. Genet Mol Biol 2023; 46:e20220158. [PMID: 36745756 PMCID: PMC9901533 DOI: 10.1590/1415-4757-gmb-2022-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/07/2022] [Indexed: 02/08/2023] Open
Abstract
The role of steroid hormones against infectious diseases has been extensively studied. From immunomodulatory action to direct inhibition of microorganism growth, hormones D3 (VD3) and 17β-estradiol (E2), and the genetic pathways modulated by them, are key targets for a better understanding pathogenesis of infectious respiratory diseases (IRD) such as tuberculosis (TB) and the coronavirus disease-19 (COVID-19). Currently, the world faces two major public health problems, the outbreak of COVID-19, accounting for more than 6 million so far, and TB, more than 1 million deaths per year. Both, although resulting from different pathogens, the Mtb and the SARS-CoV-2, respectively, are considered serious and epidemic. TB and COVID-19 present similar infection rates between men and women, however the number of complications and deaths resulting from the two infections is higher in men when compared to women in childbearing age, which may indicate a role of the sex hormone E2 in the context of these diseases. E2 and VD3 act upon key gene pathways as important immunomodulatory players and supporting molecules in IRDs. This review summarizes the main roles of these hormones (VD3 and E2) in modulating immune and inflammatory responses and their relationship with TB and COVID-19.
Collapse
Affiliation(s)
- Maria Eduarda de Albuquerque Borborema
- Universidade Federal de Pernambuco, Departamento de Genética, Laboratório de Genética e Biologia Molecular Humana (LGBMH), Recife, PE, Brazil
- Universidade Federal de Pernambuco, Laboratório de Imunopatologia Keizo Asami (LIKA), Recife, PE, Brazil
| | - Thays Maria Costa de Lucena
- Universidade Federal de Pernambuco, Departamento de Genética, Laboratório de Genética e Biologia Molecular Humana (LGBMH), Recife, PE, Brazil
- Universidade Federal de Pernambuco, Laboratório de Imunopatologia Keizo Asami (LIKA), Recife, PE, Brazil
| | - Jaqueline de Azevêdo Silva
- Universidade Federal de Pernambuco, Departamento de Genética, Laboratório de Genética e Biologia Molecular Humana (LGBMH), Recife, PE, Brazil
- Universidade Federal de Pernambuco, Laboratório de Imunopatologia Keizo Asami (LIKA), Recife, PE, Brazil
| |
Collapse
|
12
|
Ifijen IH, Atoe B, Ekun RO, Ighodaro A, Odiachi IJ. Treatments of Mycobacterium tuberculosis and Toxoplasma gondii with Selenium Nanoparticles. BIONANOSCIENCE 2023; 13:249-277. [PMID: 36687337 PMCID: PMC9838309 DOI: 10.1007/s12668-023-01059-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2023] [Indexed: 01/13/2023]
Abstract
Toxoplasma gondii and Mycobacterium tuberculosis are pathogens that are harmful to humans. When these diseases interact in humans, the result is typically fatal to the public health. Several investigations on the relationship between M. tuberculosis and T. gondii infections have found that there is a strong correlation between them with each infection having a reciprocal effect on the other. TB may contribute to the reactivation of innate toxoplasmosis or enhance susceptibility to a new infection, and toxoplasma co-infection may worsen the severity of pulmonary tuberculosis. As a consequence, there is an earnest and urgent necessity to generate novel therapeutics that can subdue these challenges. Selenium nanostructures' compelling properties have been shown to be a successful treatment for Mycobacterium TB and Toxoplasma gondii. Despite the fact that selenium (Se) offers many health advantages for people, it also has a narrow therapeutic window; therefore, consuming too much of either inorganic or organic compounds based on selenium can be hazardous. Compared to both inorganic and organic Se, Se nanoparticles (SeNPs) are less hazardous. They are biocompatible and excellent in selectively targeting specific cells. As a consequence, this review conducted a summary of the efficacy of biogenic Se NPs in the treatment of tuberculosis (TB) and toxoplasmosis. Mycobacterium tuberculosis, Toxoplasma gondii, and their co-infection were all briefly described.
Collapse
Affiliation(s)
- Ikhazuagbe H. Ifijen
- Department of Research Outreach, Rubber Research Institute of Nigeria, Iyanomo, P.M.B, 1049, Benin City, Nigeria
| | - Best Atoe
- Department of Daily Need, Worldwide Healthcare, 100, Textile Mill Road, Benin City, Edo State Nigeria
| | - Raphael O. Ekun
- grid.440833.80000 0004 0642 9705Department of Electrical Electronics, Cyprus International University, Haspolat, Lefkosa, North Cyprus Mersin 10 Turkey
| | - Augustine Ighodaro
- Depatment of Aseptic Quality, Quantum Pharmaceuticals, Quantum House, Durham, UK
| | - Ifeanyi J. Odiachi
- grid.461933.a0000 0004 0446 5040Department of Science Laboratory Technology, Delta State Polytechnic Ogwashi-Uku, Ogwashi-Uku, Nigeria
| |
Collapse
|
13
|
Simper JD, Perez E, Schlesinger LS, Azad AK. Resistance and Susceptibility Immune Factors at Play during Mycobacterium tuberculosis Infection of Macrophages. Pathogens 2022; 11:pathogens11101153. [PMID: 36297211 PMCID: PMC9611686 DOI: 10.3390/pathogens11101153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/28/2022] Open
Abstract
Tuberculosis (TB), caused by infection with Mycobacterium tuberculosis (M.tb), is responsible for >1.5 million deaths worldwide annually. Innate immune cells, especially macrophages, are the first to encounter M.tb, and their response dictates the course of infection. During infection, macrophages exert a variety of immune factors involved in either controlling or promoting the growth of M.tb. Research on this topic has been performed in both in vitro and in vivo animal models with discrepant results in some cases based on the model of study. Herein, we review macrophage resistance and susceptibility immune factors, focusing primarily on recent advances in the field. We include macrophage cellular pathways, bioeffector proteins and molecules, cytokines and chemokines, associated microbiological factors and bacterial strains, and host genetic factors in innate immune genes. Recent advances in mechanisms underlying macrophage resistance and susceptibility factors will aid in the successful development of host-directed therapeutics, a topic emphasized throughout this review.
Collapse
Affiliation(s)
- Jan D. Simper
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Department of Microbiology, Immunology and Molecular Genetics, UT Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Esteban Perez
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Translational Sciences Program, UT Health San Antonio Graduate School of Biomedical Sciences, San Antonio, TX 78229, USA
| | - Larry S. Schlesinger
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Correspondence: (L.S.S.); (A.K.A.); Tel.: +1-210-258-9578 (L.S.S.); +1-210-258-9467 (A.K.A.)
| | - Abul K. Azad
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W. Military Drive, San Antonio, TX 78227, USA
- Correspondence: (L.S.S.); (A.K.A.); Tel.: +1-210-258-9578 (L.S.S.); +1-210-258-9467 (A.K.A.)
| |
Collapse
|
14
|
Panda S, Seelan DM, Faisal S, Arora A, Luthra K, Palanichamy JK, Mohan A, Vikram NK, Gupta NK, Ramakrishnan L, Singh A. Chronic hyperglycemia drives alterations in macrophage effector function in pulmonary tuberculosis. Eur J Immunol 2022; 52:1595-1609. [PMID: 36066992 DOI: 10.1002/eji.202249839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/05/2022] [Accepted: 09/05/2022] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus (DM) alters immune responses and given the rising prevalence of DM in tuberculosis (TB) endemic countries; hyperglycemia can be a potential risk factor for active TB development. However, the impact of hyperglycemia on TB-specific innate immune response in terms of macrophage functions remains poorly addressed. We assessed macrophage effector functions in uncontrolled DM patients with or without TB infection (PTB+DM and DM), non-diabetic TB patients (PTB), and non-diabetic-uninfected controls. Phagocytic capacity against BCG and surface expression of different pattern recognition receptors (PRRs) (CD11b, CD14, CD206, MARCO, and TLR-2) were measured via flow cytometry. Effector molecules (ROS and NO) required for bacterial killing were assessed via DCFDA and Griess reaction respectively. A systematic dysregulation in phagocytic capacity with concurrent alterations in the expression pattern of key PRRs (CD11b, MARCO, and CD206) was observed in PTB+DM. These altered PRR expressions were associated with decreased phagocytic capacity of macrophages. Similarly, ROS was aberrantly higher while NO was lower in PTB+DM. These altered macrophage functions were positively correlated with increasing disease severity. Our results highlight several key patterns of immune dysregulation against TB infection under hyperglycemic conditions and highlight a negative impact of hyperglycemia with etiology and progression of TB.
