1
|
Fournier C, Mercey-Ressejac M, Derangère V, Al Kadi A, Rageot D, Charrat C, Leroy A, Vollaire J, Josserand V, Escudé M, Escaich S, Ghiringhelli F, Decaens T, Navarro FP, Jouvin-Marche E, Marche PN. Nanostructured lipid carriers based mRNA vaccine leads to a T cell-inflamed tumour microenvironment favourable for improving PD-1/PD-L1 blocking therapy and long-term immunity in a cold tumour model. EBioMedicine 2025; 112:105543. [PMID: 39793480 PMCID: PMC11774803 DOI: 10.1016/j.ebiom.2024.105543] [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: 09/04/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND mRNA-based cancer vaccines show promise in triggering antitumour immune responses. To combine them with existing immunotherapies, the intratumoral immune microenvironment needs to be deeply characterised. Here, we test nanostructured lipid carriers (NLCs), the so-called Lipidots®, for delivering unmodified mRNA encoding Ovalbumin (OVA) antigen to elicit specific antitumour responses. METHODS We evaluated whether NLC OVA mRNA complexes activate dendritic cells (DCs) in vitro and identified the involved signalling pathways using specific inhibitors. We tested the anti-tumoral impact of Ova mRNA vaccine in B16-OVA and E.G7-OVA cold tumour-bearing C57Bl6 female mice as well as its synergy effect with an anti-PD-1 therapy by following the tumour growth and performing immunophenotyping of innate and adaptive immune cells. The intratumoral vaccine-related gene signature was assessed by RNA-sequencing. The immune memory response was assessed by re-challenging surviving treated mice with tumour cells. FINDINGS Our vaccine activates DCs in vitro through the TLR4/8 and ROS signalling pathways and induces specific T cell activation while exhibits significant preventive and therapeutic antitumour efficacy in vivo. A profound intratumoral remodelling of the innate and adaptive immunity in association with an increase in the gene expression of chemokines (Cxcl10, Cxcl11, Cxcl9) involved in CD8+ T cell attraction were observed in immunised mice. The combination of vaccine and anti-PD-1 therapy improves the rates of complete responses and memory immune responses compared to monotherapies. INTERPRETATION Lipidots® are effective platform for the development of vaccines against cancer based on mRNA delivery. Their combination with immune checkpoint blockers could counter tumour resistance and promote long-term antitumour immunity. FUNDING This work was supported by Inserm Transfert, la Région Auvergne Rhône Alpes, FINOVI, and the French Ministry of Higher Education, research and innovation (LipiVAC, COROL project, funding reference N° 2102992411).
Collapse
Affiliation(s)
- Carole Fournier
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France; Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France.
| | - Marion Mercey-Ressejac
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Valentin Derangère
- INSERM U1231, Equipe TIRECS, Dijon, 21000, France; Université de Bourgogne, Dijon, 21000, France; Centre de Lutte contre le Cancer Georges François Leclerc, Plateforme de Transfert en Biologie du Cancer, Dijon, 21000, France
| | - Amal Al Kadi
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - David Rageot
- INSERM U1231, Equipe TIRECS, Dijon, 21000, France; Université de Bourgogne, Dijon, 21000, France; Centre de Lutte contre le Cancer Georges François Leclerc, Plateforme de Transfert en Biologie du Cancer, Dijon, 21000, France
| | - Christine Charrat
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Alexis Leroy
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Julien Vollaire
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Véronique Josserand
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Marie Escudé
- CEA, LETI, Technologies for Healthcare and Biology Division, Microtechnologies for Living Systems Interactions Research Unit, Univ. Grenoble Alpes, Grenoble, F-38000, France
| | - Séverine Escaich
- CEA, LETI, Technologies for Healthcare and Biology Division, Microtechnologies for Living Systems Interactions Research Unit, Univ. Grenoble Alpes, Grenoble, F-38000, France
| | - François Ghiringhelli
- INSERM U1231, Equipe TIRECS, Dijon, 21000, France; Université de Bourgogne, Dijon, 21000, France; Centre de Lutte contre le Cancer Georges François Leclerc, Plateforme de Transfert en Biologie du Cancer, Dijon, 21000, France
| | - Thomas Decaens
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France; Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| | - Fabrice P Navarro
- CEA, LETI, Technologies for Healthcare and Biology Division, Microtechnologies for Living Systems Interactions Research Unit, Univ. Grenoble Alpes, Grenoble, F-38000, France
| | - Evelyne Jouvin-Marche
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Patrice N Marche
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| |
Collapse
|
2
|
Malamud M, Brown GD. The Dectin-1 and Dectin-2 clusters: C-type lectin receptors with fundamental roles in immunity. EMBO Rep 2024; 25:5239-5264. [PMID: 39482490 PMCID: PMC11624271 DOI: 10.1038/s44319-024-00296-2] [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: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/03/2024] Open
Abstract
The ability of myeloid cells to recognize and differentiate endogenous or exogenous ligands rely on the presence of different transmembrane protein receptors. C-type lectin receptors (CLRs), defined by the presence of a conserved structural motif called C-type lectin-like domain (CTLD), are a crucial family of receptors involved in this process, being able to recognize a diverse range of ligands from glycans to proteins or lipids and capable of initiating an immune response. The Dectin-1 and Dectin-2 clusters involve two groups of CLRs, with genes genomically linked within the natural killer cluster of genes in both humans and mice, and all characterized by the presence of a single extracellular CTLD. Fundamental immune cell functions such as antimicrobial effector mechanisms as well as internalization and presentation of antigens are induced and/or regulated through activatory, or inhibitory signalling pathways triggered by these receptors after ligand binding. In this review, we will discuss the most recent concepts regarding expression, ligands, signaling pathways and functions of each member of the Dectin clusters of CLRs, highlighting the importance and diversity of their functions.
Collapse
Affiliation(s)
- Mariano Malamud
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Gordon D Brown
- Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, UK.
| |
Collapse
|
3
|
Xie D, Han C, Chen C, Liao Z, Campos de Souza S, Niu Y, Mano JF, Dong L, Wang C. A scaffold vaccine to promote tumor antigen cross-presentation via sustained toll-like receptor-2 (TLR2) activation. Bioact Mater 2024; 37:315-330. [PMID: 38694764 PMCID: PMC11061615 DOI: 10.1016/j.bioactmat.2024.03.035] [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: 01/07/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 05/04/2024] Open
Abstract
Cancer vaccination holds great promise for cancer treatment, but its effectiveness is hindered by suboptimal activation of CD8+ cytotoxic T lymphocytes, which are potent effectors to mediate anti-tumor immune responses. A possible solution is to switch antigen-presenting cells to present tumor antigens via the major histocompatibility complex class I (MHC-I) to CD8+ T cells - a process known as cross-presentation. To achieve this goal, we develop a three-dimensional (3D) scaffold vaccine to promote antigen cross-presentation by persisted toll-like receptor-2 (TLR2) activation after one injection. This vaccine comprises polysaccharide frameworks that "hook" TLR2 agonist (acGM) via tunable hydrophobic interactions and forms a 3D macroporous scaffold via click chemistry upon subcutaneous injection. Its retention-and-release of acGM enables sustained TLR2 activation in abundantly recruited dendritic cells in situ, inducing intracellular production of reactive oxygen species (ROS) in optimal kinetics that crucially promotes efficient antigen cross-presentation. The scaffold loaded with model antigen ovalbumin (OVA) or tumor specific antigen can generate potent immune responses against lung metastasis in B16-OVA-innoculated wild-type mice or spontaneous colorectal cancer in transgenic ApcMin/+ mice, respectively. Notably, it requires neither additional adjuvants nor external stimulation to function and can be adjusted to accommodate different antigens. The developed scaffold vaccine may represent a new, competent tool for next-generation personalized cancer vaccination.