Collapse
Affiliation(s)
- Sudhasini Panda
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Diravya M Seelan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Shah Faisal
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Alisha Arora
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | | | - Anant Mohan
- Department of Pulmonary Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Naval K Vikram
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neeraj Kumar Gupta
- Department of Pulmonary Medicine, VMMC and Safdarjung Hospital, New Delhi, 110029, India
| | - Lakshmy Ramakrishnan
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
15
|
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: 0] [Impact Index Per Article: 0] [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
|
16
|
Wang Y, Shi Q, Chen Q, Zhou X, Yuan H, Jia X, Liu S, Li Q, Ge L. Emerging advances in identifying signal transmission molecules involved in the interaction between Mycobacterium tuberculosis and the host. Front Cell Infect Microbiol 2022; 12:956311. [PMID: 35959378 PMCID: PMC9359464 DOI: 10.3389/fcimb.2022.956311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/30/2022] [Indexed: 11/21/2022] Open
Abstract
Tuberculosis caused by Mycobacterium tuberculosis (MTB) is an ancient chronic infectious disease and is still the leading cause of death worldwide due to a single infectious disease. MTB can achieve immune escape by interacting with host cells through its special cell structure and secreting a variety of effector proteins. Innate immunity-related pattern recognition receptors (PPR receptors) play a key role in the regulation of signaling pathways. In this review, we focus on the latest research progress on related signal transduction molecules in the interaction between MTB and the host. In addition, we provide new research ideas for the development of new anti-tuberculosis drug targets and lead compounds and provide an overview of information useful for approaching future tuberculosis host-oriented treatment research approaches and strategies, which has crucial scientific guiding significance and research value.
Collapse
Affiliation(s)
- Yue Wang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiyuan Shi
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Qi Chen
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuebin Zhou
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Huiling Yuan
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiwen Jia
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuyuan Liu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qin Li
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Qin Li, ; Lijun Ge,
| | - Lijun Ge
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Qin Li, ; Lijun Ge,
| |
Collapse
|
17
|
Shah T, Shah Z, Yasmeen N, Baloch Z, Xia X. Pathogenesis of SARS-CoV-2 and Mycobacterium tuberculosis Coinfection. Front Immunol 2022; 13:909011. [PMID: 35784278 PMCID: PMC9246416 DOI: 10.3389/fimmu.2022.909011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/23/2022] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, is an infectious disease that poses severe threats to global public health and significant economic losses. The COVID-19 global burden is rapidly increasing, with over 246.53 million COVID-19 cases and 49.97 million deaths reported in the WHO 2021 report. People with compromised immunity, such as tuberculosis (TB) patients, are highly exposed to severe COVID-19. Both COVID-19 and TB diseases spread primarily through respiratory droplets from an infected person to a healthy person, which may cause pneumonia and cytokine storms, leading to severe respiratory disorders. The COVID-19-TB coinfection could be fatal, exacerbating the current COVID-19 pandemic apart from cellular immune deficiency, coagulation activation, myocardial infarction, and other organ dysfunction. This study aimed to assess the pathogenesis of SARS-CoV-2-Mycobacterium tuberculosis coinfections. We provide a brief overview of COVID19-TB coinfection and discuss SARS-CoV-2 host cellular receptors and pathogenesis. In addition, we discuss M. tuberculosis host cellular receptors and pathogenesis. Moreover, we highlight the impact of SARS-CoV-2 on TB patients and the pathological pathways that connect SARS-CoV-2 and M. tuberculosis infection. Further, we discuss the impact of BCG vaccination on SARS-CoV-2 cases coinfected with M. tuberculosis, as well as the diagnostic challenges associated with the coinfection.
Collapse
Affiliation(s)
- Taif Shah
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Zahir Shah
- College of Veterinary Sciences, The University of Agriculture Peshawar, Peshawar, Pakistan
| | - Nafeesa Yasmeen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zulqarnain Baloch
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
18
|
Zhang XJ, Xu HS, Li CH, Fu YR, Yi ZJ. Up-regulated SAMD9L modulated by TLR2 and HIF-1α as a promising biomarker in tuberculosis. J Cell Mol Med 2022; 26:2935-2946. [PMID: 35388602 PMCID: PMC9097843 DOI: 10.1111/jcmm.17307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 11/29/2022] Open
Abstract
The aim of this study was to identify potential biomarkers of TB in blood and determine their function in Mtb-infected macrophages. First of all, WGCNA was used to analyse 9451 genes with significant changes in TB patients' whole blood. The 220 interferon-γ-related genes were identified, and then 30 key genes were screened using Cytoscape. Then, the AUC values of key genes were calculated to further narrow the gene range. Finally, we identified 9 genes from GSE19444. ROC analysis showed that SAMD9L, among 9 genes, had a high diagnostic value (AUC = 0.925) and a differential diagnostic value (AUC>0.865). To further narrow down the range of DEGs, the top 10 hub-connecting genes were screened from monocytes (GSE19443). Finally, we obtained 4 genes (SAMD9L, GBP1, GBP5 and STAT1) by intersections of genes from monocytes and whole blood. Among them, it was found that the function of SAMD9L was unknown after data review, so this paper studied this gene. Our results showed that SAMD9L is up-regulated and suppresses cell necrosis, and might be regulated by TLR2 and HIF-1α during Mtb infection. In addition, miR-181b-5p is significantly up-regulated in the peripheral blood plasma of tuberculosis patients, which has a high diagnostic value (AUC = 0.969).
Collapse
Affiliation(s)
- Xiang-Juan Zhang
- Department of Pathogen Biology, School of Basic Medicine, Weifang Medical University, Weifang, China.,School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang, China
| | - Hai-Shan Xu
- School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang, China
| | - Chong-Hui Li
- School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang, China
| | - Yu-Rong Fu
- Department of Pathogen Biology, School of Basic Medicine, Weifang Medical University, Weifang, China.,School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang, China
| | - Zheng-Jun Yi
- School of Medical Laboratory, Key Laboratory of Clinical Laboratory Diagnostics in Universities of Shandong, Weifang Medical University, Weifang, China
| |
Collapse
|
19
|
Nguyen TK, Niaz Z, Kruzel ML, Actor JK. Recombinant Human Lactoferrin Reduces Inflammation and Increases Fluoroquinolone Penetration to Primary Granulomas During Mycobacterial Infection of C57Bl/6 Mice. Arch Immunol Ther Exp (Warsz) 2022; 70:9. [PMID: 35226195 PMCID: PMC8922470 DOI: 10.1007/s00005-022-00648-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022]
Abstract
Infection with Mycobacterium tuberculosis (Mtb) results in the primary formation of a densely packed inflammatory foci that limits entry of therapeutic agents into pulmonary sites where organisms reside. No current therapeutic regimens exist that modulate host immune responses to permit increased drug penetration to regions of pathological damage during tuberculosis disease. Lactoferrin is a natural iron-binding protein previously demonstrated to modulate inflammation and granuloma cohesiveness, while maintaining control of pathogenic burden. Studies were designed to examine recombinant human lactoferrin (rHLF) to modulate histological progression of Mtb-induced pathology in a non-necrotic model using C57Bl/6 mice. The rHLF was oral administered at times corresponding to initiation of primary granulomatous response, or during granuloma maintenance. Treatment with rHLF demonstrated significant reduction in size of primary inflammatory foci following Mtb challenge, and permitted penetration of ofloxacin fluoroquinolone therapeutic to sites of pathological disruption where activated (foamy) macrophages reside. Increased drug penetration was accompanied by retention of endothelial cell integrity. Immunohistochemistry revealed altered patterns of M1-like and M2-like phenotypic cell localization post infectious challenge, with increased presence of M2-like markers found evenly distributed throughout regions of pulmonary inflammatory foci in rHLF-treated mice.
Collapse
Affiliation(s)
- Thao K.T. Nguyen
- Department of Pathology and Laboratory Medicine, UTHealth McGovern Medical School, Houston, TX, USA,The University of Texas MD Anderson Cancer Center – UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Zainab Niaz
- Department of Pathology and Laboratory Medicine, UTHealth McGovern Medical School, Houston, TX, USA
| | - Marian L. Kruzel
- Department of Pathology and Laboratory Medicine, UTHealth McGovern Medical School, Houston, TX, USA
| | - Jeffrey K. Actor
- Department of Pathology and Laboratory Medicine, UTHealth McGovern Medical School, Houston, TX, USA
| |
Collapse
|
20
|
Imidazole-Thiosemicarbazide Derivatives as Potent Anti- Mycobacterium tuberculosis Compounds with Antibiofilm Activity. Cells 2021; 10:cells10123476. [PMID: 34943984 PMCID: PMC8700351 DOI: 10.3390/cells10123476] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an intracellular pathogenic bacterium and the causative agent of tuberculosis. This disease is one of the most ancient and deadliest bacterial infections, as it poses major health, social and economic challenges at a global level, primarily in low- and middle-income countries. The lack of an effective vaccine, the long and expensive drug therapy, and the rapid spread of drug-resistant strains of Mtb have led to the re-emergence of tuberculosis as a global pandemic. Here, we assessed the in vitro activity of new imidazole-thiosemicarbazide derivatives (ITDs) against Mtb infection and their effects on mycobacterial biofilm formation. Cytotoxicity studies of the new compounds in cell lines and human monocyte-derived macrophages (MDMs) were performed. The anti-Mtb activity of ITDs was evaluated by determining minimal inhibitory concentrations of resazurin, time-kill curves, bacterial intracellular growth and the effect on biofilm formation. Mutation frequency and whole-genome sequencing of mutants that were resistant to ITDs were performed. The antimycobacterial potential of ITDs with the ability to penetrate Mtb-infected human macrophages and significantly inhibit the intracellular growth of tubercle bacilli and suppress Mtb biofilm formation was observed.
Collapse
|
21
|
Jiao J, Zheng N, Wei W, Fleming J, Wang X, Li Z, Zhang L, Liu Y, Zhang Z, Shen A, Chuanyou L, Bi L, Zhang H. M. tuberculosis CRISPR/Cas proteins are secreted virulence factors that trigger cellular immune responses. Virulence 2021; 12:3032-3044. [PMID: 34886764 PMCID: PMC8667911 DOI: 10.1080/21505594.2021.2007621] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 09/27/2021] [Accepted: 11/10/2021] [Indexed: 11/03/2022] Open
Abstract
The role of prokaryotic CRISPR/Cas system proteins as a defensive shield against invasive nucleic acids has been studied extensively. Non-canonical roles in pathogenesis involving intracellular targeting of certain virulence-associated endogenous mRNA have also been reported for some Type I and Type II CRISPR/Cas proteins, but no such roles have yet been established for Type III system proteins. Here, we demonstrate that M. tuberculosis (Type III-A system) CRISPR/Cas proteins Csm1, Csm3, Csm5, Csm6, and Cas6 are secreted and induce host immune responses. Using cell and animal experiments, we show that Cas6, in particular, provokes IFN-γ release from PBMCs from active tuberculosis (TB) patients, and its deletion markedly attenuates virulence in a murine M. tuberculosis challenge model. Recombinant MTBCas6 induces apoptosis of macrophages and lung fibroblasts, and interacts with the surface of cells in a caspase and TLR-2 independent manner. Transcriptomic and signal pathway studies using THP-1 macrophages stimulated with MTBCas6 indicated that MTBCas6 upregulates expression of genes associated with the NF-κB pathway leading to higher levels of IL-6, IL-1β, and TNF-α release, cytokines known to activate immune system cells in response to M. tuberculosis infection. Our findings suggest that, in addition to their intracellular shielding role, M. tuberculosis CRISPR/Cas proteins have non-canonical extracellular roles, functioning like a virulent sword, and activating host immune responses.
Collapse
Affiliation(s)
- Jianjian Jiao
- Key Laboratory of RNA Biology and State Key Laboratory of Biomacromolecules, Cas Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Nan Zheng
- Key Laboratory of RNA Biology and State Key Laboratory of Biomacromolecules, Cas Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wenjing Wei
- Key Laboratory of RNA Biology and State Key Laboratory of Biomacromolecules, Cas Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Joy Fleming
- Key Laboratory of RNA Biology and State Key Laboratory of Biomacromolecules, Cas Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xingyun Wang
- Key Laboratory of RNA Biology and State Key Laboratory of Biomacromolecules, Cas Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Zihui Li
- Beijing Chest Hospital, Capital Medical University; Beijing Tuberculosis and Thoracic Tumor Research Institute; Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing, China
| | - Lili Zhang
- Key Laboratory of RNA Biology and State Key Laboratory of Biomacromolecules, Cas Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yi Liu
- Beijing Chest Hospital, Capital Medical University; Beijing Tuberculosis and Thoracic Tumor Research Institute; Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing, China
| | - Zongde Zhang
- Beijing Chest Hospital, Capital Medical University; Beijing Tuberculosis and Thoracic Tumor Research Institute; Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing, China
| | - Adong Shen
- Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Li Chuanyou
- Beijing Chest Hospital, Capital Medical University; Beijing Tuberculosis and Thoracic Tumor Research Institute; Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing, China
| | - Lijun Bi
- Key Laboratory of RNA Biology and State Key Laboratory of Biomacromolecules, Cas Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hongtai Zhang
- Key Laboratory of RNA Biology and State Key Laboratory of Biomacromolecules, Cas Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
22
|
The Role of NRF2 in Mycobacterial Infection. Antioxidants (Basel) 2021; 10:antiox10121861. [PMID: 34942964 PMCID: PMC8699052 DOI: 10.3390/antiox10121861] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/11/2021] [Accepted: 11/22/2021] [Indexed: 02/04/2023] Open
Abstract
The incidence of pulmonary nontuberculous mycobacterial (NTM) infection is increasing worldwide, and its clinical outcomes with current chemotherapies are unsatisfactory. The incidence of tuberculosis (TB) is still high in Africa, and the existence of drug-resistant tuberculosis is also an important issue for treatment. To discover and develop new efficacious anti-mycobacterial treatments, it is important to understand the host-defense mechanisms against mycobacterial infection. Nuclear erythroid 2 p45-related factor-2 (NRF2) is known to be a major regulator of various antioxidant response element (ARE)-driven cytoprotective gene expressions, and its protective role has been demonstrated in infections. However, there are not many papers or reviews regarding the role of NRF2 in mycobacterial infectious disease. Therefore, this review focuses on the role of NRF2 in the pathogenesis of Mycobacterium tuberculosis and Mycobacterium avium infection.
Collapse
|
23
|
Behura A, Das M, Kumar A, Naik L, Mishra A, Manna D, Patel S, Mishra A, Singh R, Dhiman R. ESAT-6 impedes IL-18 mediated phagosome lysosome fusion via microRNA-30a upon Calcimycin treatment in mycobacteria infected macrophages. Int Immunopharmacol 2021; 101:108319. [PMID: 34740079 DOI: 10.1016/j.intimp.2021.108319] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/05/2021] [Accepted: 10/25/2021] [Indexed: 01/18/2023]
Abstract
The weaponry possessed by Mycobacterium tuberculosis (M. tb) in the form of immunodominant antigens hijack the host immune system to give a survival advantage to this intracellular fiend, but the mechanism of this control is not entirely known. Since we have previously reported the mechanism of autophagy inhibition by early secreted antigenic target 6 kDa (ESAT-6) through microRNA (miR)-30a-3p in Calcimycin treated differentiated THP-1 (dTHP-1) cells, the present study was undertaken to deduce the effect of miR-30a on the immunomodulatory profile of ESAT-6 treated cells and the mechanism involved thereof, if any. Initially, the effect of recombinant ESAT-6 (rESAT-6) on the immunomodulatory profile in Calcimycin-treated phorbol 12-myristate 13-acetate (PMA) dTHP-1 cells was checked. Later, transfection studies using miR-30a-3p inhibitor or -5p mimic highlighted the contrary roles of different arms of the same miRNA in regulating IL-18 response by ESAT-6 in dTHP-1 cells after Calcimycin treatment. By using either IL-18 neutralizing antibody or inhibitors of phosphoinositide 3-kinase (PI3K)/NF-κB/phagosome-lysosome fusion in the miRNA-30a transfected background, IL-18 mediated signaling and intracellular killing of mycobacteria was reversed in the presence of ESAT-6. Overall, the results of this study conclusively prove the contrary roles of miR-30a-3p and miR-30a-5p in regulating IL-18 signaling by ESAT-6 in dTHP-1 cells upon Calcimycin treatment that affected phagosome-lysosome fusion and intracellular survival of mycobacteria.
Collapse
Affiliation(s)
- Assirbad Behura
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Mousumi Das
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Ashish Kumar
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Lincoln Naik
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Abtar Mishra
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Debraj Manna
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Salina Patel
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan 342011, India
| | - Ramandeep Singh
- Tuberculosis Research Laboratory, Vaccine and Infectious Disease Research Centre, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurugram Expressway, PO Box # 4, Faridabad 121001, Haryana, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela 769008, Odisha, India.
| |
Collapse
|
24
|
Kalra R, Tiwari D, Dkhar HK, Bhagyaraj E, Kumar R, Bhardwaj A, Gupta P. Host factors subverted by Mycobacterium tuberculosis: Potential targets for host directed therapy. Int Rev Immunol 2021; 42:43-70. [PMID: 34678117 DOI: 10.1080/08830185.2021.1990277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Despite new approaches in the diagnosis and treatment of tuberculosis (TB), it continues to be a major health burden. Several immunotherapies that potentiate the immune response have come up as adjuncts to drug therapies against drug resistant TB strains; however, there needs to be an urgent appraisal of host specific drug targets for improving their clinical management and to curtail disease progression. Presently, various host directed therapies (HDTs) exist (repurposed drugs, nutraceuticals, monoclonal antibodies and immunomodulatory agents), but these mostly address molecules that combat disease progression. AREAS COVERED The current review discusses major Mycobacterium tuberculosis (M. tuberculosis) survival paradigms inside the host and presents a plethora of host targets subverted by M. tuberculosis which can be further explored for future HDTs. The host factors unique to M. tuberculosis infection (in humans) have also been identified through an in-silico interaction mapping. EXPERT OPINION HDTs could become the next-generation adjunct therapies in order to counter antimicrobial resistance and virulence, as well as to reduce the duration of existing TB treatments. However, current scientific efforts are largely directed toward combatants rather than host molecules co-opted by M. tuberculosis for its survival. This might drive the immune system to a hyper-inflammatory condition; therefore, we emphasize that host factors subverted by M. tuberculosis, and their subsequent neutralization, must be considered for development of better HDTs.