Collapse
Affiliation(s)
- Daping Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Congwei Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Chonghao Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Zhencheng Liao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Senio Campos de Souza
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210093, China
- National Resource Center for Mutant Mice, Nanjing, 210093, China
- Chemistry and Biomedicine Innovative Center, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
- Department of Pharmaceutical Sciences, Faculty of Health Science, University of Macau, Taipa, Macau SAR, China
- Zhuhai UM Science and Technology Research Institute (ZUMRI), University of Macau, Hengqin, China
| |
Collapse
|
4
|
Chu GE, Park JY, Park CH, Cho WG. Mitochondrial Reactive Oxygen Species in TRIF-Dependent Toll-like Receptor 3 Signaling in Bronchial Epithelial Cells against Viral Infection. Int J Mol Sci 2023; 25:226. [PMID: 38203397 PMCID: PMC10778811 DOI: 10.3390/ijms25010226] [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: 11/12/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Toll-like receptor 3 (TLR3) plays an important role in double-stranded RNA recognition and triggers the innate immune response by acting as a key receptor against viral infections. Intracellular reactive oxygen species (ROS) are involved in TLR3-induced inflammatory responses during viral infections; however, their relationship with mitochondrial ROS (mtROS) remains largely unknown. In this study, we show that polyinosinic-polycytidylic acid (poly(I:C)), a mimic of viral RNA, induced TLR3-mediated nuclear factor-kappa B (NF-κB) signaling pathway activation and enhanced mtROS generation, leading to inflammatory cytokine production. TLR3-targeted small interfering RNA (siRNA) and Mito-TEMPO inhibited inflammatory cytokine production in poly(I:C)-treated BEAS-2B cells. Poly(I:C) recruited the TLR3 adaptor molecule Toll/IL-1R domain-containing adaptor, inducing IFN (TRIF) and activated NF-κB signaling. Additionally, TLR3-induced mtROS generation suppression and siRNA-mediated TRIF downregulation attenuated mitochondrial antiviral signaling protein (MAVS) degradation. Our findings provide insights into the TLR3-TRIF signaling pathway and MAVS in viral infections, and suggest TLR3-mtROS as a therapeutic target for the treatment of airway inflammatory and viral infectious diseases.
Collapse
Affiliation(s)
- Ga Eul Chu
- Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Republic of Korea; (G.E.C.); (C.H.P.)
| | - Jun Young Park
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea;
| | - Chan Ho Park
- Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Republic of Korea; (G.E.C.); (C.H.P.)
| | - Won Gil Cho
- Department of Anatomy, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Republic of Korea; (G.E.C.); (C.H.P.)
| |
Collapse
|
5
|
Zihad SNK, Sifat N, Islam MA, Monjur-Al-Hossain A, Sikdar KYK, Sarker MMR, Shilpi JA, Uddin SJ. Role of pattern recognition receptors in sensing Mycobacterium tuberculosis. Heliyon 2023; 9:e20636. [PMID: 37842564 PMCID: PMC10570006 DOI: 10.1016/j.heliyon.2023.e20636] [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: 10/22/2022] [Revised: 09/06/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
Mycobacterium tuberculosis is one of the major invasive intracellular pathogens causing most deaths by a single infectious agent. The interaction between host immune cells and this pathogen is the focal point of the disease, Tuberculosis. Host immune cells not only mount the protective action against this pathogen but also serve as the primary niche for growth. Thus, recognition of this pathogen by host immune cells and following signaling cascades are key dictators of the disease state. Immune cells, mainly belonging to myeloid cell lineage, recognize a wide variety of Mycobacterium tuberculosis ligands ranging from carbohydrate and lipids to proteins to nucleic acids by different membrane-bound and soluble pattern recognition receptors. Simultaneous interaction between different host receptors and pathogen ligands leads to immune-inflammatory response as well as contributes to virulence. This review summarizes the contribution of pattern recognition receptors of host immune cells in recognizing Mycobacterium tuberculosis and subsequent initiation of signaling pathways to provide the molecular insight of the specific Mtb ligands interacting with specific PRR, key adaptor molecules of the downstream signaling pathways and the resultant effector functions which will aid in identifying novel drug targets, and developing novel drugs and adjuvants.
Collapse
Affiliation(s)
| | - Nazifa Sifat
- Department of Pharmacy, ASA University of Bangladesh, Dhaka, 1207, Bangladesh
| | | | | | | | - Md Moklesur Rahman Sarker
- Department of Pharmacy, State University of Bangladesh, Dhaka, 1205, Bangladesh
- Department of Pharmacy, Gono University, Nolam, Mirzanagar, Savar, Dhaka 1344, Bangladesh
| | - Jamil A. Shilpi
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | - Shaikh Jamal Uddin
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| |
Collapse
|
6
|
Bungau AF, Radu AF, Bungau SG, Vesa CM, Tit DM, Endres LM. Oxidative stress and metabolic syndrome in acne vulgaris: Pathogenetic connections and potential role of dietary supplements and phytochemicals. Biomed Pharmacother 2023; 164:115003. [PMID: 37315434 DOI: 10.1016/j.biopha.2023.115003] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/16/2023] Open
Abstract
Acne vulgaris is a highly prevalent skin condition caused by androgen-induced elevated sebum secretion, abnormal keratinization, bacterial colonization, and inflammation. Current research indicates a link between acne vulgaris and the metabolic syndrome, a group of disorders that includes obesity, insulin resistance, hypertension, and dyslipidemia. This link is thought to be modulated by excessive concentrations of oxidative stress markers and chronic inflammation, which are included in the pathophysiological mechanisms shared by both conditions. Excessive generation of reactive oxygen species damages cellular components and initiates an inflammatory response, hence promoting the development of both disorders. The current narrative review focuses on the molecular implications of inflammatory, hormonal, and environmental factors in the acne-metabolic syndrome correlation. Furthermore, it outlines the current state of knowledge related to the phyto-therapeutic approach to these conditions as an adjuvant strategy to allopathic treatment, but future multicenter and larger-scale research studies are needed establish new algorithms to be included in the future management of patients with these conditions.
Collapse
Affiliation(s)
- Alexa Florina Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| | - Andrei Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania.
| | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania.
| | - Cosmin Mihai Vesa
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Delia Mirela Tit
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Laura Maria Endres
- Department of Psycho-Neurosciences and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
7
|
Ayodele S, Kumar P, van Eyk A, Choonara YE. Advances in immunomodulatory strategies for host-directed therapies in combating tuberculosis. Biomed Pharmacother 2023; 162:114588. [PMID: 36989709 DOI: 10.1016/j.biopha.2023.114588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/14/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
Tuberculosis (TB) maintains its infamous status regarding its detrimental effect on global health, causing the highest mortality by a single infectious agent. The presence of resistance and immune compromising disease favours the disease in maintaining its footing in the health care burden despite various anti-TB drugs used to fight it. Main factors contributing to resistance and difficulty in treating disease include prolonged treatment duration (at least 6 months) and severe toxicity, which further leads to patient non-compliance, and thus a ripple effect leading to therapeutic non-efficacy. The efficacy of new regimens demonstrates that targeting host factors concomitantly with the Mycobacterium tuberculosis (M.tb) strain is urgently required. Due to the huge expenses and time required of up to 20 years for new drug research and development, drug repurposing may be the most economical, circumspective, and conveniently faster journey to embark on. Host-directed therapy (HDT) will dampen the burden of the disease by acting as an immunomodulator, allowing it to defend the body against antibiotic-resistant pathogens whilst minimizing the possibility of developing new resistance to susceptible drugs. Repurposed drugs in TB act as host-directed therapies, acclimatizing the host immune cell to the presence of TB, improving its antimicrobial activity and time taken to get rid of the disease, whilst minimizing inflammation and tissue damage. In this review, we, therefore, explore possible immunomodulatory targets, HDT immunomodulatory agents, and their ability to improve clinical outcomes whilst minimizing the risk of drug resistance, through various pathway targeting and treatment duration reduction.
Collapse
|
8
|
Morris G, Gevezova M, Sarafian V, Maes M. Redox regulation of the immune response. Cell Mol Immunol 2022; 19:1079-1101. [PMID: 36056148 PMCID: PMC9508259 DOI: 10.1038/s41423-022-00902-0] [Citation(s) in RCA: 206] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/29/2022] [Indexed: 12/20/2022] Open
Abstract
AbstractThe immune-inflammatory response is associated with increased nitro-oxidative stress. The aim of this mechanistic review is to examine: (a) the role of redox-sensitive transcription factors and enzymes, ROS/RNS production, and the activity of cellular antioxidants in the activation and performance of macrophages, dendritic cells, neutrophils, T-cells, B-cells, and natural killer cells; (b) the involvement of high-density lipoprotein (HDL), apolipoprotein A1 (ApoA1), paraoxonase-1 (PON1), and oxidized phospholipids in regulating the immune response; and (c) the detrimental effects of hypernitrosylation and chronic nitro-oxidative stress on the immune response. The redox changes during immune-inflammatory responses are orchestrated by the actions of nuclear factor-κB, HIF1α, the mechanistic target of rapamycin, the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, mitogen-activated protein kinases, 5' AMP-activated protein kinase, and peroxisome proliferator-activated receptor. The performance and survival of individual immune cells is under redox control and depends on intracellular and extracellular levels of ROS/RNS. They are heavily influenced by cellular antioxidants including the glutathione and thioredoxin systems, nuclear factor erythroid 2-related factor 2, and the HDL/ApoA1/PON1 complex. Chronic nitro-oxidative stress and hypernitrosylation inhibit the activity of those antioxidant systems, the tricarboxylic acid cycle, mitochondrial functions, and the metabolism of immune cells. In conclusion, redox-associated mechanisms modulate metabolic reprogramming of immune cells, macrophage and T helper cell polarization, phagocytosis, production of pro- versus anti-inflammatory cytokines, immune training and tolerance, chemotaxis, pathogen sensing, antiviral and antibacterial effects, Toll-like receptor activity, and endotoxin tolerance.
Collapse
|
9
|
Li W, Deng W, Zhang N, Peng H, Xu Y. Mycobacterium tuberculosis Rv2387 Facilitates Mycobacterial Survival by Silencing TLR2/p38/JNK Signaling. Pathogens 2022; 11:pathogens11090981. [PMID: 36145413 PMCID: PMC9504853 DOI: 10.3390/pathogens11090981] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) can evade antimicrobial immunity and persist within macrophages by interfering with multiple host cellular functions through its virulence factors, causing latent tuberculosis. The Rv2387 protein has been identified as a putative effector that potentially participates in Mtb pathogenicity. To explore the role of the Rv2387 protein in host–mycobacteria interactions, we established recombinant M. smegmatis strains and RAW264.7 cell lines that stably express the Rv2387 protein. We found that this protein suppresses mycobacteria infection-induced macrophage apoptosis by inactivating caspase-3/-8, thus facilitating the intracellular survival of mycobacteria. In addition, Rv2387 inhibits the production of inflammatory cytokines in macrophages by specifically suppressing TLR2-dependent stimulation of p38 and JNK MAPK pathways. Moreover, we further determined that the Rv2387 protein conferred a growth advantage over recombinant M. smegmatis and suppressed the inflammatory response in a mouse infection model. Overall, these data suggested that Rv2387 facilitates mycobacteria to escape host immunity and might be an essential virulence factor in Mtb.
Collapse
Affiliation(s)
- Wu Li
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
- Correspondence: (W.L.); (Y.X.)
| | - Wanyan Deng
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nan Zhang
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
| | - Huijuan Peng
- Key Laboratory of Regional Characteristic Agricultural Resources, College of Life Sciences, Neijiang Normal University, Neijiang 641100, China
| | - Yi Xu
- The Joint Center for Infection and Immunity, Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
- Correspondence: (W.L.); (Y.X.)
| |
Collapse
|
10
|
Li M, Vultorius C, Bethi M, Yu Y. Spatial Organization of Dectin-1 and TLR2 during Synergistic Crosstalk Revealed by Super-resolution Imaging. J Phys Chem B 2022; 126:5781-5792. [PMID: 35913832 PMCID: PMC10636754 DOI: 10.1021/acs.jpcb.2c03557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Innate immune cells recognize and elicit responses against pathogens by integrating signals from different types of cell-surface receptors. How the receptors interact in the membrane to enable their signaling crosstalk is poorly understood. Here, we reveal the nanoscale organization of TLR2 and Dectin-1, a receptor pair known to cooperate in regulating antifungal immunity, through their synergistic signaling crosstalk at macrophage cell membranes. Using super-resolution single-molecule localization microscopy, we show that discrete noncolocalized nanoclusters of Dectin-1 and TLR2 are partially overlapped during their synergistic crosstalk. Compared to when one type of receptor is activated alone, the simultaneous activation of Dectin-1 and TLR2 leads to a higher percentage of both receptors being activated by their specific ligands and consequently an increased level of tyrosine phosphorylation. Our results depict, in nanoscale detail, how Dectin-1 and TLR2 achieve synergistic signaling through the spatial organization of their receptor nanoclusters.
Collapse
Affiliation(s)
- Miao Li
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Christopher Vultorius
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Manisha Bethi
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
11
|
“The Good, the Bad and the Ugly”: Interplay of Innate Immunity and Inflammation. Cell Microbiol 2022. [DOI: 10.1155/2022/2759513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Innate immunity recognizes microorganisms through certain invariant receptors named pattern recognition receptors (PRRs) by sensing conserved pathogen-associated molecular patterns (PAMPs). Their recognition activates several signaling pathways that lead the transcription of inflammatory mediators, contributing to trigger a very rapid inflammatory cascade aiming to contain the local infection as well as activating and instructing the adaptive immunity in a specific and synchronized immune response according to the microorganism. Inflammation is a coordinated process involving the secretion of cytokines and chemokines by macrophages and neutrophils leading to the migration of other leukocytes along the endothelium into the injured tissue. Sustained inflammatory responses can cause deleterious effects by promoting the development of autoimmune disorders, allergies, cancer, and other immune pathologies, while weak signals could exacerbate the severity of the disease. Therefore, PRR-mediated signal transduction must be tightly regulated to maintain host immune homeostasis. Innate immunity deficiencies and strategies deployed by microbes to avoid inflammatory responses lead to an altered immune response that allows the pathogen to proliferate causing death or uncontrolled inflammation. This review analyzes the complexity of the immune response at the beginning of the disease focusing on COVID-19 disease and the importance of unraveling its mechanisms to be considered when treating diseases and designing vaccines.