Collapse
Affiliation(s)
- Rashi Kalra
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Drishti Tiwari
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Hedwin Kitdorlang Dkhar
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Ella Bhagyaraj
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | - Rakesh Kumar
- Bioinformatics Center, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Anshu Bhardwaj
- Bioinformatics Center, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Pawan Gupta
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh-160036, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
25
|
Sharma S, Kumar M, Kumar J, Srivastava N, Hussain MA, Shelly A, Mazumder S. M. fortuitum-induced CNS-pathology: Deciphering the role of canonical Wnt signaling, blood brain barrier components and cytokines. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 122:104111. [PMID: 33933535 DOI: 10.1016/j.dci.2021.104111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/24/2021] [Accepted: 04/24/2021] [Indexed: 06/12/2023]
Abstract
Molecular underpinning of mycobacteria-induced CNS-pathology is not well understood. In the present study, zebrafish were infected with Mycobacterium fortuitum and the prognosis of CNS-pathogenesis studied. We observed M. fortuitum triggers extensive brain-pathology. Evans blue extravasation demonstrated compromised blood-brain barrier (BBB) integrity. Further, decreased expression in tight-junction (TJ) and adherens junction complex (AJC) genes were noted in infected brain. Wnt-signaling has emerged as a major player in host-mycobacterial immunity but its involvement/role in brain-infection is not well studied. Sustained expression of wnt2, wnt3a, fzd5, lrp5/6 and β-catenin, with concordant decline in degradation complex components axin, gsk3β and β-catenin regulator capn2a were observed. The surge in ifng1 and tnfa expression preceding il10 and il4 suggested cytokine-interplay critical in M. fortuitum-induced brain-pathology. Therefore, we suggest adult zebrafish as a viable model for studying CNS-pathology and using the same, conclude that M. fortuitum infection is associated with repressed TJ-AJC gene expression and compromised BBB permeability. Our results implicate Wnt/β-catenin pathway in M. fortuitum-induced CNS-pathology wherein Th1-type signals facilitate bacterial clearance and Th2-type signals prevent the disease sequel.
Collapse
Affiliation(s)
- Shagun Sharma
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Manmohan Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Jai Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Nidhi Srivastava
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India; Department of Zoology, School of Basic and Applied Sciences, Maharaja Agrasen University, Solan, Himachal Pradesh, 174103, India
| | - Md Arafat Hussain
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Asha Shelly
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India; Faculty of Life Sciences and Biotechnology, South Asian University, Delhi, 110021, India.
| |
Collapse
|
26
|
Nontuberculous Mycobacteria, Macrophages, and Host Innate Immune Response. Infect Immun 2021; 89:e0081220. [PMID: 34097459 DOI: 10.1128/iai.00812-20] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Although nontuberculous mycobacteria (NTM) are considered opportunistic infections, incidence and prevalence of NTM infection are increasing worldwide becoming a major public health threat. Innate immunity plays an essential role in mediating the initial host response against these intracellular bacteria. Specifically, macrophages phagocytose and eliminate NTM and act as antigen-presenting cells, which trigger downstream activation of cellular and humoral adaptive immune responses. Identification of macrophage receptors, mycobacterial ligands, phagosome maturation, autophagy/necrosis, and escape mechanisms are important components of this immunity network. The role of the macrophage in mycobacterial disease has mainly been studied in tuberculosis (TB), but limited information exists on its role in NTM. In this review, we focus on NTM immunity, the role of macrophages, and host interaction in NTM infection.
Collapse
|
27
|
Mycobacterium tuberculosis Binds Human Serum Amyloid A, and the Interaction Modulates the Colonization of Human Macrophages and the Transcriptional Response of the Pathogen. Cells 2021; 10:cells10051264. [PMID: 34065319 PMCID: PMC8160739 DOI: 10.3390/cells10051264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 01/10/2023] Open
Abstract
As a very successful pathogen with outstanding adaptive properties, Mycobacterium tuberculosis (Mtb) has developed a plethora of sophisticated mechanisms to subvert host defenses and effectively enter and replicate in the harmful environment inside professional phagocytes, namely, macrophages. Here, we demonstrated the binding interaction of Mtb with a major human acute phase protein, namely, serum amyloid A (SAA1), and identified AtpA (Rv1308), ABC (Rv2477c), EspB (Rv3881c), TB 18.6 (Rv2140c), and ThiC (Rv0423c) membrane proteins as mycobacterial effectors responsible for the pathogen-host protein interplay. SAA1-opsonization of Mtb prior to the infection of human macrophages favored bacterial entry into target phagocytes accompanied by a substantial increase in the load of intracellularly multiplying and surviving bacteria. Furthermore, binding of human SAA1 by Mtb resulted in the up- or downregulation of the transcriptional response of tubercle bacilli. The most substantial changes were related to the increased expression level of the genes of two operons encoding mycobacterial transporter systems, namely, mmpL5/mmpS5 (rv0676c), and rv1217c, rv1218c. Therefore, we postulate that during infection, Mtb-SAA1 binding promotes the infection of host macrophages by tubercle bacilli and modulates the functional response of the pathogen.
Collapse
|
28
|
Foster M, Hill PC, Setiabudiawan TP, Koeken VACM, Alisjahbana B, van Crevel R. BCG-induced protection against Mycobacterium tuberculosis infection: Evidence, mechanisms, and implications for next-generation vaccines. Immunol Rev 2021; 301:122-144. [PMID: 33709421 PMCID: PMC8252066 DOI: 10.1111/imr.12965] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/20/2022]
Abstract
The tuberculosis (TB) vaccine Bacillus Calmette-Guérin (BCG) was introduced 100 years ago, but as it provides insufficient protection against TB disease, especially in adults, new vaccines are being developed and evaluated. The discovery that BCG protects humans from becoming infected with Mycobacterium tuberculosis (Mtb) and not just from progressing to TB disease provides justification for considering Mtb infection as an endpoint in vaccine trials. Such trials would require fewer participants than those with disease as an endpoint. In this review, we first define Mtb infection and disease phenotypes that can be used for mechanistic studies and/or endpoints for vaccine trials. Secondly, we review the evidence for BCG-induced protection against Mtb infection from observational and BCG re-vaccination studies, and discuss limitations and variation of this protection. Thirdly, we review possible underlying mechanisms for BCG efficacy against Mtb infection, including alternative T cell responses, antibody-mediated protection, and innate immune mechanisms, with a specific focus on BCG-induced trained immunity, which involves epigenetic and metabolic reprogramming of innate immune cells. Finally, we discuss the implications for further studies of BCG efficacy against Mtb infection, including for mechanistic research, and their relevance to the design and evaluation of new TB vaccines.
Collapse
Affiliation(s)
- Mitchell Foster
- Department of Microbiology and ImmunologyUniversity of OtagoDunedinNew Zealand
| | - Philip C. Hill
- Centre for International HealthUniversity of OtagoDunedinNew Zealand
| | - Todia Pediatama Setiabudiawan
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI)Radboud University Medical CenterNijmegenThe Netherlands
| | - Valerie A. C. M. Koeken
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI)Radboud University Medical CenterNijmegenThe Netherlands
- Department of Computational Biology for Individualised Infection MedicineCentre for Individualised Infection Medicine (CiiM) & TWINCOREJoint Ventures between The Helmholtz‐Centre for Infection Research (HZI) and The Hannover Medical School (MHH)HannoverGermany
| | - Bachti Alisjahbana
- Tuberculosis Working GroupFaculty of MedicineUniversitas PadjadjaranBandungIndonesia
| | - Reinout van Crevel
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI)Radboud University Medical CenterNijmegenThe Netherlands
| |
Collapse
|
29
|
Mehta M, Dhanjal DS, Satija S, Wadhwa R, Paudel KR, Chellappan DK, Mohammad S, Haghi M, Hansbro PM, Dua K. Advancing of Cellular Signaling Pathways in Respiratory Diseases Using Nanocarrier Based Drug Delivery Systems. Curr Pharm Des 2021; 26:5380-5392. [PMID: 33198611 DOI: 10.2174/1381612826999201116161143] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 10/01/2020] [Indexed: 11/22/2022]
Abstract
Cell Signaling pathways form an integral part of our existence that allows the cells to comprehend a stimulus and respond back. Such reactions to external cues from the environment are required and are essential to regulate the normal functioning of our body. Abnormalities in the system arise when there are errors developed in these signals, resulting in a complication or a disease. Presently, respiratory diseases contribute to being the third leading cause of morbidity worldwide. According to the current statistics, over 339 million people are asthmatic, 65 million are suffering from COPD, 2.3 million are lung cancer patients and 10 million are tuberculosis patients. This toll of statistics with chronic respiratory diseases leaves a heavy burden on society and the nation's annual health expenditure. Hence, a better understanding of the processes governing these cellular pathways will enable us to treat and manage these deadly respiratory diseases effectively. Moreover, it is important to comprehend the synergy and interplay of the cellular signaling pathways in respiratory diseases, which will enable us to explore and develop suitable strategies for targeted drug delivery. This review, in particular, focuses on the major respiratory diseases and further provides an in-depth discussion on the various cell signaling pathways that are involved in the pathophysiology of respiratory diseases. Moreover, the review also analyses the defining concepts about advanced nano-drug delivery systems involving various nanocarriers and propose newer prospects to minimize the current challenges faced by researchers and formulation scientists.