Collapse
|
12
|
Eld HMS, Johnsen PR, Nielsen EM, Jørgensen FZ, Lindstrøm-Svendsen M, Baldry M, Ingmer H, Frøkiær H. Soluble C-Type Lectin-Receptor Ligands Stimulate ROS Production in Dendritic Cells and Potentiate Killing of MRSA as Well as the MRSA Induced IL-12 Production. Front Immunol 2022; 13:845881. [PMID: 35386713 PMCID: PMC8977849 DOI: 10.3389/fimmu.2022.845881] [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: 12/30/2021] [Accepted: 02/24/2022] [Indexed: 12/04/2022] Open
Abstract
Methicillin resistant Staphylococcus aureus (MRSA) has developed resistance to most β-lactam antibiotics leaving few treatment options against infections with MRSA. Through mannose receptors, mannan potentiates IL-12 production induced by Gram-positive bacteria, a cytokine crucial in the clearance of S. aureus infection. We investigated the IL-12 potentiating effect of mannan pre-treatment of bone marrow-derived dendritic cells prior to stimulation with clinical MRSA strains. Mannan almost doubled IL-12 as well as IFN-β production in response to USA300, also when USA300 was treated with the β-lactam cefoxitin. The MRSA-induced IL-12 production was dependent on bacterial uptake and reactive oxygen species (ROS). Mannan alone induced ROS production, and in combination with USA300, the ROS produced corresponded to the sum induced by mannan and USA300. Addition of a monoclonal antibody against the mannose receptor likewise enhanced USA300-induced IL-12 and induced ROS production. Mannan addition further increased the endocytosis as well as the rate of endosomal killing of bacteria. Pre-treatment with soluble β-glucans also induced ROS and potentiated the USA300-induced IL-12 indicating that other C-type receptors may play a similar role. In the presence of the pro-inflammatory mediators, GM-CSF or IFN-γ, the mannan-enhanced IL-12 production increased further. The USA300-induced and the mannan-facilitated enhanced IFN-β and IL-12 showed same dependency on MAPK c-Jun N-terminal kinase signaling, suggesting that mannan enhances the signals already induced by the bacteria, rather than changing them. We suggest that the C-type lectin-induced ROS production is a key factor in the IFN-β and IL-12 potentiation.
Collapse
Affiliation(s)
- Helene M S Eld
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Peter R Johnsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Emilie M Nielsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Frederikke Z Jørgensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Mara Baldry
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Hanne Ingmer
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Hanne Frøkiær
- Department of Veterinary and Animal Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
13
|
Yokobori N, López B, Ritacco V. The host-pathogen-environment triad: Lessons learned through the study of the multidrug-resistant Mycobacterium tuberculosis M strain. Tuberculosis (Edinb) 2022; 134:102200. [PMID: 35339874 DOI: 10.1016/j.tube.2022.102200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/13/2022] [Accepted: 03/18/2022] [Indexed: 10/18/2022]
Abstract
Multidrug-resistant tuberculosis is one of the major obstacles that face the tuberculosis eradication efforts. Drug-resistant Mycobacterium tuberculosis clones were initially disregarded as a public health threat, because they were assumed to have paid a high fitness cost in exchange of resistance acquisition. However, some genotypes manage to overcome the impact of drug-resistance conferring mutations, retain transmissibility and cause large outbreaks. In Argentina, the HIV-AIDS epidemics fuelled the expansion of the so-called M strain in the early 1990s, which is responsible for the largest recorded multidrug-resistant tuberculosis cluster of Latin America. The aim of this work is to review the knowledge gathered after nearly three decades of multidisciplinary research on epidemiological, microbiological and immunological aspects of this highly successful strain. Collectively, our results indicate that the successful transmission of the M strain could be ascribed to its unaltered virulence, low Th1/Th17 response, a low fitness cost imposed by the resistance conferring mutations and a high resistance to host-related stress. In the early 2000s, the incident cases due to the M strain steadily declined and stabilized in the latest years. Improvements in the management, diagnosis and treatment of multidrug-resistant tuberculosis along with societal factors such as the low domestic and international mobility of the patients affected by this strain probably contributed to the outbreak containment. This stresses the importance of sustaining the public health interventions to avoid the resurgence of this conspicuous multidrug-resistant strain.
Collapse
Affiliation(s)
- Noemí Yokobori
- Servicio de Micobacterias, Instituto Nacional de Enfermedades Infecciosas, ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina.
| | - Beatriz López
- Departamento de Bacteriología, Instituto Nacional de Enfermedades Infecciosas, ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina.
| | - Viviana Ritacco
- Servicio de Micobacterias, Instituto Nacional de Enfermedades Infecciosas, ANLIS "Dr. C. G. Malbrán", Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina.
| |
Collapse
|
14
|
Liang W, Meng X, Zhen Y, Zhang Y, Hu X, Zhang Q, Zhou X, Liu B. Integration of Transcriptome and Proteome in Lymph Nodes Reveal the Different Immune Responses to PRRSV Between PRRSV-Resistant Tongcheng Pigs and PRRSV-Susceptible Large White Pigs. Front Genet 2022; 13:800178. [PMID: 35154273 PMCID: PMC8829461 DOI: 10.3389/fgene.2022.800178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is an infectious disease that seriously affects the swine industry worldwide. Understanding the interaction between the host immune response and PRRS virus (PRRSV) can provide insight into the PRRSV pathogenesis, as well as potential clues to control PRRSV infection. Here, we examined the transcriptome and proteome differences of lymph nodes between PRRSV-resistant Tongcheng (TC) pigs and PRRSV-susceptible Large White (LW) pigs in response to PRRSV infection. 2245 and 1839 differentially expressed genes (DEGs) were detected in TC and LW pigs upon PRRSV infection, respectively. Transcriptome analysis revealed genetic differences in antigen presentation and metabolism between TC pigs and LW pigs, which may lead to different immune responses to PRRSV infection. Furthermore, 678 and 1000 differentially expressed proteins (DEPs) were identified in TC and LW pigs, and DEPs were mainly enriched in the metabolism pathways. Integrated analysis of transcriptome and proteome datasets revealed antigen recognition capacity, immune activation, cell cycles, and cell metabolism are important for PRRSV clearance. In conclusion, this study provides important resources on transcriptomic and proteomic levels in lymph nodes for further revealing the interaction between the host immune response and PRRSV, which would give us new insight into molecular mechanisms related to genetic complexity against PRRSV.
Collapse
Affiliation(s)
- Wan Liang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Prevention and Control Agents for Animal Bacteriosis (Ministry of Agriculture), Animal Husbandry and Veterinary Institute, Hubei Academy of Agricultural Science, Wuhan, China
| | - Xiangge Meng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yueran Zhen
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yu Zhang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xueying Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qingde Zhang
- Laboratory Animal Center, College of Animal Science and Technology and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiang Zhou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bang Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
15
|
Yu C, Chen H, Du D, Lv W, Li S, Li D, Xu Z, Gao M, Hu H, Liu D. β-Glucan from Saccharomyces cerevisiae alleviates oxidative stress in LPS-stimulated RAW264.7 cells via Dectin-1/Nrf2/HO-1 signaling pathway. Cell Stress Chaperones 2021; 26:629-637. [PMID: 33880723 PMCID: PMC8275741 DOI: 10.1007/s12192-021-01205-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
β-Glucan from Saccharomyces cerevisiae has been described to be effective antioxidants, but the specific antioxidation mechanism of β-glucan is unclear. The objectives of this research were to determine whether the β-glucan from Saccharomyces cerevisiae could regulate oxidative stress through the Dectin-1/Nrf2/HO-1 signaling pathway in lipopolysaccharides (LPS)-stimulated RAW264.7 cells. In this study, we examined the effects of β-glucan on the enzyme activity or production of oxidative stress indicators in LPS-stimulated RAW264.7 cells by biochemical analysis and the protein expression of key factors of Dectin-1/Nrf2/HO-1 signaling pathway by immunofluorescence and western blot. The biochemical analysis results showed that β-glucan increased the LPS-induced downregulation of enzyme activity of intracellular heme oxygenase (HO), superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-Px) while decreasing the production of reactive oxygen species (ROS) and malondialdehyde (MDA). Furthermore, immunofluorescence results showed that β-glucan can activate the nuclear factor erythroid 2-related factor 2 (Nrf2). The antioxidant mechanism study indicated that β-glucan activated dendritic-cell-associated C-type lectin 1 (Dectin-1) receptors mediated Nrf2/HO-1 signaling pathway, thereby downregulating the production of ROS and thus produced the antioxidant effects in LPS-stimulated RAW 264.7 cells. In conclusion, these results indicate that β-glucan potently alleviated oxidative stress via Dectin-1/Nrf2/HO-1 in LPS-stimulated RAW 264.7 cells.