Collapse
Affiliation(s)
- Meenu Mehta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Daljeet Singh Dhanjal
- School of Biosciences and Bioengineering, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Saurabh Satija
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Ridhima Wadhwa
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Keshav Raj Paudel
- School of Life Sciences, Faculty of Science, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | - Shiva Mohammad
- School of Life Sciences, Faculty of Science, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
| | - Mehra Haghi
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Philip M Hansbro
- School of Life Sciences, Faculty of Science, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo NSW 2007, Australia
| |
Collapse
|
30
|
Gopalaswamy R, Subbian S. Corticosteroids for COVID-19 Therapy: Potential Implications on Tuberculosis. Int J Mol Sci 2021; 22:ijms22073773. [PMID: 33917321 PMCID: PMC8038708 DOI: 10.3390/ijms22073773] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022] Open
Abstract
On 11 March 2020, the World Health Organization announced the Corona Virus Disease-2019 (COVID-19) as a global pandemic, which originated in China. At the host level, COVID-19, caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), affects the respiratory system, with the clinical symptoms ranging from mild to severe or critical illness that often requires hospitalization and oxygen support. There is no specific therapy for COVID-19, as is the case for any common viral disease except drugs to reduce the viral load and alleviate the inflammatory symptoms. Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (Mtb), also primarily affects the lungs and has clinical signs similar to pulmonary SARS-CoV-2 infection. Active TB is a leading killer among infectious diseases and adds to the burden of the COVID-19 pandemic worldwide. In immunocompetent individuals, primary Mtb infection can also lead to a non-progressive, asymptomatic latency. However, latent Mtb infection (LTBI) can reactivate symptomatic TB disease upon host immune-suppressing conditions. Importantly, the diagnosis and treatment of TB are hampered and admixed with COVID-19 control measures. The US-Center for Disease Control (US-CDC) recommends using antiviral drugs, Remdesivir or corticosteroid (CST), such as dexamethasone either alone or in-combination with specific recommendations for COVID-19 patients requiring hospitalization or oxygen support. However, CSTs can cause immunosuppression, besides their anti-inflammatory properties. The altered host immunity during COVID-19, combined with CST therapy, poses a significant risk for new secondary infections and/or reactivation of existing quiescent infections, such as LTBI. This review highlights CST therapy recommendations for COVID-19, various types and mechanisms of action of CSTs, the deadly combination of two respiratory infectious diseases COVID-19 and TB. It also discusses the importance of screening for LTBI to prevent TB reactivation during corticosteroid therapy for COVID-19.
Collapse
Affiliation(s)
- Radha Gopalaswamy
- Department of Bacteriology, ICMR-National Institute for Research in Tuberculosis, Chennai 600031, India;
| | - Selvakumar Subbian
- Public Health Research Institute at New Jersey Medical School, Rutgers University, 225 Warren Street, Newark, NJ 08854, USA
- Correspondence:
| |
Collapse
|
31
|
Takehara M, Kobayashi K, Nagahama M. Toll-Like Receptor 4 Protects Against Clostridium perfringens Infection in Mice. Front Cell Infect Microbiol 2021; 11:633440. [PMID: 33763386 PMCID: PMC7982660 DOI: 10.3389/fcimb.2021.633440] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/03/2021] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptor 4 (TLR4) has been reported to protect against Gram-negative bacteria by acting as a pathogen recognition receptor that senses mainly lipopolysaccharide (LPS) from Gram-negative bacteria. However, the role of TLR4 in Gram-positive bacterial infection is less well understood. Clostridium perfringens type A is a Gram-positive bacterium that causes gas gangrene characterized by severe myonecrosis. It was previously demonstrated that C. perfringens θ-toxin is a TLR4 agonist, but the role of TLR4 in C. perfringens infection is unclear. Here, TLR4-defective C3H/HeJ mice infected with C. perfringens showed a remarkable decrease in survival rate, an increase in viable bacterial counts, and accelerated destruction of myofibrils at the infection site compared with wild-type C3H/HeN mice. These results demonstrate that TLR4 plays an important role in the elimination of C. perfringens. Remarkable increases in levels of inflammatory cytokines, such as interleukin-1β (IL-1β), interleukin-6 (IL-6), and granulocyte colony-stimulating factor (G-CSF), were observed in C. perfringens-infected C3H/HeN mice, whereas the increases were limited in C3H/HeJ mice. Generally, increased G-CSF accelerates granulopoiesis in the bone marrow and the spleen to exacerbate neutrophil production, resulting in elimination of bacteria. The number of neutrophils in the spleen was increased in C. perfringens-infected C3H/HeN mice compared with non-infected mice, while the increase was lower in C. perfringens-infected C3H/HeJ mice. Furthermore, DNA microarray analysis revealed that the mutation in TLR4 partially affects host gene expression during C. perfringens infection. Together, our results illustrate that TLR4 is crucial for the innate ability to eliminate C. perfringens.
Collapse
Affiliation(s)
- Masaya Takehara
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Japan
| | - Keiko Kobayashi
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Japan
| | - Masahiro Nagahama
- Department of Microbiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Japan
| |
Collapse
|
32
|
Lei Y, Cao X, Xu W, Yang B, Xu Y, Zhou W, Dong S, Wu Q, Rahman K, Tyagi R, Zhao S, Chen X, Cao G. Rv3722c Promotes Mycobacterium tuberculosis Survival in Macrophages by Interacting With TRAF3. Front Cell Infect Microbiol 2021; 11:627798. [PMID: 33718275 PMCID: PMC7947218 DOI: 10.3389/fcimb.2021.627798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/19/2021] [Indexed: 01/08/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb) secretes numerous proteins to interfere with host immune response for its long-term survival. As one of the top abundant M.tb secreted proteins, Rv3722c was found to be essential for bacilli growth. However, it remains elusive how this protein interferes with the host immune response and regulates M.tb survival. Here, we confirmed that Rv3722c interacted with host TRAF3 to promote M.tb replication in macrophages. Knock-down of TRAF3 attenuated the effect of Rv3722c on the intracellular M.tb survival. The interaction between Rv3722c and TRAF3 hampered MAPK and NF-κB pathways, resulting in a significant increase of IFN-β expression and decrease of IL-1β, IL-6, IL-12p40, and TNF-α expression. Our study revealed that Rv3722c interacted with TRAF3 and interrupted its downstream pathways to promote M.tb survival in macrophages. These findings facilitate further understanding of the mechanism of M.tb secreted proteins in regulating the host cell immune response and promoting its intracellular survival.
Collapse
Affiliation(s)
- Yingying Lei
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaojian Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Weize Xu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Bing Yang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yangyang Xu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wei Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shuang Dong
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qijun Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Khaista Rahman
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Rohit Tyagi
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Shuhong Zhao
- Key Laboratory of Agricultural Animal Genetics, Breeding, Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China.,College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xi Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Bio-Medical Center, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
33
|
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: 2.0] [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
|
34
|
Sphingomyelin Biosynthesis Is Essential for Phagocytic Signaling during Mycobacterium tuberculosis Host Cell Entry. mBio 2021; 12:mBio.03141-20. [PMID: 33500344 PMCID: PMC7858061 DOI: 10.1128/mbio.03141-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) invades alveolar macrophages through phagocytosis to establish infection and cause disease. The molecular mechanisms underlying Mtb entry are still poorly understood. Phagocytosis by alveolar macrophages is the obligate first step in Mycobacterium tuberculosis (Mtb) infection, yet the mechanism underlying this process is incompletely understood. Here, we show that Mtb invasion relies on an intact sphingolipid biosynthetic pathway. Inhibition or knockout of early sphingolipid biosynthetic enzymes greatly reduces Mtb uptake across multiple phagocytic cell types without affecting other forms of endocytosis. While the phagocytic receptor dectin-1 undergoes normal clustering at the pathogen contact sites, sphingolipid biosynthetic mutant cells fail to segregate the regulatory phosphatase CD45 from the clustered receptors. Blocking sphingolipid production also impairs downstream activation of Rho GTPases, actin dynamics, and phosphoinositide turnover at the nascent phagocytic cup. Moreover, we found that production of sphingomyelin, not glycosphingolipids, is essential for Mtb uptake. Collectively, our data support a critical role of sphingomyelin biosynthesis in an early stage of Mtb infection and provide novel insights into the mechanism underlying phagocytic entry of this pathogen.