Collapse
Affiliation(s)
- Chunwei Yu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Hui Chen
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Donghua Du
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Wenting Lv
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Songjian Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Dongfang Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Zixuan Xu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Min Gao
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010018, China
| | - Honglian Hu
- Inner Mongolia Academy of Agricultural and Animal Husbandry Sciences, Hohhot, 010018, China
| | - Dacheng Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
16
|
Scherlinger M, Tsokos GC. Reactive oxygen species: The Yin and Yang in (auto-)immunity. Autoimmun Rev 2021; 20:102869. [PMID: 34118461 DOI: 10.1016/j.autrev.2021.102869] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/21/2021] [Indexed: 12/24/2022]
Abstract
Reactive oxygen species (ROS) are produced by immune cells in response to antigens. They are produced mostly in the mitochondria and their levels are tightly controlled by a series of metabolic processes. ROS are necessary for the development of the immune response but the role of ROS in the development of autoimmune disease needs further clarification. Early clinical information points to the beneficial role of supplementation of antioxidant agents or the reduction of ROS production. We review recent information in the field in an effort to identify areas more studies are needed.
Collapse
Affiliation(s)
- Marc Scherlinger
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares, Est/Sud-Ouest (RESO), France; Service de rhumatologie, Centre Hospitalier Universitaire de Strasbourg, 1 avenue Molière, 67098 Strasbourg, France.
| | - George C Tsokos
- Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Innate Immune Pattern Recognition Receptors of Mycobacterium tuberculosis: Nature and Consequences for Pathogenesis of Tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1313:179-215. [PMID: 34661896 DOI: 10.1007/978-3-030-67452-6_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Innate immunity against Mycobacterium tuberculosis is a critical early response to prevent the establishment of the infection. Despite recent advances in understanding the host-pathogen dialogue in the early stages of tuberculosis (TB), much has yet to be learnt. The nature and consequences of this dialogue ultimately determine the path of infection: namely, either early clearance of M. tuberculosis, or establishment of M. tuberculosis infection leading to active TB disease and/or latent TB infection. On the frontline in innate immunity are pattern recognition receptors (PRRs), with soluble factors (e.g. collectins and complement) and cell surface factors (e.g. Toll-like receptors and other C-type lectin receptors (Dectin 1/2, Nod-like receptors, DC-SIGN, Mincle, mannose receptor, and MCL) that play a central role in recognising M. tuberculosis and facilitating its clearance. However, in a 'double-edged sword' scenario, these factors can also be involved in enhancement of pathogenesis as well. Furthermore, innate immunity is also a critical bridge in establishing the subsequent adaptive immune response, which is also responsible for granuloma formation that cordons off M. tuberculosis infection, establishing latency and acting as a reservoir for bacterial persistence and dissemination of future disease. This chapter discusses the current understanding of pattern recognition of M. tuberculosis by innate immunity and the role this plays in the pathogenesis and protection against TB.
Collapse
|
18
|
Rodrigues TS, Conti BJ, Fraga-Silva TFDC, Almeida F, Bonato VLD. Interplay between alveolar epithelial and dendritic cells and Mycobacterium tuberculosis. J Leukoc Biol 2020; 108:1139-1156. [PMID: 32620048 DOI: 10.1002/jlb.4mr0520-112r] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/09/2020] [Accepted: 05/25/2020] [Indexed: 12/20/2022] Open
Abstract
The innate response plays a crucial role in the protection against tuberculosis development. Moreover, the initial steps that drive the host-pathogen interaction following Mycobacterium tuberculosis infection are critical for the development of adaptive immune response. As alveolar Mϕs, airway epithelial cells, and dendritic cells can sense the presence of M. tuberculosis and are the first infected cells. These cells secrete mediators, which generate inflammatory signals that drive the differentiation and activation of the T lymphocytes necessary to clear the infection. Throughout this review article, we addressed the interaction between epithelial cells and M. tuberculosis, as well as the interaction between dendritic cells and M. tuberculosis. The understanding of the mechanisms that modulate those interactions is critical to have a complete view of the onset of an infection and may be useful for the development of dendritic cell-based vaccine or immunotherapies.
Collapse
Affiliation(s)
- Tamara Silva Rodrigues
- Basic and Applied Immunology Program, Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Bruno José Conti
- Basic and Applied Immunology Program, Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Thais Fernanda de Campos Fraga-Silva
- Basic and Applied Immunology Program, Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Fausto Almeida
- Basic and Applied Immunology Program, Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Vânia Luiza Deperon Bonato
- Basic and Applied Immunology Program, Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
19
|
Braganza A, Annarapu GK, Shiva S. Blood-based bioenergetics: An emerging translational and clinical tool. Mol Aspects Med 2020; 71:100835. [PMID: 31864667 PMCID: PMC7031032 DOI: 10.1016/j.mam.2019.100835] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/27/2019] [Accepted: 12/11/2019] [Indexed: 12/16/2022]
Abstract
Accumulating studies demonstrate that mitochondrial genetics and function are central to determining the susceptibility to, and prognosis of numerous diseases across all organ systems. Despite this recognition, mitochondrial function remains poorly characterized in humans primarily due to the invasiveness of obtaining viable tissue for mitochondrial studies. Recent studies have begun to test the hypothesis that circulating blood cells, which can be obtained by minimally invasive methodology, can be utilized as a biomarker of systemic bioenergetic function in human populations. Here we present the available methodologies for assessing blood cell bioenergetics and review studies that have applied these techniques to healthy and disease populations. We focus on the validation of this methodology in healthy subjects, as well as studies testing whether blood cell bioenergetics are altered in disease, correlate with clinical parameters, and compare with other methodology for assessing human mitochondrial function. Finally, we present the challenges and goals for the development of this emerging approach into a tool for translational research and personalized medicine.
Collapse
Affiliation(s)
- Andrea Braganza
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, Pittsburgh, PA, USA
| | - Gowtham K Annarapu
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, Pittsburgh, PA, USA
| | - Sruti Shiva
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, Pittsburgh, PA, USA; Department of Pharmacology & Chemical Biology, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine (C3M), University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
20
|
Thiem K, Hoeke G, Zhou E, Hijmans A, Houben T, Boels MG, Mol IM, Lutgens E, Shiri-Sverdlov R, Bussink J, Kanneganti TD, Boon MR, Stienstra R, Tack CJ, Rensen PCN, Netea MG, Berbée JFP, van Diepen JA. Deletion of haematopoietic Dectin-2 or CARD9 does not protect from atherosclerosis development under hyperglycaemic conditions. Diab Vasc Dis Res 2020; 17:1479164119892140. [PMID: 31868000 PMCID: PMC7510497 DOI: 10.1177/1479164119892140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND C-type lectin receptors, including Dectin-2, are pattern recognition receptors on monocytes and macrophages that mainly recognize sugars and sugar-like structures present on fungi. Activation of C-type lectin receptors induces downstream CARD9 signalling, leading to the production of cytokines. We hypothesized that under hyperglycaemic conditions, as is the case in diabetes mellitus, glycosylated protein (sugar-like) structures activate C-type lectin receptors, leading to immune cell activation and increased atherosclerosis development. METHODS Low-density lipoprotein receptor-deficient mice were lethally irradiated and transplanted with bone marrow from control wild-type, Dectin-2-/- or Card9-/- mice. After 6 weeks of recovery, mice received streptozotocin injections (50 mg/g BW; 5 days) to induce hyperglycaemia. After an additional 2 weeks, mice were fed a Western-type diet (0.1% cholesterol) for 10 weeks. RESULTS AND CONCLUSION Deletion of haematopoietic Dectin-2 reduced the number of circulating Ly6Chi monocytes, increased pro-inflammatory cytokine production, but did not affect atherosclerosis development. Deletion of haematopoietic CARD9 tended to reduce macrophage and collagen content in atherosclerotic lesions, again without influencing the lesion size. Deletion of haematopoietic Dectin-2 did not influence atherosclerosis development under hyperglycaemic conditions, despite some minor effects on inflammation. Deletion of haematopoietic CARD9 induced minor alterations in plaque composition under hyperglycaemic conditions, without affecting lesion size.
Collapse
MESH Headings
- Animals
- Antigens, Ly/metabolism
- Aorta/metabolism
- Aorta/pathology
- Aortic Diseases/etiology
- Aortic Diseases/genetics
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Atherosclerosis/etiology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Biomarkers/blood
- Blood Glucose/metabolism
- Bone Marrow Transplantation
- CARD Signaling Adaptor Proteins/deficiency
- CARD Signaling Adaptor Proteins/genetics
- Cells, Cultured
- Collagen/metabolism
- Cytokines/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diet, Western
- Gene Deletion
- Genetic Predisposition to Disease
- Hematopoietic Stem Cells/metabolism
- Lectins, C-Type/deficiency
- Lectins, C-Type/genetics
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/metabolism
- Monocytes/pathology
- Plaque, Atherosclerotic
- Receptors, LDL/deficiency
- Receptors, LDL/genetics
Collapse
Affiliation(s)
- Kathrin Thiem
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
- Kathrin Thiem, Department of Internal
Medicine and Radboud Institute for Molecular Life Sciences, Radboud University
Medical Center, 463, Geert Grooteplein zuid 8, 6525 GA Nijmegen, The
Netherlands.
| | - Geerte Hoeke
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Enchen Zhou
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Anneke Hijmans
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
| | - Tom Houben
- Departments of Molecular Genetics, Human
Biology and Surgery, School of Nutrition and Translational Research in Metabolism
(NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Margien G Boels
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Division of Nephrology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Isabel M Mol
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Esther Lutgens
- Division of Experimental Vascular
Biology, Department of Medical Biochemistry, Academic Medical Center, University of
Amsterdam, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention,
Ludwig Maximilians University of Munich, Munich, Germany
| | - Ronit Shiri-Sverdlov
- Departments of Molecular Genetics, Human
Biology and Surgery, School of Nutrition and Translational Research in Metabolism
(NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Johan Bussink
- Department of Radiation Oncology,
Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Mariëtte R Boon
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rinke Stienstra
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
- Division of Human Nutrition,
Wageningen University, Wageningen, The Netherlands
| | - Cees J Tack
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
| | - Patrick CN Rensen
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
- Department for Genomics and
Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn,
Bonn, Germany
| | - Jimmy FP Berbée
- Division of Endocrinology, Department of
Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental
Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Janna A van Diepen
- Department of Internal Medicine and
Radboud Institute for Molecular Life Sciences, Radboud University Medical Center,
Nijmegen, The Netherlands
| |
Collapse
|
21
|
Geometrical reorganization of Dectin-1 and TLR2 on single phagosomes alters their synergistic immune signaling. Proc Natl Acad Sci U S A 2019; 116:25106-25114. [PMID: 31754039 DOI: 10.1073/pnas.1909870116] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Receptors of innate immune cells function synergistically to detect pathogens and elicit appropriate immune responses. Many receptor pairs also appear "colocalized" on the membranes of phagosomes, the intracellular compartments for pathogen ingestion. However, the nature of the seemingly receptor colocalization and the role it plays in immune regulation are unclear, due to the inaccessibility of intracellular phagocytic receptors. Here, we report a geometric manipulation technique to directly probe the role of phagocytic receptor "colocalization" in innate immune regulation. Using particles with spatially patterned ligands as phagocytic targets, we can decouple the receptor pair, Dectin-1 and Toll-like receptor (TLR)2, to opposite sides on a single phagosome or bring them into nanoscale proximity without changing the overall membrane composition. We show that Dectin-1 enhances immune responses triggered predominantly by TLR2 when their centroid-to-centroid proximity is <500 nm, but this signaling synergy diminishes upon receptor segregation beyond this threshold distance. Our results demonstrate that nanoscale proximity, not necessarily colocalization, between Dectin-1 and TLR2 is required for their synergistic regulation of macrophage immune responses. This study elucidates the relationship between the spatial organization of phagocytic receptors and innate immune responses. It showcases a technique that allows spatial manipulation of receptors and their signal cross-talk on phagosomes inside living cells.
Collapse
|
22
|
Role of α-glucan-induced oxygen species in dendritic cells and its impact in immune response against tuberculosis. Int J Med Microbiol 2019; 309:151328. [PMID: 31324524 DOI: 10.1016/j.ijmm.2019.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/25/2019] [Accepted: 07/07/2019] [Indexed: 11/24/2022] Open
Abstract
With 10 million new cases and three million deaths estimated to occur yearly ̶ more than any time in history ̶ tuberculosis (TB) remains the single most widespread and deadly infectious disease. Until recently, it was thought that both latent and active TB was primarily related to host factors. Nonetheless, the participation of bacterial factors is becoming increasingly evident. Minimal variations in genes related to Mycobacterium tuberculosis (Mtb) virulence and pathogenesis can lead to marked differences in immunogenicity. Dendritic cells (DC) are professional antigen presenting cells whose maturation can vary depending on the cell wall composition of each particular Mtb strain being critical for the onset of the immune response against Mtb. Here we evaluated the role played by α-glucan, in the endogenous production of reactive oxygen species, ROS, and the impact on DC maturation and function. Results showed that α-glucans on Mtb induce ROS production leading to DC maturation and lymphocyte proliferation. Even more, α-glucans induced Syk activation but were not essential in non-opsonized phagocytosis. In summary, α-glucans of Mtb participates in ROS production and the subsequent DC maturation and antigen presentation, suggesting a relevant role of α-glucans for the onset of the protective immune response against TB.
Collapse
|
23
|
Kardeh S, Moein SA, Namazi MR, Kardeh B. Evidence for the Important Role of Oxidative Stress in the Pathogenesis of Acne. Galen Med J 2019; 8:e1291. [PMID: 34466486 PMCID: PMC8344136 DOI: 10.31661/gmj.v0i0.1291] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/01/2018] [Accepted: 08/18/2018] [Indexed: 02/05/2023] Open
Abstract
Acne vulgaris is a common inflammatory skin disorder which is recognizable by dermatological lesions and scars. In addition to some pathogenetic factors such as hyperkeratinization, upregulated sebum secretion, and immunoinflammatory reactions, recent studies have also connected oxidative stress to the pathogenesis of acne vulgaris. In this article, we will briefly review clinical studies that interrogated alterations in oxidative stress biomarkers by a systematic search conducted in PubMed, Web of Science, and Scopus using "acne", "oxidative stress", and "reactive oxygen species" keywords. Overall, studies have shown that oxidative biomarkers (e.g. lipid peroxidation final products) are higher in acne vulgaris lesions. A significant positive correlation has also been noted between acne severity and oxidative biomarkers. In contrast, diminished levels of antioxidant enzymes (e.g. superoxide dismutase and catalase) have been observed in acne. We propose four probable mechanisms for the role of reactive oxygen species (ROS) in acne pathogenesis. We believe that ROS can contribute significantly to the acne vulgaris pathobiology via toll-like receptor (TLR), peroxisome proliferator-activated receptor (PPAR), mTOR pathway, and innate immune system, resulting in inflammation by alterations in the generation of several proinflammatory cytokines including IL-1, IL-8, and TNF-α.