Collapse
|
35
|
Tapela K, Ochieng’ Olwal C, Quaye O. Parallels in the pathogenesis of SARS-CoV-2 and M. tuberculosis: a synergistic or antagonistic alliance? Future Microbiol 2020; 15:1691-1695. [PMID: 33404259 PMCID: PMC7789731 DOI: 10.2217/fmb-2020-0179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/08/2020] [Indexed: 01/12/2023] Open
Affiliation(s)
- Kesego Tapela
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell & Molecular Biology, College of Basic & Applied Sciences, University of Ghana, Accra, Ghana
| | - Charles Ochieng’ Olwal
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell & Molecular Biology, College of Basic & Applied Sciences, University of Ghana, Accra, Ghana
| | - Osbourne Quaye
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell & Molecular Biology, College of Basic & Applied Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
36
|
Ivanyi J. Tuberculosis vaccination needs to avoid 'decoy' immune reactions. Tuberculosis (Edinb) 2020; 126:102021. [PMID: 33254012 DOI: 10.1016/j.tube.2020.102021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 11/16/2022]
Abstract
Current search for a new effective vaccine against tuberculosis involves selected antigens, vectors and adjuvants. These are being evaluated usually by their booster inoculation following priming with Bacillus Calmette-Guerin. The purpose of this article is to point out, that despite being attenuated of virulence, priming with BCG may still involve immune mechanisms, which are not favourable for protection against active disease. It is postulated, that the responsible 'decoy' constituents selected during the evolution of pathogenic tubercle bacilli may be involved in the evasion from bactericidal host resistance and stimulate immune responses of a cytokine phenotype, which lead to the transition from latent closed granulomas to reactivation with infectious lung cavities. The decoy mechanisms appear as favourable for most infected subjects but leading in a minority of cases to pathology which can effectively transmit the infection. It is proposed that construction and development of new vaccine candidates could benefit from avoiding decoy-type immune mechanisms.
Collapse
Affiliation(s)
- Juraj Ivanyi
- Centre for Host-Microbiome Interactions, Guy's Campus of Kings College London, SE1, 1UL, United kingdom.
| |
Collapse
|
37
|
Feng JY, Su WJ, Chuang FY, Pan SW, Yeh YC, Lin YY, Chen NJ. TREM-1 enhances Mycobacterium tuberculosis-induced inflammatory responses in macrophages. Microbes Infect 2020; 23:104765. [PMID: 33049389 DOI: 10.1016/j.micinf.2020.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/09/2020] [Accepted: 10/05/2020] [Indexed: 01/06/2023]
Abstract
Triggering receptor expressed on myeloid cells 1 (TREM-1) extensively interacts with toll-like receptors and amplifies pro-inflammatory responses. The effect of TREM-1 on Mycobacterium tuberculosis (MTB)-related immune responses remains to be elucidated. We isolated bone marrow-derived macrophages (BMDMs) from wild-type mice and Trem-1 KO mice and treated them with MTB whole cell lysate and EsxA (ESAT-6). Cytokine production and mRNA expression, including Trem-1, following stimulation were evaluated. Intratracheal instillation of heat-killed MTB (HKMTB) in mice was performed and the presence of TREM-1-positive macrophages was investigated by immunohistochemistry analysis. In our study, BMDMs isolated from wild-type mice produced more pro-inflammatory cytokines and demonstrated higher inflammatory gene expression levels compared with those isolated from Trem-1 KO mice when stimulated with MTB whole cell lysate. EsxA had a synergistic effect with MTB whole cell lysate on the induction of pro-inflammatory responses. The gene expression of Trem-1 was upregulated when treated with MTB-related proteins. TREM-1-positive macrophages were identified in the lung tissues from patients with active TB and from wild-type mice treated with intratracheal instillation of HKMTB. In conclusion, in mouse macrophages, TREM-1 could enhance pro-inflammatory immune responses when stimulated with MTB-related proteins. The gene expression of Trem-1 could also be induced by MTB-related stimulation.
Collapse
Affiliation(s)
- Jia-Yih Feng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Juin Su
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Fan-Yi Chuang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Sheng-Wei Pan
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan; Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Chen Yeh
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yung-Yang Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cerebrovascular Diseases, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
38
|
Arshad A, Dayal S, Gadhe R, Mawley A, Shin K, Tellez D, Phan P, Venketaraman V. Analysis of Tuberculosis Meningitis Pathogenesis, Diagnosis, and Treatment. J Clin Med 2020; 9:E2962. [PMID: 32937808 PMCID: PMC7565176 DOI: 10.3390/jcm9092962] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/30/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Tuberculosis (TB) is the most prevalent infectious disease in the world. In recent years there has been a significant increase in the incidence of TB due to the emergence of multidrug resistant strains of Mycobacterium tuberculosis (M. tuberculosis) and the increased numbers of highly susceptible immuno-compromised individuals. Central nervous system TB, includes TB meningitis (TBM-the most common presentation), intracranial tuberculomas, and spinal tuberculous arachnoiditis. Individuals with TBM have an initial phase of malaise, headache, fever, or personality change, followed by protracted headache, stroke, meningismus, vomiting, confusion, and focal neurologic findings in two to three weeks. If untreated, mental status deteriorates into stupor or coma. Delay in the treatment of TBM results in, either death or substantial neurological morbidity. This review provides latest developments in the biomedical research on TB meningitis mainly in the areas of host immune responses, pathogenesis, diagnosis, and treatment of this disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766-1854, USA; (A.A.); (S.D.); (R.G.); (A.M.); (K.S.); (D.T.); (P.P.)
| |
Collapse
|
39
|
Kan LLY, Liu D, Chan BCL, Tsang MSM, Hou T, Leung PC, Lam CWK, Wong CK. The flavonoids of Sophora flavescens exerts anti-inflammatory activity via promoting autophagy of Bacillus Calmette-Guérin-stimulated macrophages. J Leukoc Biol 2020; 108:1615-1629. [PMID: 32794339 DOI: 10.1002/jlb.3ma0720-682rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 11/06/2022] Open
Abstract
Tuberculosis (TB), a highly infectious air-borne disease, has remained a global health problem. Conventional treatment and preventions such as antibiotics and Bacilli Calmette-Guerin (BCG) vaccine can be unreliable. In view of the increasing prevalence of anti-TB drug resistance, adjunctive therapy may be necessary to shorten the recovery time. We have previously shown that flavonoids in the medicinal herb Sophora flavescens exhibit anti-inflammatory and bactericidal activities. The aim of this study was to investigate the molecular and cellular characteristics of flavonoids of S. flavescens (FSF) in BCG-stimulated macrophages for assessing their roles in anti-inflammation and autophagy. Mouse alveolar macrophage (MH-S) cell line and primary mouse peritoneal macrophages were stimulated in vitro with heat-inactivated BCG and treated with FSF, with or without autophagy inhibitor Bafilomycin A1 (BafA1). Gene expression was analyzed using quantitative PCR, and cytokine/chemokine release was analyzed by Milliplex assay and ELISA. Autophagy-related proteins were quantified by Western blot and flow cytometry, and autophagolysosomes were detected using fluorescence microscopy. In both MH-S cell line and mouse peritoneal macrophages stimulated by heat-inactivated BCG, FSF was found to up-regulate autophagy-related proteins microtubule-associated protein 1A/1B-light chain 3 (LC3) and protein 62 (p62), and suppress the induced proinflammatory cytokine TNF-α, CCL5, and IL-6. FSF actively modulates immune processes through suppressing BCG-mediated inflammation by promoting autophagy in MH-S cells and mouse peritoneal macrophages. We suggest that FSF may be useful as an adjunctive therapeutic agent for TB infection by modulating cell survival through autophagy and reducing inflammation.
Collapse
Affiliation(s)
- Lea Ling-Yu Kan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Dehua Liu
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben Chung-Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Miranda Sin-Man Tsang
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China.,Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Tianheng Hou
- Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ping Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher Wai-Kei Lam
- Faculty of Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Chun Kwok Wong
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China.,Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.,Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
40
|
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: 1.3] [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]
|
41
|
Sachdeva K, Goel M, Sundaramurthy V. Heterogeneity in the endocytic capacity of individual macrophage in a population determines its subsequent phagocytosis, infectivity and subcellular trafficking. Traffic 2020; 21:522-533. [DOI: 10.1111/tra.12752] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Kuldeep Sachdeva
- National Center for Biological Sciences Tata Institute of Fundamental Research Bangalore India
| | - Manisha Goel
- National Center for Biological Sciences Tata Institute of Fundamental Research Bangalore India
| | | |
Collapse
|
42
|
Liao Y, Li X, Mou T, Zhou X, Li D, Wang L, Zhang Y, Dong X, Zheng H, Guo L, Liang Y, Jiang G, Fan S, Xu X, Xie Z, Chen H, Liu L, Li Q. Distinct infection process of SARS-CoV-2 in human bronchial epithelial cell lines. J Med Virol 2020; 92:2830-2838. [PMID: 32558946 PMCID: PMC7323243 DOI: 10.1002/jmv.26200] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022]
Abstract
Coronavirus disease 2019, caused by severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2), leads to a series of clinical symptoms of respiratory and pulmonary inflammatory reactions via unknown pathologic mechanisms related to the viral infection process in tracheal or bronchial epithelial cells. Investigation of this viral infection in the human bronchial epithelial cell line (16HBE) suggests that SARS‐CoV‐2 can enter these cells through interaction between its membrane‐localized S protein with the angiotensin‐converting enzyme 2 molecule on the host cell membrane. Further observation indicates distinct viral replication with a dynamic and moderate increase, whereby viral replication does not lead to a specific cytopathic effect but maintains a continuous release of progeny virions from infected cells. Although messenger RNA expression of various innate immune signaling molecules is altered in the cells, transcription of interferons‐α (IFN‐α), IFN‐β, and IFN‐γ is unchanged. Furthermore, expression of some interleukins (IL) related to inflammatory reactions, such as IL‐6, IL‐2, and IL‐8, is maintained at low levels, whereas that of ILs involved in immune regulation is upregulated. Interestingly, IL‐22, an IL that functions mainly in tissue repair, shows very high expression. Collectively, these data suggest a distinct infection process for this virus in respiratory epithelial cells, which may be linked to its clinicopathological mechanism. SARS‐CoV‐2 does not lead to a specific cytopathic effect in 16HBE cells, but maintains continuous release of progeny virions from infected cells. During infection, IL‐22, an interleukin that functions mainly in tissue repair, showed very high expression.