Collapse
Affiliation(s)
- Sina Kardeh
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Cellular and Molecular Medicine Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Arman Moein
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Cellular and Molecular Medicine Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Reza Namazi
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Kardeh
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Cellular and Molecular Medicine Student Research Group, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
24
|
Arumugam P, Shankaran D, Bothra A, Gandotra S, Rao V. The MmpS6-MmpL6 Operon Is an Oxidative Stress Response System Providing Selective Advantage toMycobacterium tuberculosisin Stress. J Infect Dis 2018; 219:459-469. [DOI: 10.1093/infdis/jiy526] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 08/30/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Prabhakar Arumugam
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, New Delhi, India
| | - Deepthi Shankaran
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, New Delhi, India
| | - Ankur Bothra
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, New Delhi, India
| | - Sheetal Gandotra
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, New Delhi, India
| | - Vivek Rao
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|
25
|
Singla B, Ghoshal P, Lin H, Wei Q, Dong Z, Csányi G. PKCδ-Mediated Nox2 Activation Promotes Fluid-Phase Pinocytosis of Antigens by Immature Dendritic Cells. Front Immunol 2018; 9:537. [PMID: 29632528 PMCID: PMC5879126 DOI: 10.3389/fimmu.2018.00537] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/02/2018] [Indexed: 12/31/2022] Open
Abstract
Aims Macropinocytosis is a major endocytic pathway by which dendritic cells (DCs) internalize antigens in the periphery. Despite the importance of DCs in the initiation and control of adaptive immune responses, the signaling mechanisms mediating DC macropinocytosis of antigens remain largely unknown. The goal of the present study was to investigate whether protein kinase C (PKC) is involved in stimulation of DC macropinocytosis and, if so, to identify the specific PKC isoform(s) and downstream signaling mechanisms involved. Methods Various cellular, molecular and immunological techniques, pharmacological approaches and genetic knockout mice were utilized to investigate the signaling mechanisms mediating DC macropinocytosis. Results Confocal laser scanning microscopy confirmed that DCs internalize fluorescent antigens (ovalbumin) using macropinocytosis. Pharmacological blockade of classical and novel PKC isoforms using calphostin C abolished both phorbol ester- and hepatocyte growth factor-induced antigen macropinocytosis in DCs. The qRT-PCR experiments identified PKCδ as the dominant PKC isoform in DCs. Genetic studies demonstrated the functional role of PKCδ in DC macropinocytosis of antigens, their subsequent maturation, and secretion of various T-cell stimulatory cytokines, including IL-1α, TNF-α and IFN-β. Additional mechanistic studies identified NADPH oxidase 2 (Nox2) and intracellular superoxide anion as important players in DC macropinocytosis of antigens downstream of PKCδ activation. Conclusion The findings of the present study demonstrate a novel mechanism by which PKCδ activation via stimulation of Nox2 activity and downstream redox signaling promotes DC macropinocytosis of antigens. PKCδ/Nox2-mediated antigen macropinocytosis stimulates maturation of DCs and secretion of T-cell stimulatory cytokines. These findings may contribute to a better understanding of the regulatory mechanisms in DC macropinocytosis and downstream regulation of T-cell-mediated responses.
Collapse
Affiliation(s)
- Bhupesh Singla
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Pushpankur Ghoshal
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Huiping Lin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Gábor Csányi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
26
|
Klassert TE, Goyal S, Stock M, Driesch D, Hussain A, Berrocal-Almanza LC, Myakala R, Sumanlatha G, Valluri V, Ahmed N, Schumann RR, Flores C, Slevogt H. AmpliSeq Screening of Genes Encoding the C-Type Lectin Receptors and Their Signaling Components Reveals a Common Variant in MASP1 Associated with Pulmonary Tuberculosis in an Indian Population. Front Immunol 2018. [PMID: 29515573 PMCID: PMC5826192 DOI: 10.3389/fimmu.2018.00242] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Tuberculosis (TB) is a multifactorial disease governed by bacterial, host and environmental factors. On the host side, growing evidence shows the crucial role that genetic variants play in the susceptibility to Mycobacterium tuberculosis (Mtb) infection. Such polymorphisms have been described in genes encoding for different cytokines and pattern recognition receptors (PRR), including numerous Toll-like receptors (TLRs). In recent years, several members of the C-type lectin receptors (CTLRs) have been identified as key PRRs in TB pathogenesis. Nevertheless, studies to date have only addressed particular genetic polymorphisms in these receptors or their related pathways in relation with TB. In the present study, we screened the main CTLR gene clusters as well as CTLR pathway-related genes for genetic variation associated with pulmonary tuberculosis (PTB). This case-control study comprised 144 newly diagnosed pulmonary TB patients and 181 healthy controls recruited at the Bhagwan Mahavir Medical Research Center (BMMRC), Hyderabad, India. A two-stage study was employed in which an explorative AmpliSeq-based screening was followed by a validation phase using iPLEX MassARRAY. Our results revealed one SNP (rs3774275) in MASP1 significantly associated with PTB in our population (joint analysis p = 0.0028). Furthermore, serum levels of MASP1 were significantly elevated in TB patients when compared to healthy controls. Moreover, in the present study we could observe an impact of increased MASP1 levels on the lectin pathway complement activity in vitro. In conclusion, our results demonstrate a significant association of MASP1 polymorphism rs3774275 and MASP1 serum levels with the development of pulmonary TB. The present work contributes to our understanding of host-Mtb interaction and reinforces the critical significance of mannose-binding lectin and the lectin-complement pathway in Mtb pathogenesis. Moreover, it proposes a MASP1 polymorphism as a potential genetic marker for TB resistance.
Collapse
Affiliation(s)
| | - Surabhi Goyal
- Institute of Microbiology and Hygiene, Charité Universitätsmedizin Berlin, Berlin, Germany
| | | | | | - Abid Hussain
- Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| | | | | | | | | | - Niyaz Ahmed
- Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| | - Ralf R Schumann
- Institute of Microbiology and Hygiene, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Carlos Flores
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.,Research Unit, Hospital Universitario N.S. de Candelaria, Universidad de La Laguna, Santa Cruz de Tenerife, Spain.,Genomics Division, Instituto Tecnológico y de Energías Renovables (ITER), Santa Cruz de Tenerife, Spain
| | | |
Collapse
|
27
|
Wagener M, Hoving JC, Ndlovu H, Marakalala MJ. Dectin-1-Syk-CARD9 Signaling Pathway in TB Immunity. Front Immunol 2018; 9:225. [PMID: 29487599 PMCID: PMC5816931 DOI: 10.3389/fimmu.2018.00225] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/26/2018] [Indexed: 12/12/2022] Open
Abstract
One of the first steps toward mounting an effective immune response to Mycobacterium tuberculosis (Mtb) is recognition of the pathogen through pattern-recognition receptors (PRRs) expressed by innate immune cells. Activation of the PRR Dectin-1 by an unknown mycobacterial ligand triggers an intracellular signaling cascade involving numerous proteins, including spleen tyrosine kinase, protein kinase C-delta, and caspase recruitment domain family member 9, some of which have been shown to influence host immune response to TB infection. Here, we review the role of Dectin-1 signaling pathway in anti-mycobacterial immunity and discuss its contribution in the control of Mtb infection, and potential applications in TB vaccine adjuvanticity.