Collapse
Affiliation(s)
- Yun Liao
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Xueqi Li
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Tangwei Mou
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Xiaofang Zhou
- Acute Infectious Disease Laboratory, Yunnan Center for Disease Control and Prevention, Kunming, Yunnan, China
| | - Dandan Li
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Lichun Wang
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Ying Zhang
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Xingqi Dong
- Infectious Disease Laboratory, Yunnan Provincial Infectious Disease Hospital, Kunming, Yunnan, China
| | - Huiwen Zheng
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Lei Guo
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Yan Liang
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Guorun Jiang
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Shengtao Fan
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Xingli Xu
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Zhongping Xie
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Hongbo Chen
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Longding Liu
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| | - Qihan Li
- Institute of Medical Biology, Chinese Academy of Medicine Science & Peking Union Medical College, Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Diseases, Kunming, Yunnan, China
| |
Collapse
|
43
|
Naqvi KF, Endsley JJ. Myeloid C-Type Lectin Receptors in Tuberculosis and HIV Immunity: Insights Into Co-infection? Front Cell Infect Microbiol 2020; 10:263. [PMID: 32582566 PMCID: PMC7283559 DOI: 10.3389/fcimb.2020.00263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
C-type lectin receptors (CLRs) are carbohydrate binding pattern recognition receptors (PRRs) which play a central role in host recognition of pathogenic microorganisms. Signaling through CLRs displayed on antigen presenting cells dictates important innate and adaptive immune responses. Several pathogens have evolved mechanisms to exploit the receptors or signaling pathways of the CLR system to gain entry or propagate in host cells. CLR responses to high priority pathogens such as Mycobacterium tuberculosis (Mtb), HIV, Ebola, and others are described and considered potential avenues for therapeutic intervention. Mtb and HIV are the leading causes of death due to infectious disease and have a synergistic relationship that further promotes aggressive disease in co-infected persons. Immune recognition through CLRs and other PRRs are important determinants of disease outcomes for both TB and HIV. Investigations of CLR responses to Mtb and HIV, to date, have primarily focused on single infection outcomes and do not account for the potential effects of co-infection. This review will focus on CLRs recognition of Mtb and HIV motifs. We will describe their respective roles in protective immunity and immune evasion or exploitation, as well as their potential as genetic determinants of disease susceptibility, and as avenues for development of therapeutic interventions. The potential convergence of CLR-driven responses of the innate and adaptive immune systems in the setting of Mtb and HIV co-infection will further be discussed relevant to disease pathogenesis and development of clinical interventions.
Collapse
Affiliation(s)
- Kubra F Naqvi
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Janice J Endsley
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
44
|
Ali MK, Zhen G, Nzungize L, Stojkoska A, Duan X, Li C, Duan W, Xu J, Xie J. Mycobacterium tuberculosis PE31 ( Rv3477) Attenuates Host Cell Apoptosis and Promotes Recombinant M. smegmatis Intracellular Survival via Up-regulating GTPase Guanylate Binding Protein-1. Front Cell Infect Microbiol 2020; 10:40. [PMID: 32117813 PMCID: PMC7020884 DOI: 10.3389/fcimb.2020.00040] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
The Mycobacterium (M.) tuberculosis comprising proline–glutamic acid (PE) subfamily proteins associate with virulence, pathogenesis, and host-immune modulations. While the functions of most of this family members are not yet explored. Here, we explore the functions of “PE only” subfamily member PE31 (Rv3477) in virulence and host-pathogen interactions. We have expressed the M. tuberculosis PE31 in non-pathogenic Mycobacterium smegmatis strain (Ms_PE31) and demonstrated that PE31 significantly altered the cell facet features including colony morphology and biofilm formation. PE31 expressing M. smegmatis showed more resistant to the low pH, diamide, H2O2 and surface stress. Moreover, Ms_PE31 showed higher intracellular survival in macrophage THP-1 cells. Ms_PE31 significantly down-regulated the production of IL-12p40 and IL-6, while up-regulates the production of IL-10 in macrophages. Ms_PE31 also induced the expression of guanylate-binding protein-1 (GBP-1) in macrophages. Further analysis demonstrates that Ms_PE31 inhibits the caspase-3 activation and reduces the macrophages apoptosis. Besides, the NF-κB signaling pathway involves the interplay between Ms_PE31 and macrophages. Collectively, our finding identified that PE31 act as a functionally relevant virulence factor of M. tuberculosis.
Collapse
Affiliation(s)
- Md Kaisar Ali
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Gong Zhen
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Lambert Nzungize
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Andrea Stojkoska
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Xiangke Duan
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Chunyan Li
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Wei Duan
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Junqi Xu
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| | - Jianping Xie
- State Key Laboratory Breeding Base of Eco-Environment and Bio-Resource of the Three Gorges Area, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, Ministry of Education, School of Life Sciences, Institute of Modern Biopharmaceuticals, Southwest University, Chongqing, China
| |
Collapse
|
45
|
Silva-García O, Valdez-Alarcón JJ, Baizabal-Aguirre VM. Wnt/β-Catenin Signaling as a Molecular Target by Pathogenic Bacteria. Front Immunol 2019; 10:2135. [PMID: 31611869 PMCID: PMC6776594 DOI: 10.3389/fimmu.2019.02135] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/27/2019] [Indexed: 12/31/2022] Open
Abstract
The Wnt/β-catenin signaling pathway is crucial to regulate cell proliferation and polarity, cell determination, and tissue homeostasis. The activation of Wnt/β-catenin signaling is based on the interaction between Wnt glycoproteins and seven transmembrane receptors-Frizzled (Fzd). This binding promotes recruitment of the scaffolding protein Disheveled (Dvl), which results in the phosphorylation of the co-receptor LRP5/6. The resultant molecular complex Wnt-Fzd-LRP5/6-Dvl forms a structural region for Axin interaction that disrupts Axin-mediated phosphorylation/degradation of the transcriptional co-activator β-catenin, thereby allowing it to stabilize and accumulate in the nucleus where it activates the expression of Wnt-dependent genes. Due to the prominent physiological function, the Wnt/β-catenin signaling must be strictly controlled because its dysregulation, which is caused by different stimuli, may lead to alterations in cell proliferation, apoptosis, and inflammation-associated cancer. The virulence factors from pathogenic bacteria such as Salmonella enterica sv Typhimurium, Helicobacter pylori, Mycobacterium tuberculosis, Pseudomonas aeruginosa, Citrobacter rodentium, Clostridium difficile, Bacteroides fragilis, Escherichia coli, Haemophilus parasuis, Lawsonia intracellularis, Shigella dysenteriae, and Staphylococcus epidermidis employ a variety of molecular strategies to alter the appropriate functioning of diverse signaling pathways. Among these, Wnt/β-catenin has recently emerged as an important target of several virulence factors produced by bacteria. The mechanisms used by these factors to interfere with the activity of Wnt/β-catenin is diverse and include the repression of Wnt inhibitors' expression by the epigenetic modification of histones, blocking Wnt-Fzd ligand binding, activation or inhibition of β-catenin nuclear translocation, down- or up-regulation of Wnt family members, and inhibition of Axin-1 expression that promotes β-catenin activity. Such a variety of mechanisms illustrate an evolutionary co-adaptation of eukaryotic molecular signaling to a battery of soluble or structural components synthesized by pathogenic bacteria. This review gathers the recent efforts to elucidate the mechanistic details through which bacterial virulence factors modulate Wnt/β-catenin signaling and its physiological consequences concerning the inflammatory response and cancer.
Collapse
Affiliation(s)
| | - Juan J Valdez-Alarcón
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| | - Víctor M Baizabal-Aguirre
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Mexico
| |
Collapse
|
46
|
Fox KA, Kirwan DE, Whittington AM, Krishnan N, Robertson BD, Gilman RH, López JW, Singh S, Porter JC, Friedland JS. Platelets Regulate Pulmonary Inflammation and Tissue Destruction in Tuberculosis. Am J Respir Crit Care Med 2019; 198:245-255. [PMID: 29420060 DOI: 10.1164/rccm.201710-2102oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
RATIONALE Platelets may interact with the immune system in tuberculosis (TB) to regulate human inflammatory responses that lead to morbidity and spread of infection. OBJECTIVES To identify a functional role of platelets in the innate inflammatory and matrix-degrading response in TB. METHODS Markers of platelet activation were examined in plasma from 50 patients with TB before treatment and 50 control subjects. Twenty-five patients were followed longitudinally. Platelet-monocyte interactions were studied in a coculture model infected with live, virulent Mycobacterium tuberculosis (M.tb) and dissected using qRT-PCR, Luminex multiplex arrays, matrix degradation assays, and colony counts. Immunohistochemistry detected CD41 (cluster of differentiation 41) expression in a pulmonary TB murine model, and secreted platelet factors were measured in BAL fluid from 15 patients with TB and matched control subjects. MEASUREMENTS AND MAIN RESULTS Five of six platelet-associated mediators were upregulated in plasma of patients with TB compared with control subjects, with concentrations returning to baseline by Day 60 of treatment. Gene expression of the monocyte collagenase MMP-1 (matrix metalloproteinase-1) was upregulated by platelets in M.tb infection. Platelets also enhanced M.tb-induced MMP-1 and -10 secretion, which drove type I collagen degradation. Platelets increased monocyte IL-1 and IL-10 and decreased IL-12 and MDC (monocyte-derived chemokine; also known as CCL-22) secretion, as consistent with an M2 monocyte phenotype. Monocyte killing of intracellular M.tb was decreased. In the lung, platelets were detected in a TB mouse model, and secreted platelet mediators were upregulated in human BAL fluid and correlated with MMP and IL-1β concentrations. CONCLUSIONS Platelets drive a proinflammatory, tissue-degrading phenotype in TB.
Collapse
Affiliation(s)
- Katharine A Fox
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Daniela E Kirwan
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Ashley M Whittington
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Nitya Krishnan
- 2 Medical Research Council Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, United Kingdom
| | - Brian D Robertson
- 2 Medical Research Council Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, United Kingdom
| | - Robert H Gilman
- 3 Department of International Health, Johns Hopkins University, Baltimore Maryland.,4 Laboratorio de Investigación en Enfermedades Infecciosas, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - José W López
- 5 Laboratorio de Bioinformática y Biología Molecular, Universidad Peruana Cayetano Heredia, Lima, Peru.,6 Instituto Nacional de Salud del Niño, Lima, Peru; and
| | - Shivani Singh
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Joanna C Porter
- 7 Centre for Inflammation and Tissue Repair, Respiratory Medicine, University College London, United Kingdom
| | - Jon S Friedland
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| |
Collapse
|
47
|
Gao X, Wu C, He W, Wang X, Li Y, Wang Y, Jia Y, Yuan R, Li H, Zhang B. DosR antigen Rv1737c induces activation of macrophages dependent on the TLR2 pathway. Cell Immunol 2019; 344:103947. [PMID: 31326120 DOI: 10.1016/j.cellimm.2019.103947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 01/22/2023]
Abstract
Latent Mycobacterium tuberculosis (Mtb) infection (LTBI) is the main clinical manifestation after Mtb exposure. During the latent phase, Mtb retards the attempts of eradication by the host immune system. The dormancy survival regulator (DosR) is held as essential for Mtb persistence. Rv1737c is predominantly expressed by the Mtb in latent infection. However, the role of Rv1737c in the immune evasion is still largely unknown. In this study, we have characterized the Rv1737c functions in the recruitment and activation of macrophages, which play a cardinal role in the innate and adaptive immunity. For the first time, we have revealed that Rv1737c induced the tolerogenic phenotype of macrophages by upregulating the expression of indoleamine 2,3-dioxygenase 1 (IDO1). Rv1737c-activated macrophages upregulated interleukin (IL)-4, IL-10, and Foxp3 T cells proliferation in vitro. Furthermore, the interaction of Rv1737c with macrophages was found to depend on the Toll-like receptor 2 (TLR2) pathway. It augmented nuclear factor κB (NF-κB) phosphorylation and co-stimulatory molecule expression. Thus, this study provides a crucial insight into a strategy adopted by Mtb to survive in the host by inducing tolerogenic macrophage expansion.
Collapse
Affiliation(s)
- Xiaoling Gao
- The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China.
| | - Cong Wu
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Wenhua He
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiaoxia Wang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Yonghong Li
- The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Yongxiang Wang
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Yanjuan Jia
- The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, China
| | - Rui Yuan
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Haojie Li
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Benzhong Zhang
- School of Public Health, Lanzhou University, Lanzhou, China.
| |
Collapse
|
48
|
Fertan E, Rodrigues GJ, Wheeler RV, Goguen D, Wong AA, James H, Stadnyk A, Brown RE, Weaver IC. Cognitive Decline, Cerebral-Spleen Tryptophan Metabolism, Oxidative Stress, Cytokine Production, and Regulation of the Txnip Gene in a Triple Transgenic Mouse Model of Alzheimer Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1435-1450. [DOI: 10.1016/j.ajpath.2019.03.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 03/01/2019] [Accepted: 03/07/2019] [Indexed: 02/06/2023]
|
49
|
Cervantes JL, Oak E, Garcia J, Liu H, Lorenzini PA, Batra D, Chhabra A, Salazar JC, Roca X. Vitamin D modulates human macrophage response to Mycobacterium tuberculosis DNA. Tuberculosis (Edinb) 2019; 116S:S131-S137. [PMID: 31085128 PMCID: PMC6626683 DOI: 10.1016/j.tube.2019.04.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is a facultative intracellular pathogen that infects macrophages where it avoids elimination by interfering with host defense mechanisms, including phago-lysosome fusion. Endosomal Toll-like receptors (TLRs) generate Type I Interferons (IFNs), which are associated with active tuberculosis (TB). We aimed to explore if DNA from different Mtb lineages lead to differences in the inflammatory response of human monocytic/macrophage cells. THP-1 cells which express two inducible reporter constructs for interferons (IFNs) as well as for NF-κB, were stimulated via endosomal delivery of Mtb DNA as a nanocomplex with PEI. DNA from different Mtb phylogenetic lineages elicited differential inflammatory responses in human macrophages. An initial relatively weak IRF-mediated response to DNA from HN878 and H37Rv increased if the cells were pre-treated with Vitamin D (Vit D) for 72 h. RNAseq of THP-1 under different transformation conditions showed that pre-treatment with Vit D upregulated several TLR9 variants, as well as genes involved in inflammatory immune response to infection, immune cell activation, Type I IFN regulation, and regulation of inflammation. Vit D appears to be important in increasing low IRF responses to DNA from certain lineages of Mtb. Variations in the IRF-mediated response to DNA derived from different Mtb genotypes are potentially important in the pathogenesis of tuberculosis since Type I IFN responses are associated with active disease. The role of Vit D in these responses could also translate into future therapeutic approaches.
Collapse
Affiliation(s)
- Jorge L Cervantes
- Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX, USA.
| | - Esther Oak
- University of New England, College of Dental Medicine, Portland, ME, USA
| | - John Garcia
- University of Connecticut Health, School of Public Health, Farmington, CT, USA
| | - Hongfei Liu
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Paolo A Lorenzini
- School of Biological Sciences, Nanyang Technological University, Singapore; Nanyang Institute of Technology in Health and Medicine, Interdisciplinary Graduate School, Nanyang Technological University, Singapore
| | - Deepika Batra
- Stem Cell Institute, Amity University Haryana, Manesar, Gurugram, Haryana, India
| | - Arvind Chhabra
- Stem Cell Institute, Amity University Haryana, Manesar, Gurugram, Haryana, India
| | - Juan C Salazar
- University of Connecticut Health, Department of Pediatrics, Farmington, CT, USA; Connecticut Children's Medical Center, Hartford, CT, USA
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
50
|
Novakowski KE, Yap NVL, Yin C, Sakamoto K, Heit B, Golding GB, Bowdish DME. Human-Specific Mutations and Positively Selected Sites in MARCO Confer Functional Changes. Mol Biol Evol 2019; 35:440-450. [PMID: 29165618 DOI: 10.1093/molbev/msx298] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Macrophage Receptor with COllagenous structure (MARCO) is a class A scavenger receptor that binds, phagocytoses, and modifies inflammatory responses to bacterial pathogens. Multiple candidate gene approach studies have shown that polymorphisms in MARCO are associated with susceptibility or resistance to Mycobacterium tuberculosis infection, but how these variants alter function is not known. To complement candidate gene approach studies, we previously used phylogenetic analyses to identify a residue, glutamine 452 (Q452), within the ligand-binding Scavenger Receptor Cysteine Rich domain as undergoing positive selection in humans. Herein, we show that Q452 is found in Denisovans, Neanderthals, and extant humans, but all other nonprimate, terrestrial, and aquatic mammals possess an aspartic acid (D452) residue. Further analysis of hominoid sequences of MARCO identified an additional human-specific mutation, phenylalanine 282 (F282), within the collagenous domain. We show that residue 282 is polymorphic in humans, but only 17% of individuals (rs6761637) possess the ancestral serine residue at position 282. We show that rs6761637 is in linkage disequilibrium with MARCO polymorphisms that have been previously linked to susceptibility to pulmonary tuberculosis. To assess the functional importance of sites Q452 and F282 in humans, we cloned the ancestral residues and loss-of-function mutations and investigated the role of these residues in binding and internalizing polystyrene microspheres and Escherichia coli. Herein, we show that the residues at sites 452 and 282 enhance receptor function.
Collapse
Affiliation(s)
- Kyle E Novakowski
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Nicholas V L Yap
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Charles Yin
- Department of Microbiology and Immunology and The Centre for Human Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Kaori Sakamoto
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA
| | - Bryan Heit
- Department of Microbiology and Immunology and The Centre for Human Immunology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - G Brian Golding
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Dawn M E Bowdish
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|