Collapse
Affiliation(s)
- Matthew Wagener
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - J. Claire Hoving
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Hlumani Ndlovu
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Mohlopheni J. Marakalala
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
28
|
Tomić S, Janjetović K, Mihajlović D, Milenković M, Kravić-Stevović T, Marković Z, Todorović-Marković B, Spitalsky Z, Micusik M, Vučević D, Čolić M, Trajković V. Graphene quantum dots suppress proinflammatory T cell responses via autophagy-dependent induction of tolerogenic dendritic cells. Biomaterials 2017; 146:13-28. [DOI: 10.1016/j.biomaterials.2017.08.040] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/20/2022]
|
29
|
Bazzi S, Modjtahedi H, Mudan S, Achkar M, Akle C, Bahr GM. Immunomodulatory effects of heat-killed Mycobacterium obuense on human blood dendritic cells. Innate Immun 2017; 23:592-605. [PMID: 28853313 DOI: 10.1177/1753425917727838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Heat-killed (HK) Mycobacterium obuense is a novel immunomodulator, currently undergoing clinical evaluation as an immunotherapeutic agent in the treatment of cancer. Here, we examined the effect of in vitro exposure to HK M. obuense on the expression of different categories of surface receptors on human blood myeloid (m) and plasmacytoid (p) DCs. Moreover, we have characterized the cytokine and chemokine secretion patterns of purified total blood DCs stimulated with HK M. obuense. HK M. obuense significantly up-regulated the expression of CD11c, CD80, CD83, CD86, CD274 and MHC class II in whole-blood mDCs and CD80, CD123 and MHC class II in whole-blood pDCs. Down-regulation of CD195 expression in both DC subpopulations was also noted. Further analysis showed that HK M. obuense up-regulated the expression of CD80, CD83 and MHC class II on purified blood DC subpopulations. TLR2 and TLR1 were also identified to be engaged in mediating the HK M. obuense-induced up-regulation of surface receptor expression on whole blood mDCs. In addition, our data demonstrated that HK M. obuense augmented the secretion of CCL4, CCL5, CCL22, CXCL8, IL-6, IL-12p40 and TNF-α by purified total blood DCs. Taken together, our data suggest that HK M. obuense exerts potent differential immunomodulatory effects on human DC subpopulations.
Collapse
Affiliation(s)
- Samer Bazzi
- 1 School of Life Sciences, Faculty of Science, Engineering and Computing, 4264 Kingston University , Kingston upon Thames, UK.,2 Biology Department, Faculty of Sciences, 54686 University of Balamand , Al Kurah, Lebanon
| | - Helmout Modjtahedi
- 1 School of Life Sciences, Faculty of Science, Engineering and Computing, 4264 Kingston University , Kingston upon Thames, UK
| | - Satvinder Mudan
- 3 St George's University of London, Imperial College, London and The Royal Marsden Hospital, London, UK
| | - Marcel Achkar
- 4 Clinical Laboratory Department, Nini Hospital, Tripoli, Lebanon
| | | | - Georges M Bahr
- 6 Faculty of Medicine and Medical Sciences, 54686 University of Balamand , Al Kurah, Lebanon
| |
Collapse
|
30
|
Pahari S, Kaur G, Aqdas M, Negi S, Chatterjee D, Bashir H, Singh S, Agrewala JN. Bolstering Immunity through Pattern Recognition Receptors: A Unique Approach to Control Tuberculosis. Front Immunol 2017; 8:906. [PMID: 28824632 PMCID: PMC5539433 DOI: 10.3389/fimmu.2017.00906] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/14/2017] [Indexed: 12/24/2022] Open
Abstract
The global control of tuberculosis (TB) presents a continuous health challenge to mankind. Despite having effective drugs, TB still has a devastating impact on human health. Contributing reasons include the emergence of drug-resistant strains of Mycobacterium tuberculosis (Mtb), the AIDS-pandemic, and the absence of effective vaccines against the disease. Indeed, alternative and effective methods of TB treatment and control are urgently needed. One such approach may be to more effectively engage the immune system; particularly the frontline pattern recognition receptor (PRR) systems of the host, which sense pathogen-associated molecular patterns (PAMPs) of Mtb. It is well known that 95% of individuals infected with Mtb in latent form remain healthy throughout their life. Therefore, we propose that clues can be found to control the remainder by successfully manipulating the innate immune mechanisms, particularly of nasal and mucosal cavities. This article highlights the importance of signaling through PRRs in restricting Mtb entry and subsequently preventing its infection. Furthermore, we discuss whether this unique therapy employing PRRs in combination with drugs can help in reducing the dose and duration of current TB regimen.
Collapse
Affiliation(s)
- Susanta Pahari
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Gurpreet Kaur
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Mohammad Aqdas
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Shikha Negi
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Deepyan Chatterjee
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Hilal Bashir
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Sanpreet Singh
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Javed N Agrewala
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh, India
| |
Collapse
|
31
|
da Silva MV, Ferreira França FB, Mota CM, de Macedo Júnior AG, Ramos ELP, Santiago FM, Mineo JR, Mineo TWP. Dectin-1 Compromises Innate Responses and Host Resistance against Neospora caninum Infection. Front Immunol 2017; 8:245. [PMID: 28326085 PMCID: PMC5339258 DOI: 10.3389/fimmu.2017.00245] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/20/2017] [Indexed: 12/29/2022] Open
Abstract
Neospora caninum is an intracellular protozoan parasite that has drawn increasing interest due to its association with worldwide repetitive bovine abortions, which cause billionaire losses to the meat and dairy industries annually. Innate immunity plays an important role in infection control, and N. caninum activates the production of inflammatory mediators through toll-like receptors, NOD-like receptors, and mitogen-activated protein kinase signaling pathways. Advances in the knowledge of initial host–parasite interactions are desirable for the design of control measures against the infection, obliterating its pathogenesis. In that sense, we here aimed to describe the role of the innate C-type lectin receptor Dectin-1 during the infection by N. caninum. With that intent, we observed that the absence of Dectin-1, observed in genetically depleted (Dectin-1−/−) mice or competitively inhibited by an inert agonist [laminarin (LAM)], rescued 50% of the mice infected with lethal doses of N. caninum. Dectin-1−/− and LAM-treated mice also presented a reduction in the parasite load during acute and chronic phases, associated with decreased inflammatory scores in the central nervous system. Among all the cell phenotypes that migrated to the initial site of infection, dendritic cells and macrophages gained subpopulations with high Dectin-1 surface expression. The impairment of the receptor in these cells led to a decreased parasite burden, as well as augmented production of IL-12p40. We also found that Dectin-1+ cells produced less reactive oxygen species (ROS) at the initial site of the infection, while mice deficient in NADPH oxidase isoform 2 (NOX2−/−) were not able to control parasite replication and produce IL-12p40, even upon LAM treatment. Interestingly, the absence of functional Dectin-1 did not alter the susceptibility of mice against closely related Toxoplasma gondii. In conclusion, the gathered data suggest that Dectin-1 is involved in the parasite-induced downmodulation of ROS, and other key molecules triggered for the control of N. caninum infection and are a promising target for future development of protocols intended for intervention against neosporosis.
Collapse
Affiliation(s)
- Murilo Vieira da Silva
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo", Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia , Uberlândia , Brazil
| | - Flávia Batista Ferreira França
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo", Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia , Uberlândia , Brazil
| | - Caroline Martins Mota
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo", Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia , Uberlândia , Brazil
| | - Arlindo Gomes de Macedo Júnior
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo", Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia , Uberlândia , Brazil
| | - Eliézer Lucas Pires Ramos
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo", Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia , Uberlândia , Brazil
| | - Fernanda Maria Santiago
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo", Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia , Uberlândia , Brazil
| | - José Roberto Mineo
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo", Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia , Uberlândia , Brazil
| | - Tiago Wilson Patriarca Mineo
- Laboratory of Immunoparasitology "Dr. Mário Endsfeldz Camargo", Department of Immunology, Institute of Biomedical Sciences, Federal University of Uberlândia , Uberlândia , Brazil
| |
Collapse
|
32
|
C-type lectin receptors in tuberculosis: what we know. Med Microbiol Immunol 2016; 205:513-535. [DOI: 10.1007/s00430-016-0470-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/21/2016] [Indexed: 12/19/2022]
|